1
|
Jiang J, Liu F, Cui D, Xu C, Chi J, Yan T, Guo F. Novel molecular mechanisms of immune evasion in hepatocellular carcinoma: NSUN2-mediated increase of SOAT2 RNA methylation. Cancer Commun (Lond) 2025. [PMID: 40227950 DOI: 10.1002/cac2.70023] [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: 03/01/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a deadly malignancy known for its ability to evade immune surveillance. NOP2/Sun RNA methyltransferase family member 2 (NSUN2), an RNA methyltransferase involved in carcinogenesis, has been associated with immune evasion and energy metabolism reprogramming. This study aimed to examine the molecular mechanisms underlying the involvement of NSUN2 in immune evasion and metabolic reprogramming of HCC. METHODS Single-cell transcriptomic sequencing was applied to examine cellular composition changes, particularly immune cell dynamics, in HCC and adjacent normal tissues. Bulk RNA-seq and proteomics identified key genes and proteins. Methylation sequencing and methylated RNA immunoprecipitation (MeRIP) were carried out to characterize the role of NSUN2 in 5-methylcytosine (m5C) modification of sterol O-acyltransferase 2 (SOAT2). Clinical samples from 30 HCC patients were analyzed using reverse transcription-quantitative polymerase chain reaction and Western blotting. Gene expression was manipulated using CRISPR/Cas9 and lentiviral vectors. In vitro co-culture models and metabolomics were used to study HCC cell-T cell interactions, energy metabolism, and immune evasion. Tumor growth in an orthotopic mouse model was monitored by bioluminescence imaging, with subsequent measurements of tumor weight, volume, and immunohistochemical staining. RESULTS Single-cell transcriptomic analysis identified a marked increase in malignant cells in HCC tissues. Cell communication analysis indicated that tumor cells might promote cancer progression by evading immune clearance. Multi-omics analyses identified NSUN2 as a key regulator in HCC development. MeRIP confirmed that NSUN2 facilitated the m5C modification of SOAT2. Analysis of human HCC tissue samples demonstrated pronounced upregulation of NSUN2 and SOAT2, along with elevated m5C levels in HCC tissues. In vitro experiments uncovered that NSUN2 augmented the reprogramming of energy metabolism and repressed the activity and cytotoxicity of CD8+ T cells, contributing to immune evasion. In vivo studies further substantiated the role of NSUN2 in fostering immune evasion and tumor formation of HCC by modulating the m5C modification of SOAT2. CONCLUSIONS The findings highlight the critical role of NSUN2 in driving HCC progression through the regulation of m5C modification on SOAT2. These findings present potential molecular markers for HCC diagnosis and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Feng Liu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Caixia Xu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Jiachang Chi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Tinghua Yan
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Fang Guo
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
2
|
Neth BJ, Huynh K, Giles C, Wang T, Mellett NA, Duong T, Blach C, Schimmel L, Register TC, Blennow K, Zetterberg H, Batra R, Schweickart A, Dilmore AH, Martino C, Arnold M, Krumsiek J, Han X, Dorrestein PC, Knight R, Meikle PJ, Craft S, Kaddurah-Daouk R. Consuming a modified Mediterranean ketogenic diet reverses the peripheral lipid signature of Alzheimer's disease in humans. COMMUNICATIONS MEDICINE 2025; 5:11. [PMID: 39779882 PMCID: PMC11711287 DOI: 10.1038/s43856-024-00682-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/15/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a major neurodegenerative disorder with significant environmental factors, including diet and lifestyle, influencing its onset and progression. Although previous studies have suggested that certain diets may reduce the incidence of AD, the underlying mechanisms remain unclear. METHOD In this post-hoc analysis of a randomized crossover study of 20 elderly adults, we investigated the effects of a modified Mediterranean ketogenic diet (MMKD) on the plasma lipidome in the context of AD biomarkers, analyzing 784 lipid species across 47 classes using a targeted lipidomics platform. RESULTS Here we identified substantial changes in response to MMKD intervention, aside from metabolic changes associated with a ketogenic diet, we identified a a global elevation across all plasmanyl and plasmenyl ether lipid species, with many changes linked to clinical and biochemical markers of AD. We further validated our findings by leveraging our prior clinical studies into lipid related changeswith AD (n = 1912), and found that the lipidomic signature with MMKD was inversely associated with the lipidomic signature of prevalent and incident AD. CONCLUSIONS Intervention with a MMKD was able to alter the plasma lipidome in ways that contrast with AD-associated patterns. Given its low risk and cost, MMKD could be a promising approach for prevention or early symptomatic treatment of AD.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Tingting Wang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
| | - Natalie A Mellett
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
| | - Thy Duong
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
| | - Colette Blach
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Leyla Schimmel
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Thomas C Register
- Department of Pathology - Comparative Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Richa Batra
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Annalise Schweickart
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Cameron Martino
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rob Knight
- Departments of Pediatrics, Computer Science and Engineering, Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Suzanne Craft
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
3
|
González-Ramírez EJ, García-Arribas AB, Artetxe I, Shaw WA, Goñi FM, Alonso A, Jiménez-Rojo N. (1-Deoxy)ceramides in bilayers containing sphingomyelin and cholesterol. Colloids Surf B Biointerfaces 2024; 243:114155. [PMID: 39137529 DOI: 10.1016/j.colsurfb.2024.114155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
The discovery of a novel sphingolipid subclass, the (1-deoxy)sphingolipids, which lack the 1-hydroxy group, attracted considerable attention in the last decade, mainly due to their involvement in disease. They differed in their physico-chemical properties from the canonical (or 1-hydroxy) sphingolipids and they were more toxic when accumulated in cells, inducing neurodegeneration and other dysfunctions. (1-Deoxy)ceramides, (1-deoxy)dihydroceramides, and (1- deoxymethyl)dihydroceramides, the latter two containing a saturated sphingoid chain, have been studied in this work using differential scanning calorimetry, confocal fluorescence and atomic force microscopy, to evaluate their behavior in bilayers composed of mixtures of three or four lipids. When compared to canonical ceramides (Cer), a C16:0 (1-deoxy)Cer shows a lower miscibility in mixtures of the kind C16:0 sphingomyelin/cholesterol/XCer, where XCer is any (1-deoxy)ceramide, giving rise to the coexistence of a liquid-ordered phase and a gel phase. The latter resembles, in terms of thermotropic behavior and nanomechanical resistance, the gel phase of the C16:0 sphingomyelin/cholesterol/C16:0 Cer mixture [Busto et al., Biophys. J. 2014, 106, 621-630]. Differences are seen between the various C16:0 XCer under study in terms of nanomechanical resistance, bilayer thickness and bilayer topography. When examined in a more fluid environment (bilayers based on C24:1 SM), segregated gel phases are still present. Probably related to such lateral separation, XCer preserve the capacity for membrane permeation, but their effects are significantly lower than those of canonical ceramides. Moreover, C24:1 XCer show significantly lower membrane permeation capacity than their C16:0 counterparts. The above data may be relevant in the pathogenesis of certain sphingolipid-related diseases, including certain neuropathies, diabetes, and glycogen storage diseases.
Collapse
Affiliation(s)
- E J González-Ramírez
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain
| | - A B García-Arribas
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain
| | - I Artetxe
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain
| | - W A Shaw
- Avanti Polar Lipids, Alabaster, AL, USA
| | - F M Goñi
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain
| | - A Alonso
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain.
| | - N Jiménez-Rojo
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain.
| |
Collapse
|
4
|
Xia J, Yin S, Yu J, Wang J, Jin X, Wang Y, Liu H, Sun G. Improvement in Glycolipid Metabolism Parameters After Supplementing Fish Oil-Derived Omega-3 Fatty Acids Is Associated with Gut Microbiota and Lipid Metabolites in Type 2 Diabetes Mellitus. Nutrients 2024; 16:3755. [PMID: 39519588 PMCID: PMC11547733 DOI: 10.3390/nu16213755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to investigate the effects of fish oil-derived omega-3 polyunsaturated fatty acids (omega-3 PUFAs) on gut microbiota and serum lipid metabolites in T2DM. METHODS In a three-month, randomized, double-blind, placebo-controlled study, 110 T2DM patients received either fish oil (n = 55) or corn oil (n = 55) capsules daily. Serum lipids, glycemic parameters, gut microbiota diversity, and lipidomics were assessed. RESULTS This study found that fish oil-derived omega-3 PUFAs intervention did not significantly lower the fasting plasma glucose levels when compared with the baseline level (p > 0.05). However, serum fasting blood glucose (p = 0.039), glycosylated hemoglobin levels (p = 0.048), HOMA-IR (p = 0.022), total cholesterol (p < 0.001), triglyceride (p = 0.034), LDL cholesterol (p = 0.048), and non-HDL levels (p = 0.046) were significantly lower in the fish oil group compared with the corn oil group after three months of intervention. Also, it altered glycerophospholipid metabolism and gut microbiota. After three months, the fish oil group showed a significantly lower abundance of Desulfobacterota compared with the corn oil control group (p = 0.003), with reduced levels of Colidextribacter (p = 0.002), Ralstonia (p = 0.021), and Klebsiella (p = 0.013). Conversely, the abundance of Limosilactobacillus (p = 0.017), Lactobacillus (p = 0.011), and Haemophilus (p = 0.018) increased significantly. In addition, relevant glycolipid metabolism indicators showed significant correlations with the altered profiles of serum lipid metabolites, intestinal bacteria, and fungi. CONCLUSIONS This study highlights the impact of fish oil-derived omega-3 PUFAs on intestinal microbiota structure and function in patients with type 2 diabetes. The observed decrease in pathogenic bacterial species and the enhancement of beneficial species may have significant implications for gut health and systemic inflammation, both of which are pivotal in managing diabetes. Further research is warranted to comprehensively elucidate the long-term benefits and underlying mechanisms of these microbiota alterations.
Collapse
Affiliation(s)
- Jiayue Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Shiyu Yin
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Junhui Yu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jiongnan Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xingyi Jin
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yuanyuan Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Hechun Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| |
Collapse
|
5
|
Sinturel F, Chera S, Brulhart-Meynet MC, Montoya JP, Lefai E, Jornayvaz FR, D'Angelo G, Jung MK, Pataky Z, Riezman H, Dibner C. Alterations of lipid homeostasis in morbid obese patients are partly reversed by bariatric surgery. iScience 2024; 27:110820. [PMID: 39297163 PMCID: PMC11409037 DOI: 10.1016/j.isci.2024.110820] [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: 03/15/2024] [Revised: 06/25/2024] [Accepted: 08/22/2024] [Indexed: 09/21/2024] Open
Abstract
Besides its beneficial effect on weight loss, gastric bypass surgery (GBS) may impact the circulating levels of phospho- and sphingolipids. However, long-term effects have not been explored. To investigate alterations in lipidomic signatures associated with massive weight loss following GBS, we conducted direct infusion tandem mass spectrometry on serum and subcutaneous adipose tissue (SAT) samples collected in a longitudinal cohort of morbid obese patients prior to GBS and 1 year following the surgery. A tissue-specific rearrangement of 13% among over 400 phospholipid and sphingolipid species quantified in serum and SAT was observed 1 year following GBS, with a substantial reduction of ceramide levels and increased amount of hexosylceramides detected in both tissues. The comparison of these new lipidomic profiles with the serum and SAT lipidomes established from an independent cohort of lean and morbid obese subjects revealed that GBS partly restored the lipid alterations associated with morbid obesity.
Collapse
Affiliation(s)
- Flore Sinturel
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland
| | - Simona Chera
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marie-Claude Brulhart-Meynet
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Jonathan Paz Montoya
- Proteomics Core Facility, EPFL, 1015 Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Etienne Lefai
- INRA, Unité de Nutrition Humaine, Université Clermont Auvergne, Paris, France
| | - François R Jornayvaz
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Unit of therapeutic patient education, WHO Collaborating Centre, Department of Medicine, University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Giovanni D'Angelo
- Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Minoa Karin Jung
- Division of Visceral Surgery, Department of Surgery, University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Zoltan Pataky
- Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Unit of therapeutic patient education, WHO Collaborating Centre, Department of Medicine, University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Charna Dibner
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland
| |
Collapse
|
6
|
Xie X, Zhang M, Luo H. Regulation of metabolism by circadian rhythms: Support from time-restricted eating, intestinal microbiota & omics analysis. Life Sci 2024; 351:122814. [PMID: 38857654 DOI: 10.1016/j.lfs.2024.122814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/05/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Circadian oscillatory system plays a key role in coordinating the metabolism of most organisms. Perturbation of genetic effects and misalignment of circadian rhythms result in circadian dysfunction and signs of metabolic disorders. The eating-fasting cycle can act on the peripheral circadian clocks, bypassing the photoperiod. Therefore, time-restricted eating (TRE) can improve metabolic health by adjusting eating rhythms, a process achieved through reprogramming of circadian genomes and metabolic programs at different tissue levels or remodeling of the intestinal microbiota, with omics technology allowing visualization of the regulatory processes. Here, we review recent advances in circadian regulation of metabolism, focus on the potential application of TRE for rescuing circadian dysfunction and metabolic disorders with the contribution of intestinal microbiota in between, and summarize the significance of omics technology.
Collapse
Affiliation(s)
- Ximei Xie
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China
| | - Mengjie Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China.
| |
Collapse
|
7
|
Zhan T, Zou Y, Han Z, Tian X, Chen M, Liu J, Yang X, Zhu Q, Liu M, Chen W, Chen M, Huang X, Tan J, Liu W, Tian X. Single-cell sequencing combined with spatial transcriptomics reveals that the IRF7 gene in M1 macrophages inhibits the occurrence of pancreatic cancer by regulating lipid metabolism-related mechanisms. Clin Transl Med 2024; 14:e1799. [PMID: 39118300 PMCID: PMC11310283 DOI: 10.1002/ctm2.1799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
AIM The main focus of this study is to explore the molecular mechanism of IRF7 regulation on RPS18 transcription in M1-type macrophages in pancreatic adenocarcinoma (PAAD) tissue, as well as the transfer of RPS18 by IRF7 via exosomes to PAAD cells and the regulation of ILF3 expression. METHODS By utilising single-cell RNA sequencing (scRNA-seq) data and spatial transcriptomics (ST) data from the Gene Expression Omnibus database, we identified distinct cell types with significant expression differences in PAAD tissue. Among these cell types, we identified those closely associated with lipid metabolism. The differentially expressed genes within these cell types were analysed, and target genes relevant to prognosis were identified. Flow cytometry was employed to assess the expression levels of target genes in M1 and M2 macrophages. Cell lines with target gene knockout were constructed using CRISPR/Cas9 editing technology, and cell lines with target gene knockdown and overexpression were established using lentiviral vectors. Additionally, a co-culture model of exosomes derived from M1 macrophages with PAAD cells was developed. The impact of M1 macrophage-derived exosomes on the lipid metabolism of PAAD cells in the model was evaluated through metabolomics analysis. The effects of M1 macrophage-derived exosomes on the viability, proliferation, division, migration and apoptosis of PAAD cells were assessed using MTT assay, flow cytometry, EdU assay, wound healing assay, Transwell assay and TUNEL staining. Furthermore, a mouse PAAD orthotopic implantation model was established, and bioluminescence imaging was utilised to assess the influence of M1 macrophage-derived exosomes on the intratumoural formation capacity of PAAD cells, as well as measuring tumour weight and volume. The expression of proliferation-associated proteins in tumour tissues was examined using immunohistochemistry. RESULTS Through combined analysis of scRNA-seq and ST technologies, we discovered a close association between M1 macrophages in PAAD samples and lipid metabolism signals, as well as a negative correlation between M1 macrophages and cancer cells. The construction of a prognostic risk score model identified RPS18 and IRF7 as two prognostically relevant genes in M1 macrophages, exhibiting negative and positive correlations, respectively. Mechanistically, it was found that IRF7 in M1 macrophages can inhibit the transcription of RPS18, reducing the transfer of RPS18 to PAAD cells via exosomes, consequently affecting the expression of ILF3 in PAAD cells. IRF7/RPS18 in M1 macrophages can also suppress lipid metabolism, cell viability, proliferation, migration, invasion and intratumoural formation capacity of PAAD cells, while promoting cell apoptosis. CONCLUSION Overexpression of IRF7 in M1 macrophages may inhibit RPS18 transcription, reduce the transfer of RPS18 from M1 macrophage-derived exosomes to PAAD cells, thereby suppressing ILF3 expression in PAAD cells, inhibiting the lipid metabolism pathway, and curtailing the viability, proliferation, migration, invasion of PAAD cells, as well as enhancing cell apoptosis, ultimately inhibiting tumour formation in PAAD cells in vivo. Targeting IRF7/RPS18 in M1 macrophages could represent a promising immunotherapeutic approach for PAAD in the future.
Collapse
Affiliation(s)
- Ting Zhan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Yanli Zou
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Zheng Han
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - XiaoRong Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Mengge Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jiaxi Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiulin Yang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qingxi Zhu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Meng Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Wei Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Mingtao Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xiaodong Huang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jie Tan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Weijie Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xia Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| |
Collapse
|
8
|
Barranco-Altirriba M, Alonso N, Weber RJM, Lloyd GR, Hernandez M, Yanes O, Capellades J, Jankevics A, Winder C, Falguera M, Franch-Nadal J, Dunn WB, Perera-Lluna A, Castelblanco E, Mauricio D. Lipidome characterisation and sex-specific differences in type 1 and type 2 diabetes mellitus. Cardiovasc Diabetol 2024; 23:109. [PMID: 38553758 PMCID: PMC10981308 DOI: 10.1186/s12933-024-02202-5] [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] [Received: 12/07/2023] [Accepted: 03/14/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND In this study, we evaluated the lipidome alterations caused by type 1 diabetes (T1D) and type 2 diabetes (T2D), by determining lipids significantly associated with diabetes overall and in both sexes, and lipids associated with the glycaemic state. METHODS An untargeted lipidomic analysis was performed to measure the lipid profiles of 360 subjects (91 T1D, 91 T2D, 74 with prediabetes and 104 controls (CT)) without cardiovascular and/or chronic kidney disease. Ultra-high performance liquid chromatography-electrospray ionization mass spectrometry (UHPLC-ESI-MS) was conducted in two ion modes (positive and negative). We used multiple linear regression models to (1) assess the association between each lipid feature and each condition, (2) determine sex-specific differences related to diabetes, and (3) identify lipids associated with the glycaemic state by considering the prediabetes stage. The models were adjusted by sex, age, hypertension, dyslipidaemia, body mass index, glucose, smoking, systolic blood pressure, triglycerides, HDL cholesterol, LDL cholesterol, alternate Mediterranean diet score (aMED) and estimated glomerular filtration rate (eGFR); diabetes duration and glycated haemoglobin (HbA1c) were also included in the comparison between T1D and T2D. RESULTS A total of 54 unique lipid subspecies from 15 unique lipid classes were annotated. Lysophosphatidylcholines (LPC) and ceramides (Cer) showed opposite effects in subjects with T1D and subjects with T2D, LPCs being mainly up-regulated in T1D and down-regulated in T2D, and Cer being up-regulated in T2D and down-regulated in T1D. Also, Phosphatidylcholines were clearly down-regulated in subjects with T1D. Regarding sex-specific differences, ceramides and phosphatidylcholines exhibited important diabetes-associated differences due to sex. Concerning the glycaemic state, we found a gradual increase of a panel of 1-deoxyceramides from normoglycemia to prediabetes to T2D. CONCLUSIONS Our findings revealed an extensive disruption of lipid metabolism in both T1D and T2D. Additionally, we found sex-specific lipidome changes associated with diabetes, and lipids associated with the glycaemic state that can be linked to previously described molecular mechanisms in diabetes.
Collapse
Affiliation(s)
- Maria Barranco-Altirriba
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Departament d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, B2SLab, Barcelona, Spain
- Networking Biomedical Research Centre in the subject area of Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN), Madrid, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Núria Alonso
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain
- Servicio de Endocrinología y Nutrición, Hospital Universitario e Instituto de Investigación en Ciencias de la Salud Germans Trias i Pujol, Badalona, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Ralf J M Weber
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Gavin R Lloyd
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Marta Hernandez
- Department of Endocrinology & Nutrition, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Oscar Yanes
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain
- Department of Electronic Engineering, Universitat Rovira i Virgili, IISPV, Tarragona, Spain
| | - Jordi Capellades
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Andris Jankevics
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Catherine Winder
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L69 7ZB, Liverpool, UK
| | - Mireia Falguera
- Institut d'Investigació Biomèdica, Centre Atenció Primària Cervera, Gerència d'Atenció Primària, Universitat de Lleida, Institut Català de la Salut, Lleida, Spain
| | - Josep Franch-Nadal
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain
- DAP-Cat Group, Unitat de Suport a La Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
| | - Warwick B Dunn
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L69 7ZB, Liverpool, UK
| | - Alexandre Perera-Lluna
- Departament d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, B2SLab, Barcelona, Spain
- Networking Biomedical Research Centre in the subject area of Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN), Madrid, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Esmeralda Castelblanco
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, 63110, St. Louis, MO, USA.
- Unitat de Suport a la Recerca Barcelona, Institut Universitari d'Investigació en Atenció Primària Jordi Gol i Gurina, 08007, Barcelona, Spain.
| | - Didac Mauricio
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain.
- Institut d'Investigació Biomèdica Sant Pau (IR Sant Pau), 08041, Barcelona, Spain.
- Faculty of Medicine, University of Vic, Vic, Spain.
| |
Collapse
|
9
|
Frendi S, Martineau C, Cazier H, Nicolle R, Chassac A, Albuquerque M, Raffenne J, Le Faouder J, Paradis V, Cros J, Couvelard A, Rebours V. Role of the fatty pancreatic infiltration in pancreatic oncogenesis. Sci Rep 2024; 14:6582. [PMID: 38503902 PMCID: PMC10951200 DOI: 10.1038/s41598-024-57294-6] [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: 10/24/2023] [Accepted: 03/16/2024] [Indexed: 03/21/2024] Open
Abstract
Although pancreatic precancerous lesions are known to be related to obesity and fatty pancreatic infiltration, the mechanisms remain unclear. We assessed the role of fatty infiltration in the process of pancreatic oncogenesis and obesity. A combined transcriptomic, lipidomic and pathological approach was used to explore neoplastic transformations. Intralobular (ILF) and extralobular (ELF) lipidomic profiles were analyzed to search for lipids associated with pancreatic intraepithelial neoplasia (PanINs) and obesity; the effect of ILF and ELF on acinar tissue and the histopathological aspects of pancreatic parenchyma changes in obese (OB) and non-obese patients. This study showed that the lipid composition of ILF was different from that of ELF. ILF was related to obesity and ELF-specific lipids were correlated to PanINs. Acinar cells were shown to have different phenotypes depending on the presence and proximity to ILF in OB patients. Several lipid metabolic pathways, oxidative stress and inflammatory pathways were upregulated in acinar tissue during ILF infiltration in OB patients. Early acinar transformations, called acinar nodules (AN) were linked to obesity but not ELF or ILF suggesting that they are the first reversible precancerous pancreatic lesions to occur in OB patients. On the other hand, the number of PanINs was higher in OB patients and was positively correlated to ILF and ELF scores as well as to fibrosis. Our study suggests that two types of fat infiltration must be distinguished, ELF and ILF. ILF plays a major role in acinar modifications and the development of precancerous lesions associated with obesity, while ELF may play a role in the progression of PDAC.
Collapse
Affiliation(s)
- Sonia Frendi
- Inflammation Research Center (CRI), INSERM, U1149, Paris-Cité University, 75018, Paris, France
| | - Chloé Martineau
- Pancreatology and digestive oncology Department - DMU Digest, Beaujon Hospital, AP-HP, Paris-Cité University, 100 Boulevard du Général Leclerc, 92110, Clichy, France
| | - Hélène Cazier
- Inflammation Research Center (CRI), INSERM, U1149, Paris-Cité University, 75018, Paris, France
| | - Rémy Nicolle
- INSERM U1149, CNRS ERL 8252, Inflammation Research Center (CRI), Paris-Cité University, 75018, Paris, France
| | - Anaïs Chassac
- Pathology Department, Bichat Hospital, AP-HP, Paris-Cité University, Paris, France
| | - Miguel Albuquerque
- Inflammation Research Center (CRI), INSERM, U1149, Paris-Cité University, 75018, Paris, France
- Pathology Department, FHU MOSAIC, AP-HP, Beaujon Hospital, Clichy, France
| | | | - Julie Le Faouder
- Inflammation Research Center (CRI), INSERM, U1149, Paris-Cité University, 75018, Paris, France
| | - Valérie Paradis
- Inflammation Research Center (CRI), INSERM, U1149, Paris-Cité University, 75018, Paris, France
- Pathology Department, FHU MOSAIC, AP-HP, Beaujon Hospital, Clichy, France
| | - Jérôme Cros
- Inflammation Research Center (CRI), INSERM, U1149, Paris-Cité University, 75018, Paris, France
- Pathology Department, FHU MOSAIC, AP-HP, Beaujon Hospital, Clichy, France
| | - Anne Couvelard
- Inflammation Research Center (CRI), INSERM, U1149, Paris-Cité University, 75018, Paris, France
- Pathology Department, Bichat Hospital, AP-HP, Paris-Cité University, Paris, France
| | - Vinciane Rebours
- Inflammation Research Center (CRI), INSERM, U1149, Paris-Cité University, 75018, Paris, France.
- Pancreatology and digestive oncology Department - DMU Digest, Beaujon Hospital, AP-HP, Paris-Cité University, 100 Boulevard du Général Leclerc, 92110, Clichy, France.
| |
Collapse
|
10
|
Saprina TV, Bashirova AS, Ivanov VV, Pekov SI, Popov IA, Bashirov SR, Vasilyeva EA, Pavlenko OA, Krinitskii DV, Chen M. Lipidomic markers of obesity and their dynamics after bariatric surgery. BULLETIN OF SIBERIAN MEDICINE 2024; 22:174-187. [DOI: 10.20538/1682-0363-2023-4-174-187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Obesity is considered as a chronic progressive disease, heterogeneous in its etiology and clinical manifestations, and characterized by excess in body fat mass and its deposition in the body. The term “morbid obesity” refers to excessive deposition of adipose tissue with a body mass index (BMI) ≥40 kg / m2 or with a BMI ≥ 35 kg / m2 in the presence of serious complications associated with obesity. Along with obesity, the frequency of type 2 diabetes mellitus and cardiovascular diseases closely associated with it has increased. It results from the progression of metabolic disorders, including insulin resistance, which is inextricably linked with the accumulation of visceral fat and plays a key role in the pathogenesis of obesity-related diseases.The study of lipidomic signatures in obesity and associated conditions is a promising branch of fundamental medicine, which makes it possible to significantly and at a new conceptual level stratify a cohort of obese patients into various phenotypes, including a metabolically healthy and metabolically unhealthy obesity phenotypes. Dynamic changes in the lipidome both in the context of diet, drug treatment, and after various bariatric surgeries are of great interest for developing personalized strategies for the treatment of this disease. Currently available studies and their results suggest that we are only at the very start of studying this promising biomedical field.
Collapse
Affiliation(s)
| | | | | | - S. I. Pekov
- Siberian State Medical University;
Skolkovo Institute of Science and Technology;
Moscow Institute of Physics and Technology
| | - I. A. Popov
- Siberian State Medical University;
Moscow Institute of Physics and Technology
| | | | | | | | | | - M. Chen
- Siberian State Medical University
| |
Collapse
|
11
|
Jiménez-Sánchez C, Sinturel F, Mezza T, Loizides-Mangold U, Montoya JP, Li L, Di Giuseppe G, Quero G, Guessous I, Jornayvaz F, Schrauwen P, Stenvers DJ, Alfieri S, Giaccari A, Berishvili E, Compagnon P, Bosco D, Riezman H, Dibner C, Maechler P. Lysophosphatidylinositols Are Upregulated After Human β-Cell Loss and Potentiate Insulin Release. Diabetes 2024; 73:93-107. [PMID: 37862465 DOI: 10.2337/db23-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
In this study, we identified new lipid species associated with the loss of pancreatic β-cells triggering diabetes. We performed lipidomics measurements on serum from prediabetic mice lacking β-cell prohibitin-2 (a model of monogenic diabetes) patients without previous history of diabetes but scheduled for pancreaticoduodenectomy resulting in the acute reduction of their β-cell mass (∼50%), and patients with type 2 diabetes (T2D). We found lysophosphatidylinositols (lysoPIs) were the main circulating lipid species altered in prediabetic mice. The changes were confirmed in the patients with acute reduction of their β-cell mass and in those with T2D. Increased lysoPIs significantly correlated with HbA1c (reflecting glycemic control), fasting glycemia, and disposition index, and did not correlate with insulin resistance or obesity in human patients with T2D. INS-1E β-cells as well as pancreatic islets isolated from nondiabetic mice and human donors exposed to exogenous lysoPIs showed potentiated glucose-stimulated and basal insulin secretion. Finally, addition of exogenous lysoPIs partially rescued impaired glucose-stimulated insulin secretion in islets from mice and humans in the diabetic state. Overall, lysoPIs appear to be lipid species upregulated in the prediabetic stage associated with the loss of β-cells and that support the secretory function of the remaining β-cells. ARTICLE HIGHLIGHTS Circulating lysophosphatidylinositols (lysoPIs) are increased in situations associated with β-cell loss in mice and humans such as (pre-)diabetes, and hemipancreatectomy. Pancreatic islets isolated from nondiabetic mice and human donors, as well as INS-1E β-cells, exposed to exogenous lysoPIs exhibited potentiated glucose-stimulated and basal insulin secretion. Addition of exogenous lysoPIs partially rescued impaired glucose-stimulated insulin secretion in islets from mice and humans in the diabetic state. LysoPIs appear as lipid species being upregulated already in the prediabetic stage associated with the loss of β-cells and supporting the function of the remaining β-cells.
Collapse
Affiliation(s)
- Cecilia Jiménez-Sánchez
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Flore Sinturel
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Teresa Mezza
- Pancreas Unit, Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli, Institute of Hospitalization and Scientific Care (IRCCS), Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ursula Loizides-Mangold
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Jonathan Paz Montoya
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lingzi Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Gianfranco Di Giuseppe
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Giuseppe Quero
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Chirurgia Digestiva, Fondazione Policlinico Universitario Gemelli IRCSS Università Cattolica del Sacro Cuore, Rome, Italy
| | - Idris Guessous
- Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - François Jornayvaz
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Division of Endocrinology, Diabetes, Nutrition and Patient Education, Department of Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands
| | - Sergio Alfieri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Chirurgia Digestiva, Fondazione Policlinico Universitario Gemelli IRCSS Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Giaccari
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Ekaterine Berishvili
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Compagnon
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Domenico Bosco
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, National Centre of Competence in Research Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| |
Collapse
|
12
|
Sinturel F, Chera S, Brulhart-Meynet MC, Montoya JP, Stenvers DJ, Bisschop PH, Kalsbeek A, Guessous I, Jornayvaz FR, Philippe J, Brown SA, D'Angelo G, Riezman H, Dibner C. Circadian organization of lipid landscape is perturbed in type 2 diabetic patients. Cell Rep Med 2023; 4:101299. [PMID: 38016481 PMCID: PMC10772323 DOI: 10.1016/j.xcrm.2023.101299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/26/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
Lipid homeostasis in humans follows a diurnal pattern in muscle and pancreatic islets, altered upon metabolic dysregulation. We employ tandem and liquid-chromatography mass spectrometry to investigate daily regulation of lipid metabolism in subcutaneous white adipose tissue (SAT) and serum of type 2 diabetic (T2D) and non-diabetic (ND) human volunteers (n = 12). Around 8% of ≈440 lipid metabolites exhibit diurnal rhythmicity in serum and SAT from ND and T2D subjects. The spectrum of rhythmic lipids differs between ND and T2D individuals, with the most substantial changes observed early morning, as confirmed by lipidomics in an independent cohort of ND and T2D subjects (n = 32) conducted at a single morning time point. Strikingly, metabolites identified as daily rhythmic in both serum and SAT from T2D subjects exhibit phase differences. Our study reveals massive temporal and tissue-specific alterations of human lipid homeostasis in T2D, providing essential clues for the development of lipid biomarkers in a temporal manner.
Collapse
Affiliation(s)
- Flore Sinturel
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland
| | - Simona Chera
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland; Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Marie-Claude Brulhart-Meynet
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Jonathan Paz Montoya
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands; Laboratory for Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Arts and Sciences (KNAW), Amsterdam, 1105 BA, the Netherlands
| | - Idris Guessous
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - François R Jornayvaz
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Department of Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Jacques Philippe
- Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Department of Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Giovanni D'Angelo
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Charna Dibner
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland.
| |
Collapse
|
13
|
Harayama T. Metabolic bias: Lipid structures as determinants of their metabolic fates. Biochimie 2023; 215:34-41. [PMID: 37769936 DOI: 10.1016/j.biochi.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/06/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
Cellular lipids have an enormous diversity in their chemical structures, which affect the physicochemical properties of lipids and membranes, as well as their regulatory roles on protein functions. Here, I review additional roles of lipid structures. Multiple studies show that structural differences affect how lipids, even from the same class, are metabolically converted via distinct pathways. I propose the name "structure-guided metabolic bias" for this phenomenon, and discuss its biological relevance. This metabolic bias seems implicated in the buildup of basic cellular lipid compositions, as well as genetic predisposition to diseases. Thus, guiding metabolic biases is an important function of lipid structures, while having the characteristic of being difficult to study by in vitro biochemical reconstitutions.
Collapse
Affiliation(s)
- Takeshi Harayama
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de La Recherche Scientifique and Université Côte D'Azur, 660 Route des Lucioles, 06560, Valbonne, France.
| |
Collapse
|
14
|
Mietus-Snyder M, Perak AM, Cheng S, Hayman LL, Haynes N, Meikle PJ, Shah SH, Suglia SF. Next Generation, Modifiable Cardiometabolic Biomarkers: Mitochondrial Adaptation and Metabolic Resilience: A Scientific Statement From the American Heart Association. Circulation 2023; 148:1827-1845. [PMID: 37902008 DOI: 10.1161/cir.0000000000001185] [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] [Indexed: 10/31/2023]
Abstract
Cardiometabolic risk is increasing in prevalence across the life span with disproportionate ramifications for youth at socioeconomic disadvantage. Established risk factors and associated disease progression are harder to reverse as they become entrenched over time; if current trends are unchecked, the consequences for individual and societal wellness will become untenable. Interrelated root causes of ectopic adiposity and insulin resistance are understood but identified late in the trajectory of systemic metabolic dysregulation when traditional cardiometabolic risk factors cross current diagnostic thresholds of disease. Thus, children at cardiometabolic risk are often exposed to suboptimal metabolism over years before they present with clinical symptoms, at which point life-long reliance on pharmacotherapy may only mitigate but not reverse the risk. Leading-edge indicators are needed to detect the earliest departure from healthy metabolism, so that targeted, primordial, and primary prevention of cardiometabolic risk is possible. Better understanding of biomarkers that reflect the earliest transitions to dysmetabolism, beginning in utero, ideally biomarkers that are also mechanistic/causal and modifiable, is critically needed. This scientific statement explores emerging biomarkers of cardiometabolic risk across rapidly evolving and interrelated "omic" fields of research (the epigenome, microbiome, metabolome, lipidome, and inflammasome). Connections in each domain to mitochondrial function are identified that may mediate the favorable responses of each of the omic biomarkers featured to a heart-healthy lifestyle, notably to nutritional interventions. Fuller implementation of evidence-based nutrition must address environmental and socioeconomic disparities that can either facilitate or impede response to therapy.
Collapse
|
15
|
Beyene HB, Giles C, Huynh K, Wang T, Cinel M, Mellett NA, Olshansky G, Meikle TG, Watts GF, Hung J, Hui J, Cadby G, Beilby J, Blangero J, Moses EK, Shaw JE, Magliano DJ, Meikle PJ. Metabolic phenotyping of BMI to characterize cardiometabolic risk: evidence from large population-based cohorts. Nat Commun 2023; 14:6280. [PMID: 37805498 PMCID: PMC10560260 DOI: 10.1038/s41467-023-41963-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/26/2023] [Indexed: 10/09/2023] Open
Abstract
Obesity is a risk factor for type 2 diabetes and cardiovascular disease. However, a substantial proportion of patients with these conditions have a seemingly normal body mass index (BMI). Conversely, not all obese individuals present with metabolic disorders giving rise to the concept of "metabolically healthy obese". We use lipidomic-based models for BMI to calculate a metabolic BMI score (mBMI) as a measure of metabolic dysregulation associated with obesity. Using the difference between mBMI and BMI (mBMIΔ), we identify individuals with a similar BMI but differing in their metabolic health and disease risk profiles. Exercise and diet associate with mBMIΔ suggesting the ability to modify mBMI with lifestyle intervention. Our findings show that, the mBMI score captures information on metabolic dysregulation that is independent of the measured BMI and so provides an opportunity to assess metabolic health to identify "at risk" individuals for targeted intervention and monitoring.
Collapse
Affiliation(s)
- Habtamu B Beyene
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Tingting Wang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Michelle Cinel
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | | | - Thomas G Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, WA, Australia
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, WA, Australia
| | - Joseph Hung
- School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Jennie Hui
- PathWest Laboratory Medicine of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- School of Population and Global Health, University of Western Australia, Crawley, WA, Australia
| | - Gemma Cadby
- School of Population and Global Health, University of Western Australia, Crawley, WA, Australia
| | - John Beilby
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - John Blangero
- South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric K Moses
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Jonathan E Shaw
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia.
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia.
| |
Collapse
|
16
|
Ding L, Yang J, Guo H, Cong P, Xu J, Xue C, Mao X, Zhang T, Wang Y. Dietary Eicosapentaenoic Acid Containing Phosphoethanolamine Plasmalogens Remodels the Lipidome of White Adipose Tissue and Suppresses High-Fat Diet Induced Obesity in Mice. Mol Nutr Food Res 2023; 67:e2200321. [PMID: 37439463 DOI: 10.1002/mnfr.202200321] [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: 05/16/2022] [Revised: 04/26/2023] [Indexed: 07/14/2023]
Abstract
SCOPE Dietary supplementation of docosahexaenoic acid (DHA)/eicosapentaenoic acid (EPA) can alter the lipidome profiles of adipocytes, thereby counteract obesity. DHA/EPA in the form of phospholipids demonstrates higher bioavailability than triglyceride or ethyl ester (EE), but their effects on the lipidome and metabolic changes during obesity are still unknown. METHODS AND RESULTS High-fat diet-induced obese mice are treated with different molecular forms of EPA, and EPA supplemented as phosphoethanolamine plasmalogens (PlsEtn) has a superior effect on reducing fat mass accumulation than phosphatidylcholine (PC) or EE. The lipidomics analysis indicates that EPA in form of PlsEtn but not PC or EE significantly decreases total PC and sphingomyelin content in white adipose tissue (WAT). Some specific polyunsaturated fatty acid -containing PCs and ether phospholipids are increased in EPA-PlsEtn-fed mice, which may attribute to the upregulation of unsaturated fatty acid biosynthesis and fatty acid elongation reactions in WAT. In addition, the expression of genes related to fatty acid catabolism is also promoted by EPA-PlsEtn supplementation, which may cause the decreased content of saturated and monounsaturated fatty acid-containing PCs. CONCLUSIONS EPA-PlsEtn supplementation is demonstrated to remodel lipidome and regulate the fatty acid metabolic process in WAT, indicating it may serve as a new strategy for obesity treatment in the future.
Collapse
Affiliation(s)
- Lin Ding
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou, 215123, P. R. China
| | - Jinyue Yang
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Haoran Guo
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou, 215123, P. R. China
| | - Peixu Cong
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Xiangzhao Mao
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Tiantian Zhang
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Yuming Wang
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong, 266237, P. R. China
| |
Collapse
|
17
|
Rosarda JD, Giles S, Harkins-Perry S, Mills EA, Friedlander M, Wiseman RL, Eade KT. Imbalanced unfolded protein response signaling contributes to 1-deoxysphingolipid retinal toxicity. Nat Commun 2023; 14:4119. [PMID: 37433773 DOI: 10.1038/s41467-023-39775-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 06/23/2023] [Indexed: 07/13/2023] Open
Abstract
The accumulation of atypical, cytotoxic 1-deoxysphingolipids (1-dSLs) has been linked to retinal diseases such as diabetic retinopathy and Macular Telangiectasia Type 2. However, the molecular mechanisms by which 1-dSLs induce toxicity in retinal cells remain poorly understood. Here, we integrate bulk and single-nucleus RNA-sequencing to define biological pathways that modulate 1-dSL toxicity in human retinal organoids. Our results demonstrate that 1-dSLs differentially activate signaling arms of the unfolded protein response (UPR) in photoreceptor cells and Müller glia. Using a combination of pharmacologic activators and inhibitors, we show that sustained PERK signaling through the integrated stress response (ISR) and deficiencies in signaling through the protective ATF6 arm of the UPR are implicated in 1-dSL-induced photoreceptor toxicity. Further, we demonstrate that pharmacologic activation of ATF6 mitigates 1-dSL toxicity without impacting PERK/ISR signaling. Collectively, our results identify new opportunities to intervene in 1-dSL linked diseases through targeting different arms of the UPR.
Collapse
Affiliation(s)
- Jessica D Rosarda
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Sarah Giles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Lowy Medical Research Institute, La Jolla, CA, 92037, USA
| | - Sarah Harkins-Perry
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Lowy Medical Research Institute, La Jolla, CA, 92037, USA
| | - Elizabeth A Mills
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Lowy Medical Research Institute, La Jolla, CA, 92037, USA
| | - Martin Friedlander
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Lowy Medical Research Institute, La Jolla, CA, 92037, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Kevin T Eade
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Lowy Medical Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
18
|
Petrenko V, Sinturel F, Riezman H, Dibner C. Lipid metabolism around the body clocks. Prog Lipid Res 2023; 91:101235. [PMID: 37187314 DOI: 10.1016/j.plipres.2023.101235] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/06/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
Lipids play important roles in energy metabolism along with diverse aspects of biological membrane structure, signaling and other functions. Perturbations of lipid metabolism are responsible for the development of various pathologies comprising metabolic syndrome, obesity, and type 2 diabetes. Accumulating evidence suggests that circadian oscillators, operative in most cells of our body, coordinate temporal aspects of lipid homeostasis. In this review we summarize current knowledge on the circadian regulation of lipid digestion, absorption, transportation, biosynthesis, catabolism, and storage. Specifically, we focus on the molecular interactions between functional clockwork and biosynthetic pathways of major lipid classes comprising cholesterol, fatty acids, triacylglycerols, glycerophospholipids, glycosphingolipids, and sphingomyelins. A growing body of epidemiological studies associate a socially imposed circadian misalignment common in modern society with growing incidence of metabolic disorders, however the disruption of lipid metabolism rhythms in this connection has only been recently revealed. Here, we highlight recent studies that unravel the mechanistic link between intracellular molecular clocks, lipid homeostasis and development of metabolic diseases based on animal models of clock disruption and on innovative translational studies in humans. We also discuss the perspectives of manipulating circadian oscillators as a potentially powerful approach for preventing and managing metabolic disorders in human patients.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva 1211, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), Geneva 1211, Switzerland
| | - Flore Sinturel
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva 1211, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), Geneva 1211, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, NCCR Chemical Biology, University of Geneva, Geneva 1211, Switzerland
| | - Charna Dibner
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva 1211, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), Geneva 1211, Switzerland.
| |
Collapse
|
19
|
Tiwari A, Rathor P, Trivedi PK, Ch R. Multi-Omics Reveal Interplay between Circadian Dysfunction and Type2 Diabetes. BIOLOGY 2023; 12:301. [PMID: 36829576 PMCID: PMC9953493 DOI: 10.3390/biology12020301] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
Type 2 diabetes is one of the leading threats to human health in the 21st century. It is a metabolic disorder characterized by a dysregulated glucose metabolism resulting from impaired insulin secretion or insulin resistance. More recently, accumulated epidemiological and animal model studies have confirmed that circadian dysfunction caused by shift work, late meal timing, and sleep loss leads to type 2 diabetes. Circadian rhythms, 24-h endogenous biological oscillations, are a fundamental feature of nearly all organisms and control many physiological and cellular functions. In mammals, light synchronizes brain clocks and feeding is a main stimulus that synchronizes the peripheral clocks in metabolic tissues, such as liver, pancreas, muscles, and adipose tissues. Circadian arrhythmia causes the loss of synchrony of the clocks of these metabolic tissues and leads to an impaired pancreas β-cell metabolism coupled with altered insulin secretion. In addition to these, gut microbes and circadian rhythms are intertwined via metabolic regulation. Omics approaches play a significant role in unraveling how a disrupted circadian metabolism causes type 2 diabetes. In the present review, we emphasize the discoveries of several genes, proteins, and metabolites that contribute to the emergence of type 2 diabetes mellitus (T2D). The implications of these discoveries for comprehending the circadian clock network in T2D may lead to new therapeutic solutions.
Collapse
Affiliation(s)
- Ashutosh Tiwari
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
| | - Priya Rathor
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
| | - Prabodh Kumar Trivedi
- Department of Biotechnology, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
- Academy of Council of Scientific and Industrial Research (ACSIR), Gaziabad 201002, India
| | - Ratnasekhar Ch
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
- Academy of Council of Scientific and Industrial Research (ACSIR), Gaziabad 201002, India
- School of Biological Sciences, Queen’s University Belfast, Belfast BT9 5DL, UK
| |
Collapse
|
20
|
Persson PB. Who may receive the next Acta Physiologica Award of US$ 100 000? Acta Physiol (Oxf) 2023; 237:e13901. [PMID: 36314053 DOI: 10.1111/apha.13901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 01/18/2023]
Affiliation(s)
- Pontus B Persson
- Corporate member of Freie Universität Berlin, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Translational Physiology, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
21
|
Prola A, Pilot-Storck F. Cardiolipin Alterations during Obesity: Exploring Therapeutic Opportunities. BIOLOGY 2022; 11:1638. [PMID: 36358339 PMCID: PMC9687765 DOI: 10.3390/biology11111638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 08/13/2023]
Abstract
Cardiolipin is a specific phospholipid of the mitochondrial inner membrane that participates in many aspects of its organization and function, hence promoting proper mitochondrial ATP production. Here, we review recent data that have investigated alterations of cardiolipin in different tissues in the context of obesity and the related metabolic syndrome. Data relating perturbations of cardiolipin content or composition are accumulating and suggest their involvement in mitochondrial dysfunction in tissues from obese patients. Conversely, cardiolipin modulation is a promising field of investigation in a search for strategies for obesity management. Several ways to restore cardiolipin content, composition or integrity are emerging and may contribute to the improvement of mitochondrial function in tissues facing excessive fat storage. Inversely, reduction of mitochondrial efficiency in a controlled way may increase energy expenditure and help fight against obesity and in this perspective, several options aim at targeting cardiolipin to achieve a mild reduction of mitochondrial coupling. Far from being just a victim of the deleterious consequences of obesity, cardiolipin may ultimately prove to be a possible weapon to fight against obesity in the future.
Collapse
Affiliation(s)
- Alexandre Prola
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Fanny Pilot-Storck
- Team Relaix, INSERM, IMRB, Université Paris-Est Créteil, F-94010 Créteil, France
- EnvA, IMRB, F-94700 Maisons-Alfort, France
| |
Collapse
|
22
|
Circadian rhythm of lipid metabolism. Biochem Soc Trans 2022; 50:1191-1204. [PMID: 35604112 DOI: 10.1042/bst20210508] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Lipids comprise a diverse group of metabolites that are indispensable as energy storage molecules, cellular membrane components and mediators of inter- and intra-cellular signaling processes. Lipid homeostasis plays a crucial role in maintaining metabolic health in mammals including human beings. A growing body of evidence suggests that the circadian clock system ensures temporal orchestration of lipid homeostasis, and that perturbation of such diurnal regulation leads to the development of metabolic disorders comprising obesity and type 2 diabetes. In view of the emerging role of circadian regulation in maintaining lipid homeostasis, in this review, we summarize the current knowledge on lipid metabolic pathways controlled by the mammalian circadian system. Furthermore, we review the emerging connection between the development of human metabolic diseases and changes in lipid metabolites that belong to major classes of lipids. Finally, we highlight the mechanisms underlying circadian organization of lipid metabolic rhythms upon the physiological situation, and the consequences of circadian clock dysfunction for dysregulation of lipid metabolism.
Collapse
|
23
|
Effects of Isocaloric Fructose Restriction on Ceramide Levels in Children with Obesity and Cardiometabolic Risk: Relation to Hepatic De Novo Lipogenesis and Insulin Sensitivity. Nutrients 2022; 14:nu14071432. [PMID: 35406045 PMCID: PMC9002884 DOI: 10.3390/nu14071432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
Sugar intake, particularly fructose, is implicated as a factor contributing to insulin resistance via hepatic de novo lipogenesis (DNL). A nine-day fructose reduction trial, controlling for other dietary factors and weight, in children with obesity and metabolic syndrome, decreased DNL and mitigated cardiometabolic risk (CMR) biomarkers. Ceramides are bioactive sphingolipids whose dysregulated metabolism contribute to lipotoxicity, insulin resistance, and CMR. We evaluated the effect of fructose reduction on ceramides and correlations between changes observed and changes in traditional CMR biomarkers in this cohort. Analyses were completed on data from 43 participants. Mean weight decreased (−0.9 ± 1.1 kg). The majority of total and subspecies ceramide levels also decreased significantly, including dihydroceramides, deoxyceramides and ceramide-1-phoshates. Change in each primary ceramide species correlated negatively with composite insulin sensitivity index (CISI). Change in deoxyceramides positively correlated with change in DNL. These results suggest that ceramides decrease in response to dietary fructose restriction, negatively correlate with insulin sensitivity, and may represent an intermediary link between hepatic DNL, insulin resistance, and CMR.
Collapse
|
24
|
Groeneveld K. Skeletal muscles do more than the loco-motion. Acta Physiol (Oxf) 2022; 234:e13791. [PMID: 35094479 DOI: 10.1111/apha.13791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Kathrin Groeneveld
- ThIMEDOP, Thüringer Innovationszentrum für Medizintechnik Lösungen Universitätsklinikum Jena Jena Germany
| |
Collapse
|
25
|
Lange M, Angelidou G, Ni Z, Criscuolo A, Schiller J, Blüher M, Fedorova M. AdipoAtlas: A reference lipidome for human white adipose tissue. Cell Rep Med 2021; 2:100407. [PMID: 34755127 PMCID: PMC8561168 DOI: 10.1016/j.xcrm.2021.100407] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/29/2021] [Accepted: 08/26/2021] [Indexed: 01/16/2023]
Abstract
Obesity, characterized by expansion and metabolic dysregulation of white adipose tissue (WAT), has reached pandemic proportions and acts as a primer for a wide range of metabolic disorders. Remodeling of WAT lipidome in obesity and associated comorbidities can explain disease etiology and provide valuable diagnostic and prognostic markers. To support understanding of WAT lipidome remodeling at the molecular level, we provide in-depth lipidomics profiling of human subcutaneous and visceral WAT of lean and obese individuals. We generate a human WAT reference lipidome by performing tissue-tailored preanalytical and analytical workflows, which allow accurate identification and semi-absolute quantification of 1,636 and 737 lipid molecular species, respectively. Deep lipidomic profiling allows identification of main lipid (sub)classes undergoing depot-/phenotype-specific remodeling. Previously unanticipated diversity of WAT ceramides is now uncovered. AdipoAtlas reference lipidome serves as a data-rich resource for the development of WAT-specific high-throughput methods and as a scaffold for systems medicine data integration.
Collapse
Affiliation(s)
- Mike Lange
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Georgia Angelidou
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Zhixu Ni
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Angela Criscuolo
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
- Thermo Fisher Scientific, Dreieich, Germany
| | - Jürgen Schiller
- Institute of Medical Physics and Biophysics, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
26
|
Karsai G, Steiner R, Kaech A, Lone MA, von Eckardstein A, Hornemann T. Metabolism of HSAN1- and T2DM-associated 1-deoxy-sphingolipids inhibits the migration of fibroblasts. J Lipid Res 2021; 62:100122. [PMID: 34563520 PMCID: PMC8521209 DOI: 10.1016/j.jlr.2021.100122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 11/03/2022] Open
Abstract
Hereditary sensory neuropathy type 1 (HSAN1) is a rare axonopathy, characterized by a progressive loss of sensation (pain, temperature, and vibration), neuropathic pain, and wound healing defects. HSAN1 is caused by several missense mutations in the serine palmitoyltransferase long-chain base subunit 1 and serine palmitoyltransferase long-chain base subunit 2 of the enzyme serine palmitoyltransferase-the key enzyme for the synthesis of sphingolipids. The mutations change the substrate specificity of serine palmitoyltransferase, which then forms an atypical class of 1-deoxy-sphinglipids (1-deoxySLs). Similarly, patients with type 2 diabetes mellitus also present with elevated 1-deoxySLs and a comparable clinical phenotype. The effect of 1-deoxySLs on neuronal cells was investigated in detail, but their impact on other cell types remains elusive. Here, we investigated the consequences of externally added 1-deoxySLs on the migration of fibroblasts in a scratch assay as a simplified cellular wound-healing model. We showed that 1-deoxy-sphinganine (1-deoxySA) inhibits the migration of NIH-3T3 fibroblasts in a dose- and time-dependent manner. This was not seen for a non-native, L-threo stereoisomer. Supplemented 1-deoxySA was metabolized to 1-deoxy-(dihydro)ceramide and downstream to 1-deoxy-sphingosine. Inhibiting downstream metabolism by blocking N-acylation rescued the migration phenotype. In contrast, adding 1-deoxy-sphingosine had a lesser effect on cell migration but caused the massive formation of intracellular vacuoles. Further experiments showed that the effect on cell migration was primarily mediated by 1-deoxy-dihydroceramides rather than by the free base or 1-deoxyceramides. Based on these findings, we suggest that limiting the N-acylation of 1-deoxySA could be a therapeutic approach to improve cell migration and wound healing in patients with HSAN1 and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Regula Steiner
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zürich, Zürich, Switzerland
| | - Museer A Lone
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | | | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland.
| |
Collapse
|
27
|
Tran D, Myers S, McGowan C, Henstridge D, Eri R, Sonda S, Caruso V. 1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts. Front Endocrinol (Lausanne) 2021; 12:772925. [PMID: 35002962 PMCID: PMC8739520 DOI: 10.3389/fendo.2021.772925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/03/2021] [Indexed: 01/18/2023] Open
Abstract
Metabolic dysfunction, dysregulated differentiation, and atrophy of skeletal muscle occur as part of a cluster of abnormalities associated with the development of Type 2 diabetes mellitus (T2DM). Recent interest has turned to the attention of the role of 1-deoxysphingolipids (1-DSL), atypical class of sphingolipids which are found significantly elevated in patients diagnosed with T2DM but also in the asymptomatic population who later develop T2DM. In vitro studies demonstrated that 1-DSL have cytotoxic properties and compromise the secretion of insulin from pancreatic beta cells. However, the role of 1-DSL on the functionality of skeletal muscle cells in the pathophysiology of T2DM still remains unclear. This study aimed to investigate whether 1-DSL are cytotoxic and disrupt the cellular processes of skeletal muscle precursors (myoblasts) and differentiated cells (myotubes) by performing a battery of in vitro assays including cell viability adenosine triphosphate assay, migration assay, myoblast fusion assay, glucose uptake assay, and immunocytochemistry. Our results demonstrated that 1-DSL significantly reduced the viability of myoblasts in a concentration and time-dependent manner, and induced apoptosis as well as cellular necrosis. Importantly, myoblasts were more sensitive to the cytotoxic effects induced by 1-DSL rather than by saturated fatty acids, such as palmitate, which are critical mediators of skeletal muscle dysfunction in T2DM. Additionally, 1-DSL significantly reduced the migration ability of myoblasts and the differentiation process of myoblasts into myotubes. 1-DSL also triggered autophagy in myoblasts and significantly reduced insulin-stimulated glucose uptake in myotubes. These findings demonstrate that 1-DSL directly compromise the functionality of skeletal muscle cells and suggest that increased levels of 1-DSL observed during the development of T2DM are likely to contribute to the pathophysiology of muscle dysfunction detected in this disease.
Collapse
Affiliation(s)
- Duyen Tran
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Stephen Myers
- School of Health Science, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Courtney McGowan
- School of Health Science, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
- Sport Performance Optimization Research Team, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Darren Henstridge
- School of Health Science, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Rajaraman Eri
- School of Health Science, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Sabrina Sonda
- School of Health Science, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Vanni Caruso
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
- Institute for Research on Pain, Istituto di Formazione e Ricerca in Scienze Algologiche (ISAL) Foundation, Rimini, Italy
- *Correspondence: Vanni Caruso,
| |
Collapse
|