1
|
Hilsabeck TAU, Narayan VP, Wilson KA, Carrera EM, Raftery D, Promislow D, Brem RB, Campisi J, Kapahi P. Systems biology approaches identify metabolic signatures of dietary lifespan and healthspan across species. Nat Commun 2024; 15:9330. [PMID: 39472442 PMCID: PMC11522498 DOI: 10.1038/s41467-024-52909-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/18/2024] [Indexed: 11/02/2024] Open
Abstract
Dietary restriction (DR) is a potent method to enhance lifespan and healthspan, but individual responses are influenced by genetic variations. Understanding how metabolism-related genetic differences impact longevity and healthspan are unclear. To investigate this, we used metabolites as markers to reveal how different genotypes respond to diet to influence longevity and healthspan traits. We analyzed data from Drosophila Genetic Reference Panel (DGRP) strains raised under AL and DR conditions, combining metabolomic, phenotypic, and genome-wide information. We employed two computational and complementary methods across species-random forest modeling within the DGRP as our primary analysis and Mendelian randomization in human cohorts as a secondary analysis. We pinpointed key traits with cross-species relevance as well as underlying heterogeneity and pleiotropy that influence lifespan and healthspan. Notably, orotate was linked to parental age at death in humans and blocked the DR lifespan extension in flies, while threonine supplementation extended lifespan, in a strain- and sex-specific manner. Thus, utilizing natural genetic variation data from flies and humans, we employed a systems biology approach to elucidate potential therapeutic pathways and metabolomic targets for diet-dependent changes in lifespan and healthspan.
Collapse
Affiliation(s)
- Tyler A U Hilsabeck
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Davis School of Gerontology, University of Southern California, University Park, University Park, Los Angeles, CA, 90089, USA
- Computational Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Vikram P Narayan
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Department of Biology & Chemistry, Embry-Riddle Aeronautical University, Prescott, AZ, 86301, USA
| | - Kenneth A Wilson
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Davis School of Gerontology, University of Southern California, University Park, University Park, Los Angeles, CA, 90089, USA
| | - Enrique M Carrera
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Dominican University of California, San Rafael, CA, 94901, USA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Daniel Promislow
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
- Department of Biology, University of Washington, Seattle, WA, 98195, USA
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Rachel B Brem
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Davis School of Gerontology, University of Southern California, University Park, University Park, Los Angeles, CA, 90089, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA, 94720, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Davis School of Gerontology, University of Southern California, University Park, University Park, Los Angeles, CA, 90089, USA.
| |
Collapse
|
2
|
Wu Y, Foollee A, Chan AY, Hille S, Hauke J, Challis MP, Johnson JL, Yaron TM, Mynard V, Aung OH, Cleofe MAS, Huang C, Lim Kam Sian TCC, Rahbari M, Gallage S, Heikenwalder M, Cantley LC, Schittenhelm RB, Formosa LE, Smith GC, Okun JG, Müller OJ, Rusu PM, Rose AJ. Phosphoproteomics-directed manipulation reveals SEC22B as a hepatocellular signaling node governing metabolic actions of glucagon. Nat Commun 2024; 15:8390. [PMID: 39333498 PMCID: PMC11436942 DOI: 10.1038/s41467-024-52703-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
The peptide hormone glucagon is a fundamental metabolic regulator that is also being considered as a pharmacotherapeutic option for obesity and type 2 diabetes. Despite this, we know very little regarding how glucagon exerts its pleiotropic metabolic actions. Given that the liver is a chief site of action, we performed in situ time-resolved liver phosphoproteomics to reveal glucagon signaling nodes. Through pathway analysis of the thousands of phosphopeptides identified, we reveal "membrane trafficking" as a dominant signature with the vesicle trafficking protein SEC22 Homolog B (SEC22B) S137 phosphorylation being a top hit. Hepatocyte-specific loss- and gain-of-function experiments reveal that SEC22B was a key regulator of glycogen, lipid and amino acid metabolism, with SEC22B-S137 phosphorylation playing a major role in glucagon action. Mechanistically, we identify several protein binding partners of SEC22B affected by glucagon, some of which were differentially enriched with SEC22B-S137 phosphorylation. In summary, we demonstrate that phosphorylation of SEC22B is a hepatocellular signaling node mediating the metabolic actions of glucagon and provide a rich resource for future investigations on the biology of glucagon action.
Collapse
Affiliation(s)
- Yuqin Wu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Ashish Foollee
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Andrea Y Chan
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Susanne Hille
- Department of Internal Medicine V, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Jana Hauke
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Matthew P Challis
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Department of Cell Biology, Harvard Medical School, Boston, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Tomer M Yaron
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Victoria Mynard
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Okka H Aung
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Maria Almira S Cleofe
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Cheng Huang
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, Victoria, Australia
| | | | - Mohammad Rahbari
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Department of Surgery, Theodor-Kutzer-Ufer 1-3, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
| | - Suchira Gallage
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard-Karls University, Tübingen, Germany
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Department of Cell Biology, Harvard Medical School, Boston, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Ralf B Schittenhelm
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, Victoria, Australia
| | - Luke E Formosa
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Greg C Smith
- School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Jürgen G Okun
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Oliver J Müller
- Department of Internal Medicine V, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Patricia M Rusu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia.
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia.
| |
Collapse
|
3
|
Kim SQ, Spann RA, Khan MSH, Berthoud HR, Münzberg H, Albaugh VL, He Y, McDougal DH, Soto P, Yu S, Morrison CD. FGF21 as a mediator of adaptive changes in food intake and macronutrient preference in response to protein restriction. Neuropharmacology 2024; 255:110010. [PMID: 38797244 PMCID: PMC11156534 DOI: 10.1016/j.neuropharm.2024.110010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Free-feeding animals navigate complex nutritional landscapes in which food availability, cost, and nutritional value can vary markedly. Animals have thus developed neural mechanisms that enable the detection of nutrient restriction, and these mechanisms engage adaptive physiological and behavioral responses that limit or reverse this nutrient restriction. This review focuses specifically on dietary protein as an essential and independently defended nutrient. Adequate protein intake is required for life, and ample evidence exists to support an active defense of protein that involves behavioral changes in food intake, food preference, and food motivation, likely mediated by neural changes that increase the reward value of protein foods. Available evidence also suggests that the circulating hormone fibroblast growth factor 21 (FGF21) acts in the brain to coordinate these adaptive changes in food intake, making it a unique endocrine signal that drives changes in macronutrient preference in the context of protein restriction. This article is part of the Special Issue on "Food intake and feeding states".
Collapse
Affiliation(s)
- Sora Q Kim
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Redin A Spann
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | | | | - Heike Münzberg
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Vance L Albaugh
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA; Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - David H McDougal
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Paul Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, 70810, USA
| | - Sangho Yu
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | |
Collapse
|
4
|
Fulton TL, Johnstone JN, Tan JJ, Balagopal K, Dedman A, Chan AY, Johnson TK, Mirth CK, Piper MDW. Transiently restricting individual amino acids protects Drosophila melanogaster against multiple stressors. Open Biol 2024; 14:240093. [PMID: 39106944 PMCID: PMC11303031 DOI: 10.1098/rsob.240093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 08/09/2024] Open
Abstract
Nutrition and resilience are linked, though it is not yet clear how diet confers stress resistance or the breadth of stressors that it can protect against. We have previously shown that transiently restricting an essential amino acid can protect Drosophila melanogaster against nicotine poisoning. Here, we sought to characterize the nature of this dietary-mediated protection and determine whether it was sex, amino acid and/or nicotine specific. When we compared between sexes, we found that isoleucine deprivation increases female, but not male, nicotine resistance. Surprisingly, we found that this protection afforded to females was not replicated by dietary protein restriction and was instead specific to individual amino acid restriction. To understand whether these beneficial effects of diet were specific to nicotine or were generalizable across stressors, we pre-treated flies with amino acid restriction diets and exposed them to other types of stress. We found that some of the diets that protected against nicotine also protected against oxidative and starvation stress, and improved survival following cold shock. Interestingly, we found that a diet lacking isoleucine was the only diet to protect against all these stressors. These data point to isoleucine as a critical determinant of robustness in the face of environmental challenges.
Collapse
Affiliation(s)
- Tahlia L. Fulton
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Joshua N. Johnstone
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Jing J. Tan
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Krithika Balagopal
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Amy Dedman
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Andrea Y. Chan
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria3800, Australia
| | - Travis K. Johnson
- School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria3086, Australia
| | - Christen K. Mirth
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Matthew D. W. Piper
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| |
Collapse
|
5
|
Zhuang Z, Jia W, Wu L, Li Y, Lu Y, Xu M, Bai H, Bi Y, Wang Z, Chen S, Jiang Y, Chang G. Threonine Deficiency Increases Triglyceride Deposition in Primary Duck Hepatocytes by Reducing STAT3 Phosphorylation. Int J Mol Sci 2024; 25:8142. [PMID: 39125712 PMCID: PMC11312044 DOI: 10.3390/ijms25158142] [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: 06/02/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 08/12/2024] Open
Abstract
Liver lipid metabolism disruption significantly contributes to excessive fat buildup in waterfowl. Research suggests that the supplementation of Threonine (Thr) in the diet can improve liver lipid metabolism disorder, while Thr deficiency can lead to such metabolic disorders in the liver. The mechanisms through which Thr regulates lipid metabolism remain unclear. STAT3 (signal transducer and activator of transcription 3), a crucial transcription factor in the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway, participates in various biological processes, including lipid and energy metabolism. This research investigates the potential involvement of STAT3 in the increased lipid storage seen in primary duck hepatocytes as a result of a lack of Thr. Using small interfering RNA and Stattic, a specific STAT3 phosphorylation inhibitor, we explored the impact of STAT3 expression patterns on Thr-regulated lipid synthesis metabolism in hepatocytes. Through transcriptome sequencing, we uncovered pathways related to lipid synthesis and metabolism jointly regulated by Thr and STAT3. The results showed that Thr deficiency increases lipid deposition in primary duck hepatocytes (p < 0.01). The decrease in protein and phosphorylation levels of STAT3 directly caused this deposition (p < 0.01). Transcriptomic analysis revealed that Thr deficiency and STAT3 knockdown jointly altered the mRNA expression levels of pathways related to long-chain fatty acid synthesis and energy metabolism (p < 0.05). Thr deficiency, through mediating STAT3 inactivation, upregulated ELOVL7, PPARG, MMP1, MMP13, and TIMP4 mRNA levels, and downregulated PTGS2 mRNA levels (p < 0.01). In summary, these results suggest that Thr deficiency promotes lipid synthesis, reduces lipid breakdown, and leads to lipid metabolism disorders and triglyceride deposition by downregulating STAT3 activity in primary duck hepatocytes.
Collapse
Affiliation(s)
- Zhong Zhuang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Wenqian Jia
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Lei Wu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yongpeng Li
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yijia Lu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Minghong Xu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Hao Bai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China;
| | - Yulin Bi
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Zhixiu Wang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Shihao Chen
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yong Jiang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Guobin Chang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| |
Collapse
|
6
|
Ambroszkiewicz J, Gajewska J, Szamotulska K, Rowicka G, Klemarczyk W, Strucińska M, Chełchowska M. Comparative Analysis of Myokines and Bone Metabolism Markers in Prepubertal Vegetarian and Omnivorous Children. Nutrients 2024; 16:2009. [PMID: 38999757 PMCID: PMC11243178 DOI: 10.3390/nu16132009] [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: 06/03/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The role of bone and muscle as endocrine organs may be important contributing factors for children's growth and development. Myokines, secreted by muscle cells, play a role in regulating bone metabolism, either directly or indirectly. Conversely, markers of bone metabolism, reflecting the balance between bone formation and bone resorption, can also influence myokine secretion. This study investigated a panel of serum myokines and their relationships with bone metabolism markers in children following vegetarian and omnivorous diets. A cohort of sixty-eight healthy prepubertal children, comprising 44 vegetarians and 24 omnivores, participated in this study. Anthropometric measurements, dietary assessments, and biochemical analyses were conducted. To evaluate the serum concentrations of bone markers and myokines, an enzyme-linked immunosorbent assay (ELISA) was used. The studied children did not differ regarding their serum myokine levels, except for a higher concentration of decorin in the vegetarian group (p = 0.020). The vegetarians demonstrated distinct pattern of bone metabolism markers compared to the omnivores, with lower levels of N-terminal propeptide of type I procollagen (P1NP) (p = 0.001) and elevated levels of C-terminal telopeptide of type I collagen (CTX-I) (p = 0.018). Consequently, the P1NP/CTX-I ratio was significantly decreased in the vegetarians. The children following a vegetarian diet showed impaired bone metabolism with reduced bone formation and increased bone resorption. Higher levels of decorin, a myokine involved in collagen fibrillogenesis and essential for tissue structure and function, may suggest a potential compensatory mechanism contributing to maintaining bone homeostasis in vegetarians. The observed significant positive correlations between myostatin and bone metabolism markers, including P1NP and soluble receptor activator of nuclear factor kappa-B ligand (sRANKL), suggest an interplay between muscle and bone metabolism, potentially through the RANK/RANKL/OPG signaling pathway.
Collapse
Affiliation(s)
- Jadwiga Ambroszkiewicz
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland; (J.G.); (M.C.)
| | - Joanna Gajewska
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland; (J.G.); (M.C.)
| | - Katarzyna Szamotulska
- Department of Epidemiology and Biostatistics, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland;
| | - Grażyna Rowicka
- Department of Nutrition, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland; (G.R.); (W.K.); (M.S.)
| | - Witold Klemarczyk
- Department of Nutrition, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland; (G.R.); (W.K.); (M.S.)
| | - Małgorzata Strucińska
- Department of Nutrition, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland; (G.R.); (W.K.); (M.S.)
| | - Magdalena Chełchowska
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland; (J.G.); (M.C.)
| |
Collapse
|
7
|
Zaman K, Mun HC, Solon-Biet SM, Senior AM, Raubenheimer D, Simpson SJ, Conigrave AD. Mice Regulate Dietary Amino Acid Balance and Energy Intake by Selecting between Complementary Protein Sources. J Nutr 2024; 154:1766-1780. [PMID: 38583524 DOI: 10.1016/j.tjnut.2024.04.007] [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: 10/18/2023] [Revised: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND A balanced intake of protein and constituent amino acids (AAs) requires adjustments to total food intake (protein leverage [PL]) and food selection to balance deficits and excesses (complementary feeding). We provided mice with choices of casein and whey, 2 protein sources that are complementary in AA balance, across a range of protein concentrations (P%) of digestible energy (DE). OBJECTIVES We aimed to determine if: 1) PL operates similarly for casein and whey; 2) one protein source is preferred at control P%; 3) the preference changes as P% falls; and 4) AA intakes under control and low P% levels identify AAs that drive changes in protein selection. METHODS Food intake and plasma fibroblast growth factor-21 (FGF21) concentrations were measured in mice at various P% (P7.5%-P33%). For direct comparisons, defined diets were used in which the protein source was either casein or whey. In food choice studies, mice had access to foods in which both casein and whey were provided at the same P% level at the same time. RESULTS PL operated at different P% thresholds in casein (13%)- and whey (10%)-based diets, and the magnitude of PL was greater for casein. Although mice preferred casein under control conditions (P23%), a pronounced preference shift to whey occurred as P% fell to P13% and P10%. At low P%, increases in food intake were accompanied by increases in plasma FGF21, a protein hunger signal. Among AAs deficient in casein and enriched in whey, the intake of Cys was the most invariant as P% changed between P23% and P10%, appearing to drive the switch in protein preference. CONCLUSIONS Mice selected between complementary protein sources, casein and whey, achieving stable total energy intake and regulated intake of AAs as P% varied. Supplementation of low P% casein diets with one whey-enriched AA, Cys, suppressed plasma FGF21 and total food intake.
Collapse
Affiliation(s)
- Kamrul Zaman
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Hee-Chang Mun
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Alistair M Senior
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - David Raubenheimer
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Arthur D Conigrave
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Liang Z, He Y, Wei D, Fu P, Li Y, Wang H, Yang D, Hou X. Tree peony seed oil alleviates hyperlipidemia and hyperglycemia by modulating gut microbiota and metabolites in high-fat diet mice. Food Sci Nutr 2024; 12:4421-4434. [PMID: 38873446 PMCID: PMC11167153 DOI: 10.1002/fsn3.4108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 06/15/2024] Open
Abstract
With the changes of people's lifestyle, hyperlipidemia and hyperglycemia which were induced from a diet high in both fat and sugar have become serious health concerns. Tree peony seed oil (PSO) is a novel kind of edible oil that shows great potential in the food industry because of its high constituent of unsaturated fatty acids. Based 16S rRNA and gut untargeted metabolomics, this study elucidated that the mechanism of PSO regulating blood glucose (Glu) and lipids. The impact of PSO on gut microbiota balance and gut metabolites of mice with a high-fat diet (HFD) was evaluated. The findings indicated that PSO decreased HFD mice's body weight and fat accumulation, ameliorating the levels of blood lipid, reduced liver fat vacuole levels. What's more PSO modulated the proportion of gut microbiota in HFD mice and enhanced the abundance of probiotics. Furthermore, untargeted metabolomic analysis revealed that PSO not only impacted the generation of short-chain fatty acids (SCFAs) by gut microorganism and altered metabolic pathway but exerted influence on secondary bile acids (BA), amino acid metabolism, and various other metabolites. These results suggested that PSO has the potential function for mitigating HFD-induced hyperlipidemia and hyperglycemia by regulating gut microbiota and host metabolism.
Collapse
Affiliation(s)
- Ziyue Liang
- College of Agriculture/Tree PeonyHenan University of Science and TechnologyLuoyangChina
- Key Laboratory of Efficient Cultivation and Comprehensive Utilization of Tree Peony in Henan ProvinceLuoyangChina
| | - Yinglong He
- College of Agriculture/Tree PeonyHenan University of Science and TechnologyLuoyangChina
- Key Laboratory of Efficient Cultivation and Comprehensive Utilization of Tree Peony in Henan ProvinceLuoyangChina
| | - Dongfeng Wei
- College of Urban Construction, Luoyang Vocational and Technical CollegeLuoyangChina
| | - Peixin Fu
- College of Agriculture/Tree PeonyHenan University of Science and TechnologyLuoyangChina
- Key Laboratory of Efficient Cultivation and Comprehensive Utilization of Tree Peony in Henan ProvinceLuoyangChina
| | - Yuying Li
- College of Agriculture/Tree PeonyHenan University of Science and TechnologyLuoyangChina
- Key Laboratory of Efficient Cultivation and Comprehensive Utilization of Tree Peony in Henan ProvinceLuoyangChina
| | - Hao Wang
- College of Agriculture/Tree PeonyHenan University of Science and TechnologyLuoyangChina
- Key Laboratory of Efficient Cultivation and Comprehensive Utilization of Tree Peony in Henan ProvinceLuoyangChina
| | - Di Yang
- College of Agriculture/Tree PeonyHenan University of Science and TechnologyLuoyangChina
- Key Laboratory of Efficient Cultivation and Comprehensive Utilization of Tree Peony in Henan ProvinceLuoyangChina
| | - Xiaogai Hou
- College of Agriculture/Tree PeonyHenan University of Science and TechnologyLuoyangChina
- Key Laboratory of Efficient Cultivation and Comprehensive Utilization of Tree Peony in Henan ProvinceLuoyangChina
| |
Collapse
|
9
|
Li N, Pan C, Lu G, Pan H, Han Y, Wang K, Jin P, Liu Q, Jiang J. Hydrophobic Trinuclear Copper Cluster-Containing Organic Framework for Synergetic Electrocatalytic Synthesis of Amino Acids. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311023. [PMID: 38050947 DOI: 10.1002/adma.202311023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Electrocatalytic synthesis of amino acids provides a promising green and efficient pathway to manufacture the basic substances of life. Herein, reaction of 2,5-perfluroalkyl-terepthalohydrazide and tris(4-µ2 -O-carboxaldehyde-pyrazolato-N, N')-tricopper affords a crystalline trinuclear copper cluster-containing organic framework, named F-Cu3 -OF. Incorporation of abundant hydrophobic perfluroalkyl groups inside the channels of F-Cu3 -OF is revealed to successfully suppress the hydrogen evolution reaction via preventing H+ cation with large polarity from the framework of F-Cu3 -OF and in turn increasing the adsorption of other substrates with relatively small polarity like NO3 - and keto acids on the active sites. The copper atoms with short distance in the trinuclear copper clusters of F-Cu3 -OF enable simultaneous activization of NO3 - and keto acids, facilitating the following synergistic and efficient C─N coupling on the basis of in situ spectroscopic investigations together with theoretical calculation. Combination of these effects leads to efficient electroproduction of various amino acids including glycine, alanine, leucine, valine, and phenylalanine from NO3 - and keto acids with a Faraday efficiency of 42%-71% and a yield of 187-957 µmol cm-2 h-1 , representing the thus far best performance. This work shall be helpful for developing economical, eco-friendly, and high-efficiency strategy for the production of amino acids and other life substances.
Collapse
Affiliation(s)
- Ning Li
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Chenliang Pan
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300130, China
| | - Guang Lu
- College of Chemical and Biological Engineering, Shandong University of Science and Technology, Qingdao, 266590, China
| | - Houhe Pan
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Yuesheng Han
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Kang Wang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Peng Jin
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300130, China
| | - Qingyun Liu
- College of Chemical and Biological Engineering, Shandong University of Science and Technology, Qingdao, 266590, China
| | - Jianzhuang Jiang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| |
Collapse
|
10
|
Rose AJ, Rusu PM. A leucine-macrophage mTORC1 connection drives increased risk of atherosclerosis with high-protein diets. Nat Metab 2024; 6:203-204. [PMID: 38409322 DOI: 10.1038/s42255-023-00952-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Affiliation(s)
- Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Dept. Of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.
| | - Patricia M Rusu
- Nutrient Metabolism & Signalling Laboratory, Dept. Of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Zhang X, Kapoor D, Jeong SJ, Fappi A, Stitham J, Shabrish V, Sergin I, Yousif E, Rodriguez-Velez A, Yeh YS, Park A, Yurdagul A, Rom O, Epelman S, Schilling JD, Sardiello M, Diwan A, Cho J, Stitziel NO, Javaheri A, Lodhi IJ, Mittendorfer B, Razani B. Identification of a leucine-mediated threshold effect governing macrophage mTOR signalling and cardiovascular risk. Nat Metab 2024; 6:359-377. [PMID: 38409323 PMCID: PMC11448845 DOI: 10.1038/s42255-024-00984-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 01/09/2024] [Indexed: 02/28/2024]
Abstract
High protein intake is common in western societies and is often promoted as part of a healthy lifestyle; however, amino-acid-mediated mammalian target of rapamycin (mTOR) signalling in macrophages has been implicated in the pathogenesis of ischaemic cardiovascular disease. In a series of clinical studies on male and female participants ( NCT03946774 and NCT03994367 ) that involved graded amounts of protein ingestion together with detailed plasma amino acid analysis and human monocyte/macrophage experiments, we identify leucine as the key activator of mTOR signalling in macrophages. We describe a threshold effect of high protein intake and circulating leucine on monocytes/macrophages wherein only protein in excess of ∼25 g per meal induces mTOR activation and functional effects. By designing specific diets modified in protein and leucine content representative of the intake in the general population, we confirm this threshold effect in mouse models and find ingestion of protein in excess of ∼22% of dietary energy requirements drives atherosclerosis in male mice. These data demonstrate a mechanistic basis for the adverse impact of excessive dietary protein on cardiovascular risk.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Divya Kapoor
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Alan Fappi
- Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Medicine and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Jeremiah Stitham
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Vasavi Shabrish
- Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Medicine and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Ismail Sergin
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Eman Yousif
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | | | - Yu-Sheng Yeh
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Slava Epelman
- Peter Munk Cardiac Center and University Health Network, University of Toronto, Toronto, Canada
| | - Joel D Schilling
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Marco Sardiello
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St Louis, MO, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Nathan O Stitziel
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Ali Javaheri
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St Louis, MO, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Bettina Mittendorfer
- Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Departments of Medicine and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA.
| | - Babak Razani
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA.
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Wu T, Baatar D, O' Connor AE, O'Bryan MK, Stringer JM, Hutt KJ, Malimige Aponso M, Monro K, Luo J, Zhu Y, Ernst A, Swindells EOK, Alesi LR, Tho Tony Nguyen N, Piper MDW, Bennett LE. Exome-informed formulations of food proteins enhance body growth and feed conversion efficiency in ad libitum-fed mice. Food Res Int 2024; 176:113819. [PMID: 38163720 DOI: 10.1016/j.foodres.2023.113819] [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: 07/06/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
Meeting requirements for dietary proteins, especially of essential amino acids (EAAs), is critical for the life-long health of living organisms. However, defining EAA targets for preparing biologically-matched nutrition that satisfies metabolic requirements for protein remains challenging. Previous research has shown the advantages of 'exome matching' in representing the specific requirement of dietary AAs, where the target dietary AA profile was derived from in silico translation of the genome of an organism, specifically responsible for protein expression (the 'exome'). However, past studies have assessed these effects in only one sex, for few parameters (body mass and composition), and have used purified diets in which protein is supplied as a mixture of individual AAs. Here, for the first time, we utilise a computational method to guide the formulation of custom protein blends and test if exome matching can be achieved at the intact protein level, through blending standard protein ingredients, ultimately leading to optimal growth, longevity and reproductive function. Mice were provided ad libitum (ad lib) access to one of the four iso-energetic protein-limited diets, two matched and two mis-matched to the mouse exome target, and fed at a fixed protein energy level of 6.2%. During or following 13-weeks of feeding, the food intake, body growth, composition and reproductive functions were measured. Compared to the two mis-matched diets, male and female animals on the exome-matched diet with protein digestibility correction applied, exhibited significantly improved growth rates and final body mass. The feed conversion efficiency in the same diet was also increased by 62% and 40% over the worst diets for males and females, respectively. Male, not female, exhibited higher accretion of lean body mass with the matched, digestibility-corrected diet. All reproductive function measures in both sexes were comparable among diets, with the exception of testicular daily sperm production in males, which was higher in the two matched diets versus the mis-matched diets. The results collectively demonstrate the pronounced advantages of exome-matching in supporting body growth and improving feed conversion efficiency in both sexes. However, the potential impact of this approach in enhancing fertility needs further investigation.
Collapse
Affiliation(s)
- Tong Wu
- School of Chemistry, Faculty of Science, Monash University, Clayton, Victoria 3800, Australia
| | - Davaatseren Baatar
- School of Mathematics, Faculty of Science, Monash University, Clayton, Victoria 3800, Australia
| | - Anne E O' Connor
- School of BioScience and the Bio21 Institute, The University of Melbourne, Parkville, Australia
| | - Moira K O'Bryan
- School of BioScience and the Bio21 Institute, The University of Melbourne, Parkville, Australia
| | - Jessica M Stringer
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Karla J Hutt
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Minoli Malimige Aponso
- School of Chemistry, Faculty of Science, Monash University, Clayton, Victoria 3800, Australia
| | - Keyne Monro
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Victoria 3800, Australia
| | - Jiaqiang Luo
- School of Agriculture and Food, The University of Melbourne, Parkville, Australia
| | - Yingchun Zhu
- College of Food Science and Engineering, Shanxi Agricultural University, Shanxi, China
| | - Andreas Ernst
- School of Mathematics, Faculty of Science, Monash University, Clayton, Victoria 3800, Australia
| | - Elyse O K Swindells
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Lauren R Alesi
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Ngoc Tho Tony Nguyen
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Matthew D W Piper
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Victoria 3800, Australia
| | - Louise E Bennett
- School of Chemistry, Faculty of Science, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
13
|
Solon-Biet SM, Clark X, Bell-Anderson K, Rusu PM, Perks R, Freire T, Pulpitel T, Senior AM, Hoy AJ, Aung O, Le Couteur DG, Raubenheimer D, Rose AJ, Conigrave AD, Simpson SJ. Toward reconciling the roles of FGF21 in protein appetite, sweet preference, and energy expenditure. Cell Rep 2023; 42:113536. [PMID: 38060447 DOI: 10.1016/j.celrep.2023.113536] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2023] [Accepted: 11/17/2023] [Indexed: 12/30/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21), an endocrine signal robustly increased by protein restriction independently of an animal's energy status, exerts profound effects on feeding behavior and metabolism. Here, we demonstrate that considering the nutritional contexts within which FGF21 is elevated can help reconcile current controversies over its roles in mediating macronutrient preference, food intake, and energy expenditure. We show that FGF21 is primarily a driver of increased protein intake in mice and that the effect of FGF21 on sweet preference depends on the carbohydrate balance of the animal. Under no-choice feeding, FGF21 infusion either increased or decreased energy expenditure depending on whether the animal was fed a high- or low-energy diet, respectively. We show that while the role of FGF21 in mediating feeding behavior is complex, its role in promoting protein appetite is robust and that the effects on sweet preference and energy expenditure are macronutrient-state-dependent effects of FGF21.
Collapse
Affiliation(s)
- Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia; School of Medicine, The University of Notre Dame, Darlinghurst, NSW 2010, Australia.
| | - Ximonie Clark
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Kim Bell-Anderson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Patricia M Rusu
- Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Ruth Perks
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Therese Freire
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2006, Australia
| | - Tamara Pulpitel
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Andrew J Hoy
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2006, Australia
| | - Okka Aung
- Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - David G Le Couteur
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2006, Australia; Ageing and Alzheimer's Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord, NSW 2139, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Adam J Rose
- Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Arthur D Conigrave
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW 2050 Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
14
|
Hilsabeck TAU, Narayan VP, Wilson KA, Carrera E, Raftery D, Promislow D, Brem RB, Campisi J, Kapahi P. Systems biology and machine learning approaches identify metabolites that influence dietary lifespan and healthspan responses across flies and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.09.548232. [PMID: 37503266 PMCID: PMC10369897 DOI: 10.1101/2023.07.09.548232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Dietary restriction (DR) is a potent method to enhance lifespan and healthspan, but individual responses are influenced by genetic variations. Understanding how metabolism-related genetic differences impact longevity and healthspan are unclear. To investigate this, we used metabolites as markers to reveal how different genotypes respond to diet to influence longevity and healthspan traits. We analyzed data from Drosophila Genetic Reference Panel strains raised under AL and DR conditions, combining metabolomic, phenotypic, and genome-wide information. Employing two computational methods across species-random forest modeling within the DGRP and Mendelian randomization in the UK Biobank-we pinpointed key traits with cross-species relevance that influence lifespan and healthspan. Notably, orotate was linked to parental age at death in humans and counteracted DR effects in flies, while threonine extended lifespan, in a strain- and sex-specific manner. Thus, utilizing natural genetic variation data from flies and humans, we employed a systems biology approach to elucidate potential therapeutic pathways and metabolomic targets for diet-dependent changes in lifespan and healthspan.
Collapse
|
15
|
Trautman ME, Braucher LN, Elliehausen C, Zhu WG, Zelenovskiy E, Green M, Sonsalla MM, Yeh CY, Hornberger TA, Konopka AR, Lamming DW. Resistance exercise protects mice from protein-induced fat accretion. eLife 2023; 12:RP91007. [PMID: 38019262 PMCID: PMC10686620 DOI: 10.7554/elife.91007] [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: 11/30/2023] Open
Abstract
Low-protein (LP) diets extend the lifespan of diverse species and are associated with improved metabolic health in both rodents and humans. Paradoxically, many athletes and bodybuilders consume high-protein (HP) diets and protein supplements, yet are both fit and metabolically healthy. Here, we examine this paradox using weight pulling, a validated progressive resistance exercise training regimen, in mice fed either an LP diet or an isocaloric HP diet. We find that despite having lower food consumption than the LP group, HP-fed mice gain significantly more fat mass than LP-fed mice when not exercising, while weight pulling protected HP-fed mice from this excess fat accretion. The HP diet augmented exercise-induced hypertrophy of the forearm flexor complex, and weight pulling ability increased more rapidly in the exercised HP-fed mice. Surprisingly, exercise did not protect from HP-induced changes in glycemic control. Our results confirm that HP diets can augment muscle hypertrophy and accelerate strength gain induced by resistance exercise without negative effects on fat mass, and also demonstrate that LP diets may be advantageous in the sedentary. Our results highlight the need to consider both dietary composition and activity, not simply calories, when taking a precision nutrition approach to health.
Collapse
Affiliation(s)
- Michaela E Trautman
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Nutrition and Metabolism Graduate Program, University of Wisconsin- MadisonMadisonUnited States
| | - Leah N Braucher
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Christian Elliehausen
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Wenyuan G Zhu
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Esther Zelenovskiy
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Madelyn Green
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Troy A Hornberger
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- School of Veterinary Medicine, University of Wisconsin-MadisonMadisonUnited States
| | - Adam R Konopka
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Nutrition and Metabolism Graduate Program, University of Wisconsin- MadisonMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- University of Wisconsin Carbone Cancer CenterMadisonUnited States
| |
Collapse
|
16
|
Green CL, Trautman ME, Chaiyakul K, Jain R, Alam YH, Babygirija R, Pak HH, Sonsalla MM, Calubag MF, Yeh CY, Bleicher A, Novak G, Liu TT, Newman S, Ricke WA, Matkowskyj KA, Ong IM, Jang C, Simcox J, Lamming DW. Dietary restriction of isoleucine increases healthspan and lifespan of genetically heterogeneous mice. Cell Metab 2023; 35:1976-1995.e6. [PMID: 37939658 PMCID: PMC10655617 DOI: 10.1016/j.cmet.2023.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023]
Abstract
Low-protein diets promote health and longevity in diverse species. Restriction of the branched-chain amino acids (BCAAs) leucine, isoleucine, and valine recapitulates many of these benefits in young C57BL/6J mice. Restriction of dietary isoleucine (IleR) is sufficient to promote metabolic health and is required for many benefits of a low-protein diet in C57BL/6J males. Here, we test the hypothesis that IleR will promote healthy aging in genetically heterogeneous adult UM-HET3 mice. We find that IleR improves metabolic health in young and old HET3 mice, promoting leanness and glycemic control in both sexes, and reprograms hepatic metabolism in a sex-specific manner. IleR reduces frailty and extends the lifespan of male and female mice, but to a greater degree in males. Our results demonstrate that IleR increases healthspan and longevity in genetically diverse mice and suggests that IleR, or pharmaceuticals that mimic this effect, may have potential as a geroprotective intervention.
Collapse
Affiliation(s)
- Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Krittisak Chaiyakul
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Raghav Jain
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yasmine H Alam
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Anneliese Bleicher
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Grace Novak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Teresa T Liu
- George M. O'Brien Center of Research Excellence, Department of Urology, University of Wisconsin, Madison, WI 93705, USA
| | - Sarah Newman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Will A Ricke
- George M. O'Brien Center of Research Excellence, Department of Urology, University of Wisconsin, Madison, WI 93705, USA
| | - Kristina A Matkowskyj
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| | - Irene M Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA; University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53705, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA; Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA; University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
17
|
Yap YW, Rusu PM, Foollee A, Rose AJ. Post-nursing early life macronutrient balance promotes persistent and malleable biometric and metabolic traits in mice. J Physiol 2023; 601:3813-3824. [PMID: 37535037 DOI: 10.1113/jp281185] [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: 06/15/2023] [Accepted: 07/20/2023] [Indexed: 08/04/2023] Open
Abstract
It is known that dietary factors within the gestational and nursing period affect early life and stably affect later life traits in animals. However, there is very little understanding of whether dietary factors within the early life period from post-nursing to adulthood affect traits in adulthood. To address this, we conducted studies on male C57Bl/6J mice fed from 3 weeks (immediately post-nursing) until 12 weeks (full maturity) using nine different diets varying in all three major macronutrients to parse out the effects of individual macronutrients. Early life macronutrient balance affected body composition and glucose homeostasis in early adulthood, with dietary protein and fat showing major effects. Despite this, mice showed rapid reversal of the effects on body composition and glucose homeostasis of early life diet feeding, upon standard diet feeding in adulthood. However, some traits were persistent, with early life low dietary protein levels stably affecting lean and muscle mass, and early life dietary fat levels stably affecting serum and liver triglyceride levels. In summary, macronutrient balance in the post-nursing early life period does not stably affect adiposity or glucose homeostasis but does impact muscle mass and lipid homeostasis in adulthood, with prominent effects of both protein and fat levels. KEY POINTS: Early life dietary low protein and high fat levels lowered and heightened body mass, respectively. These effects did not substantially persist into adulthood with rapid catch-up growth on a normal diet. Early life protein (negative) and fat (positive) levels affected fat mass. Early life low protein levels negatively affected lean mass. Low protein effects on lower lean and muscle mass persisted into adulthood. Early life macronutrient balance effects did not affect later life glucose homeostasis but early life high fat level affected later life dyslipidaemia. Effects of dietary carbohydrate levels in early and later life were minor.
Collapse
Affiliation(s)
- Yann W Yap
- Nutrient Metabolism & Signalling Laboratory, Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Patricia M Rusu
- Nutrient Metabolism & Signalling Laboratory, Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Ashish Foollee
- Nutrient Metabolism & Signalling Laboratory, Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| |
Collapse
|
18
|
Abstract
Amino acids derived from protein digestion are important nutrients for the growth and maintenance of organisms. Approximately half of the 20 proteinogenic amino acids can be synthesized by mammalian organisms, while the other half are essential and must be acquired from the nutrition. Absorption of amino acids is mediated by a set of amino acid transporters together with transport of di- and tripeptides. They provide amino acids for systemic needs and for enterocyte metabolism. Absorption is largely complete at the end of the small intestine. The large intestine mediates the uptake of amino acids derived from bacterial metabolism and endogenous sources. Lack of amino acid transporters and peptide transporter delays the absorption of amino acids and changes sensing and usage of amino acids by the intestine. This can affect metabolic health through amino acid restriction, sensing of amino acids, and production of antimicrobial peptides.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, Australian National University, Canberra, Australia;
| |
Collapse
|
19
|
Jonsson WO, Borowik AK, Pranay A, Kinter MT, Mirek ET, Levy JL, Snyder EM, Miller BF, Anthony TG. Kinetic proteomics identifies targeted changes in liver metabolism and the ribo-interactome by dietary sulfur amino acid restriction. GeroScience 2023; 45:2425-2441. [PMID: 36976488 PMCID: PMC10651627 DOI: 10.1007/s11357-023-00758-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/19/2023] [Indexed: 03/29/2023] Open
Abstract
Dietary sulfur amino acid restriction (SAAR) protects against diet-induced obesity, extends healthspan, and coincides with an overall reduction in hepatic protein synthesis. To explore the underpinnings of SAAR-induced slowed growth and its impact on liver metabolism and proteostasis, we resolved changes in hepatic mRNA and protein abundances and compared synthesis rates of individual liver proteins. To achieve this, adult male mice were provided deuterium-labeled drinking water while freely consuming either a regular-fat or high-fat diet that was SAA restricted. Livers from these mice and their respective dietary controls were used to conduct transcriptomic, proteomic, and kinetic proteomic analyses. We found that remodeling of the transcriptome by SAAR was largely agnostic to dietary fat content. Shared signatures included activation of the integrated stress response alongside alterations in metabolic processes impacting lipids, fatty acids, and amino acids. Changes to the proteome correlated poorly with the transcriptome, and yet, functional clustering of kinetic proteomic changes in the liver during SAAR revealed that the management of fatty acids and amino acids were altered to support central metabolism and redox balance. Dietary SAAR also strongly influenced the synthesis rates of ribosomal proteins and ribosome-interacting proteins regardless of dietary fat. Taken together, dietary SAAR alters the transcriptome and proteome in the liver to safely manage increased fatty acid flux and energy use and couples this with targeted changes in the ribo-interactome to support proteostasis and slowed growth.
Collapse
Affiliation(s)
- William O Jonsson
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, 59 Dudley Road - Foran Hall, Room 166, New Brunswick, NJ, 08901, USA
| | - Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael T Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Emily T Mirek
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, 59 Dudley Road - Foran Hall, Room 166, New Brunswick, NJ, 08901, USA
| | - Jordan L Levy
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, 59 Dudley Road - Foran Hall, Room 166, New Brunswick, NJ, 08901, USA
| | - Elizabeth M Snyder
- Department of Animal Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City VA, Oklahoma City, OK, USA
| | - Tracy G Anthony
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, 59 Dudley Road - Foran Hall, Room 166, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
20
|
Mihaylova MM, Chaix A, Delibegovic M, Ramsey JJ, Bass J, Melkani G, Singh R, Chen Z, Ja WW, Shirasu-Hiza M, Latimer MN, Mattison JA, Thalacker-Mercer AE, Dixit VD, Panda S, Lamming DW. When a calorie is not just a calorie: Diet quality and timing as mediators of metabolism and healthy aging. Cell Metab 2023; 35:1114-1131. [PMID: 37392742 PMCID: PMC10528391 DOI: 10.1016/j.cmet.2023.06.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/07/2023] [Accepted: 06/13/2023] [Indexed: 07/03/2023]
Abstract
An epidemic of obesity has affected large portions of the world, increasing the risk of developing many different age-associated diseases, including cancer, cardiovascular disease, and diabetes. In contrast with the prevailing notion that "a calorie is just a calorie," there are clear differences, within and between individuals, in the metabolic response to different macronutrient sources. Recent findings challenge this oversimplification; calories from different macronutrient sources or consumed at different times of day have metabolic effects beyond their value as fuel. Here, we summarize discussions conducted at a recent NIH workshop that brought together experts in calorie restriction, macronutrient composition, and time-restricted feeding to discuss how dietary composition and feeding schedule impact whole-body metabolism, longevity, and healthspan. These discussions may provide insights into the long-sought molecular mechanisms engaged by calorie restriction to extend lifespan, lead to novel therapies, and potentially inform the development of a personalized food-as-medicine approach to healthy aging.
Collapse
Affiliation(s)
- Maria M Mihaylova
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA; The Ohio State University, Comprehensive Cancer Center, Wexner Medical Center, Arthur G. James Cancer Hospital, Columbus, OH, USA.
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Mirela Delibegovic
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill Health Campus, Aberdeen, UK
| | - Jon J Ramsey
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Girish Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rajat Singh
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - William W Ja
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Michele Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Mary N Latimer
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julie A Mattison
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Anna E Thalacker-Mercer
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vishwa Deep Dixit
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA; Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA; Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
| | - Satchidananda Panda
- Regulatory Biology Lab, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
21
|
Rafiq T, Stearns JC, Shanmuganathan M, Azab SM, Anand SS, Thabane L, Beyene J, Williams NC, Morrison KM, Teo KK, Britz-McKibbin P, de Souza RJ. Integrative multiomics analysis of infant gut microbiome and serum metabolome reveals key molecular biomarkers of early onset childhood obesity. Heliyon 2023; 9:e16651. [PMID: 37332914 PMCID: PMC10272340 DOI: 10.1016/j.heliyon.2023.e16651] [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: 12/13/2022] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023] Open
Abstract
Evidence supports a complex interplay of gut microbiome and host metabolism as regulators of obesity. The metabolic phenotype and microbial metabolism of host diet may also contribute to greater obesity risk in children early in life. This study aimed to identify features that discriminated overweight/obese from normal weight infants by integrating gut microbiome and serum metabolome profiles. This prospective analysis included 50 South Asian children living in Canada, selected from the SouTh Asian biRth cohorT (START). Serum metabolites were measured by multisegment injection-capillary electrophoresis-mass spectrometry and the relative abundance of bacterial 16S rRNA gene amplicon sequence variant was evaluated at 1 year. Cumulative body mass index (BMIAUC) and skinfold thickness (SSFAUC) scores were calculated from birth to 3 years as the total area under the growth curve (AUC). BMIAUC and/or SSFAUC >85th percentile was used to define overweight/obesity. Data Integration Analysis for Biomarker discovery using Latent cOmponent (DIABLO) was used to identify discriminant features associated with childhood overweight/obesity. The associations between identified features and anthropometric measures were examined using logistic regression. Circulating metabolites including glutamic acid, acetylcarnitine, carnitine, and threonine were positively, whereas γ-aminobutyric acid (GABA), symmetric dimethylarginine (SDMA), and asymmetric dimethylarginine (ADMA) were negatively associated with childhood overweight/obesity. The abundance of the Pseudobutyrivibrio and Lactobacillus genera were positively, and Clostridium sensu stricto 1 and Akkermansia were negatively associated with childhood overweight/obesity. Integrative analysis revealed that Akkermansia was positively whereas Lactobacillus was inversely correlated with GABA and SDMA, and Pseudobutyrivibrio was inversely correlated with GABA. This study provides insights into metabolic and microbial signatures which may regulate satiety, energy metabolism, inflammatory processes, and/or gut barrier function, and therefore, obesity trajectories in childhood. Understanding the functional capacity of these molecular features and potentially modifiable risk factors such as dietary exposures early in life may offer a novel approach for preventing childhood obesity.
Collapse
Affiliation(s)
- Talha Rafiq
- Medical Sciences Graduate Program, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
| | - Jennifer C. Stearns
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Meera Shanmuganathan
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Sandi M. Azab
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pharmacognosy, Alexandria University, Alexandria 21521, Egypt
| | - Sonia S. Anand
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Lehana Thabane
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biostatistics Unit, Father Sean O’Sullivan Research Centre, The Research Institute, St Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
- Faculty of Health Sciences, University of Johannesburg, Johannesburg 524, South Africa
| | - Joseph Beyene
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | | - Katherine M. Morrison
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Koon K. Teo
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Russell J. de Souza
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
22
|
Flores V, Spicer AB, Sonsalla MM, Richardson NE, Yu D, Sheridan GE, Trautman ME, Babygirija R, Cheng EP, Rojas JM, Yang SE, Wakai MH, Hubbell R, Kasza I, Tomasiewicz JL, Green CL, Dantoin C, Alexander CM, Baur JA, Malecki KC, Lamming DW. Regulation of metabolic health by dietary histidine in mice. J Physiol 2023; 601:2139-2163. [PMID: 36086823 PMCID: PMC9995620 DOI: 10.1113/jp283261] [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: 04/26/2022] [Accepted: 09/01/2022] [Indexed: 11/08/2022] Open
Abstract
Low-protein (LP) diets are associated with a decreased risk of diabetes in humans, and promote leanness and glycaemic control in both rodents and humans. While the effects of an LP diet on glycaemic control are mediated by reduced levels of the branched-chain amino acids, we have observed that reducing dietary levels of the other six essential amino acids leads to changes in body composition. Here, we find that dietary histidine plays a key role in the response to an LP diet in male C57BL/6J mice. Specifically reducing dietary levels of histidine by 67% reduces the weight gain of young, lean male mice, reducing both adipose and lean mass without altering glucose metabolism, and rapidly reverses diet-induced obesity and hepatic steatosis in diet-induced obese male mice, increasing insulin sensitivity. This normalization of metabolic health was associated not with caloric restriction or increased activity, but with increased energy expenditure. Surprisingly, the effects of histidine restriction do not require the energy balance hormone Fgf21. Histidine restriction that was started in midlife promoted leanness and glucose tolerance in aged males but not females, but did not affect frailty or lifespan in either sex. Finally, we demonstrate that variation in dietary histidine levels helps to explain body mass index differences in humans. Overall, our findings demonstrate that dietary histidine is a key regulator of weight and body composition in male mice and in humans, and suggest that reducing dietary histidine may be a translatable option for the treatment of obesity. KEY POINTS: Protein restriction (PR) promotes metabolic health in rodents and humans and extends rodent lifespan. Restriction of specific individual essential amino acids can recapitulate the benefits of PR. Reduced histidine promotes leanness and increased energy expenditure in male mice. Reduced histidine does not extend the lifespan of mice when begun in midlife. Dietary levels of histidine are positively associated with body mass index in humans.
Collapse
Affiliation(s)
- Victoria Flores
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alexandra B. Spicer
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Michelle M. Sonsalla
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nicole E. Richardson
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Deyang Yu
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Grace E. Sheridan
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Michaela E. Trautman
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Reji Babygirija
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eunhae P. Cheng
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jennifer M. Rojas
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shany E. Yang
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Matthew H. Wakai
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ryan Hubbell
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Cara L. Green
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Claudia Dantoin
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Caroline M. Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristen C. Malecki
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Dudley W. Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
- Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI 53706, USA
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA
| |
Collapse
|
23
|
Weber SL, Hustedt K, Schnepel N, Visscher C, Muscher-Banse AS. Modulation of GCN2/eIF2α/ATF4 Pathway in the Liver and Induction of FGF21 in Young Goats Fed a Protein- and/or Phosphorus-Reduced Diet. Int J Mol Sci 2023; 24:ijms24087153. [PMID: 37108315 PMCID: PMC10138370 DOI: 10.3390/ijms24087153] [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: 03/07/2023] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Mammals respond to amino acid (AA) deficiency by initiating an AA response pathway (AAR) that involves the activation of general control nonderepressible 2 (GCN2), phosphorylation of eukaryotic translation initiation factor 2α (eIF2α), and activation of transcription factor 4 (ATF4). In this study, the effects of protein (N) and/or phosphorus (P) restriction on the GCN2/eIF2α/ATF4 pathway in the liver and the induction of fibroblast growth factor 21 (FGF21) in young goats were investigated. An N-reduced diet resulted in a decrease in circulating essential AA (EAA) and an increase in non-essential AA (NEAA), as well as an increase in hepatic mRNA expression of GCN2 and ATF4 and protein expression of GCN2. Dietary N restriction robustly increased both hepatic FGF21 mRNA expression and circulating FGF21 levels. Accordingly, numerous significant correlations demonstrated the effects of the AA profile on the AAR pathway and confirmed an association. Furthermore, activation of the AAR pathway depended on the sufficient availability of P. When dietary P was restricted, the GCN2/eIF2α/ATF4 pathway was not initiated, and no increase in FGF21 was observed. These results illustrate how the AAR pathway responds to N- and/or P-reduced diets in ruminants, thus demonstrating the complexity of dietary component changes.
Collapse
Affiliation(s)
- Sarah L Weber
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany
| | - Karin Hustedt
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany
| | - Nadine Schnepel
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany
| | - Christian Visscher
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, 30173 Hannover, Germany
| | - Alexandra S Muscher-Banse
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany
| |
Collapse
|
24
|
Hope DCD, Tan TMM. Glucagon and energy expenditure; Revisiting amino acid metabolism and implications for weight loss therapy. Peptides 2023; 162:170962. [PMID: 36736539 DOI: 10.1016/j.peptides.2023.170962] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Glucagon receptor (GCGR)-targeted multi-agonists are being developed for the treatment of obesity and metabolic disease. GCGR activity is utilised for its favourable weight loss and metabolic properties, including increased energy expenditure (EE) and hepatic lipid metabolism. GLP1R and GIPR activities are increasingly present in a multi-agonist strategy. Due to the compound effect of increased satiety, reduced food intake and increased energy expenditure, the striking weight loss effects of these multi-agonists has been demonstrated in pre-clinical models of obesity. The precise contribution and mechanism of GCGR activity to enhanced energy expenditure and weight loss in both rodents and humans is not fully understood. In this review, our understanding of glucagon-mediated EE is explored, and an amino acid-centric paradigm contributing to this phenomenon is presented. The current progress of GCGR-targeted multi-agonists in development is also highlighted with a focus on the implications of glucagon-stimulated hypoaminoacidemia.
Collapse
Affiliation(s)
- D C D Hope
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - T M-M Tan
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| |
Collapse
|
25
|
Surugihalli C, Muralidaran V, Ryan CE, Patel K, Zhao D, Sunny NE. Branched-chain amino acids alter cellular redox to induce lipid oxidation and reduce de novo lipogenesis in the liver. Am J Physiol Endocrinol Metab 2023; 324:E299-E313. [PMID: 36791321 PMCID: PMC10042599 DOI: 10.1152/ajpendo.00307.2022] [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: 11/17/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
Metabolic and molecular interactions between branched-chain amino acid (BCAA) and lipid metabolism are evident in insulin-resistant tissues. However, it remains unclear whether insulin resistance is a prerequisite for these relationships and whether BCAAs or their metabolic intermediates can modulate hepatic lipid oxidation and synthesis. We hypothesized that BCAAs can alter hepatic oxidative function and de novo lipogenesis, independent of them being anaplerotic substrates for the mitochondria. Mice (C57BL/6NJ) were reared on a low-fat (LF), LF diet plus 1.5X BCAAs (LB), high-fat (HF) or HF diet plus 1.5X BCAAs (HB) for 12 wk. Hepatic metabolism was profiled utilizing stable isotopes coupled to mass spectrometry and nuclear magnetic resonance, together with fed-to-fasted changes in gene and protein expression. A greater induction of lipid oxidation and ketogenesis on fasting was evident in the BCAA-supplemented, insulin-sensitive livers from LB mice, whereas their rates of hepatic de novo lipogenesis remained lower than their LF counterparts. Onset of insulin resistance in HF and HB mice livers blunted these responses. Whole body turnover of BCAAs and their ketoacids, their serum concentrations, and the ketogenic flux from BCAA catabolism, all remained similar between fasted LF and LB mice. This suggested that the impact of BCAAs on lipid metabolism can occur independent of them or their degradation products fueling anaplerosis through the liver mitochondria. Furthermore, the greater induction of lipid oxidation in the LB livers accompanied higher mitochondrial NADH/NAD+ ratio and higher fed-to-fasting phosphorylation of AMPKα and ACC. Taken together, our results provide evidence that BCAA supplementation, under conditions of insulin sensitivity, improved the feeding-to-fasting induction of hepatic lipid oxidation through changes in cellular redox, thus providing a favorable biochemical environment for flux through β-oxidation and lower de novo lipogenesis.NEW & NOTEWORTHY Branched-chain amino acids (BCAAs) have been shown to modulate lipid metabolic networks in various tissues, especially during insulin resistance. In this study we show that the dietary supplementation of BCAAs to normal, insulin-sensitive mice resulted in higher mitochondrial NADH:NAD+ ratios and AMPK activation in the liver. This change in the cellular redox status provided an optimal metabolic milieu to increase fatty acid oxidation while keeping the rates of de novo lipogenesis lower in the BCAA-supplemented mice livers.
Collapse
Affiliation(s)
- Chaitra Surugihalli
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States
| | - Vaishna Muralidaran
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States
| | - Caitlin E Ryan
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States
| | - Kruti Patel
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States
| | - David Zhao
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States
| | - Nishanth E Sunny
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States
| |
Collapse
|
26
|
Hodge SH, Krauss MZ, Kaymak I, King JI, Howden AJ, Panic G, Grencis RK, Swann JR, Sinclair LV, Hepworth MR. Amino acid availability acts as a metabolic rheostat to determine the magnitude of ILC2 responses. J Exp Med 2023; 220:e20221073. [PMID: 36571761 PMCID: PMC9794837 DOI: 10.1084/jem.20221073] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/15/2022] [Accepted: 12/16/2022] [Indexed: 12/27/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2) are functionally poised, tissue-resident lymphocytes that respond rapidly to damage and infection at mucosal barrier sites. ILC2 reside within complex microenvironments where they are subject to cues from both the diet and invading pathogens-including helminths. Emerging evidence suggests ILC2 are acutely sensitive not only to canonical activating signals but also perturbations in nutrient availability. In the context of helminth infection, we identify amino acid availability as a nutritional cue in regulating ILC2 responses. ILC2 are found to be uniquely preprimed to import amino acids via the large neutral amino acid transporters Slc7a5 and Slc7a8. Cell-intrinsic deletion of these transporters individually impaired ILC2 expansion, while concurrent loss of both transporters markedly impaired the proliferative and cytokine-producing capacity of ILC2. Mechanistically, amino acid uptake determined the magnitude of ILC2 responses in part via tuning of mTOR. These findings implicate essential amino acids as a metabolic requisite for optimal ILC2 responses within mucosal barrier tissues.
Collapse
Affiliation(s)
- Suzanne H. Hodge
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Maria Z. Krauss
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Irem Kaymak
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James I. King
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Andrew J.M. Howden
- Cell Signalling and Immunology Division, School of Life Sciences, University of Dundee, Dundee, UK
| | - Gordana Panic
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, South Kensington, UK
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Richard K. Grencis
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Jonathan R. Swann
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, South Kensington, UK
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Linda V. Sinclair
- Cell Signalling and Immunology Division, School of Life Sciences, University of Dundee, Dundee, UK
| | - Matthew R. Hepworth
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
27
|
Wu J, Wen L, Chen J, Chang Y, Huang R, Lin Y, Shen G, Feng J. Discover boy specific-biomarkers and reveal gender-related metabolic differences in central precocious puberty. J Steroid Biochem Mol Biol 2023; 231:106305. [PMID: 36997004 DOI: 10.1016/j.jsbmb.2023.106305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
The incidence of central precocious puberty (CPP) in boys is rising, but lack of effective molecular biomarkers often leads to delayed treatment and thus the terrible clinical complications in adulthood. This study aims to identify the specific-biomarkers of CPP boys and understand the gender-related differences in metabolic characteristics of CPP. The specific-biomarkers of CPP boys were identified from serum and their combination was optimized by cross-metabolomics combined with linear discriminant analysis effect size analysis after age correction. The differences in metabolic characteristics between boys and girls with CPP were explored by cross-metabolomics and weighted gene co-expression network analysis. Results show that CPP activated in advance the HPG axis and induced gender-related clinical phenotypes. Seven serum metabolites were identified as specific-biomarkers of CPP boys, including acetoacetate, aspartate, choline, creatinine, myo-inositol, N,N-dimethylglycine and N-Acetyl-glycoprotein. The combination of aspartate, choline, myo-inositol and creatinine achieved an optimized diagnosis, where AUC is 0.949, prediction accuracy for CPP boys is 91.1%, and the average accuracy is 0.865. The metabolic disorders of CPP boys mainly involve in glycerophospholipid metabolism, and synthesis and degradation of ketone bodies. Betaine, glutamine, isoleucine, lactate, leucine, lysine, pyruvate, α-&β-glucose were identified as gender-related biomarkers for CPP, and they are mainly involved in glycolysis/gluconeogenesis, pyruvate metabolism, and alanine, aspartate and glutamate metabolism. Biomarkers combination provides a promising diagnostic potential for CPP boy with a favorite sensitivity and specificity. In addition, the differences of metabolic characteristics between boys and girls with CPP will contribute to the development of individualized clinical treatments in CPP.
Collapse
|
28
|
Lysine or Threonine Deficiency Decreases Body Weight Gain in Growing Rats despite an Increase in Food Intake without Increasing Energy Expenditure in Response to FGF21. Nutrients 2022; 15:nu15010197. [PMID: 36615854 PMCID: PMC9824894 DOI: 10.3390/nu15010197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023] Open
Abstract
The objective of this study is to evaluate the effects of a strictly essential amino acid (lysine or threonine; EAA) deficiency on energy metabolism in growing rats. Rats were fed for three weeks severely (15% and 25% of recommendation), moderately (40% and 60%), and adequate (75% and 100%) lysine or threonine-deficient diets. Food intake and body weight were measured daily and indirect calorimetry was performed the week three. At the end of the experimentation, body composition, gene expression, and biochemical analysis were performed. Lysine and threonine deficiency induced a lower body weight gain and an increase in relative food intake. Lysine or threonine deficiency induced liver FGF21 synthesis and plasma release. However, no changes in energy expenditure were observed for lysine deficiency, unlike threonine deficiency, which leads to a decrease in total and resting energy expenditure. Interestingly, threonine severe deficiency, but not lysine deficiency, increase orexigenic and decreases anorexigenic hypothalamic neuropeptides expression, which could explain the higher food intake. Our results show that the deficiency in one EAA, induces a decrease in body weight gain, despite an increased relative food intake, without any increase in energy expenditure despite an induction of FGF21.
Collapse
|
29
|
Fulton TL, Mirth CK, Piper MDW. Restricting a single amino acid cross-protects Drosophila melanogaster from nicotine poisoning through mTORC1 and GCN2 signalling. Open Biol 2022; 12:220319. [PMID: 36514979 PMCID: PMC9748770 DOI: 10.1098/rsob.220319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Dietary interventions that restrict protein intake have repeatedly been shown to offer beneficial health outcomes to the consumer. Benefits such as increased stress tolerance can be observed when individual amino acids are restricted, thus mimicking dietary protein restriction. Here, we sought to further understand the relationship between dietary amino acids and stress tolerance using Drosophila melanogaster. Using a chemically defined medium for Drosophila, we found that transiently restricting adult flies of a single essential amino acid generally protects against a lethal dose of the naturally occurring insecticide, nicotine. This protection varied with the identity of the focal amino acid and depended on the duration and intensity of its restriction. To understand the molecular basis of these effects, we modified the signalling of two cellular sensors of amino acids, GCN2 and mTORC1, in combination with amino acid restriction. We found that GCN2 was necessary for diets to protect against nicotine, whereas the suppression of mTORC1 was sufficient to induce nicotine resistance. This finding implies that amino acid restriction acts via amino acid signalling to cross-protect against seemingly unrelated stressors. Altogether, our study offers new insights into the physiological responses to restriction of individual amino acids that confer stress tolerance.
Collapse
Affiliation(s)
- Tahlia L. Fulton
- School of Biological Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Christen K. Mirth
- School of Biological Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Matthew D. W. Piper
- School of Biological Sciences, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
30
|
Hope DCD, Hinds CE, Lopes T, Vincent ML, Shrewsbury JV, Yu ATC, Davies I, Scott R, Jones B, Murphy KG, Minnion JS, Sardini A, Carling D, Lutz TA, Bloom SR, Tan TMM, Owen BM. Hypoaminoacidemia underpins glucagon-mediated energy expenditure and weight loss. Cell Rep Med 2022; 3:100810. [PMID: 36384093 PMCID: PMC9729826 DOI: 10.1016/j.xcrm.2022.100810] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 09/26/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022]
Abstract
Glucagon analogs show promise as components of next-generation, multi-target, anti-obesity therapeutics. The biology of chronic glucagon treatment, in particular, its ability to induce energy expenditure and weight loss, remains poorly understood. Using a long-acting glucagon analog, G108, we demonstrate that glucagon-mediated body weight loss is intrinsically linked to the hypoaminoacidemia associated with its known amino acid catabolic action. Mechanistic studies reveal an energy-consuming response to low plasma amino acids in G108-treated mice, prevented by dietary amino acid supplementation and mimicked by a rationally designed low amino acid diet. Therefore, low plasma amino acids are a pre-requisite for G108-mediated energy expenditure and weight loss. However, preventing hypoaminoacidemia with additional dietary protein does not affect the ability of G108 to improve glycemia or hepatic steatosis in obese mice. These studies provide a mechanism for glucagon-mediated weight loss and confirm the hepatic glucagon receptor as an attractive molecular target for metabolic disease therapeutics.
Collapse
Affiliation(s)
- David C D Hope
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Charlotte E Hinds
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tatiana Lopes
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Matthew L Vincent
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jed V Shrewsbury
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Arthur T C Yu
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Iona Davies
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Rebecca Scott
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ben Jones
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Kevin G Murphy
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - James S Minnion
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alessandro Sardini
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - David Carling
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Stephen R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M M Tan
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Bryn M Owen
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
31
|
Jin T. Fibroblast growth factor 21 and dietary interventions: what we know and what we need to know next. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:524-530. [PMID: 37724164 PMCID: PMC10388781 DOI: 10.1515/mr-2022-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/10/2022] [Indexed: 09/20/2023]
Abstract
Dietary interventions include the change of dietary styles, such as fasting and dietary or nutrient restrictions; or the addition of plant-derived compounds (such as polyphenols known as curcumin, resveratrol, or anthocyanin, or other nutraceuticals) into the diet. During the past a few decades, large number of studies have demonstrated therapeutic activities of these dietary interventions on metabolic and other diseases in human subjects or various animal models. Mechanisms underlying those versatile therapeutic activities, however, remain largely unclear. Interestingly, recent studies have shown that fibroblast growth factor 21 (FGF21), a liver-derived hormone or hepatokine, mediates metabolic beneficial effects of certain dietary polyphenols as well as protein restriction. Here I have briefly summarized functions of FGF21, highlighted related dietary interventions, and presented literature discussions on role of FGF21 in mediating function of dietary polyphenol intervention and protein restriction. This is followed by presenting my perspective view, with the involvement of gut microbiota. It is anticipated that further breakthroughs in this field in the near future will facilitate conceptual merge of classical medicine and modern medicine.
Collapse
Affiliation(s)
- Tianru Jin
- Division of Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, TorontoCanada
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, TorontoCanada
| |
Collapse
|
32
|
Multiomics assessment of dietary protein titration reveals altered hepatic glucose utilization. Cell Rep 2022; 40:111187. [PMID: 35977507 DOI: 10.1016/j.celrep.2022.111187] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 06/17/2021] [Accepted: 07/20/2022] [Indexed: 12/13/2022] Open
Abstract
Dietary protein restriction (PR) has rapid effects on metabolism including improved glucose and lipid homeostasis, via multiple mechanisms. Here, we investigate responses of fecal microbiome, hepatic transcriptome, and hepatic metabolome to six diets with protein from 18% to 0% of energy in mice. PR alters fecal microbial composition, but metabolic effects are not transferable via fecal transplantation. Hepatic transcriptome and metabolome are significantly altered in diets with lower than 10% energy from protein. Changes upon PR correlate with calorie restriction but with a larger magnitude and specific changes in amino acid (AA) metabolism. PR increases steady-state aspartate, serine, and glutamate and decreases glucose and gluconeogenic intermediates. 13C6 glucose and glycerol tracing reveal increased fractional enrichment in aspartate, serine, and glutamate. Changes remain intact in hepatic ATF4 knockout mice. Together, this demonstrates an ATF4-independent shift in gluconeogenic substrate utilization toward specific AAs, with compensation from glycerol to promote a protein-sparing response.
Collapse
|
33
|
Anderson EM, Rozowsky JM, Fazzone BJ, Schmidt EA, Stevens BR, O’Malley KA, Scali ST, Berceli SA. Temporal Dynamics of the Intestinal Microbiome Following Short-Term Dietary Restriction. Nutrients 2022; 14:2785. [PMID: 35889742 PMCID: PMC9318361 DOI: 10.3390/nu14142785] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 12/04/2022] Open
Abstract
Short-term dietary restriction has been proposed as an intriguing pre-operative conditioning strategy designed to attenuate the surgical stress response and improve outcomes. However, it is unclear how this nutritional intervention influences the microbiome, which is known to modulate the systemic condition. Healthy individuals were recruited to participate in a four-day, 70% protein-restricted, 30% calorie-restricted diet, and stool samples were collected at baseline, after the restricted diet, and after resuming normal food intake. Taxonomy and functional pathway analysis was performed via shotgun metagenomic sequencing, prevalence filtering, and differential abundance analysis. High prevalence species were altered by the dietary intervention but quickly returned to baseline after restarting a regular diet. Composition and functional changes after the restricted diet included the decreased relative abundance of commensal bacteria and a catabolic phenotype. Notable species changes included Faecalibacterium prausnitzii and Roseburia intestinalis, which are major butyrate producers within the colon and are characteristically decreased in many disease states. The macronutrient components of the diet might have influenced these changes. We conclude that short-term dietary restriction modulates the ecology of the gut microbiome, with this modulation being characterized by a relative dysbiosis.
Collapse
Affiliation(s)
- Erik M. Anderson
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Jared M. Rozowsky
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Brian J. Fazzone
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Emilie A. Schmidt
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Bruce R. Stevens
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA;
| | - Kerri A. O’Malley
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Salvatore T. Scali
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Scott A. Berceli
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| |
Collapse
|
34
|
Jonsson WO, Mirek ET, Wek RC, Anthony TG. Activation and execution of the hepatic integrated stress response by dietary essential amino acid deprivation is amino acid specific. FASEB J 2022; 36:e22396. [PMID: 35690926 PMCID: PMC9204950 DOI: 10.1096/fj.202200204rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/16/2022] [Accepted: 05/24/2022] [Indexed: 12/30/2022]
Abstract
Dietary removal of an essential amino acid (EAA) triggers the integrated stress response (ISR) in liver. Herein, we explored the mechanisms that activate the ISR and execute changes in transcription and translation according to the missing EAA. Wild‐type mice and mice lacking general control nonderepressible 2 (Gcn2) were fed an amino acid complete diet or a diet devoid of either leucine or sulfur amino acids (methionine and cysteine). Serum and liver leucine concentrations were significantly reduced within the first 6 h of feeding a diet lacking leucine, corresponding with modest, GCN2‐dependent increases in Atf4 mRNA translation and induction of selected ISR target genes (Fgf21, Slc7a5, Slc7a11). In contrast, dietary removal of the sulfur amino acids lowered serum methionine, but not intracellular methionine, and yet hepatic mRNA abundance of Atf4, Fgf21, Slc7a5, Slc7a11 substantially increased regardless of GCN2 status. Liver tRNA charging levels did not correlate with intracellular EAA concentrations or GCN2 status and remained similar to mice fed a complete diet. Furthermore, loss of Gcn2 increased the occurrence of ribosome collisions in liver and derepressed mechanistic target of rapamycin complex 1 signal transduction, but these changes did not influence execution of the ISR. We conclude that ISR activation is directed by intracellular EAA concentrations, but ISR execution is not. Furthermore, a diet devoid of sulfur amino acids does not require GCN2 for the ISR to execute changes to the transcriptome.
Collapse
Affiliation(s)
- William O Jonsson
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey, USA
| | - Emily T Mirek
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey, USA
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tracy G Anthony
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
35
|
Kosakamoto H, Okamoto N, Aikawa H, Sugiura Y, Suematsu M, Niwa R, Miura M, Obata F. Sensing of the non-essential amino acid tyrosine governs the response to protein restriction in Drosophila. Nat Metab 2022; 4:944-959. [PMID: 35879463 DOI: 10.1038/s42255-022-00608-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 06/15/2022] [Indexed: 11/08/2022]
Abstract
The intake of dietary protein regulates growth, metabolism, fecundity and lifespan across various species, which makes amino acid (AA)-sensing vital for adaptation to the nutritional environment. The general control nonderepressible 2 (GCN2)-activating transcription factor 4 (ATF4) pathway and the mechanistic target of rapamycin complex 1 (mTORC1) pathway are involved in AA-sensing. However, it is not fully understood which AAs regulate these two pathways in living animals and how they coordinate responses to protein restriction. Here we show in Drosophila that the non-essential AA tyrosine (Tyr) is a nutritional cue in the fat body necessary and sufficient for promoting adaptive responses to a low-protein diet, which entails reduction of protein synthesis and mTORC1 activity and increased food intake. This adaptation is regulated by dietary Tyr through GCN2-independent induction of ATF4 target genes in the fat body. This study identifies the Tyr-ATF4 axis as a regulator of the physiological response to a low-protein diet and sheds light on the essential function of a non-essential nutrient.
Collapse
Affiliation(s)
- Hina Kosakamoto
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- RIKEN Center for Biosystems and Dynamics Research, Kobe, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Hide Aikawa
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Fumiaki Obata
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- RIKEN Center for Biosystems and Dynamics Research, Kobe, Japan.
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan.
- Laboratory of Molecular Cell Biology and Development, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| |
Collapse
|
36
|
Srivastava A, Lu J, Gadalla DS, Hendrich O, Grönke S, Partridge L. The Role of GCN2 Kinase in Mediating the Effects of Amino Acids on Longevity and Feeding Behaviour in Drosophila. FRONTIERS IN AGING 2022; 3:944466. [PMID: 35821827 PMCID: PMC9261369 DOI: 10.3389/fragi.2022.944466] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 05/30/2022] [Indexed: 02/03/2023]
Abstract
Restriction of amino acids in the diet can extend lifespan in diverse species ranging from flies to mammals. However, the role of individual amino acids and the underlying molecular mechanisms are only partially understood. The evolutionarily conserved serine/threonine kinase General Control Nonderepressible 2 (GCN2) is a key sensor of amino acid deficiency and has been implicated in the response of lifespan to dietary restriction (DR). Here, we generated a novel Drosophila GCN2 null mutant and analyzed its response to individual amino acid deficiency. We show that GCN2 function is essential for fly development, longevity and feeding behaviour under long-term, but not short-term, deprivation of all individual essential amino acids (EAAs) except for methionine. GCN2 mutants were longer-lived than control flies and showed normal feeding behaviour under methionine restriction. Thus, in flies at least two systems regulate these responses to amino acid deprivation. Methionine deprivation acts via a GCN2-independent mechanism, while all other EAA are sensed by GCN2. Combined deficiency of methionine and a second EAA blocked the response of GCN2 mutants to methionine, suggesting that these two pathways are interconnected. Wild type flies showed a short-term rejection of food lacking individual EAA, followed by a long-term compensatory increase in food uptake. GCN2 mutants also showed a short-term rejection of food deprived of individual EAA, but were unable to mount the compensatory long-term increase in food uptake. Over-expression of the downstream transcription factor ATF4 partially rescued the response of feeding behaviour in GCN2 mutants to amino acid deficiency. Phenotypes of GCN2 mutants induced by leucine and tryptophan, but not isoleucine, deficiency were partially rescued by ATF4 over-expression. The exact function of GCN2 as an amino acid sensor in vivo and the downstream action of its transcription factor effector ATF4 are thus context-specific with respect to the EAA involved.
Collapse
Affiliation(s)
| | - Jiongming Lu
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Oliver Hendrich
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, United Kingdom
| |
Collapse
|
37
|
Raubenheimer D, Senior AM, Mirth C, Cui Z, Hou R, Le Couteur DG, Solon-Biet SM, Léopold P, Simpson SJ. An integrative approach to dietary balance across the life course. iScience 2022; 25:104315. [PMID: 35602946 PMCID: PMC9117877 DOI: 10.1016/j.isci.2022.104315] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Animals require specific blends of nutrients that vary across the life course and with circumstances, e.g., health and activity levels. Underpinning and complicating these requirements is that individual traits may be optimized on different dietary compositions leading to nutrition-mediated trade-offs among outcomes. Additionally, the food environment may constrain which nutrient mixtures are achievable. Natural selection has equipped animals for solving such multi-dimensional, dynamic challenges of nutrition, but little is understood about the details and their theoretical and practical implications. We present an integrative framework, nutritional geometry, which models complex nutritional interactions in the context of multiple nutrients and across levels of biological organization (e.g., cellular, individual, and population) and levels of analysis (e.g., mechanistic, developmental, ecological, and evolutionary). The framework is generalizable across different situations and taxa. We illustrate this using examples spanning insects to primates and settings (laboratory, and the wild), and demonstrate its relevance for human health.
Collapse
Affiliation(s)
- David Raubenheimer
- The University of Sydney, Charles Perkins Centre and School of Life and Environmental Sciences, Sydney, Australia
- Zhengzhou University, Centre for Nutritional Ecology and Centre for Sport Nutrition and Health, Zhengzhou, China
| | - Alistair M. Senior
- The University of Sydney, Charles Perkins Centre and School of Life and Environmental Sciences, Sydney, Australia
- The University of Sydney, School of Mathematics and Statistics, Sydney, Australia
| | - Christen Mirth
- Monash University, School of Biological Science, Melbourne, Australia
| | - Zhenwei Cui
- Zhengzhou University, Centre for Nutritional Ecology and Centre for Sport Nutrition and Health, Zhengzhou, China
| | - Rong Hou
- Northwest University, Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Xi’an, China
| | - David G. Le Couteur
- The University of Sydney, Charles Perkins Centre and Faculty of Medicine and Health, Concord Clinical School, ANZAC Research Institute, Centre for Education and Research on Ageing, Sydney, Australia
| | - Samantha M. Solon-Biet
- The University of Sydney, Charles Perkins Centre and School of Medical Sciences, Sydney, Australia
| | - Pierre Léopold
- Institut Curie, PSL Research University, CNRS UMR3215, INSERM U934, UPMC Paris-Sorbonne, Paris, France
| | - Stephen J. Simpson
- The University of Sydney, Charles Perkins Centre and School of Life and Environmental Sciences, Sydney, Australia
| |
Collapse
|
38
|
Paulusma CC, Lamers W, Broer S, van de Graaf SFJ. Amino acid metabolism, transport and signalling in the liver revisited. Biochem Pharmacol 2022; 201:115074. [PMID: 35568239 DOI: 10.1016/j.bcp.2022.115074] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/02/2022]
Abstract
The liver controls the systemic exposure of amino acids entering via the gastro-intestinal tract. For most amino acids except branched chain amino acids, hepatic uptake is very efficient. This implies that the liver orchestrates amino acid metabolism and also controls systemic amino acid exposure. Although many amino acid transporters have been identified, cloned and investigated with respect to substrate specificity, transport mechanism, and zonal distribution, which of these players are involved in hepatocellular amino acid transport remains unclear. Here, we aim to provide a review of current insight into the molecular machinery of hepatic amino acid transport. Furthermore, we place this information in a comprehensive overview of amino acid transport, signalling and metabolism.
Collapse
Affiliation(s)
- Coen C Paulusma
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Wouter Lamers
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - Stefan Broer
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Research School of Biology, Australian National University, Canberra, Australia
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
39
|
Gautrey SL, Simons MJP. Amino acid availability is not essential for lifespan extension by dietary restriction in the fly. J Gerontol A Biol Sci Med Sci 2022; 77:2181-2185. [PMID: 35486979 DOI: 10.1093/gerona/glac100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Indexed: 11/12/2022] Open
Abstract
Dietary restriction (DR) is one of the most potent ways to extend health- and lifespan. Key progress in understanding the mechanisms of DR, and ageing more generally, was made when dietary protein, and more specifically essential amino acids (EAA), were identified as the dietary component to restrict to obtain DR's health and lifespan benefits. This role of dietary amino acids has influenced work on ageing mechanisms, especially in nutrient sensing, e.g. Tor and insulin(-like) signalling networks. Experimental biology in Drosophila melanogaster has been instrumental in generating and confirming the hypothesis that EAA availability is important in ageing. Here, we expand on previous work testing the involvement of EAA in DR through large scale (N=6,238) supplementation experiments across four diets and two genotypes in female flies. Surprisingly, we find that EAA are not essential to DR's lifespan benefits. Importantly, we do identify the fecundity benefits of EAA supplementation suggesting the supplemented EAA were bioavailable. Furthermore, we find that the effects of amino acids on lifespan vary by diet and genetic line studied and that at our most restricted diet fecundity is constrained by other nutrients than EAA. We suggest that DR for optimal health is a concert of nutritional effects, orchestrated by genetic, dietary and other environmental interactions. Our results question the universal importance of amino acid availability in the biology of ageing and DR.
Collapse
Affiliation(s)
- Sarah L Gautrey
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, UK
| | - Mirre J P Simons
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, UK
| |
Collapse
|
40
|
FGF21 is required for protein restriction to extend lifespan and improve metabolic health in male mice. Nat Commun 2022; 13:1897. [PMID: 35393401 PMCID: PMC8991228 DOI: 10.1038/s41467-022-29499-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Dietary protein restriction is increasingly recognized as a unique approach to improve metabolic health, and there is increasing interest in the mechanisms underlying this beneficial effect. Recent work indicates that the hormone FGF21 mediates the metabolic effects of protein restriction in young mice. Here we demonstrate that protein restriction increases lifespan, reduces frailty, lowers body weight and adiposity, improves physical performance, improves glucose tolerance, and alters various metabolic markers within the serum, liver, and adipose tissue of wildtype male mice. Conversely, mice lacking FGF21 fail to exhibit metabolic responses to protein restriction in early life, and in later life exhibit early onset of age-related weight loss, reduced physical performance, increased frailty, and reduced lifespan. These data demonstrate that protein restriction in aging male mice exerts marked beneficial effects on lifespan and metabolic health and that a single metabolic hormone, FGF21, is essential for the anti-aging effect of this dietary intervention.
Collapse
|
41
|
Choi W, Kim J, Ko JW, Choi A, Kwon YH. Effects of maternal branched-chain amino acid and alanine supplementation on growth and biomarkers of protein metabolism in dams fed a low-protein diet and their offspring. Amino Acids 2022; 54:977-988. [PMID: 35353249 DOI: 10.1007/s00726-022-03157-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/13/2022] [Indexed: 11/24/2022]
Abstract
A considerable number of studies have reported that maternal protein restriction may disturb fetal growth and organ development due to a lower availability of amino acids. Leucine, one of branched-chain amino acid (BCAA) promotes protein synthesis through mechanistic target of rapamycin signaling. Here, we investigated the effects of BCAA supplementation in the dams fed a low-protein diet on serum and hepatic biochemical parameters of protein metabolism of dams and their offspring. Female ICR mice were fed a control (20% casein), a low-protein (10% casein), a low-protein with 2% BCAAs or a low-protein with 2% alanine diet for 2 weeks before mating and then throughout pregnancy and lactation. Alanine was used as an amino nitrogen control for the BCAA. Dams and their male offspring were sacrificed at postnatal day 21. There were no changes in body weight and fat mass in low-protein fed dams; however, BCAA supplementation significantly increased fat mass and serum leptin levels. Low-protein diet consumption reduced maternal protein synthesis based on biochemical analysis of serum albumin and hepatic protein levels and immunoblotting of S6 protein, which were increased by BCAA and alanine supplementation. Offspring from dams fed a low-protein diet exhibited lower body and organ weights. Body weight and hepatic protein levels of the offspring were increased by alanine supplementation. However, the decreased serum biochemical parameters, including glucose, triglyceride, total protein and albumin levels in the low-protein offspring group were not changed in response to BCAA or alanine supplementation. A reduced density of the hepatic vessel system in the offspring from dams fed a low-protein diet was restored in the offspring from dams fed either BCAA and alanine-supplemented diet. These results suggest that supplementation of amino nitrogen per se may be responsible for inducing hepatic protein synthesis in the dams fed a low-protein diet and alleviating the distorted growth and liver development of their offspring.
Collapse
Affiliation(s)
- Wooseon Choi
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.,Department of Pharmacology, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Juhae Kim
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Je Won Ko
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Alee Choi
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea. .,Research Institute of Human Ecology, Seoul National University, Seoul, South Korea.
| |
Collapse
|
42
|
Abstract
BACKGROUND Obesity develops due to an imbalance in energy homeostasis, wherein energy intake exceeds energy expenditure. Accumulating evidence shows that manipulations of dietary protein and their component amino acids affect the energy balance, resulting in changes in fat mass and body weight. Amino acids are not only the building blocks of proteins but also serve as signals regulating multiple biological pathways. SCOPE OF REVIEW We present the currently available evidence regarding the effects of dietary alterations of a single essential amino acid (EAA) on energy balance and relevant signaling mechanisms at both central and peripheral levels. We summarize the association between EAAs and obesity in humans and the clinical use of modifying the dietary EAA composition for therapeutic intervention in obesity. Finally, similar mechanisms underlying diets varying in protein levels and diets altered of a single EAA are described. The current review would expand our understanding of the contribution of protein and amino acids to energy balance control, thus helping discover novel therapeutic approaches for obesity and related diseases. MAJOR CONCLUSIONS Changes in circulating EAA levels, particularly increased branched-chain amino acids (BCAAs), have been reported in obese human and animal models. Alterations in dietary EAA intake result in improvements in fat and weight loss in rodents, and each has its distinct mechanism. For example, leucine deprivation increases energy expenditure, reduces food intake and fat mass, primarily through regulation of the general control nonderepressible 2 (GCN2) and mammalian target of rapamycin (mTOR) signaling. Methionine restriction by 80% decreases fat mass and body weight while developing hyperphagia, primarily through fibroblast growth factor 21 (FGF-21) signaling. Some effects of diets with different protein levels on energy homeostasis are mediated by similar mechanisms. However, reports on the effects and underlying mechanisms of dietary EAA imbalances on human body weight are few, and more investigations are needed in future.
Collapse
Affiliation(s)
- Fei Xiao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China.
| |
Collapse
|
43
|
Chen Y, Wu X, Zhang J, Pan G, Wang X, Guo X, Wang J, Cui X, Gao H, Cheng M, Yang J, Zhang C, Jiang F. Amino acid starvation-induced LDLR trafficking accelerates lipoprotein endocytosis and LDL clearance. EMBO Rep 2022; 23:e53373. [PMID: 34994492 PMCID: PMC8892268 DOI: 10.15252/embr.202153373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
Mammalian cells utilize Akt‐dependent signaling to deploy intracellular Glut4 toward cell surface to facilitate glucose uptake. Low‐density lipoprotein receptor (LDLR) is the cargo receptor mediating endocytosis of apolipoprotein B‐containing lipoproteins. However, signaling‐controlled regulation of intracellular LDLR trafficking remains elusive. Here, we describe a unique amino acid stress response, which directs the deployment of intracellular LDLRs, causing enhanced LDL endocytosis, likely via Ca2+ and calcium/calmodulin‐dependent protein kinase II‐mediated signalings. This response is independent of induction of autophagy. Amino acid stress‐induced increase in LDL uptake in vitro is comparable to that by pravastatin. In vivo, acute AAS challenge for up to 72 h enhanced the rate of hepatic LDL uptake without changing the total expression level of LDLR. Reducing dietary amino acids by 50% for 2 to 4 weeks ameliorated high fat diet‐induced hypercholesterolemia in heterozygous LDLR‐deficient mice, with reductions in both LDL and VLDL fractions. We suggest that identification of signaling‐controlled regulation of intracellular LDLR trafficking has advanced our understanding of the LDLR biology, and may benefit future development of additional therapeutic strategies for treating hypercholesterolemia.
Collapse
Affiliation(s)
- Ye Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China.,The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guopin Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyun Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaosun Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianli Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaopei Cui
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiqing Gao
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mei Cheng
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingwen Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China.,The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fan Jiang
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
44
|
Bröer S. Amino acid transporters as modulators of glucose homeostasis. Trends Endocrinol Metab 2022; 33:120-135. [PMID: 34924221 DOI: 10.1016/j.tem.2021.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/01/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022]
Abstract
Amino acids modulate glucose homeostasis. Cytosolic levels of amino acids are regulated by amino acid transporters, modulating insulin release, protein synthesis, cell proliferation, cell fate, and metabolism. In β-cells, amino acid transporters modulate incretin-stimulated insulin release. In the liver, amino acid transporters provide glutamine and alanine for gluconeogenesis. Intestinal amino acid transporters facilitate the intake of amino acids causing protein restriction when inactive. Adipocyte development is regulated by amino acid transporters through activation of mechanistic target of rapamycin (mTORC1) and amino acid-related metabolites. The accumulation and metabolism of branched-chain amino acids (BCAAs) in muscle depends on transporters. The integration between amino acid metabolism and transport is critical for the maintenance and function of tissues and cells involved in glucose homeostasis.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, Australian National University, Acton 2601, Australia.
| |
Collapse
|
45
|
Wu CT, Chaffin AT, Ryan KK. Fibroblast Growth Factor 21 Facilitates the Homeostatic Control of Feeding Behavior. J Clin Med 2022; 11:580. [PMID: 35160033 PMCID: PMC8836936 DOI: 10.3390/jcm11030580] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a stress hormone that is released from the liver in response to nutritional and metabolic challenges. In addition to its well-described effects on systemic metabolism, a growing body of literature now supports the notion that FGF21 also acts via the central nervous system to control feeding behavior. Here we review the current understanding of FGF21 as a hormone regulating feeding behavior in rodents, non-human primates, and humans. First, we examine the nutritional contexts that induce FGF21 secretion. Initial reports describing FGF21 as a 'starvation hormone' have now been further refined. FGF21 is now better understood as an endocrine mediator of the intracellular stress response to various nutritional manipulations, including excess sugars and alcohol, caloric deficits, a ketogenic diet, and amino acid restriction. We discuss FGF21's effects on energy intake and macronutrient choice, together with our current understanding of the underlying neural mechanisms. We argue that the behavioral effects of FGF21 function primarily to maintain systemic macronutrient homeostasis, and in particular to maintain an adequate supply of protein and amino acids for use by the cells.
Collapse
Affiliation(s)
| | | | - Karen K. Ryan
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA 95616, USA; (C.-T.W.); (A.T.C.)
| |
Collapse
|
46
|
Solon-Biet SM, Griffiths L, Fosh S, Le Couteur DG, Simpson SJ, Senior AM. Meta-analysis links dietary branched-chain amino acids to metabolic health in rodents. BMC Biol 2022; 20:19. [PMID: 35031039 PMCID: PMC8760763 DOI: 10.1186/s12915-021-01201-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Background The role of dietary branched chain amino acids (BCAAs) and their effect on metabolic health is complex. How dietary BCAA levels and their interaction with background nutrition affect health is unclear. Here, we used meta-analysis and meta-regression, together with the nutritional modelling, to analyse the results of rodent studies that increased the level of dietary BCAAs and measured circulating levels, outcomes related to metabolic health, body mass and food intake. Results Across all studies, increasing dietary BCAAs resulted in increased levels of circulating BCAAs. These effects, however, were heavily moderated by background dietary levels whereby on high BCAA diets, further increases were not reflected in the blood. Impaired glucose tolerance was associated with elevated dietary BCAAs, with the greatest effect occurring with a simultaneous increase in total protein intake. Effects of dietary BCAAs on plasma glucose, insulin, or HOMA emerged only when dietary macronutrient background was considered. We found that elevated dietary BCAAs increases % body fat, with largest increases in adiposity occurring when BCAAs are increased on a high protein, low carbohydrate dietary background. Finally, we found that increased dietary BCAAs were associated with increased food intake when the background diet was low in BCAAs. Conclusion Our data highlights the interaction between BCAAs and background nutrition. We show that the effects of BCAAs on metabolic health cannot be studied in isolation but must be considered as part of complex mixture of dietary components. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01201-2.
Collapse
Affiliation(s)
- Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia. .,School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia.
| | - Lucy Griffiths
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Sophie Fosh
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - David G Le Couteur
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia.,Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia.,Ageing and Alzheimers Institute and Centre for Education and Research on Ageing, Concord Hospital, Sydney, NSW, Australia.,ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia. .,School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia. .,School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
47
|
Green CL, Lamming DW, Fontana L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat Rev Mol Cell Biol 2022; 23:56-73. [PMID: 34518687 PMCID: PMC8692439 DOI: 10.1038/s41580-021-00411-4] [Citation(s) in RCA: 243] [Impact Index Per Article: 121.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 02/08/2023]
Abstract
Dietary restriction with adequate nutrition is the gold standard for delaying ageing and extending healthspan and lifespan in diverse species, including rodents and non-human primates. In this Review, we discuss the effects of dietary restriction in these mammalian model organisms and discuss accumulating data that suggest that dietary restriction results in many of the same physiological, metabolic and molecular changes responsible for the prevention of multiple ageing-associated diseases in humans. We further discuss how different forms of fasting, protein restriction and specific reductions in the levels of essential amino acids such as methionine and the branched-chain amino acids selectively impact the activity of AKT, FOXO, mTOR, nicotinamide adenine dinucleotide (NAD+), AMP-activated protein kinase (AMPK) and fibroblast growth factor 21 (FGF21), which are key components of some of the most important nutrient-sensing geroprotective signalling pathways that promote healthy longevity.
Collapse
Affiliation(s)
- Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
- Department of Clinical and Experimental Sciences, Brescia University School of Medicine, Brescia, Italy.
| |
Collapse
|
48
|
Guan Z, Li Y, Hu S, Mo C, He D, Huang Z, Liao M. Screening and identification of differential metabolites in serum and urine of bamaxiang pigs bitten by trimeresurus stejnegeri based on UPLC-Q-TOF/MS metabolomics technology. J Toxicol Sci 2022; 47:389-407. [PMID: 36104186 DOI: 10.2131/jts.47.389] [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: 11/02/2022]
Abstract
Trimeresurus stejnegeri is one of the top ten venomous snakes in China, and its bite causes acute and severe diseases. Elucidating the metabolic changes of the body caused by Trimeresurus stejnegeri bite will be beneficial to the diagnosis and treatment of snakebite. Thus, an animal pig model of Trimeresurus stejnegeri bite was established, and then the metabolites of serum and urine were subsequently screened and identified in both ESI+ and ESI- modes identified by ultra-performance liquid chromatography-quadrupole-time of flight-mass spectrometry (UPLC-Q-TOF-MS) methods. There are 9 differential metabolites in serum, including Oleic acid, Lithocholic acid, Deoxycholic acid, Hypoxanthine, etc. There are 11 differential metabolites in urine, including Dopamine, Thiocysteine, Arginine, Indoleacetaldehyde, etc. Serum enrichment pathway analysis showed that 5 metabolic pathways, including Tryptophanuria, Liver disease due to cystic fibrosis, Hartnup disease, Hyperbaric oxygen exposure and Biliary cirrhosis, the core metabolites in these pathways, including deoxycholic acid, lithocholic acid, tryptophan and hypoxanthine, changed significantly. Urine enrichment pathway analysis showed that 4 metabolic pathways, including Aromatic L-Amino Acid Decarboxylase, Vitiligo, Blue Diaper Syndrome and Hyperargininemia, the core metabolites in these pathways including dopamine, 5-hydroxyindole acetic acid and arginine. Taken together, the current study has successfully established an animal model of Trimeresurus stejnegeri bite, and identified the metabolic markers and metabolic pathways of Trimeresurus stejnegeri bite. These metabolites and pathways may have potential application value and provide a therapeutic basis for the treatment of Trimeresurus stejnegeri bite.
Collapse
Affiliation(s)
- ZheZhe Guan
- Institute of Life Sciences of Guangxi Medical University, China
| | - YaLan Li
- Institute of Life Sciences of Guangxi Medical University, China
| | - ShaoCong Hu
- Institute of Life Sciences of Guangxi Medical University, China
| | - CaiFeng Mo
- Institute of Life Sciences of Guangxi Medical University, China
| | - DongLing He
- Institute of Life Sciences of Guangxi Medical University, China
| | - Zhi Huang
- Institute of Life Sciences of Guangxi Medical University, China
| | - Ming Liao
- Institute of Life Sciences of Guangxi Medical University, China
| |
Collapse
|
49
|
Wang D, Ye J, Shi R, Zhao B, Liu Z, Lin W, Liu X. Dietary protein and amino acid restriction: Roles in metabolic health and aging-related diseases. Free Radic Biol Med 2022; 178:226-242. [PMID: 34890767 DOI: 10.1016/j.freeradbiomed.2021.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022]
Abstract
The prevalence of obesity is a worldwide phenomenon in all age groups and is associated with aging-related diseases such as type 2 diabetes, as well metabolic and cardiovascular diseases. The use of dietary restriction (DR) while avoiding malnutrition has many profound beneficial effects on aging and metabolic health, and dietary protein or specific amino acid (AA) restrictions, rather than overall calorie intake, are considered to play key roles in the effects of DR on host health. Whereas comprehensive reviews of the underlying mechanisms are limited, protein restriction and methionine (Met) restriction improve metabolic health and aging-related neurodegenerative diseases, and may be associated with FGF21, mTOR and autophagy, improved mitochondrial function and oxidative stress. Circulating branched-chain amino acids (BCAAs) are inversely correlated with metabolic health, and BCAAs and leucine (Leu) restriction promote metabolic homeostasis in rodents. Although tryptophan (Trp) restriction extends the lifespan of rodents, the Trp-restricted diet is reported to increase inflammation in aged mice, while severe Trp restriction has side effects such as anorexia. Furthermore, inadequate protein intake in the elderly increases the risk of muscle-centric health. Therefore, the restriction of specific AAs may be an effective and executable dietary manipulation for metabolic and aging-related health in humans, which warrants further investigation to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Beita Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Wei Lin
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China.
| |
Collapse
|
50
|
Zhou Q, Kerbl-Knapp J, Zhang F, Korbelius M, Kuentzel KB, Vujić N, Akhmetshina A, Hörl G, Paar M, Steyrer E, Kratky D, Madl T. Metabolomic Profiles of Mouse Tissues Reveal an Interplay between Aging and Energy Metabolism. Metabolites 2021; 12:17. [PMID: 35050139 PMCID: PMC8779655 DOI: 10.3390/metabo12010017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/16/2022] Open
Abstract
Energy metabolism, including alterations in energy intake and expenditure, is closely related to aging and longevity. Metabolomics studies have recently unraveled changes in metabolite composition in plasma and tissues during aging and have provided critical information to elucidate the molecular basis of the aging process. However, the metabolic changes in tissues responsible for food intake and lipid storage have remained unexplored. In this study, we aimed to investigate aging-related metabolic alterations in these tissues. To fill this gap, we employed NMR-based metabolomics in several tissues, including different parts of the intestine (duodenum, jejunum, ileum) and brown/white adipose tissues (BAT, WAT), of young (9-10 weeks) and old (96-104 weeks) wild-type (mixed genetic background of 129/J and C57BL/6) mice. We, further, included plasma and skeletal muscle of the same mice to verify previous results. Strikingly, we found that duodenum, jejunum, ileum, and WAT do not metabolically age. In contrast, plasma, skeletal muscle, and BAT show a strong metabolic aging phenotype. Overall, we provide first insights into the metabolic changes of tissues essential for nutrient uptake and lipid storage and have identified biomarkers for metabolites that could be further explored, to study the molecular mechanisms of aging.
Collapse
Affiliation(s)
- Qishun Zhou
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
| | - Jakob Kerbl-Knapp
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
| | - Fangrong Zhang
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350001, China
| | - Melanie Korbelius
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
| | - Katharina Barbara Kuentzel
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
| | - Nemanja Vujić
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
| | - Alena Akhmetshina
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
| | - Gerd Hörl
- Otto-Loewi Research Center, Physiological Chemistry, Medical University of Graz, 8010 Graz, Austria; (G.H.); (M.P.)
| | - Margret Paar
- Otto-Loewi Research Center, Physiological Chemistry, Medical University of Graz, 8010 Graz, Austria; (G.H.); (M.P.)
| | - Ernst Steyrer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
| | - Dagmar Kratky
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
- BioTechMed-Graz, 8010 Graz, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (Q.Z.); (J.K.-K.); (F.Z.); (M.K.); (K.B.K.); (N.V.); (A.A.); (E.S.); (D.K.)
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|