1
|
Fan X, Li C, Luo J, Wang R, Zhang H. Lipid composition and its molecular classes of milk fat globule membranes derived from yak, buffalo, and holstein cow milk were characterized based on UHPLC-MS/MS and untargeted-lipidomics. Lebensm Wiss Technol 2025; 219:117563. [DOI: 10.1016/j.lwt.2025.117563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
|
2
|
Nenadovic A, Kovacevic S, Stankovic A, Popovic T, Debeljak Martacic J, Rankovic S, De Luka SR, Milasin J, Nesovic Ostojic J. Organ-Specific Responses to Chronic High-Fat Diets in Mice: Insights into Phospholipid Fatty Acid Distribution. Nutrients 2025; 17:821. [PMID: 40077693 PMCID: PMC11901986 DOI: 10.3390/nu17050821] [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: 02/05/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: This research aimed to investigate phospholipid fatty acid (PLFA) distribution in the brain, kidneys, and white adipose tissue (WAT) and lipid profiles in response to high-fat diets. Methods: Adult female C57BL/6 mice were fed high-fat diets containing 25% linseed, palm, or sunflower oil for 100 days. The fatty acid composition of dietary oils and tissue PL were analyzed using gas-liquid chromatography. Results: Linseed oil increased n-3 polyunsaturated fatty acids (PUFAs) with subsequent conversion into long-chain n-3 PUFAs in the brain and kidney PL, while only alpha-linolenic acid was elevated in WAT. Palm and sunflower oils resulted in unique PLFA distributions in the kidneys and WAT. Palm oil raised linoleic acid without conversion to pro-inflammatory n-6 PUFAs. Sunflower oil increased saturated palmitic acid, as opposed to the rise in monounsaturated oleic acid. Linseed oil also significantly improved lipid profiles, reducing LDL and increasing HDL levels while enhancing cardiovascular indices. Conclusions: This study demonstrates that dietary oils significantly impact organ-specific PLFA profiles, with linseed oil enriching brain and renal n-3 PUFAs, while palm and sunflower oils induce distinct modifications in the kidney and WAT. Moreover, linseed oil offers notable cardioprotective benefits due to the favorable lipid profile changes. These findings highlight the importance of dietary fat selection in achieving balanced lipid metabolism and suggest that diverse oil combinations may be essential for optimizing health outcomes.
Collapse
Affiliation(s)
- Aleksandra Nenadovic
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (A.N.); (S.K.); (A.S.); (S.R.D.L.)
| | - Sanjin Kovacevic
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (A.N.); (S.K.); (A.S.); (S.R.D.L.)
| | - Anica Stankovic
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (A.N.); (S.K.); (A.S.); (S.R.D.L.)
| | - Tamara Popovic
- Centre of Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (T.P.); (J.D.M.); (S.R.)
| | - Jasmina Debeljak Martacic
- Centre of Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (T.P.); (J.D.M.); (S.R.)
| | - Slavica Rankovic
- Centre of Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (T.P.); (J.D.M.); (S.R.)
| | - Silvio R. De Luka
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (A.N.); (S.K.); (A.S.); (S.R.D.L.)
| | - Jelena Milasin
- Department of Human Genetics, Faculty of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Jelena Nesovic Ostojic
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (A.N.); (S.K.); (A.S.); (S.R.D.L.)
| |
Collapse
|
3
|
Tian H, Qiao H, Han F, Kong X, Zhu S, Xing F, Duan H, Li W, Wang W, Zhang D, Wu Y. Genome-wide DNA methylation analysis of body composition in Chinese monozygotic twins. Eur J Clin Invest 2023; 53:e14055. [PMID: 37392072 DOI: 10.1111/eci.14055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND Little is currently known about epigenetic alterations associated with body composition in obesity. Thus, we aimed to explore epigenetic relationships between genome-wide DNA methylation levels and three common traits of body composition as measured by body fat percentage (BF%), fat mass (FM) and lean body mass (LBM) among Chinese monozygotic twins. METHODS Generalized estimated equation model was used to regress the methylation level of CpG sites on body composition. Inference about Causation Through Examination Of Familial Confounding was used to explore the evidence of a causal relationship. Gene expression analysis was further performed to validate the results of differentially methylated genes. RESULTS We identified 32, 22 and 28 differentially methylated CpG sites (p < 10-5 ) as well as 20, 17 and eight differentially methylated regions (slk-corrected p < 0.05) significantly associated with BF%, FM and LBM which were annotated to 65 genes, showing partially overlapping. Causal inference demonstrated bidirectional causality between DNA methylation and body composition (p < 0.05). Gene expression analysis revealed significant correlations between expression levels of five differentially methylated genes and body composition (p < 0.05). CONCLUSIONS These DNA methylation signatures will contribute to increased knowledge about the epigenetic basis of body composition and provide new strategies for early prevention and treatment of obesity and its related diseases.
Collapse
Affiliation(s)
- Huimin Tian
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Haofei Qiao
- Qingdao Mental Health Centre, Qingdao, China
| | - Fulei Han
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Xiangjie Kong
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Shuai Zhu
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Fangjie Xing
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Haiping Duan
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Weilong Li
- Population Research Unit, Faculty of Social Sciences, University of Helsinki, Helsinki, Finland
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Yili Wu
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Shannon CE, Ní Chathail MB, Mullin SM, Meehan A, McGillicuddy FC, Roche HM. Precision nutrition for targeting pathophysiology of cardiometabolic phenotypes. Rev Endocr Metab Disord 2023; 24:921-936. [PMID: 37402955 PMCID: PMC10492734 DOI: 10.1007/s11154-023-09821-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2023] [Indexed: 07/06/2023]
Abstract
Obesity is a heterogenous disease accompanied by a broad spectrum of cardiometabolic risk profiles. Traditional paradigms for dietary weight management do not address biological heterogeneity between individuals and have catastrophically failed to combat the global pandemic of obesity-related diseases. Nutritional strategies that extend beyond basic weight management to instead target patient-specific pathophysiology are warranted. In this narrative review, we provide an overview of the tissue-level pathophysiological processes that drive patient heterogeneity to shape distinct cardiometabolic phenotypes in obesity. Specifically, we discuss how divergent physiology and postprandial phenotypes can reveal key metabolic defects within adipose, liver, or skeletal muscle, as well as the integrative involvement of the gut microbiome and the innate immune system. Finally, we highlight potential precision nutritional approaches to target these pathways and discuss recent translational evidence concerning the efficacy of such tailored dietary interventions for different obesity phenotypes, to optimise cardiometabolic benefits.
Collapse
Affiliation(s)
- Christopher E Shannon
- Nutrigenomics Research Group, UCD Conway Institute, and Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Republic of Ireland
- School of Medicine, University College Dublin, Dublin, Republic of Ireland
- Division of Diabetes, Department of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Méabh B Ní Chathail
- Nutrigenomics Research Group, UCD Conway Institute, and Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Republic of Ireland
| | - Sinéad M Mullin
- Nutrigenomics Research Group, UCD Conway Institute, and Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Republic of Ireland
| | - Andrew Meehan
- School of Medicine, University College Dublin, Dublin, Republic of Ireland
| | | | - Helen M Roche
- Nutrigenomics Research Group, UCD Conway Institute, and Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Republic of Ireland.
- Institute for Global Food Security, Queen's University Belfast, Belfast, Northern Ireland.
| |
Collapse
|
5
|
Iacucci M, Jeffery L, Acharjee A, Grisan E, Buda A, Nardone OM, Smith SCL, Labarile N, Zardo D, Ungar B, Hunter S, Mao R, Cannatelli R, Shivaji UN, Parigi TL, Reynolds GM, Gkoutos GV, Ghosh S. Computer-Aided Imaging Analysis of Probe-Based Confocal Laser Endomicroscopy With Molecular Labeling and Gene Expression Identifies Markers of Response to Biological Therapy in IBD Patients: The Endo-Omics Study. Inflamm Bowel Dis 2023; 29:1409-1420. [PMID: 36378498 PMCID: PMC10472745 DOI: 10.1093/ibd/izac233] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND We aimed to predict response to biologics in inflammatory bowel disease (IBD) using computerized image analysis of probe confocal laser endomicroscopy (pCLE) in vivo and assess the binding of fluorescent-labeled biologics ex vivo. Additionally, we investigated genes predictive of anti-tumor necrosis factor (TNF) response. METHODS Twenty-nine patients (15 with Crohn's disease [CD], 14 with ulcerative colitis [UC]) underwent colonoscopy with pCLE before and 12 to 14 weeks after starting anti-TNF or anti-integrin α4β7 therapy. Biopsies were taken for fluorescein isothiocyanate-labeled infliximab and vedolizumab staining and gene expression analysis. Computer-aided quantitative image analysis of pCLE was performed. Differentially expressed genes predictive of response were determined and validated in a public cohort. RESULTS In vivo, vessel tortuosity, crypt morphology, and fluorescein leakage predicted response in UC (area under the receiver-operating characteristic curve [AUROC], 0.93; accuracy 85%, positive predictive value [PPV] 89%; negative predictive value [NPV] 75%) and CD (AUROC, 0.79; accuracy 80%; PPV 75%; NPV 83%) patients. Ex vivo, increased binding of labeled biologic at baseline predicted response in UC (UC) (AUROC, 83%; accuracy 77%; PPV 89%; NPV 50%) but not in Crohn's disease (AUROC 58%). A total of 325 differentially expressed genes distinguished responders from nonresponders, 86 of which fell within the most enriched pathways. A panel including ACTN1, CXCL6, LAMA4, EMILIN1, CRIP2, CXCL13, and MAPKAPK2 showed good prediction of anti-TNF response (AUROC >0.7). CONCLUSIONS Higher mucosal binding of the drug target is associated with response to therapy in UC. In vivo, mucosal and microvascular changes detected by pCLE are associated with response to biologics in inflammatory bowel disease. Anti-TNF-responsive UC patients have a less inflamed and fibrotic state pretreatment. Chemotactic pathways involving CXCL6 or CXCL13 may be novel targets for therapy in nonresponders.
Collapse
Affiliation(s)
- Marietta Iacucci
- National Institute for Health Research Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK
- Gastroenterology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Louisa Jeffery
- Gastroenterology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- National Institute for Health Research Surgical Reconstruction, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Enrico Grisan
- Department of Information Engineering, University of Padova, Padova, Italy
- School of Engineering Computer Science and Informatics, London South Bank University, London, UK
| | - Andrea Buda
- Gastroenterology Unit, Department of Gastrointestinal Oncological Surgery, S. Maria del Prato Hospital, Feltre, Italy
| | - Olga M Nardone
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Samuel C L Smith
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Nunzia Labarile
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Davide Zardo
- Gastroenterology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Bella Ungar
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Stuart Hunter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rosanna Cannatelli
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Uday N Shivaji
- Gastroenterology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | - Gary M Reynolds
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Georgios V Gkoutos
- National Institute for Health Research Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Subrata Ghosh
- National Institute for Health Research Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK
- Gastroenterology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
6
|
Bernstock JD, Willis CM, Garcia-Segura ME, Gaude E, Anni D, Lee YJ, Thomas LW, Casey A, Vicario N, Leonardi T, Nicaise AM, Gessler FA, Izzy S, Buffelli MR, Seidlitz J, Srinivasan S, Murphy MP, Ashcroft M, Cambiaghi M, Hallenbeck JM, Peruzzotti-Jametti L. Integrative transcriptomic and metabolic analyses of the mammalian hibernating brain identifies a key role for succinate dehydrogenase in ischemic tolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534718. [PMID: 37205496 PMCID: PMC10187245 DOI: 10.1101/2023.03.29.534718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Ischemic stroke results in a loss of tissue homeostasis and integrity, the underlying pathobiology of which stems primarily from the depletion of cellular energy stores and perturbation of available metabolites 1 . Hibernation in thirteen-lined ground squirrels (TLGS), Ictidomys tridecemlineatus , provides a natural model of ischemic tolerance as these mammals undergo prolonged periods of critically low cerebral blood flow without evidence of central nervous system (CNS) damage 2 . Studying the complex interplay of genes and metabolites that unfolds during hibernation may provide novel insights into key regulators of cellular homeostasis during brain ischemia. Herein, we interrogated the molecular profiles of TLGS brains at different time points within the hibernation cycle via RNA sequencing coupled with untargeted metabolomics. We demonstrate that hibernation in TLGS leads to major changes in the expression of genes involved in oxidative phosphorylation and this is correlated with an accumulation of the tricarboxylic acid (TCA) cycle intermediates citrate, cis-aconitate, and α-ketoglutarate-αKG. Integration of the gene expression and metabolomics datasets led to the identification of succinate dehydrogenase (SDH) as the critical enzyme during hibernation, uncovering a break in the TCA cycle at that level. Accordingly, the SDH inhibitor dimethyl malonate (DMM) was able to rescue the effects of hypoxia on human neuronal cells in vitro and in mice subjected to permanent ischemic stroke in vivo . Our findings indicate that studying the regulation of the controlled metabolic depression that occurs in hibernating mammals may lead to novel therapeutic approaches capable of increasing ischemic tolerance in the CNS.
Collapse
|
7
|
Garcia-Segura ME, Durainayagam BR, Liggi S, Graça G, Jimenez B, Dehghan A, Tzoulaki I, Karaman I, Elliott P, Griffin JL. Pathway-based integration of multi-omics data reveals lipidomics alterations validated in an Alzheimer's disease mouse model and risk loci carriers. J Neurochem 2023; 164:57-76. [PMID: 36326588 PMCID: PMC10107183 DOI: 10.1111/jnc.15719] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/06/2022]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative disorder. Despite increasing evidence of the importance of metabolic dysregulation in AD, the underlying metabolic changes that may impact amyloid plaque formation are not understood, particularly for late-onset AD. This study analyzed genome-wide association studies (GWAS), transcriptomics, and proteomics data obtained from several data repositories to obtain differentially expressed (DE) multi-omics elements in mouse models of AD. We characterized the metabolic modulation in these data sets using gene ontology, transcription factor, pathway, and cell-type enrichment analyses. A predicted lipid signature was extracted from genome-scale metabolic networks (GSMN) and subsequently validated in a lipidomic data set derived from cortical tissue of ABCA-7 null mice, a mouse model of one of the genes associated with late-onset AD. Moreover, a metabolome-wide association study (MWAS) was performed to further characterize the association between dysregulated lipid metabolism in human blood serum and genes associated with AD risk. We found 203 DE transcripts, 164 DE proteins, and 58 DE GWAS-derived mouse orthologs associated with significantly enriched metabolic biological processes. Lipid and bioenergetic metabolic pathways were significantly over-represented across the AD multi-omics data sets. Microglia and astrocytes were significantly enriched in the lipid-predominant AD-metabolic transcriptome. We also extracted a predicted lipid signature that was validated and robustly modeled class separation in the ABCA7 mice cortical lipidome, with 11 of these lipid species exhibiting statistically significant modulations. MWAS revealed 298 AD single nucleotide polymorphisms-metabolite associations, of which 70% corresponded to lipid classes. These results support the importance of lipid metabolism dysregulation in AD and highlight the suitability of mapping AD multi-omics data into GSMNs to identify metabolic alterations.
Collapse
Affiliation(s)
- Monica Emili Garcia-Segura
- Department of Brain Sciences, Imperial College London, London, UK.,Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Brenan R Durainayagam
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
| | - Sonia Liggi
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Gonçalo Graça
- Section of Bioinformatics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Beatriz Jimenez
- Section of Bioanalytical Chemistry and the National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Abbas Dehghan
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK.,MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Ioanna Tzoulaki
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK.,National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, UK.,Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Ibrahim Karaman
- Section of Bioinformatics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Paul Elliott
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK.,MRC Centre for Environment and Health, Imperial College London, London, UK.,National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, UK
| | - Julian L Griffin
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK.,Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK.,The Rowett Institute, University of Aberdeen, Aberdeen, Scotland
| |
Collapse
|
8
|
Vomhof-DeKrey EE, Singhal S, Singhal SK, Stover AD, Rajpathy O, Preszler E, Garcia L, Basson MD. RNA Sequencing of Intestinal Enterocytes Pre- and Post-Roux-en-Y Gastric Bypass Reveals Alteration in Gene Expression Related to Enterocyte Differentiation, Restitution, and Obesity with Regulation by Schlafen 12. Cells 2022; 11:3283. [PMID: 36291149 PMCID: PMC9601224 DOI: 10.3390/cells11203283] [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: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The intestinal lining renews itself in a programmed fashion that can be affected by adaptation to surgical procedures such as gastric bypass. METHODS To assess adaptive mechanisms in the human intestine after Roux-en-Y gastric bypass (RYGB), we biopsied proximal jejunum at the anastomotic site during surgery to establish a baseline and endoscopically re-biopsied the same area 6-9 months after bypass for comparison. Laser microdissection was performed on pre- and post-RYGB biopsies to isolate enterocytes for RNA sequencing. RESULTS RNA sequencing suggested significant decreases in gene expression associated with G2/M DNA damage checkpoint regulation of the cell cycle pathway, and significant increases in gene expression associated with the CDP-diacylglycerol biosynthesis pathway TCA cycle II pathway, and pyrimidine ribonucleotide salvage pathway after RYGB. Since Schlafen 12 (SLFN12) is reported to influence enterocytic differentiation, we stained mucosa for SLFN12 and observed increased SLFN12 immunoreactivity. We investigated SLFN12 overexpression in HIEC-6 and FHs 74 Int intestinal epithelial cells and observed similar increased expression of the following genes that were also increased after RYGB: HES2, CARD9, SLC19A2, FBXW7, STXBP4, SPARCL1, and UTS. CONCLUSIONS Our data suggest that RYGB promotes SLFN12 protein expression, cellular mechanism and replication pathways, and genes associated with differentiation and restitution (HES2, CARD9, SLC19A2), as well as obesity-related genes (FBXW7, STXBP4, SPARCL1, UTS).
Collapse
Affiliation(s)
- Emilie E. Vomhof-DeKrey
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Sonalika Singhal
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Sandeep K. Singhal
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Allie D. Stover
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Odele Rajpathy
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Elizabeth Preszler
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Luis Garcia
- Sanford Health Clinic, Sioux Falls, ND 57117, USA
| | - Marc D. Basson
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| |
Collapse
|
9
|
Fan M, Choi YJ, Wedamulla NE, Tang Y, Han KI, Hwang JY, Kim EK. Heat-Killed Enterococcus faecalis EF-2001 Attenuate Lipid Accumulation in Diet-Induced Obese (DIO) Mice by Activating AMPK Signaling in Liver. Foods 2022; 11:575. [PMID: 35206052 PMCID: PMC8870772 DOI: 10.3390/foods11040575] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023] Open
Abstract
To explore the inhibitory mechanism of heat-killed Enterococcus faecalis, EF-2001 on hepatic lipid deposition, a diet-induced obese (DIO) animal model was established by high-fat diet (HFD). The DIO C57BL/6 mice were divided into four groups: the normal group without HFD (ND, n = 8), obesity group (HFD, n = 8), experimental group (HFD + EF-2001, 200 mg/kg, n = 8), and positive control group (HFD + Orlistat, 60 mg/kg, n = 8). After 4 weeks, liver and adipose tissue were fixed in 10% paraformaldehyde, followed by embedding in paraffin for tissue sectioning. The differences in body mass, body fat ratio, fatty cell area, and lipid profiling of the liver (TC, LDL, and HDL) were also determined. Moreover, Western blot was performed to analyze the expression of lipid accumulation-related proteins, including AMPK, PPARγ, SREBP-1, ACC, and FAS. Compared with the HFD group, the HFD + EF-2001 group exhibited decreased fat mass, liver index, adipocyte area, TC, and LDL, and an increased level of HDL. The results of liver hematoxylin and eosin (H&E), and oil red O staining showed that the mice in each intervention group were improved on hepatic lipid accumulation, and the mice in the HFD + EF-2001 group were the most similar to those in the normal group when compared with the HFD group. From the Western blot results, we proved that EF-2001 activated the AMPK signaling pathway. EF-2001 significantly upregulated the expressions of p-AMPK and p-ACC and downregulated PPARγ, SREBP-1, and FAS in murine liver. Taken together, these results suggest that EF-2001 decrease lipid accumulation in the DIO model mice through the AMPK pathway and ameliorate liver damage by HFD.
Collapse
Affiliation(s)
- Meiqi Fan
- Division of Food Bioscience, College of Biomedical and Health Sciences, Konkuk University, Chungju 27478, Korea;
| | - Young-Jin Choi
- Department of Food Science and Nutrition, College of Health Science, Dong-A University, Busan 49315, Korea; (Y.-J.C.); (N.E.W.)
- Center for Silver-Targeted Biomaterials, Brain Busan 21 Plus Program, Dong-A University, Busan 49315, Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Korea
| | - Nishala Erandi Wedamulla
- Department of Food Science and Nutrition, College of Health Science, Dong-A University, Busan 49315, Korea; (Y.-J.C.); (N.E.W.)
- Center for Silver-Targeted Biomaterials, Brain Busan 21 Plus Program, Dong-A University, Busan 49315, Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Korea
- Department of Export Agriculture, Faculty of Animal Science and Export Agriculture, Uva Wellassa University, Badulla 90000, Sri Lanka
| | - Yujiao Tang
- School of Bio-Science and Food Engineering, Changchun University of Science and Technology, Changchun 130600, China;
| | | | - Ji-Young Hwang
- Department of Food Science & Technology, Dong-Eui University, Busan 47340, Korea;
| | - Eun-Kyung Kim
- Department of Food Science and Nutrition, College of Health Science, Dong-A University, Busan 49315, Korea; (Y.-J.C.); (N.E.W.)
- Center for Silver-Targeted Biomaterials, Brain Busan 21 Plus Program, Dong-A University, Busan 49315, Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Korea
- Department of Export Agriculture, Faculty of Animal Science and Export Agriculture, Uva Wellassa University, Badulla 90000, Sri Lanka
- Center for Food & Bio Innovation, Dong-A University, Busan 49315, Korea
| |
Collapse
|
10
|
Palavicini JP, Chavez-Velazquez A, Fourcaudot M, Tripathy D, Pan M, Norton L, DeFronzo RA, Shannon CE. The Insulin-Sensitizer Pioglitazone Remodels Adipose Tissue Phospholipids in Humans. Front Physiol 2021; 12:784391. [PMID: 34925073 PMCID: PMC8674727 DOI: 10.3389/fphys.2021.784391] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022] Open
Abstract
The insulin-sensitizer pioglitazone exerts its cardiometabolic benefits in type 2 diabetes (T2D) through a redistribution of body fat, from ectopic and visceral areas to subcutaneous adipose depots. Whereas excessive weight gain and lipid storage in obesity promotes insulin resistance and chronic inflammation, the expansion of subcutaneous adipose by pioglitazone is associated with a reversal of these immunometabolic deficits. The precise events driving this beneficial remodeling of adipose tissue with pioglitazone remain unclear, and whether insulin-sensitizers alter the lipidomic composition of human adipose has not previously been investigated. Using shotgun lipidomics, we explored the molecular lipid responses in subcutaneous adipose tissue following 6months of pioglitazone treatment (45mg/day) in obese humans with T2D. Despite an expected increase in body weight following pioglitazone treatment, no robust effects were observed on the composition of storage lipids (i.e., triglycerides) or the content of lipotoxic lipid species (e.g., ceramides and diacylglycerides) in adipose tissue. Instead, pioglitazone caused a selective remodeling of the glycerophospholipid pool, characterized by a decrease in lipids enriched for arachidonic acid, such as plasmanylethanolamines and phosphatidylinositols. This contributed to a greater overall saturation and shortened chain length of fatty acyl groups within cell membrane lipids, changes that are consistent with the purported induction of adipogenesis by pioglitazone. The mechanism through which pioglitazone lowered adipose tissue arachidonic acid, a major modulator of inflammatory pathways, did not involve alterations in phospholipase gene expression but was associated with a reduction in its precursor linoleic acid, an effect that was also observed in skeletal muscle samples from the same subjects. These findings offer important insights into the biological mechanisms through which pioglitazone protects the immunometabolic health of adipocytes in the face of increased lipid storage.
Collapse
Affiliation(s)
- Juan P. Palavicini
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alberto Chavez-Velazquez
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Marcel Fourcaudot
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Devjit Tripathy
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Meixia Pan
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Luke Norton
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ralph A. DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Christopher E. Shannon
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
11
|
Liśkiewicz AD, Marczak Ł, Bogus K, Liśkiewicz D, Przybyła M, Lewin-Kowalik J. Proteomic and Structural Manifestations of Cardiomyopathy in Rat Models of Obesity and Weight Loss. Front Endocrinol (Lausanne) 2021; 12:568197. [PMID: 33716957 PMCID: PMC7945951 DOI: 10.3389/fendo.2021.568197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity cardiomyopathy increases the risk of heart failure and death. Obesity is curable, leading to the restoration of the heart phenotype, but it is not clear if there are any after-effects of obesity present after weight loss. We characterize the proteomic landscape of obesity cardiomyopathy with an evaluation of whether the cardiac phenotype is still shaped after weight loss. Cardiomyopathy was validated by cardiac hypertrophy, fibrosis, oversized myocytes, and mTOR upregulation in a rat model of cafeteria diet-induced developmental obesity. By global proteomic techniques (LC-MS/MS) a plethora of molecular changes was observed in the heart and circulation of obese animals, suggesting abnormal utilization of metabolic substrates. This was confirmed by increased levels of cardiac ACSL-1, a key enzyme for fatty acid degradation and decreased GLUT-1, a glucose transporter in obese rats. Calorie restriction and weight loss led to the normalization of the heart's size, but fibrosis was still excessive. The proteomic compositions of cardiac tissue and plasma were different after weight loss as compared to control. In addition to morphological consequences, obesity cardiomyopathy involves many proteomic changes. Weight loss provides for a partial repair of the heart's architecture, but the trace of fibrotic deposition and proteomic alterations may occur.
Collapse
Affiliation(s)
- Arkadiusz D. Liśkiewicz
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Łukasz Marczak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Katarzyna Bogus
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Daniela Liśkiewicz
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
- Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marta Przybyła
- Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Joanna Lewin-Kowalik
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
12
|
Acharjee A, Larkman J, Xu Y, Cardoso VR, Gkoutos GV. A random forest based biomarker discovery and power analysis framework for diagnostics research. BMC Med Genomics 2020; 13:178. [PMID: 33228632 PMCID: PMC7685541 DOI: 10.1186/s12920-020-00826-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 11/15/2020] [Indexed: 11/25/2022] Open
Abstract
Background Biomarker identification is one of the major and important goal of functional genomics and translational medicine studies. Large scale –omics data are increasingly being accumulated and can provide vital means for the identification of biomarkers for the early diagnosis of complex disease and/or for advanced patient/diseases stratification. These tasks are clearly interlinked, and it is essential that an unbiased and stable methodology is applied in order to address them. Although, recently, many, primarily machine learning based, biomarker identification approaches have been developed, the exploration of potential associations between biomarker identification and the design of future experiments remains a challenge. Methods In this study, using both simulated and published experimentally derived datasets, we assessed the performance of several state-of-the-art Random Forest (RF) based decision approaches, namely the Boruta method, the permutation based feature selection without correction method, the permutation based feature selection with correction method, and the backward elimination based feature selection method. Moreover, we conducted a power analysis to estimate the number of samples required for potential future studies. Results We present a number of different RF based stable feature selection methods and compare their performances using simulated, as well as published, experimentally derived, datasets. Across all of the scenarios considered, we found the Boruta method to be the most stable methodology, whilst the Permutation (Raw) approach offered the largest number of relevant features, when allowed to stabilise over a number of iterations. Finally, we developed and made available a web interface (https://joelarkman.shinyapps.io/PowerTools/) to streamline power calculations thereby aiding the design of potential future studies within a translational medicine context. Conclusions We developed a RF-based biomarker discovery framework and provide a web interface for our framework, termed PowerTools, that caters the design of appropriate and cost-effective subsequent future omics study.
Collapse
Affiliation(s)
- Animesh Acharjee
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, B15 2TT, UK. .,Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, Birmingham, B15 2TT, UK. .,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospital Birmingham, Birmingham, B15 2WB, UK.
| | - Joseph Larkman
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, B15 2TT, UK.,Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, Birmingham, B15 2TT, UK
| | - Yuanwei Xu
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, B15 2TT, UK.,Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, Birmingham, B15 2TT, UK
| | - Victor Roth Cardoso
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, B15 2TT, UK.,Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, Birmingham, B15 2TT, UK.,MRC Health Data Research UK (HDR UK), London, UK
| | - Georgios V Gkoutos
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, B15 2TT, UK.,Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, Birmingham, B15 2TT, UK.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospital Birmingham, Birmingham, B15 2WB, UK.,MRC Health Data Research UK (HDR UK), London, UK.,NIHR Experimental Cancer Medicine Centre, Birmingham, B15 2TT, UK.,NIHR Biomedical Research Centre, University Hospital Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|