1
|
Gharagozlou S, Wright NM, Murguila-Favela L, Eshleman J, Midgley J, Saygili S, Mathew G, Lesmana H, Makkoukdji N, Gans M, Saba JD. Sphingosine phosphate lyase insufficiency syndrome as a primary immunodeficiency state. Adv Biol Regul 2024; 94:101058. [PMID: 39454238 DOI: 10.1016/j.jbior.2024.101058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Sphingosine phosphate lyase insufficiency syndrome (SPLIS) is a genetic disease associated with renal, endocrine, neurological, skin and immune defects. SPLIS is caused by inactivating mutations in SGPL1, which encodes sphingosine phosphate lyase (SPL). SPL catalyzes the irreversible degradation of the bioactive sphingolipid sphingosine-1-phosphate (S1P), a key regulator of lymphocyte egress. The SPL reaction represents the only exit point of sphingolipid metabolism, and SPL insufficiency causes widespread sphingolipid derangements that could additionally contribute to immunodeficiency. Herein, we review SPLIS, the sphingolipid metabolic pathway, and various roles sphingolipids play in immunity. We then explore SPLIS-related immunodeficiency by analyzing data available in the published literature supplemented by medical record reviews in ten SPLIS children. We found 93% of evaluable SPLIS patients had documented evidence of immunodeficiency. Many of the remainder of cases were unevaluable due to lack of available immunological data. Most commonly, SPLIS patients exhibited lymphopenia and T cell-specific lymphopenia, consistent with the established role of the S1P/S1P1/SPL axis in lymphocyte egress. However, low B and NK cell counts, hypogammaglobulinemia, and opportunistic infections with bacterial, viral and fungal pathogens were observed. Diminished responses to childhood vaccinations were less frequently observed. Screening blood tests quantifying recent thymic emigrants identified some lymphopenic SPLIS patients in the newborn period. Lymphopenia has been reported to improve after cofactor supplementation in some SPLIS patients, indicating upregulation of SPL activity. A variety of treatments including immunoglobulin replacement, prophylactic antimicrobials and special preparation of blood products prior to transfusion have been employed in SPLIS. The diverse immune consequences in SPLIS patients suggest that aberrant S1P signaling may not fully explain the extent of immunodeficiency. Further study will be required to fully elucidate the complex mechanisms underlying SPLIS immunodeficiency and determine the most effective prophylaxis against infection.
Collapse
Affiliation(s)
- Saber Gharagozlou
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| | - NicolaA M Wright
- Department of Pediatrics, Cummings School of Medicine, University of Calgary, Alberta, Canada.
| | - Luis Murguila-Favela
- Department of Pediatrics, Cummings School of Medicine, University of Calgary, Alberta, Canada.
| | - Juliette Eshleman
- Department of Pediatrics, Cummings School of Medicine, University of Calgary, Alberta, Canada.
| | - Julian Midgley
- Department of Pediatrics, Cummings School of Medicine, University of Calgary, Alberta, Canada.
| | - Seha Saygili
- Department of Pediatric Nephrology, Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Istanbul, Turkey.
| | - Georgie Mathew
- Division of Pediatric Nephrology, Christian Medical College, Vellore, India.
| | - Harry Lesmana
- Department of Medical Genetics and Genomics, Department of Pediatric Hematology/Oncology and BMT, Cleveland Clinic, Cleveland, OH, USA.
| | - Nadia Makkoukdji
- Department of Pediatrics, Division of Allergy & Immunology University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, FL, USA.
| | - Melissa Gans
- Department of Pediatrics, Division of Allergy & Immunology University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, FL, USA.
| | - Julie D Saba
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Foran D, Antoniades C, Akoumianakis I. Emerging Roles for Sphingolipids in Cardiometabolic Disease: A Rational Therapeutic Target? Nutrients 2024; 16:3296. [PMID: 39408263 PMCID: PMC11478599 DOI: 10.3390/nu16193296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality. New research elucidates increasingly complex relationships between cardiac and metabolic health, giving rise to new possible therapeutic targets. Sphingolipids are a heterogeneous class of bioactive lipids with critical roles in normal human physiology. They have also been shown to play both protective and deleterious roles in the pathogenesis of cardiovascular disease. Ceramides are implicated in dysregulating insulin signalling, vascular endothelial function, inflammation, oxidative stress, and lipoprotein aggregation, thereby promoting atherosclerosis and vascular disease. Ceramides also advance myocardial disease by enhancing pathological cardiac remodelling and cardiomyocyte death. Glucosylceramides similarly contribute to insulin resistance and vascular inflammation, thus playing a role in atherogenesis and cardiometabolic dysfunction. Sphingosing-1-phosphate, on the other hand, may ameliorate some of the pathological functions of ceramide by protecting endothelial barrier integrity and promoting cell survival. Sphingosine-1-phosphate is, however, implicated in the development of cardiac fibrosis. This review will explore the roles of sphingolipids in vascular, cardiac, and metabolic pathologies and will evaluate the therapeutic potential in targeting sphingolipids with the aim of prevention and reversal of cardiovascular disease in order to improve long-term cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.F.); (C.A.)
| |
Collapse
|
3
|
Huang Y, Sulek K, Stinson SE, Holm LA, Kim M, Trost K, Hooshmand K, Lund MAV, Fonvig CE, Juel HB, Nielsen T, Ängquist L, Rossing P, Thiele M, Krag A, Holm JC, Legido-Quigley C, Hansen T. Lipid profiling identifies modifiable signatures of cardiometabolic risk in children and adolescents with obesity. Nat Med 2024:10.1038/s41591-024-03279-x. [PMID: 39304782 DOI: 10.1038/s41591-024-03279-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024]
Abstract
Pediatric obesity is a progressive, chronic disease that can lead to serious cardiometabolic complications. Here we investigated the peripheral lipidome in 958 children and adolescents with overweight or obesity and 373 with normal weight, in a cross-sectional study. We also implemented a family-based, personalized program to assess the effects of obesity management on 186 children and adolescents in a clinical setting. Using mass spectrometry-based lipidomics, we report an increase in ceramides, alongside a decrease in lysophospholipids and omega-3 fatty acids with obesity metabolism. Ceramides, phosphatidylethanolamines and phosphatidylinositols were associated with insulin resistance and cardiometabolic risk, whereas sphingomyelins showed inverse associations. Additionally, a panel of three lipids predicted hepatic steatosis as effectively as liver enzymes. Lipids partially mediated the association between obesity and cardiometabolic traits. The nonpharmacological management reduced levels of ceramides, phospholipids and triglycerides, indicating that lowering the degree of obesity could partially restore a healthy lipid profile in children and adolescents.
Collapse
Affiliation(s)
- Yun Huang
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Sara E Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Paediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
| | - Min Kim
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Kajetan Trost
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | | | - Morten Asp Vonsild Lund
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Paediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cilius E Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Paediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helene Bæk Juel
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Trine Nielsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Medical Department, Zealand University Hospital, Roskilde, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maja Thiele
- Center for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Aleksander Krag
- Center for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Paediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Cristina Legido-Quigley
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark.
- Institute of Pharmaceutical Science, King's College London, London, United Kingdom.
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Delcheva G, Stefanova K, Stankova T. Ceramides-Emerging Biomarkers of Lipotoxicity in Obesity, Diabetes, Cardiovascular Diseases, and Inflammation. Diseases 2024; 12:195. [PMID: 39329864 PMCID: PMC11443555 DOI: 10.3390/diseases12090195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024] Open
Abstract
Abnormalities in lipid homeostasis have been associated with many human diseases, and the interrelation between lipotoxicity and cellular dysfunction has received significant attention in the past two decades. Ceramides (Cers) are bioactive lipid molecules that serve as precursors of all complex sphingolipids. Besides their function as structural components in cell and mitochondrial membranes, Cers play a significant role as key mediators in cell metabolism and are involved in numerous cellular processes, such as proliferation, differentiation, inflammation, and induction of apoptosis. The accumulation of various ceramides in tissues causes metabolic and cellular disturbances. Recent studies suggest that Cer lipotoxicity has an important role in obesity, metabolic syndrome, type 2 diabetes, atherosclerosis, and cardiovascular diseases (CVDs). In humans, elevated plasma ceramide levels are associated with insulin resistance and impaired cardiovascular and metabolic health. In this review, we summarize the role of ceramides as key mediators of lipotoxicity in obesity, diabetes, cardiovascular diseases, and inflammation and their potential as a promising diagnostic tool.
Collapse
Affiliation(s)
- Ginka Delcheva
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Katya Stefanova
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Teodora Stankova
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| |
Collapse
|
5
|
Brandao CFC, Krempf M, Giolo de Carvalho F, Aguesse A, Junqueira-Franco MVM, Batitucci G, de Freitas EC, Noronha NY, Rodrigues GDS, Junqueira GP, Borba DA, Billon-Crossouard S, Croyal M, Marchini JS. Sphingolipid and Trimethylamine-N-Oxide (TMAO) Levels in Women with Obesity after Combined Physical Training. Metabolites 2024; 14:398. [PMID: 39195494 DOI: 10.3390/metabo14080398] [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: 06/01/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 08/29/2024] Open
Abstract
Obesity causes metabolic changes, such as the development of cardiovascular diseases. Moreover, physical exercise promotes protection against these diseases. Thus, the objective of the present study was to evaluate whether combined physical training can improve the metabolic system of women with obesity, reducing plasma concentrations of trimethylamine N-oxide (TMAO) and sphingolipids, regardless of weight loss. Fourteen obese women (BMI 30-40 kg/m2), aged 20-40 years, sedentary, were submitted to 8 weeks of combined physical training (strength and aerobic exercises). The training was performed three times/week, 55 min/session, at 75-90% maximum heart rate. All participants were evaluated pre- and post-exercise intervention, and their body composition, plasma TMAO, creatinine, lipid profile, and sphingolipid concentrations were recorded. Maximum oxygen consumption (VO2max), Speed lactate threshold 1 (SpeedLT1), and Speed lactate threshold 2 (SpeedLT2) evaluated physical performance. Results: After combined exercise, it did not change body composition, but TMAO, total cholesterol, and sphingolipid concentrations significantly decreased (p < 0.05). There was an increase in physical performance by improving VO2max, SpeedLT1, and SpeedLT2 (p < 0.05). The combined physical exercise could induce cardiovascular risk protection by decreasing TMAO in obese women, parallel to physical performance improvement, independent of weight loss.
Collapse
Affiliation(s)
- Camila Fernanda Cunha Brandao
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
- Department of Physical Education, State University of Minas Gerais, Divinópolis 35500-000, Minas Gerais, Brazil
| | - Michel Krempf
- NUN, INRA, The Research Unit of the Thorax Institute, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, F-44000 Nantes, France
| | - Flávia Giolo de Carvalho
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| | - Audrey Aguesse
- NUN, INRA, The Research Unit of the Thorax Institute, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, F-44000 Nantes, France
| | | | - Gabriela Batitucci
- Department of Food and Nutrition, School of Pharmaceutical Sciences of Araraquara, State University of São Paulo, Rod. Araraquara-Jau Km 1, Araraquara 14800-000, São Paulo, Brazil
| | - Ellen Cristini de Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
- Department of Food and Nutrition, School of Pharmaceutical Sciences of Araraquara, State University of São Paulo, Rod. Araraquara-Jau Km 1, Araraquara 14800-000, São Paulo, Brazil
| | - Natalia Yumi Noronha
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| | - Guilherme da Silva Rodrigues
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| | - Gizela Pedroso Junqueira
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| | - Diego Alcantara Borba
- Department of Physical Education, State University of Minas Gerais, Divinópolis 35500-000, Minas Gerais, Brazil
| | - Stéphanie Billon-Crossouard
- NUN, INRA, The Research Unit of the Thorax Institute, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, F-44000 Nantes, France
| | - Mikael Croyal
- NUN, INRA, The Research Unit of the Thorax Institute, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, F-44000 Nantes, France
| | - Julio Sergio Marchini
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| |
Collapse
|
6
|
Klandorf H, Dartigue V. Oxidative stress and plasma ceramides in broiler chickens. Front Physiol 2024; 15:1411332. [PMID: 39077757 PMCID: PMC11284268 DOI: 10.3389/fphys.2024.1411332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/19/2024] [Indexed: 07/31/2024] Open
Abstract
The selection for rapid growth in chickens has rendered meat-type (broiler) chickens susceptible to develop metabolic syndrome and thus inflammation. The sphingolipid ceramide has been linked as a marker of oxidative stress in mammals, however, the relationship between sphingolipid ceramide supply and oxidative stress in broiler chickens has not been investigated. Therefore, we employed a lipidomic approach to investigate the changes in circulating sphingolipid ceramides in context of allopurinol-induced oxidative stress in birds. Day zero hatched chicks (n = 60) were equally divided into six groups; an unsupplemented control, an allopurinol group (25 mg/kg body weight), a conjugated linoleic acid (CLA) group where half of the oil used in the control diet was substituted for a CLA oil mixture, a CLA and an allopurinol group utilizing the same dose of CLA and allopurinol, a berberine (BRB) group consisting of berberine supplementation (200 mg/kg feed), and a BRB and allopurinol group, utilizing the same dose of BRB and allopurinol. Conjugated linoleic acid and berberine were utilized to potentially enhance antioxidant activity and suppress the oxidative stress induced by allopurinol treatment. Body weight, plasma uric acid, nonesterified fatty acids (NEFA) and sphingolipid ceramides were quantified. Allopurinol induced an inflammatory state as measured by a significant reduction in plasma uric acid - an antioxidant in birds as well as a metabolic waste product. Results showed that both total and saturated sphingolipid ceramides declined (p < 0.05) with age in unsupplemented chicks, although plasma ceramides C16:0 and 18:0 increased in concentration over the study period. Simple total and saturated sphingolipid ceremide's were further decreased (p < 0.05) with allopurinol supplementation, however, this may be an indirect consequence of inducing an inflammatory state. Neither CLA or BRB were able to significantly attenuate the decline. The administration of allopurinol specifically targets the liver which in birds, is the primary organ for fatty acids synthesis. For this reason, sphingolipid ceramide production might have been unwittingly affected by the addition of allopurinol.
Collapse
|
7
|
Wilkerson JL, Tatum SM, Holland WL, Summers SA. Ceramides are fuel gauges on the drive to cardiometabolic disease. Physiol Rev 2024; 104:1061-1119. [PMID: 38300524 PMCID: PMC11381030 DOI: 10.1152/physrev.00008.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/02/2024] Open
Abstract
Ceramides are signals of fatty acid excess that accumulate when a cell's energetic needs have been met and its nutrient storage has reached capacity. As these sphingolipids accrue, they alter the metabolism and survival of cells throughout the body including in the heart, liver, blood vessels, skeletal muscle, brain, and kidney. These ceramide actions elicit the tissue dysfunction that underlies cardiometabolic diseases such as diabetes, coronary artery disease, metabolic-associated steatohepatitis, and heart failure. Here, we review the biosynthesis and degradation pathways that maintain ceramide levels in normal physiology and discuss how the loss of ceramide homeostasis drives cardiometabolic pathologies. We highlight signaling nodes that sense small changes in ceramides and in turn reprogram cellular metabolism and stimulate apoptosis. Finally, we evaluate the emerging therapeutic utility of these unique lipids as biomarkers that forecast disease risk and as targets of ceramide-lowering interventions that ameliorate disease.
Collapse
Affiliation(s)
- Joseph L Wilkerson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
8
|
Norris MK, Tippetts TS, Wilkerson JL, Nicholson RJ, Maschek JA, Levade T, Medin JA, Summers SA, Holland WL. Adiponectin overexpression improves metabolic abnormalities caused by acid ceramidase deficiency but does not prolong lifespan in a mouse model of Farber Disease. Mol Genet Metab Rep 2024; 39:101077. [PMID: 38595987 PMCID: PMC11002753 DOI: 10.1016/j.ymgmr.2024.101077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/23/2024] [Indexed: 04/11/2024] Open
Abstract
Farber Disease is a debilitating and lethal childhood disease of ceramide accumulation caused by acid ceramidase deficiency. The potent induction of a ligand-gated neutral ceramidase activity promoted by adiponectin may provide sufficient lowering of ceramides to allow for the treatment of Farber Disease. In vitro, adiponectin or adiponectin receptor agonist treatments lowered total ceramide concentrations in human fibroblasts from a patient with Farber Disease. However, adiponectin overexpression in a Farber Disease mouse model did not improve lifespan or immune infiltration. Intriguingly, mice heterozygous for the Farber Disease mutation were more prone to glucose intolerance and insulin resistance when fed a high-fat diet, and adiponectin overexpression protected from these metabolic perturbations. These studies suggest that adiponectin evokes a ceramidase activity that is not reliant on the functional expression of acid ceramidase, but indicates that additional strategies are required to ameliorate outcomes of Farber Disease.
Collapse
Affiliation(s)
- Marie K. Norris
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| | - Trevor S. Tippetts
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph L. Wilkerson
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| | - Rebekah J. Nicholson
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| | - J. Alan Maschek
- Metabolomics Core Facility, University of Utah, Salt Lake City, UT, USA
| | - Thierry Levade
- Laboratoire de Biochimie Métabolique, CHU Toulouse and INSERM U1037, Centre de Recherches en Cancérologie de Toulouse, Université Paul Sabatier, 31037 Toulouse, France
| | - Jeffrey A. Medin
- Departments of Pediatrics and Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott A. Summers
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| | - William L. Holland
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
9
|
Nehus EJ, Sheanon NM, Zhang W, Marcovina SM, Setchell KDR, Mitsnefes MM. Urinary sphingolipids in adolescents and young adults with youth-onset diabetes. Pediatr Nephrol 2024; 39:1875-1883. [PMID: 38172468 DOI: 10.1007/s00467-023-06257-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/17/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND This study evaluated urinary sphingolipids as a marker of diabetic kidney disease (DKD) in adolescents and young adults with youth-onset type 1 and type 2 diabetes. METHODS A comprehensive panel of urinary sphingolipids, including sphingomyelin (SM), glucosylceramide (GC), ceramide (Cer), and lactosylceramide (LC) species, was performed in patients with youth-onset diabetes from the SEARCH for Diabetes in Youth cohort. Sphingolipid levels, normalized to urine creatinine, were compared in 57 adolescents and young adults with type 1 diabetes, 59 with type 2 diabetes, and 44 healthy controls. The association of sphingolipids with albumin-to-creatinine (ACR) ratio and estimated glomerular filtration rate (eGFR) was evaluated. RESULTS The median age (interquartile range [IQR]) of participants was 23.1 years (20.9, 24.9) and the median duration of diabetes was 9.3 (8.5, 10.2) years. Urinary sphingolipid concentrations in patients with and without DKD (ACR ≥ 30 mg/g) were significantly elevated compared to healthy controls. There were no significant differences in sphingolipid levels between participants with type 1 and type 2 diabetes. In multivariable analysis, many sphingolipid species were positively correlated with ACR. Most significant associations were evident for the following species: C18 SM, C24:1 SM, C24:1 GC, and C24:1 Cer (all p < 0.001). Sphingolipid levels were not associated with eGFR. However, several interaction terms (diabetes type*sphingolipid) were significant, indicating diabetes type may modify the association of sphingolipids with eGFR. CONCLUSION Urinary sphingolipids are elevated in adolescents and young adults with youth-onset diabetes and correlate with ACR. Urinary sphingolipids may therefore represent an early biomarker of DKD.
Collapse
Affiliation(s)
- Edward J Nehus
- Department of Pediatrics, West Virginia University School of Medicine Charleston Campus, Charleston, WV, USA.
| | - Nicole M Sheanon
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wujuan Zhang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Clinical Mass Spectroscopy Facility, Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Kenneth D R Setchell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Clinical Mass Spectroscopy Facility, Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mark M Mitsnefes
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Nephrology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
10
|
Wajapeyee N, Beamon TC, Gupta R. Roles and therapeutic targeting of ceramide metabolism in cancer. Mol Metab 2024; 83:101936. [PMID: 38599378 PMCID: PMC11031839 DOI: 10.1016/j.molmet.2024.101936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Ceramides are sphingolipids that act as signaling molecules involved in regulating cellular processes including apoptosis, proliferation, and metabolism. Deregulation of ceramide metabolism contributes to cancer development and progression. Therefore, regulation of ceramide levels in cancer cells is being explored as a new approach for cancer therapy. SCOPE OF THE REVIEW This review discusses the multiple roles of ceramides in cancer cells and strategies to modulate ceramide levels for cancer therapy. Ceramides attenuate cell survival signaling and metabolic pathways, while activating apoptotic mechanisms, making them tumor-suppressive. Approaches to increase ceramide levels in cancer cells include using synthetic analogs, inhibiting ceramide degradation, and activating ceramide synthesis. We also highlight combination therapies such as use of ceramide modulators with chemotherapies, immunotherapies, apoptosis inducers, and anti-angiogenics, which offer synergistic antitumor effects. Additionally, we also describe ongoing clinical trials evaluating ceramide nanoliposomes and analogs. Finally, we discuss the challenges of these therapeutic approaches including the complexity of ceramide metabolism, targeted delivery, cancer heterogeneity, resistance mechanisms, and long-term safety. MAJOR CONCLUSIONS Ceramide-based therapy is a potentially promising approach for cancer therapy. However, overcoming hurdles in pharmacokinetics, specificity, and resistance is needed to optimize its efficacy and safety. This requires comprehensive preclinical/clinical studies into ceramide signaling, formulations, and combination therapies. Ceramide modulation offers opportunities for developing novel cancer treatments, but a deeper understanding of ceramide biology is vital to advance its clinical applications.
Collapse
Affiliation(s)
- Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| | - Teresa Chiyanne Beamon
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
11
|
Drinković N, Beus M, Barbir R, Debeljak Ž, Tariba Lovaković B, Kalčec N, Ćurlin M, Bekavac A, Gorup D, Mamić I, Mandić D, Micek V, Turčić P, Günday-Türeli N, Türeli E, Vinković Vrček I. Novel PLGA-based nanoformulation decreases doxorubicin-induced cardiotoxicity. NANOSCALE 2024. [PMID: 38650478 DOI: 10.1039/d3nr06269d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Nanotechnology has the potential to provide formulations of antitumor agents with increased selectivity towards cancer tissue thereby decreasing systemic toxicity. This in vivo study evaluated the potential of novel nanoformulation based on poly(lactic-co-glycolic acid) (PLGA) to reduce the cardiotoxic potential of doxorubicin (DOX). In vivo toxicity of PLGADOX was compared with clinically approved non-PEGylated, liposomal nanoformulation of DOX (LipoDOX) and conventional DOX form (ConvDOX). The study was performed using Wistar Han rats of both sexes that were treated intravenously for 28 days with 5 doses of tested substances at intervals of 5 days. Histopathological analyses of heart tissues showed the presence of myofiber necrosis, degeneration processes, myocytolysis, and hemorrhage after treatment with ConvDOX, whereas only myofiber degeneration and hemorrhage were present after the treatment with nanoformulations. All DOX formulations caused an increase in the troponin T with the greatest increase caused by convDOX. qPCR analyses revealed an increase in the expression of inflammatory markers IL-6 and IL-8 after ConvDOX and an increase in IL-8 expression after lipoDOX treatments. The mass spectra imaging (MSI) of heart tissue indicates numerous metabolic and lipidomic changes caused by ConvDOX, while less severe cardiac damages were found after treatment with nanoformulations. In the case of LipoDOX, autophagy and apoptosis were still detectable, whereas PLGADOX induced only detectable mitochondrial toxicity. Cardiotoxic effects were frequently sex-related with the greater risk of cardiotoxicity observed mostly in male rats.
Collapse
Affiliation(s)
| | - Maja Beus
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Rinea Barbir
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Željko Debeljak
- JJ Strossmayer University of Osijek, Faculty of Medicine, Osijek, Croatia
- University Hospital Osijek, Osijek, Croatia
| | | | - Nikolina Kalčec
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | | | - Ana Bekavac
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Dunja Gorup
- Department of Neuroradiology, Klinik für Neuroradiology, Universitätspital Zürich Universitätsspital Zürich, 8006 Zürich, Switzerland
| | - Ivan Mamić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | | | - Vedran Micek
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Petra Turčić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | | | | | - Ivana Vinković Vrček
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
- University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
12
|
Nakamura M. Lipotoxicity as a therapeutic target in obesity and diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12568. [PMID: 38706718 PMCID: PMC11066298 DOI: 10.3389/jpps.2024.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Unhealthy sources of fats, ultra-processed foods with added sugars, and a sedentary lifestyle make humans more susceptible to developing overweight and obesity. While lipids constitute an integral component of the organism, excessive and abnormal lipid accumulation that exceeds the storage capacity of lipid droplets disrupts the intracellular composition of fatty acids and results in the release of deleterious lipid species, thereby giving rise to a pathological state termed lipotoxicity. This condition induces endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory responses, and cell death. Recent advances in omics technologies and analytical methodologies and clinical research have provided novel insights into the mechanisms of lipotoxicity, including gut dysbiosis, epigenetic and epitranscriptomic modifications, dysfunction of lipid droplets, post-translational modifications, and altered membrane lipid composition. In this review, we discuss the recent knowledge on the mechanisms underlying the development of lipotoxicity and lipotoxic cardiometabolic disease in obesity, with a particular focus on lipotoxic and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, United States
| |
Collapse
|
13
|
Flori L, Piragine E, Calderone V, Testai L. Role of hydrogen sulfide in the regulation of lipid metabolism: Implications on cardiovascular health. Life Sci 2024; 341:122491. [PMID: 38336275 DOI: 10.1016/j.lfs.2024.122491] [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: 12/06/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The World Health Organization (WHO) defines obesity as an urgency for health and a social emergency. Today around 39 % of people is overweight, of these over 13 % is obese. It is well-consolidated that the adipose cells are deputy to lipid storage under caloric excess; however, despite the classical idea that adipose tissue has exclusively a passive function, now it is known to be deeply involved in the regulation of systemic metabolism in physiological as well as under obesogenic conditions, with consequences on cardiovascular health. Beside two traditional types of adipose cells (white and brown), recently the beige one has been highlighted as the consequence of the healthy remodeling of white adipocytes, confirming their metabolic adaptability. In this direction, pharmacological, nutraceutical and nutrient-based approaches are addressed to positively influence inflammation and metabolism, thus contributing to reduce the obese-associated cardiovascular risk. In this scenario, hydrogen sulfide emerges as a new mediator that may regulate crucial targets involved in the regulation of metabolism. The current evidence demonstrates that hydrogen sulfide may induce peroxisome proliferator activated receptor γ (PPARγ), a crucial mediator of adipogenesis, inhibit the phosphorylation of perlipin-1 (plin-1), a protein implicated in the lipolysis, and finally promote browning process, through the release of irisin from skeletal muscle. The results summarized in this review suggest an important role of hydrogen sulfide in the regulation of metabolism and in the prevention/treatment of obese-associated cardiovascular diseases and propose new insight on the putative mechanisms underlying the release of hydrogen sulfide or its biosynthesis, delineating a further exciting field of application.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Eugenia Piragine
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Lara Testai
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| |
Collapse
|
14
|
Al-Rashed F, Arefanian H, Madhoun AA, Bahman F, Sindhu S, AlSaeed H, Jacob T, Thomas R, Al-Roub A, Alzaid F, Malik MDZ, Nizam R, Thanaraj TA, Al-Mulla F, Hannun YA, Ahmad R. Neutral Sphingomyelinase 2 Inhibition Limits Hepatic Steatosis and Inflammation. Cells 2024; 13:463. [PMID: 38474427 PMCID: PMC10931069 DOI: 10.3390/cells13050463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is manifested by hepatic steatosis, insulin resistance, hepatocyte death, and systemic inflammation. Obesity induces steatosis and chronic inflammation in the liver. However, the precise mechanism underlying hepatic steatosis in the setting of obesity remains unclear. Here, we report studies that address this question. After 14 weeks on a high-fat diet (HFD) with high sucrose, C57BL/6 mice revealed a phenotype of liver steatosis. Transcriptional profiling analysis of the liver tissues was performed using RNA sequencing (RNA-seq). Our RNA-seq data revealed 692 differentially expressed genes involved in processes of lipid metabolism, oxidative stress, immune responses, and cell proliferation. Notably, the gene encoding neutral sphingomyelinase, SMPD3, was predominantly upregulated in the liver tissues of the mice displaying a phenotype of steatosis. Moreover, nSMase2 activity was elevated in these tissues of the liver. Pharmacological and genetic inhibition of nSMase2 prevented intracellular lipid accumulation and TNFα-induced inflammation in in-vitro HepG2-steatosis cellular model. Furthermore, nSMase2 inhibition ameliorates oxidative damage by rescuing PPARα and preventing cell death associated with high glucose/oleic acid-induced fat accumulation in HepG2 cells. Collectively, our findings highlight the prominent role of nSMase2 in hepatic steatosis, which could serve as a potential therapeutic target for NAFLD and other hepatic steatosis-linked disorders.
Collapse
Affiliation(s)
- Fatema Al-Rashed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Hossein Arefanian
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Ashraf Al Madhoun
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (S.S.)
| | - Fatemah Bahman
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Sardar Sindhu
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (S.S.)
| | - Halemah AlSaeed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Texy Jacob
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Reeby Thomas
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Areej Al-Roub
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Fawaz Alzaid
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France;
| | - MD Zubbair Malik
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (M.Z.M.); (R.N.); (T.A.T.); (F.A.-M.)
| | - Rasheeba Nizam
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (M.Z.M.); (R.N.); (T.A.T.); (F.A.-M.)
| | - Thangavel Alphonse Thanaraj
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (M.Z.M.); (R.N.); (T.A.T.); (F.A.-M.)
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (M.Z.M.); (R.N.); (T.A.T.); (F.A.-M.)
| | - Yusuf A. Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| |
Collapse
|
15
|
Klemetti MM, Pettersson ABV, Ahmad Khan A, Ermini L, Porter TR, Litvack ML, Alahari S, Zamudio S, Illsley NP, Röst H, Post M, Caniggia I. Lipid profile of circulating placental extracellular vesicles during pregnancy identifies foetal growth restriction risk. J Extracell Vesicles 2024; 13:e12413. [PMID: 38353485 PMCID: PMC10865917 DOI: 10.1002/jev2.12413] [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: 08/15/2023] [Revised: 12/18/2023] [Accepted: 01/13/2024] [Indexed: 02/16/2024] Open
Abstract
Small-for-gestational age (SGA) neonates exhibit increased perinatal morbidity and mortality, and a greater risk of developing chronic diseases in adulthood. Currently, no effective maternal blood-based screening methods for determining SGA risk are available. We used a high-resolution MS/MSALL shotgun lipidomic approach to explore the lipid profiles of small extracellular vesicles (sEV) released from the placenta into the circulation of pregnant individuals. Samples were acquired from 195 normal and 41 SGA pregnancies. Lipid profiles were determined serially across pregnancy. We identified specific lipid signatures of placental sEVs that define the trajectory of a normal pregnancy and their changes occurring in relation to maternal characteristics (parity and ethnicity) and birthweight centile. We constructed a multivariate model demonstrating that specific lipid features of circulating placental sEVs, particularly during early gestation, are highly predictive of SGA infants. Lipidomic-based biomarker development promises to improve the early detection of pregnancies at risk of developing SGA, an unmet clinical need in obstetrics.
Collapse
Affiliation(s)
- Miira M. Klemetti
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoOntarioCanada
- Department of Obstetrics & GynecologyUniversity of TorontoTorontoOntarioCanada
| | - Ante B. V. Pettersson
- Program in Translational Medicine, Peter Gilgan Centre for Research and LearningHospital for Sick ChildrenTorontoOntarioCanada
| | - Aafaque Ahmad Khan
- Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoCanada
| | - Leonardo Ermini
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoOntarioCanada
| | - Tyler R. Porter
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoOntarioCanada
| | - Michael L. Litvack
- Program in Translational Medicine, Peter Gilgan Centre for Research and LearningHospital for Sick ChildrenTorontoOntarioCanada
| | - Sruthi Alahari
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoOntarioCanada
| | | | | | - Hannes Röst
- Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoCanada
| | - Martin Post
- Program in Translational Medicine, Peter Gilgan Centre for Research and LearningHospital for Sick ChildrenTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department PhysiologyUniversity of TorontoTorontoOntarioCanada
| | - Isabella Caniggia
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoOntarioCanada
- Department of Obstetrics & GynecologyUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department PhysiologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
16
|
Park AY, Leney-Greene M, Lynberg M, Gabrielski JQ, Xu X, Schwarz B, Zheng L, Balasubramaniyam A, Ham H, Chao B, Zhang Y, Matthews HF, Cui J, Yao Y, Kubo S, Chanchu JM, Morawski AR, Cook SA, Jiang P, Ravell JC, Cheng YH, George A, Faruqi A, Pagalilauan AM, Bergerson JRE, Ganesan S, Chauvin SD, Aluri J, Edwards-Hicks J, Bohrnsen E, Tippett C, Omar H, Xu L, Butcher GW, Pascall J, Karakoc-Aydiner E, Kiykim A, Maecker H, Tezcan İ, Esenboga S, Heredia RJ, Akata D, Tekin S, Kara A, Kuloglu Z, Unal E, Kendirli T, Dogu F, Karabiber E, Atkinson TP, Cochet C, Filhol O, Bosio CM, Davis MM, Lifton RP, Pearce EL, Daumke O, Aytekin C, Şahin GE, Aksu AÜ, Uzel G, Koneti Rao V, Sari S, Dalgıç B, Boztug K, Cagdas D, Haskologlu S, Ikinciogullari A, Schwefel D, Vilarinho S, Baris S, Ozen A, Su HC, Lenardo MJ. GIMAP5 deficiency reveals a mammalian ceramide-driven longevity assurance pathway. Nat Immunol 2024; 25:282-293. [PMID: 38172257 PMCID: PMC11151279 DOI: 10.1038/s41590-023-01691-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/26/2023] [Indexed: 01/05/2024]
Abstract
Preserving cells in a functional, non-senescent state is a major goal for extending human healthspans. Model organisms reveal that longevity and senescence are genetically controlled, but how genes control longevity in different mammalian tissues is unknown. Here, we report a new human genetic disease that causes cell senescence, liver and immune dysfunction, and early mortality that results from deficiency of GIMAP5, an evolutionarily conserved GTPase selectively expressed in lymphocytes and endothelial cells. We show that GIMAP5 restricts the pathological accumulation of long-chain ceramides (CERs), thereby regulating longevity. GIMAP5 controls CER abundance by interacting with protein kinase CK2 (CK2), attenuating its ability to activate CER synthases. Inhibition of CK2 and CER synthase rescues GIMAP5-deficient T cells by preventing CER overaccumulation and cell deterioration. Thus, GIMAP5 controls longevity assurance pathways crucial for immune function and healthspan in mammals.
Collapse
Affiliation(s)
- Ann Y Park
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael Leney-Greene
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew Lynberg
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Justin Q Gabrielski
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Xijin Xu
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Schwarz
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Lixin Zheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arasu Balasubramaniyam
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Structural Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Hyoungjun Ham
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brittany Chao
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yu Zhang
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Helen F Matthews
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jing Cui
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yikun Yao
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Satoshi Kubo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jean Michel Chanchu
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aaron R Morawski
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah A Cook
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ping Jiang
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juan C Ravell
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Internal Medicine, Hackensack Meridian School of Medicine, Nutley, NJ, USA
| | - Yan H Cheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex George
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aiman Faruqi
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alison M Pagalilauan
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jenna R E Bergerson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sundar Ganesan
- Biological Imaging Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Samuel D Chauvin
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jahnavi Aluri
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joy Edwards-Hicks
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Eric Bohrnsen
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Caroline Tippett
- Section of Digestive Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Habib Omar
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leilei Xu
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Geoffrey W Butcher
- Laboratory of Lymphocyte Signaling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - John Pascall
- Laboratory of Lymphocyte Signaling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - Elif Karakoc-Aydiner
- Division of Pediatric Allergy and Immunology, Marmara University, School of Medicine Pendik, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Marmara University, Pendik, Istanbul, Turkey
| | - Ayca Kiykim
- Division of Pediatric Allergy and Immunology, Marmara University, School of Medicine Pendik, Istanbul, Turkey
| | - Holden Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - İlhan Tezcan
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Saliha Esenboga
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Raul Jimenez Heredia
- St Anna Children's Cancer Research Institute, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Deniz Akata
- Department of Radiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Saban Tekin
- Department of Basic Medical Sciences, Hamidiye Faculty of Medicine, Division of Medical Biology, University of Health Sciences, İstanbul, Turkey
| | - Altan Kara
- TUBITAK Marmara Research Center, Gene Engineering and Biotechnology Institute, Gebze, Turkey
| | - Zarife Kuloglu
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Ankara University School of Medicine, Ankara, Türkiye
| | - Emel Unal
- Department of Pediatric Oncology, Ankara University Medical School, Ankara, Turkey
| | - Tanıl Kendirli
- Department of Pediatric Intensive Care Unit, Ankara University Medical School, Ankara, Turkey
| | - Figen Dogu
- Department of Pediatric Immunology and Allergy, Ankara University Medical School, Ankara, Turkey
| | - Esra Karabiber
- Department of Chest Diseases, Faculty of Medicine, Division of Adult Allergy-Immunology, Marmara University, Istanbul, Turkey
| | - T Prescott Atkinson
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Claude Cochet
- University Grenoble Alpes, INSERM, CEA, UMR Biosanté, Grenoble, France
| | - Odile Filhol
- University Grenoble Alpes, INSERM, CEA, UMR Biosanté, Grenoble, France
| | - Catherine M Bosio
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, Stanford University, Palo Alto, CA, USA
| | - Richard P Lifton
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Oliver Daumke
- Department of Structural Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Caner Aytekin
- Department of Pediatric Immunology, Dr Sami Ulus Maternity and Children's Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Gülseren Evirgen Şahin
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, University of Health Sciences, Dr Sami Ulus Maternity and Children's Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Aysel Ünlüsoy Aksu
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, University of Health Sciences, Dr Sami Ulus Maternity and Children's Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - V Koneti Rao
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sinan Sari
- Department of Pediatric Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Buket Dalgıç
- Department of Pediatric Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Kaan Boztug
- St Anna Children's Cancer Research Institute, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- St Anna Children's Hospital, Vienna, Austria
| | - Deniz Cagdas
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Sule Haskologlu
- Department of Pediatric Immunology and Allergy, Ankara University Medical School, Ankara, Turkey
| | - Aydan Ikinciogullari
- Department of Pediatric Immunology and Allergy, Ankara University Medical School, Ankara, Turkey
| | - David Schwefel
- Department of Structural Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Bioanalytics Unit, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Silvia Vilarinho
- Section of Digestive Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Safa Baris
- Division of Pediatric Allergy and Immunology, Marmara University, School of Medicine Pendik, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Marmara University, Pendik, Istanbul, Turkey
| | - Ahmet Ozen
- Division of Pediatric Allergy and Immunology, Marmara University, School of Medicine Pendik, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Marmara University, Pendik, Istanbul, Turkey
| | - Helen C Su
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
17
|
Gu W, Chai Y, Huang Y, Cai Z, Li R, Chen R, Liu C, Sun Q. Desipramine ameliorates fine particulate matter-induced hepatic insulin resistance by modulating the ceramide metabolism in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115849. [PMID: 38134639 DOI: 10.1016/j.ecoenv.2023.115849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
Recent research has highlighted a correlation between exposure to ambient fine particulate matter (PM2.5) and the development of systemic insulin resistance (IR) along with an elevated risk of diabetes. Ceramide has emerged as one of the pathogenic mechanisms contributing to IR. The inhibition of acid sphingomyelinase (ASMase) activity by desipramine (DES) has been shown to effectively reduce ceramide levels. In the present study, 24 female C57BL/6 N mice were randomized into one of the four groups: the filtered air exposure (FA) group, the concentrated PM2.5 exposure (PM) group, the concentrated PM2.5 treated with low-dose DES (DL) group, and the concentrated PM2.5 treated with high-dose DES (DH) group. The PM, DL and DH groups were exposed to PM2.5 for an 8-week period within a whole-body exposure system. The study encompassed extensive examinations of glucose homeostasis, liver lipid profile, ceramide pathway, and insulin signaling pathway. Our results demonstrated that PM2.5 exposure caused impaired glucose tolerance, elevated ceramide levels, increased phosphorylation PP2A, reduced Akt phosphorylation, and hindered GLUT2 expression. Remarkably, DES administration mitigated PM2.5-induced IR by effectively lowering ceramide levels. In conclusion, the reduction of ceramide levels by DES may be a promising therapeutic strategy for coping PM2.5-induced IR.
Collapse
Affiliation(s)
- Weijia Gu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Yanxi Chai
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuxin Huang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ziwei Cai
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Rucheng Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China.
| |
Collapse
|
18
|
Lopes-Virella MF, Hammad SM, Baker NL, Klein RL, Hunt KJ. Circulating Lipoprotein Sphingolipids in Chronic Kidney Disease with and without Diabetes. Biomedicines 2024; 12:190. [PMID: 38255295 PMCID: PMC10813484 DOI: 10.3390/biomedicines12010190] [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: 11/22/2023] [Revised: 12/25/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Abnormalities of sphingolipid metabolism play an important role in diabetes. We compared sphingolipid levels in plasma and in isolated lipoproteins between healthy control subjects and two groups of patients, one with chronic kidney disease without diabetes (ND-CKD), and the other with type 2 diabetes and macroalbuminuria (D-MA). Ceramides, sphingomyelins, and sphingoid bases and their phosphates in LDL were higher in ND-CKD and in D-MA patients compared to controls. However, ceramides and sphingoid bases in HDL2 and HDL3 were lower in ND-CKD and in D-MA patients than in controls. Sphingomyelins in HDL2 and HDL3 were lower in D-MA patients than in controls but were normal in ND-CKD patients. Compared to controls, lactosylceramides in LDL and VLDL were higher in ND-CKD patients but not in D-MA patients. However, lactosylceramides in HDL2 and HDL3 were lower in both ND-CKD and D-MA patients than in controls. Plasma hexosylceramides in ND-CKD patients were increased and sphingoid bases decreased in both ND-CKD and D-MA patients. However, hexosylceramides in LDL, HDL2, and HDL3 were higher in ND-CKD patients than in controls. In D-MA patients, only C16:0 hexosylceramide in LDL was higher than in controls. The data suggest that sphingolipid measurement in lipoproteins, rather than in whole plasma, is crucial to decipher the role of sphingolipids in kidney disease.
Collapse
Affiliation(s)
- Maria F. Lopes-Virella
- Department of Medicine, Division of Diabetes, Endocrinology and Medical Genetics, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA;
| | - Samar M. Hammad
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nathaniel L. Baker
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Richard L. Klein
- Department of Medicine, Division of Diabetes, Endocrinology and Medical Genetics, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA;
| | - Kelly J. Hunt
- Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA;
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
19
|
Norris AC, Yazlovitskaya EM, Zhu L, Rose BS, May JC, Gibson-Corley KN, McLean JA, Stafford JM, Graham TR. Deficiency of the lipid flippase ATP10A causes diet-induced dyslipidemia in female mice. Sci Rep 2024; 14:343. [PMID: 38172157 PMCID: PMC10764864 DOI: 10.1038/s41598-023-50360-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Genetic association studies have linked ATP10A and closely related type IV P-type ATPases (P4-ATPases) to insulin resistance and vascular complications, such as atherosclerosis. ATP10A translocates phosphatidylcholine and glucosylceramide across cell membranes, and these lipids or their metabolites play important roles in signal transduction pathways regulating metabolism. However, the influence of ATP10A on lipid metabolism in mice has not been explored. Here, we generated gene-specific Atp10A knockout mice and show that Atp10A-/- mice fed a high-fat diet did not gain excess weight relative to wild-type littermates. However, Atp10A-/- mice displayed female-specific dyslipidemia characterized by elevated plasma triglycerides, free fatty acids and cholesterol, as well as altered VLDL and HDL properties. We also observed increased circulating levels of several sphingolipid species along with reduced levels of eicosanoids and bile acids. The Atp10A-/- mice also displayed hepatic insulin resistance without perturbations to whole-body glucose homeostasis. Thus, ATP10A has a sex-specific role in regulating plasma lipid composition and maintaining hepatic liver insulin sensitivity in mice.
Collapse
Affiliation(s)
- Adriana C Norris
- Department of Biological Sciences, Vanderbilt University, 465 21St Ave S, Nashville, TN, 37212, USA
| | - Eugenia M Yazlovitskaya
- Department of Biological Sciences, Vanderbilt University, 465 21St Ave S, Nashville, TN, 37212, USA
| | - Lin Zhu
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bailey S Rose
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA
| | - Jody C May
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA
| | - Katherine N Gibson-Corley
- Division of Comparative Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John A McLean
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA
| | - John M Stafford
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Todd R Graham
- Department of Biological Sciences, Vanderbilt University, 465 21St Ave S, Nashville, TN, 37212, USA.
| |
Collapse
|
20
|
Syed-Abdul MM. Lipid Metabolism in Metabolic-Associated Steatotic Liver Disease (MASLD). Metabolites 2023; 14:12. [PMID: 38248815 PMCID: PMC10818604 DOI: 10.3390/metabo14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Metabolic-associated steatotic liver disease (MASLD) is a cluster of pathological conditions primarily developed due to the accumulation of ectopic fat in the hepatocytes. During the severe form of the disease, i.e., metabolic-associated steatohepatitis (MASH), accumulated lipids promote lipotoxicity, resulting in cellular inflammation, oxidative stress, and hepatocellular ballooning. If left untreated, the advanced form of the disease progresses to fibrosis of the tissue, resulting in irreversible hepatic cirrhosis or the development of hepatocellular carcinoma. Although numerous mechanisms have been identified as significant contributors to the development and advancement of MASLD, altered lipid metabolism continues to stand out as a major factor contributing to the disease. This paper briefly discusses the dysregulation in lipid metabolism during various stages of MASLD.
Collapse
Affiliation(s)
- Majid Mufaqam Syed-Abdul
- Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
21
|
Fenizia S, Gaggini M, Vassalle C. Interplay between Vitamin D and Sphingolipids in Cardiometabolic Diseases. Int J Mol Sci 2023; 24:17123. [PMID: 38069444 PMCID: PMC10706901 DOI: 10.3390/ijms242317123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/28/2023] [Accepted: 12/03/2023] [Indexed: 12/18/2023] Open
Abstract
Sphingolipids (SLs) are structural, bioactive molecules with several key cellular roles, whereas 1,25-dihydroxyvitamin D (1,25(OH)D), the active form of vitamin D, is considered the major regulator of calcium homeostasis, although it also exerts other extraskeletal effects. Many studies reported the physiological connection between vitamin D and SLs, highlighting not only the effects of vitamin D on SL metabolism and signaling but also the influence of SLs on vitamin D levels and function, thus strongly suggesting a crosstalk between these molecules. After a brief description of 1,25(OH)D and SL metabolism, this review aims to discuss the preclinical and clinical evidence on the crosstalk between SLs and 1,25(OH)D, with a special focus on cardiometabolic diseases.
Collapse
Affiliation(s)
- Simona Fenizia
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Corso Trieste 15/A, I-28100 Novara, Italy;
- Department of Translational Medicine, University of Piemonte Orientale, Corso Trieste 15/A, I-28100 Novara, Italy
| | - Melania Gaggini
- Istituto di Fisiologia Clinica, Italian National Research Council, Via Moruzzi 1, I-56124 Pisa, Italy;
| | - Cristina Vassalle
- Fondazione CNR-Regione Toscana G. Monasterio, Via Moruzzi 1, I-56124 Pisa, Italy
| |
Collapse
|
22
|
Hülsmeier AJ, Toelle SP, Bellstedt P, Wentzel C, Bahr A, Kolokotronis K, Hornemann T. The atypical sphingolipid SPB 18:1(14Z);O2 is a biomarker for DEGS1 related hypomyelinating leukodystrophy. J Lipid Res 2023; 64:100464. [PMID: 37890668 PMCID: PMC10696257 DOI: 10.1016/j.jlr.2023.100464] [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: 05/11/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Sphingolipids (SL) represent a structurally diverse class of lipids that are central to cellular physiology and neuronal development and function. Defects in the sphingolipid metabolism are typically associated with nervous system disorders. The C4-dihydroceramide desaturase (DEGS1) catalyzes the conversion of dihydroceramide to ceramide, the final step in the SL de-novo synthesis. Loss of function mutations in DEGS1 cause a hypomyelinating leukodystrophy, which is associated with increased plasma dihydrosphingolipids (dhSL) and with the formation of an atypical SPB 18:1(14Z);O2 metabolite. Here, we characterize two novel DEGS1 variants of unknown significance (VUS), provide a structural model with a predicted substrate binding site, and propose a regulatory link between DEGS1 and fatty acid desaturase 3 (FADS3). Both VUS involve single amino acid substitutions near the C-terminus within conserved regions of the enzyme. Patient 1 (p.R311K variant) shows severe progressive tetraspasticity, intellectual disability, and epilepsy in combination with brain magnetic resonance imaging (MRI) findings, typical for DEGS1-related leukodystrophy. Patient 2 (p.G270E variant) presents with delayed psychomotor development, oculomotor apraxia, and a normal brain MRI. Plasma from the p.R311K carrier showed a significantly elevated dhSL species and the presence of SPB 18:1(14Z);O2, while the plasma SL profile for the p.G270E variant was not altered. This suggests the p.R331K variant is pathogenic, while the p.G270E appears benign. As an increase in dihydroSL species is also seen in other pathological disorders of the SL metabolism, the SPB 18:1(14Z);O2 seems to be a more specific biomarker to discriminate between pathogenic and benign DEGS1 variants.
Collapse
Affiliation(s)
- Andreas J Hülsmeier
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Sandra P Toelle
- Department of Pediatric Neurology, University Children's Hospital, Zurich, University of Zurich, Zurich, Switzerland
| | - Peter Bellstedt
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christian Wentzel
- Department of Women's and Children's Health, Pediatric Oncological and Neurological Research, Uppsala University, Uppsala, Sweden
| | - Angela Bahr
- Institute of Medical Genetics, University of Zurich, Schlieren, Zurich, Switzerland
| | | | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Dai T, Yan F, Gong J, Liu S. Anti-heart failure mechanism of saponin extract of black ginseng based on metabolomics. J Pharm Biomed Anal 2023; 236:115738. [PMID: 37742504 DOI: 10.1016/j.jpba.2023.115738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
OBJECTIVE This study aimed to explore the mechanism of total saponin of black ginseng (TSBG) in treating heart failure (HF) in DOX-induced HF model rats. METHODS Rats with HF induced by the intraperitoneal injection of DOX were treated with TSBG (low dose, 30 mg/kg/day; medium dose, 60 mg/kg/day; high dose, 120 mg/kg/day) and shakubar trivalsartan (80 mg/kg/day, positive control) for four weeks. Serum BNP and ANP levels were tested by ELISA, and pathological tissue sections were examined. Serum metabolites were measured using nontargeted metabolomic techniques. The expression of Akt/mTOR autophagy-associated proteins in heart tissue was detected using Western blot, including Beclin1, p62, LCII and LC3I. RESULTS Compared with the model group, rats in the TSBG-H group had a significantly lower heart index (p < 0.05), significantly lower serum levels of BNP (p < 0.01) and ANP (p < 0.01) and significantly fewer cardiac histopathological changes. Metabolomic results showed that TSBG significantly back-regulated 12 metabolites (p < 0.05), including cholesterol, histamine, sphinganine, putrescine, arachidonic acid, 3-sulfinoalanine, hypotaurine, gluconic acid and lysoPC (18:0:0). These metabolite changes were involved in taurine and hypotaurine metabolism, arachidonic acid metabolism, sphingolipid metabolism, etc. The protein expression level of p-Akt/Akt and p-mTOR/mTOR was significantly up-regulated (p < 0.001), whereas that of Beclin1, p62 (p < 0.001) and LCII/LC3I was down-regulated (p < 0.05). CONCLUSION TSBG has an excellent therapeutic effect on DOX-induced HF in rats, probably by regulating the Akt/mTOR autophagy signalling pathway, resulting in the improvement of taurine and hypotaurine metabolism, arachidonic acid metabolism and sphingolipid metabolism, which may provide a reference for elucidating the potential mechanism of action of TSBG against HF.
Collapse
Affiliation(s)
- Tingting Dai
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Fuyuan Yan
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jiyu Gong
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Shuying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
24
|
Xu J, Jin Y, Song C, Chen G, Li Q, Yuan H, Wei S, Yang M, Li S, Jin S. Comparative analysis of the synergetic effects of Diwuyanggan prescription on high fat diet-induced non-alcoholic fatty liver disease using untargeted metabolomics. Heliyon 2023; 9:e22151. [PMID: 38045182 PMCID: PMC10692813 DOI: 10.1016/j.heliyon.2023.e22151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 12/05/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic liver disorders worldwide and had no approved pharmacological treatments. Diwuyanggan prescription (DWYG) is a traditional Chinese medicine preparation composed of 5 kinds of herbs, which has been used for treating chronic liver diseases in clinic. Whereas, the synergistic mechanism of this prescription for anti-NAFLD remains unclear. In this study, we aimed to demonstrate the synergetic effect of DWYG by using the disassembled prescriptions and untargeted metabolomics research strategies. The therapeutic effects of the whole prescription of DWYG and the individual herb were divided into six groups according to the strategy of disassembled prescriptions, including DWYG, Artemisia capillaris Thunb. (AC), Curcuma longa L. (CL), Schisandra chinensis Baill. (SC), Rehmannia glutinosa Libosch. (RG) and Glycyrrhiza uralensis Fisch. (GU) groups. The high fat diets-induced NAFLD mice model was constructed to evaluate the efficacy effects of DWYG. An untargeted metabolomics based on the UPLC-QTOF-MS/MS approach was carried out to make clear the synergetic effect on the regulation of metabolites dissecting the united mechanisms. Experimental results on animals revealed that the anti-NAFLD effect of DWYG prescription was better than the individual herb group in reducing liver lipid deposition and restoring the abnormality of lipidemia. In addition, further metabolomics analysis indicated that 23 differential metabolites associated with the progression of NAFLD were identified and 19 of them could be improved by DWYG. Compared with five single herbs, DWYG showed the most extensive regulatory effects on metabolites and their related pathways, which were related to lipid and amino acid metabolisms. Besides, each individual herb in DWYG was found to show different degrees of regulatory effects on NAFLD and metabolic pathways. SC and CL possessed the highest relationship in the regulation of NAFLD. Altogether, these results provided an insight into the synergetic mechanisms of DWYG from the metabolic perspective, and also supported a scientific basis for the rationality of clinical use of this prescription.
Collapse
Affiliation(s)
- Jinlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Department of Pharmacy, Ezhou Central Hospital, Ezhou 436000, China
| | - Yuehui Jin
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Chengwu Song
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Guangya Chen
- Department of Pharmacy, Ezhou Central Hospital, Ezhou 436000, China
| | - Qiaoyu Li
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Hao Yuan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Department of Pharmacy, Ezhou Central Hospital, Ezhou 436000, China
| | - Sha Wei
- School of Basic Medicine Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Min Yang
- School of Basic Medicine Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Sen Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuna Jin
- School of Basic Medicine Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| |
Collapse
|
25
|
Kasai S, Kokubu D, Mizukami H, Itoh K. Mitochondrial Reactive Oxygen Species, Insulin Resistance, and Nrf2-Mediated Oxidative Stress Response-Toward an Actionable Strategy for Anti-Aging. Biomolecules 2023; 13:1544. [PMID: 37892226 PMCID: PMC10605809 DOI: 10.3390/biom13101544] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Reactive oxygen species (ROS) are produced mainly by mitochondrial respiration and function as signaling molecules in the physiological range. However, ROS production is also associated with the pathogenesis of various diseases, including insulin resistance (IR) and type 2 diabetes (T2D). This review focuses on the etiology of IR and early events, especially mitochondrial ROS (mtROS) production in insulin-sensitive tissues. Importantly, IR and/or defective adipogenesis in the white adipose tissues (WAT) is thought to increase free fatty acid and ectopic lipid deposition to develop into systemic IR. Fatty acid and ceramide accumulation mediate coenzyme Q reduction and mtROS production in IR in the skeletal muscle, while coenzyme Q synthesis downregulation is also involved in mtROS production in the WAT. Obesity-related IR is associated with the downregulation of mitochondrial catabolism of branched-chain amino acids (BCAAs) in the WAT, and the accumulation of BCAA and its metabolites as biomarkers in the blood could reliably indicate future T2D. Transcription factor NF-E2-related factor 2 (Nrf2), which regulates antioxidant enzyme expression in response to oxidative stress, is downregulated in insulin-resistant tissues. However, Nrf2 inducers, such as sulforaphane, could restore Nrf2 and target gene expression and attenuate IR in multiple tissues, including the WAT.
Collapse
Affiliation(s)
- Shuya Kasai
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| | - Daichi Kokubu
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
- Diet & Well-being Research Institute, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara 329-2762, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| | - Ken Itoh
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| |
Collapse
|
26
|
Anh NH, Min YJ, Thi My Nhung T, Long NP, Han S, Kim SJ, Jung CW, Yoon YC, Kang YP, Park SK, Kwon SW. Unveiling potentially convergent key events related to adverse outcome pathways induced by silver nanoparticles via cross-species omics-scale analysis. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132208. [PMID: 37544172 DOI: 10.1016/j.jhazmat.2023.132208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/19/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
The adverse effects of silver nanoparticles (AgNPs) have been studied in various models. However, there has been discordance between molecular responses across the literature, attributed to methodological biases and the physicochemical variability of AgNPs. In this study, a gene pathway meta-analysis was conducted to identify convergent and divergent key events (KEs) associated with AgNPs and explore common patterns of these KEs across species. We performed a cross-species analysis of transcriptomic data from multiple studies involving various AgNPs exposure. Pathway enrichment analysis revealed a set of pathways linked to oxidative stress, apoptosis, and metabolite and lipid metabolism, which are considered potentially conserved KEs across species. Subsequently, experiments confirmed that oxidative stress responses could be early KEs in both Caenorhabditis elegans and HepG2 cells. Moreover, AgNPs preferentially impaired the mitochondria, as evidenced by mitochondrial fragmentation and dysfunction. Furthermore, disruption of amino acids, nucleotides, sulfur compounds, glycerolipids, and glycerophospholipids metabolism were in good agreement with gene pathway shreds of evidence. Our findings imply that, although there may be organism-specific responses, potentially conserved events could exist regardless of species and physicochemical factors. These results provide valuable insights into the development of adverse outcome pathways of AgNPs across species and the regulatory toxicity of AgNPs.
Collapse
Affiliation(s)
- Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, the Republic of Korea
| | - Young Jin Min
- College of Pharmacy, Seoul National University, Seoul 08826, the Republic of Korea
| | - Truong Thi My Nhung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, the Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, the Republic of Korea
| | - Seunghyeon Han
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, the Republic of Korea
| | - Sun Jo Kim
- College of Pharmacy, Seoul National University, Seoul 08826, the Republic of Korea
| | - Cheol Woon Jung
- College of Pharmacy, Seoul National University, Seoul 08826, the Republic of Korea
| | - Young Cheol Yoon
- College of Pharmacy, Seoul National University, Seoul 08826, the Republic of Korea
| | - Yun Pyo Kang
- College of Pharmacy, Seoul National University, Seoul 08826, the Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, the Republic of Korea
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, the Republic of Korea; Plant Genomics and Breeding Institute, Seoul National University, Seoul 08826, the Republic of Korea.
| |
Collapse
|
27
|
Leiherer A, Muendlein A, Saely CH, Geiger K, Brandtner EM, Heinzle C, Gaenger S, Mink S, Laaksonen R, Fraunberger P, Drexel H. Coronary Event Risk Test (CERT) as a Risk Predictor for the 10-Year Clinical Outcome of Patients with Peripheral Artery Disease. J Clin Med 2023; 12:6151. [PMID: 37834795 PMCID: PMC10573503 DOI: 10.3390/jcm12196151] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/30/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
(1) Background: Ceramides are a new kind of lipid biomarker and have already been demonstrated to be valuable risk predictors in coronary patients. Patients with peripheral artery disease (PAD) are a population with a worse prognosis and higher mortality risk compared to coronary artery disease (CAD) patients. However, the value of ceramides for risk prediction in PAD patients is still vague, as addressed in the present study. (2)Methods: This observational study included 379 PAD patients. The primary endpoint was all-cause mortality at 10 years of follow-up. A set of ceramides was measured by LC-MS/MS and combined according to the Coronary Event Risk Test (CERT) score, which categorizes patients into one of four risk groups (low risk, moderate risk, high risk, very high risk). (3) Results: Kaplan-Meier survival curves revealed that the overall survival of patients decreased with the increasing risk predicted by the four CERT categories, advancing from low risk to very high risk. Cox regression analysis demonstrated that each one-category increase resulted in a 35% rise in overall mortality risk (HR = 1.35 [1.16-1.58]). Multivariable adjustment, including, among others, age, LDL-cholesterol, type 2 diabetes, and statin treatment before the baseline, did not abrogate this significant association (HR = 1.22 [1.04-1.43]). Moreover, we found that the beneficial effect of statin treatment is significantly stronger in patients with a higher risk, according to CERT. (4) Conclusions: We conclude that the ceramide-based risk score CERT is a strong predictor of the 10-year mortality risk in patients with PAD.
Collapse
Affiliation(s)
- Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, A-6800 Feldkirch, Austria; (A.M.); (K.G.); (E.-M.B.); (S.G.); (H.D.)
- Private University of the Principality of Liechtenstein, FL-9495 Triesen, Liechtenstein; (S.M.); (P.F.)
- Medical Central Laboratories, A-6800 Feldkirch, Austria
| | - Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, A-6800 Feldkirch, Austria; (A.M.); (K.G.); (E.-M.B.); (S.G.); (H.D.)
- Private University of the Principality of Liechtenstein, FL-9495 Triesen, Liechtenstein; (S.M.); (P.F.)
| | - Christoph H. Saely
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, A-6800 Feldkirch, Austria; (A.M.); (K.G.); (E.-M.B.); (S.G.); (H.D.)
- Private University of the Principality of Liechtenstein, FL-9495 Triesen, Liechtenstein; (S.M.); (P.F.)
- Department of Internal Medicine III, Academic Teaching Hospital Feldkirch, A-6800 Feldkirch, Austria
| | - Kathrin Geiger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, A-6800 Feldkirch, Austria; (A.M.); (K.G.); (E.-M.B.); (S.G.); (H.D.)
- Medical Central Laboratories, A-6800 Feldkirch, Austria
| | - Eva-Maria Brandtner
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, A-6800 Feldkirch, Austria; (A.M.); (K.G.); (E.-M.B.); (S.G.); (H.D.)
| | - Christine Heinzle
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, A-6800 Feldkirch, Austria; (A.M.); (K.G.); (E.-M.B.); (S.G.); (H.D.)
- Medical Central Laboratories, A-6800 Feldkirch, Austria
| | - Stella Gaenger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, A-6800 Feldkirch, Austria; (A.M.); (K.G.); (E.-M.B.); (S.G.); (H.D.)
| | - Sylvia Mink
- Private University of the Principality of Liechtenstein, FL-9495 Triesen, Liechtenstein; (S.M.); (P.F.)
- Medical Central Laboratories, A-6800 Feldkirch, Austria
| | - Reijo Laaksonen
- Finnish Cardiovascular Research Center, University of Tampere, FI-33014 Tampere, Finland;
- Zora Biosciences, FI-02150 Espoo, Finland
| | - Peter Fraunberger
- Private University of the Principality of Liechtenstein, FL-9495 Triesen, Liechtenstein; (S.M.); (P.F.)
- Medical Central Laboratories, A-6800 Feldkirch, Austria
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, A-6800 Feldkirch, Austria; (A.M.); (K.G.); (E.-M.B.); (S.G.); (H.D.)
- Private University of the Principality of Liechtenstein, FL-9495 Triesen, Liechtenstein; (S.M.); (P.F.)
- Vorarlberger Landeskrankenhausbetriebsgesellschaft, Academic Teaching Hospital Feldkirch, A-6800 Feldkirch, Austria
- Drexel University College of Medicine, Philadelphia, PA 19129, USA
| |
Collapse
|
28
|
Hong BV, Rhodes CH, Agus JK, Tang X, Zhu C, Zheng JJ, Zivkovic AM. A single 36-h water-only fast vastly remodels the plasma lipidome. Front Cardiovasc Med 2023; 10:1251122. [PMID: 37745091 PMCID: PMC10513913 DOI: 10.3389/fcvm.2023.1251122] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Background Prolonged fasting, characterized by restricting caloric intake for 24 h or more, has garnered attention as a nutritional approach to improve lifespan and support healthy aging. Previous research from our group showed that a single bout of 36-h water-only fasting in humans resulted in a distinct metabolomic signature in plasma and increased levels of bioactive metabolites, which improved macrophage function and lifespan in C. elegans. Objective This secondary outcome analysis aimed to investigate changes in the plasma lipidome associated with prolonged fasting and explore any potential links with markers of cardiometabolic health and aging. Method We conducted a controlled pilot study with 20 male and female participants (mean age, 27.5 ± 4.4 years; mean BMI, 24.3 ± 3.1 kg/m2) in four metabolic states: (1) overnight fasted (baseline), (2) 2-h postprandial fed state (fed), (3) 36-h fasted state (fasted), and (4) 2-h postprandial refed state 12 h after the 36-h fast (refed). Plasma lipidomic profiles were analyzed using liquid chromatography and electrospray ionization mass spectrometry. Results Several lipid classes, including lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), phosphatidylethanolamine, and triacylglycerol were significantly reduced in the 36-h fasted state, while free fatty acids, ceramides, and sphingomyelin were significantly increased compared to overnight fast and fed states (P < 0.05). After correction for multiple testing, 245 out of 832 lipid species were significantly altered in the fasted state compared to baseline (P < 0.05). Random forest models revealed that several lipid species, such as LPE(18:1), LPC(18:2), and FFA(20:1) were important features in discriminating the fasted state from both the overnight fasted and postprandial state. Conclusion Our findings indicate that prolonged fasting vastly remodels the plasma lipidome and markedly alters the concentrations of several lipid species, which may be sensitive biomarkers of prolonged fasting. These changes in lipid metabolism during prolonged fasting have important implications for the management of cardiometabolic health and healthy aging, and warrant further exploration and validation in larger cohorts and different population groups.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Angela M. Zivkovic
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
29
|
Norris AC, Yazlovitskaya EM, Zhu L, Rose BS, May JC, Gibson-Corley KN, McLean JA, Stafford JM, Graham TR. Deficiency of the lipid flippase ATP10A causes diet-induced dyslipidemia in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545392. [PMID: 37398141 PMCID: PMC10312798 DOI: 10.1101/2023.06.16.545392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Genetic association studies have linked ATP10A and closely related type IV P-type ATPases (P4-ATPases) to insulin resistance and vascular complications, such as atherosclerosis. ATP10A translocates phosphatidylcholine and glucosylceramide across cell membranes, and these lipids or their metabolites play important roles in signal transduction pathways regulating metabolism. However, the influence of ATP10A on lipid metabolism in mice has not been explored. Here, we generated gene-specific Atp10A knockout mice and show that Atp10A-/- mice fed a high-fat diet did not gain excess weight relative to wild-type littermates. However, Atp10A-/- mice displayed female-specific dyslipidemia characterized by elevated plasma triglycerides, free fatty acids and cholesterol, as well as altered VLDL and HDL properties. We also observed increased circulating levels of several sphingolipid species along with reduced levels of eicosanoids and bile acids. The Atp10A-/- mice also displayed hepatic insulin resistance without perturbations to whole-body glucose homeostasis. Thus, ATP10A has a sex-specific role in regulating plasma lipid composition and maintaining hepatic liver insulin sensitivity in mice.
Collapse
Affiliation(s)
- Adriana C. Norris
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Lin Zhu
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, USA
| | - Bailey S. Rose
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA
| | - Jody C. May
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA
| | - Katherine N. Gibson-Corley
- Division of Comparative Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John A. McLean
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA
| | - John M. Stafford
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Tennessee, USA
| | - Todd R. Graham
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
30
|
Li D, Tian L, Nan P, Zhang J, Zheng Y, Jia X, Gong Y, Wu Z. CerS6 triggered by high glucose activating the TLR4/IKKβ pathway regulates ferroptosis of LO2 cells through mitochondrial oxidative stress. Mol Cell Endocrinol 2023; 572:111969. [PMID: 37230220 DOI: 10.1016/j.mce.2023.111969] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 05/27/2023]
Abstract
Lipid metabolism disorders and mitochondrial dysfunction contribute to the progression of diabetes and chronic liver disease (CLD). Ferroptosis, as a form of cell death centered on reactive oxygen species (ROS) accumulation and lipid peroxidation, is closely related to mitochondrial dysfunction. However, whether there exists mechanistic links between these processes remains unknown. Here, to explore the molecular mechanism of diabetes complicated with CLD, we showed that high glucose could restrain the activity of antioxidant enzymes, promote mitochondrial ROS (mtROS) production, and induce a state of oxidative stress in the mitochondria of human normal liver (LO2) cells. We demonstrated that high glucose induced ferroptosis and promoted the development of CLD, which was reversed by the ferroptosis inhibitor Ferrostatin-1 (Fer-1). In addition, the mitochondria-targeting antioxidant Mito-TEMPO was used to intervene LO2 cells in high-glucose culture, and ferroptosis was found to be inhibited, whereas markers of liver injury and fibrosis improved. Furthermore, high glucose could promote ceramide synthetase 6 (CerS6) synthesis through the TLR4/IKKβ pathway. The knockout of CerS6 in LO2 cells showed that mitochondrial oxidative stress was attenuated, ferroptosis was inhibited, and markers of liver injury and fibrosis were ameliorated. In contrast, the overexpression of CerS6 in LO2 cells showed the opposite changes and these changes were inhibited by Mito-TEMPO. In short, we positioned the study of lipid metabolism to a specific enzyme CerS6, with a high degree of specificity. Our findings revealed the mechanism by which the mitochondria act as a bridge linking CerS6 and ferroptosis, confirming that under high glucose conditions, CerS6 promotes ferroptosis through mitochondrial oxidative stress, eventually leading to CLD.
Collapse
Affiliation(s)
- Dan Li
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Ling Tian
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Ping Nan
- Department of Obster & Gynecol, Shengli Oilfield Central Hospital, 31 Jinan Road, Dongying, 257000, Shandong, China
| | - Jun Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yin Zheng
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xinxin Jia
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yihui Gong
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Zhongming Wu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
31
|
Yu Y, Yu Y, Wang Y, Chen Y, Wang N, Wang B, Lu Y. Nonalcoholic fatty liver disease and type 2 diabetes: an observational and Mendelian randomization study. Front Endocrinol (Lausanne) 2023; 14:1156381. [PMID: 37223039 PMCID: PMC10200946 DOI: 10.3389/fendo.2023.1156381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/10/2023] [Indexed: 05/25/2023] Open
Abstract
Introduction Nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) are both chronic multisystem diseases that cause tremendous health burdens worldwide. Previous epidemiological studies have found a bidirectional relationship between these two diseases; however, their causality remains largely unknown. We aim to examine the causal relationship between NAFLD and T2DM. Methods The observational analysis included 2,099 participants from the SPECT-China study and 502,414 participants from the UK Biobank. Logistic regression and Cox regression models were used to examine the bidirectional association between NAFLD and T2DM. Two-sample Mendelian randomization (MR) analyses were conducted to investigate the causal effects of the two diseases using summary statistics of genome-wide association studies from the UK Biobank for T2DM and the FinnGen study for NAFLD. Results During the follow-up, 129 T2DM cases and 263 NAFLD cases were observed in the SPECT-China study, and 30,274 T2DM cases and 4,896 NAFLD cases occurred in the UK Biobank cohort. Baseline NAFLD was associated with an increased risk of incident T2DM in both studies (SPECT-China: OR: 1.74 (95% confidence interval (CI): 1.12-2.70); UK Biobank: HR: 2.16 (95% CI: 1.82-2.56)), while baseline T2DM was associated with incident NAFLD in the UK Biobank study only (HR: 1.58). Bidirectional MR analysis showed that genetically determined NAFLD was significantly associated with an increased risk of T2DM (OR: 1.003 (95% CI: 1.002-1.004, p< 0.001)); however, there was no evidence of an association between genetically determined T2DM and NAFLD (OR: 28.1 (95% CI: 0.7-1,143.0)). Conclusions Our study suggested the causal effect of NAFLD on T2DM development. The lack of a causal association between T2DM and NAFLD warrants further verification.
Collapse
Affiliation(s)
| | | | | | | | - Ningjian Wang
- *Correspondence: Ningjian Wang, ; Bin Wang, ; Yingli Lu,
| | - Bin Wang
- *Correspondence: Ningjian Wang, ; Bin Wang, ; Yingli Lu,
| | - Yingli Lu
- *Correspondence: Ningjian Wang, ; Bin Wang, ; Yingli Lu,
| |
Collapse
|
32
|
Jin Z, Ji Y, Su W, Zhou L, Wu X, Gao L, Guo J, Liu Y, Zhang Y, Wen X, Xia ZY, Xia Z, Lei S. The role of circadian clock-controlled mitochondrial dynamics in diabetic cardiomyopathy. Front Immunol 2023; 14:1142512. [PMID: 37215098 PMCID: PMC10196400 DOI: 10.3389/fimmu.2023.1142512] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Diabetes mellitus is a metabolic disease with a high prevalence worldwide, and cardiovascular complications are the leading cause of mortality in patients with diabetes. Diabetic cardiomyopathy (DCM), which is prone to heart failure with preserved ejection fraction, is defined as a cardiac dysfunction without conventional cardiac risk factors such as coronary heart disease and hypertension. Mitochondria are the centers of energy metabolism that are very important for maintaining the function of the heart. They are highly dynamic in response to environmental changes through mitochondrial dynamics. The disruption of mitochondrial dynamics is closely related to the occurrence and development of DCM. Mitochondrial dynamics are controlled by circadian clock and show oscillation rhythm. This rhythm enables mitochondria to respond to changing energy demands in different environments, but it is disordered in diabetes. In this review, we summarize the significant role of circadian clock-controlled mitochondrial dynamics in the etiology of DCM and hope to play a certain enlightening role in the treatment of DCM.
Collapse
Affiliation(s)
- Zhenshuai Jin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanwei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wating Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Gao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junfan Guo
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yutong Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuefu Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Escalante-Covarrubias Q, Mendoza-Viveros L, González-Suárez M, Sitten-Olea R, Velázquez-Villegas LA, Becerril-Pérez F, Pacheco-Bernal I, Carreño-Vázquez E, Mass-Sánchez P, Bustamante-Zepeda M, Orozco-Solís R, Aguilar-Arnal L. Time-of-day defines NAD + efficacy to treat diet-induced metabolic disease by synchronizing the hepatic clock in mice. Nat Commun 2023; 14:1685. [PMID: 36973248 PMCID: PMC10043291 DOI: 10.1038/s41467-023-37286-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
The circadian clock is an endogenous time-tracking system that anticipates daily environmental changes. Misalignment of the clock can cause obesity, which is accompanied by reduced levels of the clock-controlled, rhythmic metabolite NAD+. Increasing NAD+ is becoming a therapy for metabolic dysfunction; however, the impact of daily NAD+ fluctuations remains unknown. Here, we demonstrate that time-of-day determines the efficacy of NAD+ treatment for diet-induced metabolic disease in mice. Increasing NAD+ prior to the active phase in obese male mice ameliorated metabolic markers including body weight, glucose and insulin tolerance, hepatic inflammation and nutrient sensing pathways. However, raising NAD+ immediately before the rest phase selectively compromised these responses. Remarkably, timed NAD+ adjusted circadian oscillations of the liver clock until completely inverting its oscillatory phase when increased just before the rest period, resulting in misaligned molecular and behavioral rhythms in male and female mice. Our findings unveil the time-of-day dependence of NAD+-based therapies and support a chronobiology-based approach.
Collapse
Affiliation(s)
- Quetzalcoatl Escalante-Covarrubias
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Lucía Mendoza-Viveros
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Laboratorio de Cronobiología y Metabolismo, Instituto Nacional de Medicina Genómica, 14610, Mexico City, Mexico
| | - Mirna González-Suárez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Román Sitten-Olea
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Laura A Velázquez-Villegas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, 14080, Mexico City, Mexico
| | - Fernando Becerril-Pérez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Ignacio Pacheco-Bernal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Erick Carreño-Vázquez
- Laboratorio de Cronobiología y Metabolismo, Instituto Nacional de Medicina Genómica, 14610, Mexico City, Mexico
| | - Paola Mass-Sánchez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Marcia Bustamante-Zepeda
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Ricardo Orozco-Solís
- Laboratorio de Cronobiología y Metabolismo, Instituto Nacional de Medicina Genómica, 14610, Mexico City, Mexico
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados, 14330, Mexico City, Mexico
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
34
|
Martynova E, Khaibullin T, Salafutdinov I, Markelova M, Laikov A, Lopukhov L, Liu R, Sahay K, Goyal M, Baranwal M, Rizvanov AA, Khaiboullina S. Seasonal Changes in Serum Metabolites in Multiple Sclerosis Relapse. Int J Mol Sci 2023; 24:3542. [PMID: 36834957 PMCID: PMC9959388 DOI: 10.3390/ijms24043542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Multiple sclerosis (MS) is a debilitating chronic disease of unknown etiology. There are limited treatment options due to an incomplete understanding of disease pathology. The disease is shown to have seasonal exacerbation of clinical symptoms. The mechanisms of such seasonal worsening of symptoms remains unknown. In this study, we applied targeted metabolomics analysis of serum samples using LC-MC/MC to determine seasonal changes in metabolites throughout the four seasons. We also analyzed seasonal serum cytokine alterations in patients with relapsed MS. For the first time, we can demonstrate seasonal changes in various metabolites in MS compared to the control. More metabolites were affected in MS in the fall season followed by spring, while summer MS was characterized by the smallest number of affected metabolites. Ceramides were activated in all seasons, suggesting their central role in the disease pathogenesis. Substantial changes in glucose metabolite levels were found in MS, indicating a potential shift to glycolysis. An increased serum level of quinolinic acid was demonstrated in winter MS. Histidine pathways were affected, suggesting their role in relapse of MS in the spring and fall. We also found that spring and fall seasons had a higher number of overlapping metabolites affected in MS. This could be explained by patients having a relapse of symptoms during these two seasons.
Collapse
Affiliation(s)
- Ekaterina Martynova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Timur Khaibullin
- Republican Clinical Neurological Center, Republic of Tatarstan, 420021 Kazan, Russia
| | - Ilnur Salafutdinov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
- Department of Medical Biology and Genetic, Kazan State Medical University, 420088 Kazan, Russia
| | - Maria Markelova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Alexander Laikov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Leonid Lopukhov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Rongzeng Liu
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
| | - Kritika Sahay
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Mehendi Goyal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Svetlana Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| |
Collapse
|
35
|
Heim S, Teav T, Gallart-Ayala H, Ivanisevic J, Salamin N. Divergence in metabolomic profile in clownfish and damselfish skin mucus. Front Ecol Evol 2023. [DOI: 10.3389/fevo.2023.1050083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
IntroductionThe clownfish - sea anemone mutualism was suggested to have triggered the adaptive radiation of clownfishes, but the origin of clownfish resistance to stinging tentacles of host anemones remains unclear. The presence of specific compounds in the mucus of clownfishes conferring them the unique ability to prevent nematocyst discharge from their hosts has been the most supported hypothesis. Yet the mystery regarding the types of compounds found in clownfish skin mucus remains unsolved.MethodsWe analyzed the chemical composition of clownfish and damselfish mucus using an untargeted metabolomics (HILIC-HRMS) and lipidomics (RPLC-HRMS) approach.Results and DiscussionThe polar and lipid metabolome signatures were highly specific and allowed to discriminate between the clownfish and damselfish clades. The most discriminative part of the signature was the sphingolipid profile, displaying a broader diversity of ceramides present in significantly higher levels in clownfish mucus. Importantly, the inter-specific variability of metabolic signature was significantly higher in clownfishes, although their diversification is evolutionarily more recent, thus implying the impact of symbiosis on metabolic variability and adaptation. Furthermore, specialists and generalists clownfish species displayed distinctive metabolite signature. Two strict clownfish specialists, which are phylogenetically distant but share the same host species, clustered together based on their molecular signature, suggesting a link with their mutualistic nature. Overall, comparative analyses of metabolic signatures highlight differences in chemical composition of clownfish mucus and provide insight into biochemical pathways potentially implicated in clownfish adaptation to inhabit sea anemones and consequently diversify.
Collapse
|
36
|
Begemann K, Heyde I, Witt P, Inderhees J, Leinweber B, Koch CE, Jöhren O, Oelkrug R, Liskiewicz A, Müller TD, Oster H. Rest phase snacking increases energy resorption and weight gain in male mice. Mol Metab 2023; 69:101691. [PMID: 36746332 PMCID: PMC9950950 DOI: 10.1016/j.molmet.2023.101691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Snacking, i.e., the intake of small amounts of palatable food items, is a common behavior in modern societies, promoting overeating and obesity. Shifting food intake into the daily rest phase disrupts circadian rhythms and is also known to stimulate weight gain. We therefore hypothesized that chronic snacking in the inactive phase may promote body weight gain and that this effect is based on disruption of circadian clocks. METHODS Male mice were fed a daily chocolate snack either during their rest or their active phase and body weight development and metabolic parameters were investigated. Snacking experiments were repeated in constant darkness and in clock-deficient mutant mice to examine the role of external and internal time cues in mediating the metabolic effects of snacking. RESULTS Chronic snacking in the rest phase increased body weight gain and disrupted metabolic circadian rhythms in energy expenditure, body temperature, and locomotor activity. Additionally, these rest phase snacking mice assimilated more energy during the inactive phase. Body weight remained increased in rest phase snacking wildtype mice in constant darkness as well as in clock-deficient mutant mice under a regular light-dark cycle compared to mice snacking in the active phase. Weight gain effects were abolished in clock-deficient mice in constant darkness. CONCLUSIONS Our data suggest that mistimed snacking increases energy resorption and promotes body weight gain. This effect requires a functional circadian clock at least under constant darkness conditions.
Collapse
Affiliation(s)
- Kimberly Begemann
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| | - Isabel Heyde
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Pia Witt
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Julica Inderhees
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Bioanalytic Core Facility, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Brinja Leinweber
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Christiane E. Koch
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Olaf Jöhren
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Bioanalytic Core Facility, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Rebecca Oelkrug
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Institute for Endocrinology and Diabetes, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany,Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice 40-752, Poland
| | - Timo D. Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
37
|
Intermittent Fasting Activates AMP-Kinase to Restructure Right Ventricular Lipid Metabolism and Microtubules. JACC Basic Transl Sci 2023; 8:239-254. [PMID: 37034280 PMCID: PMC10077124 DOI: 10.1016/j.jacbts.2022.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 02/22/2023]
Abstract
Intermittent fasting (IF) extends life span via pleotropic mechanisms, but one important molecular mediator is adenosine monophosphate-activated protein kinase (AMPK). AMPK enhances lipid metabolism and modulates microtubule dynamics. Dysregulation of these molecular pathways causes right ventricular (RV) failure in patients with pulmonary arterial hypertension. In rodent pulmonary arterial hypertension, IF activates RV AMPK, which restores mitochondrial and peroxisomal morphology and restructures mitochondrial and peroxisomal lipid metabolism protein regulation. In addition, IF increases electron transport chain protein abundance and activity in the right ventricle. Echocardiographic and hemodynamic measures of RV function are positively associated with fatty acid oxidation and electron transport chain protein levels. IF also combats heightened microtubule density, which normalizes transverse tubule structure.
Collapse
|
38
|
Metabolic Profiling Reveals Changes in Serum Predictive of Venous Ulcer Healing. Ann Surg 2023; 277:e467-e474. [PMID: 35916649 PMCID: PMC9831039 DOI: 10.1097/sla.0000000000004933] [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] [Indexed: 01/14/2023]
Abstract
OBJECTIVE The aim of this study was to identify potential biomarkers predictive of healing or failure to heal in a population with venous leg ulceration. SUMMARY BACKGROUND DATA Venous leg ulceration presents important physical, psychological, social and financial burdens. Compression therapy is the main treatment, but it can be painful and time-consuming, with significant recurrence rates. The identification of a reliable biochemical signature with the ability to identify nonhealing ulcers has important translational applications for disease prognostication, personalized health care and the development of novel therapies. METHODS Twenty-eight patients were assessed at baseline and at 20 weeks. Untargeted metabolic profiling was performed on urine, serum, and ulcer fluid, using mass spectrometry and nuclear magnetic resonance spectroscopy. RESULTS A differential metabolic phenotype was identified in healing (n = 15) compared to nonhealing (n = 13) venous leg ulcer patients. Analysis of the assigned metabolites found ceramide and carnitine metabolism to be relevant pathways. In this pilot study, only serum biofluids could differentiate between healing and nonhealing patients. The ratio of carnitine to ceramide was able to differentiate between healing phenotypes with 100% sensitivity, 79% specificity, and 91% accuracy. CONCLUSIONS This study reports a metabolic signature predictive of healing in venous leg ulceration and presents potential translational applications for disease prognostication and development of targeted therapies.
Collapse
|
39
|
Tang C, Zhao H, Kong L, Meng F, Zhou L, Lu Z, Lu Y. Probiotic Yogurt Alleviates High-Fat Diet-Induced Lipid Accumulation and Insulin Resistance in Mice via the Adiponectin Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1464-1476. [PMID: 36695046 DOI: 10.1021/acs.jafc.2c05670] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
A high-fat diet (HFD) easily contributes to the pathogenesis of obesity and insulin resistance. Obesity and insulin resistance have been clinical and public health challenges all over the world. Probiotic-fermented yogurt is one type of popular and functional beverage in people's daily lives. This study mainly explored the lipid- and glucose-lowering effects of Lactobacillus acidophilus NX2-6-fermented yogurt (LA-Y) in HFD-fed mice. The results showed that LA-Y administration improved the lipid profile in the serum and liver, reduced fasting blood glucose levels, and enhanced insulin sensitivity. Protein analysis showed that LA-Y treatment promoted fatty acid oxidation and suppressed de novo lipogenesis in the adipose tissue and liver. LA-Y effectively alleviated glucose metabolism disorders by activating the insulin signaling pathway, suppressing gluconeogenesis in the liver and muscle, reducing the concentration of pro-inflammatory cytokines in the serum, and promoting glycolysis and gluconeogenesis in the small intestine. LA-Y supplementation also promoted fat browning via the adiponectin/AMPKα/PGC-1α/UCP1 pathway and enhanced mitochondrial biogenesis in the liver and muscle by activating the adiponectin/AdipoR1/APPL1/AMPKα/PGC-1α pathway, leading to increased energy expenditure. Therefore, LA-Y may be a functional dairy food for preventing and alleviating diet-induced metabolic disorders.
Collapse
Affiliation(s)
- Chao Tang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Hongyuan Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Liangyu Kong
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Fanqiang Meng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Libang Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
40
|
Differential Regulation of Glucosylceramide Synthesis and Efflux by Golgi and Plasma Membrane Bound ABCC10. Nutrients 2023; 15:nu15020346. [PMID: 36678216 PMCID: PMC9862172 DOI: 10.3390/nu15020346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 01/12/2023] Open
Abstract
Glucosylceramide (GlcCer) synthesis by the enzyme glucosylceramide synthase (GCS) occurs on the cytosolic leaflet of the Golgi and is the first important step for the synthesis of complex glycosphingolipids (GSLs) that takes place inside the lumen. Apart from serving as a precursor for glycosylation, newly synthesized GlcCer is also transported to the plasma membrane and secreted onto HDL in the circulation. The mechanism by which GlcCer is transported to HDL remains unclear. Recently, we showed that ATP-binding cassette transporter protein C10 (ABCC10) plays an important role in the synthesis and efflux of GlcCer in Huh-7 cells. In this study, we found that treatment of Huh-7 cells with an ABCC10 inhibitor, sorafenib, decreased the synthesis and efflux of GlcCer. However, treatment of cells with cepharanthine reduced only the efflux, but not synthesis, of GlcCer. These results indicate that ABCC10 may regulate the synthesis and efflux of GlcCer differentially in liver cells.
Collapse
|
41
|
Tang Y, Chen B, Huang X, He X, Yi J, Zhao H, Tian F, Liu Y, Liu B. Fu brick tea alleviates high fat induced non-alcoholic fatty liver disease by remodeling the gut microbiota and liver metabolism. Front Nutr 2022; 9:1062323. [PMID: 36618677 PMCID: PMC9815510 DOI: 10.3389/fnut.2022.1062323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Fu brick tea (FBT) and its extracts have good lipid-lowering effects and have been used in the treatment of obesity in previous studies. Unfortunately, the therapeutic effect of FBT on non-alcoholic fatty liver disease (NAFLD) has not been thoroughly studied. In this study, we explored the mechanism by which FBT alleviates NAFLD from the perspective of the gut microbiota and liver metabolites. The results showed that FBT could reduce the body weight, liver weight and abdominal fat of NAFLD mice, and improve liver pathological morphology, liver lipid deposition, blood lipids and liver function. Moreover, FBT improved the diversity of the gut microbiota and changed the profile of liver metabolism in NAFLD mice. Further studies showed that FBT could ameliorate the cecum barrier, and regulate the effects of factors related to lipid synthesis in the cecum and liver of NAFLD mice. In conclusion, the present study confirmed that FBT can alleviate high fat induced NAFLD by regulating the homeostasis of the gut microbiota and liver metabolites.
Collapse
Affiliation(s)
- Yan Tang
- Department of Basic Medicine, Yiyang Medical College, Yiyang, China
| | - Bowei Chen
- The First Hospital, Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xin Huang
- Department of Clinical Medicine, Yiyang Medical College, Yiyang, China
| | - Xu He
- Department of Basic Medicine, Yiyang Medical College, Yiyang, China
| | - Jian Yi
- The First Hospital, Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Hong Zhao
- Department of Basic Medicine, Yiyang Medical College, Yiyang, China
| | - Fengming Tian
- The First Hospital, Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yingfei Liu
- The First Hospital, Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Baiyan Liu
- Hunan Academy of Chinese Medicine, Changsha, China
| |
Collapse
|
42
|
Kundu P, Paraiso IL, Choi J, Miranda CL, Kioussi C, Maier CS, Bobe G, Stevens JF, Raber J. Xanthohumol improves cognition in farnesoid X receptor-deficient mice on a high-fat diet. Dis Model Mech 2022; 15:dmm049820. [PMID: 36353888 PMCID: PMC9713832 DOI: 10.1242/dmm.049820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/26/2022] [Indexed: 08/18/2023] Open
Abstract
Xanthohumol (XN) improves cognition of wild-type rodents on a high-fat diet (HFD). Bile acids and ceramide levels in the liver and hippocampus might be linked to these effects. XN modulates activity of the nuclear farnesoid X receptor (FXR; also known as NR1H4), the primary receptor for bile acids. To determine the role of FXR in the liver and intestine in mediating the effects of XN on cognitive performance, mice with intestine- and liver-specific FXR ablation (FXRIntestine-/- and FXRLiver-/-, respectively) on an HFD or an HFD containing XN were cognitively tested. XN improved cognitive performance in a genotype- and sex-dependent manner, with improved task learning in females (specifically wild-type), reversal learning in males (specifically wild-type and FXRIntestine-/- mutant) and spatial learning (both sexes). XN increased hippocampal diacylglycerol and sphingomyelin levels in females but decreased them in males. XN increased the ratio of shorter-chain to longer-chain ceramides and hexaceramides. Higher diacylglycerol and lower longer-chain ceramide and hexaceramide levels were linked to improved cognitive performance. Thus, the beneficial sex-dependent cognitive effects of XN are linked to changes in hippocampal diacylglycerol and ceramide levels. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ines L. Paraiso
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Cristobal L. Miranda
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
43
|
ATP-Binding Cassette Transporter Family C Protein 10 Participates in the Synthesis and Efflux of Hexosylceramides in Liver Cells. Nutrients 2022; 14:nu14204401. [PMID: 36297086 PMCID: PMC9610179 DOI: 10.3390/nu14204401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 11/19/2022] Open
Abstract
In addition to sphingomyelin and ceramide, sugar derivatives of ceramides, hexosylceramides (HexCer) are the major circulating sphingolipids. We have shown that silencing of ABCA1 transmembrane protein function for instance in cases of loss of function of ABCA1 gene results in low levels of HDL as well as a concomitant reduction in plasma HexCer levels. However, proteins involved in hepatic synthesis and egress of HexCer from cells is not well known although ABCA1 seems to be indirectly controlling the HexCer plasma levels by supporting HDL synthesis. In this study, we hypothesized that protein(s) other than ABCA1 are involved in the transport of HexCer to HDL. Using an unbiased knockdown approach, we found that ATP-binding cassette transporter protein C10 (ABCC10) participates in the synthesis of HexCer and thereby affects egress to HDL in human hepatoma Huh-7 cells. Furthermore, livers from ABCC10 deficient mice had significantly lower levels of HexCer compared to wild type livers. These studies suggest that ABCC10 partakes in modulating the synthesis and subsequent efflux of HexCer to HDL in liver cells.
Collapse
|
44
|
Lee CG, Lee SJ, Park S, Choi SE, Song MW, Lee HW, Kim HJ, Kang Y, Lee KW, Kim HM, Kwak JY, Lee IJ, Jeon JY. In Vivo Two-Photon Imaging Analysis of Dynamic Degradation of Hepatic Lipid Droplets in MS-275-Treated Mouse Liver. Int J Mol Sci 2022; 23:ijms23179978. [PMID: 36077368 PMCID: PMC9456374 DOI: 10.3390/ijms23179978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 12/03/2022] Open
Abstract
The accumulation of hepatic lipid droplets (LDs) is a hallmark of non-alcoholic fatty liver disease (NAFLD). Appropriate degradation of hepatic LDs and oxidation of complete free fatty acids (FFAs) are important for preventing the development of NAFLD. Histone deacetylase (HDAC) is involved in the impaired lipid metabolism seen in high-fat diet (HFD)-induced obese mice. Here, we evaluated the effect of MS-275, an inhibitor of HDAC1/3, on the degradation of hepatic LDs and FFA oxidation in HFD-induced NAFLD mice. To assess the dynamic degradation of hepatic LDs and FFA oxidation in fatty livers of MS-275-treated HFD C57BL/6J mice, an intravital two-photon imaging system was used and biochemical analysis was performed. The MS-275 improved hepatic metabolic alterations in HFD-induced fatty liver by increasing the dynamic degradation of hepatic LDs and the interaction between LDs and lysozyme in the fatty liver. Numerous peri-droplet mitochondria, lipolysis, and lipophagy were observed in the MS-275-treated mouse fatty liver. Biochemical analysis revealed that the lipolysis and autophagy pathways were activated in MS-275 treated mouse liver. In addition, MS-275 reduced the de novo lipogenesis, but increased the mitochondrial oxidation and the expression levels of oxidation-related genes, such as PPARa, MCAD, CPT1b, and FGF21. Taken together, these results suggest that MS-275 stimulates the degradation of hepatic LDs and mitochondrial free fatty acid oxidation, thus protecting against HFD-induced NAFLD.
Collapse
Affiliation(s)
- Chang-Gun Lee
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
| | - Soo-Jin Lee
- Three-Dimensional Immune System Imaging Core Facility, Ajou University, Suwon 16499, Gyeonggi-do, Korea
| | - Seokho Park
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
- Department of Biomedical Science, The Graduate School, Ajou University, Suwon 16499, Gyeonggi-do, Korea
| | - Sung-E Choi
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
| | - Min-Woo Song
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
| | - Hyo Won Lee
- Department of Energy Systems Research, Ajou University, Suwon 16499, Gyeonggi-do, Korea
- Department of Chemistry, Ajou University, Suwon 16499, Gyeonggi-do, Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
| | - Kwan Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
| | - Hwan Myung Kim
- Department of Energy Systems Research, Ajou University, Suwon 16499, Gyeonggi-do, Korea
- Department of Chemistry, Ajou University, Suwon 16499, Gyeonggi-do, Korea
| | - Jong-Young Kwak
- Three-Dimensional Immune System Imaging Core Facility, Ajou University, Suwon 16499, Gyeonggi-do, Korea
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
- Correspondence: (J.-Y.K.); (J.Y.J.); Tel.: +82-31-219-4487 (J.-Y.K.); +82-31-219-7459 (J.Y.J.); Fax: +82-31-219-5069 (J.-Y.K.); +82-31-219-4497 (J.Y.J.)
| | - In-Jeong Lee
- Three-Dimensional Immune System Imaging Core Facility, Ajou University, Suwon 16499, Gyeonggi-do, Korea
| | - Ja Young Jeon
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, Korea
- Correspondence: (J.-Y.K.); (J.Y.J.); Tel.: +82-31-219-4487 (J.-Y.K.); +82-31-219-7459 (J.Y.J.); Fax: +82-31-219-5069 (J.-Y.K.); +82-31-219-4497 (J.Y.J.)
| |
Collapse
|
45
|
Zhu Y, Wei YL, Karras I, Cai PJ, Xiao YH, Jia CL, Qian XL, Zhu SY, Zheng LJ, Hu X, Sun AD. Modulation of the gut microbiota and lipidomic profiles by black chokeberry ( Aronia melanocarpa L.) polyphenols via the glycerophospholipid metabolism signaling pathway. Front Nutr 2022; 9:913729. [PMID: 35990329 PMCID: PMC9387202 DOI: 10.3389/fnut.2022.913729] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Black chokeberry (Aronia melanocarpa L.) is rich in polyphenols with various physiological and pharmacological activities. However, the relationship between the modulation effect of black chokeberry polyphenols on obesity and the alteration of lipid metabolism is not clearly understood. This study aimed to investigate the beneficial effects of the black chokeberry polyphenols (BCPs) treatment on the structure of gut microbiota, lipid metabolism, and associated mechanisms in high-fat diet (HFD)-induced obese rats. Here, we found that a high-fat diet promoted body weight gain and lipid accumulation in rats, while oral BCPs supplementation reduced body weight, liver, and white adipose tissue weight and alleviated dyslipidemia and hepatic steatosis in HFD-induced obese rats. In addition, BCPs supplementation prevented gut microbiota dysbiosis by increasing the relative abundance of Bacteroides, Prevotella, Romboutsia, and Akkermansia and decreasing the relative abundance of Desulfovibrio and Clostridium. Furthermore, 64 lipids were identified as potential lipid biomarkers through lipidomics analysis after BCPs supplementation, especially PE (16:0/22:6), PE (18:0/22:6), PC (20:3/19:0), LysoPE (24:0), LysoPE (24:1), and LysoPC (20:0). Moreover, our studies provided new evidence that composition of gut microbiota was closely related to the alteration of lipid profiles after BCPs supplementation. Additionally, BCPs treatment could ameliorate the disorder of lipid metabolism by regulating the mRNA and protein expression of genes related to the glycerophospholipid metabolism signaling pathway in HFD-induced obese rats. The mRNA and protein expression of PPARα, CPT1α, EPT1, and LCAT were significantly altered after BCPs treatment. In conclusion, the results of this study indicated that BCPs treatment alleviated HFD-induced obesity by modulating the composition and function of gut microbiota and improving the lipid metabolism disorder via the glycerophospholipid metabolism signaling pathway.
Collapse
Affiliation(s)
- Yue Zhu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Yu-Long Wei
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Ioanna Karras
- College of Agricultural, Consumer and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Peng-Ju Cai
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Yu-Hang Xiao
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Cheng-Li Jia
- Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Xiao-Lin Qian
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Shi-Yu Zhu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Lu-Jie Zheng
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Xin Hu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Ai-Dong Sun
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| |
Collapse
|
46
|
Iqbal J, Suarez MD, Yadav PK, Walsh MT, Li Y, Wu Y, Huang Z, James AW, Escobar V, Mokbe A, Brickman AM, Luchsinger JA, Dai K, Moreno H, Hussain MM. ATP-binding cassette protein ABCA7 deficiency impairs sphingomyelin synthesis, cognitive discrimination, and synaptic plasticity in the entorhinal cortex. J Biol Chem 2022; 298:102411. [PMID: 36007616 PMCID: PMC9513280 DOI: 10.1016/j.jbc.2022.102411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 08/06/2022] [Accepted: 08/10/2022] [Indexed: 12/22/2022] Open
Abstract
Sphingomyelin (SM) is an abundant plasma membrane and plasma lipoprotein sphingolipid. We previously reported that ATP-binding cassette family A protein 1 (ABCA1) deficiency in humans and mice decreases plasma SM levels. However, overexpression, induction, downregulation, inhibition, and knockdown of ABCA1 in human hepatoma Huh7 cells did not decrease SM efflux. Using unbiased siRNA screening, here we identified that ABCA7 plays a role in the biosynthesis and efflux of SM without affecting cellular uptake and metabolism. Since loss of function mutations in the ABCA7 gene exhibit strong associations with late-onset Alzheimer's disease (LOAD) across racial groups, we also studied the effects of ABCA7 deficiency in the mouse brain. Brains of ABCA7-deficient (KO) mice, compared with wild type (WT), had significantly lower levels of several SM species with long chain fatty acids. In addition, we observed that older KO mice exhibited behavioral deficits in cognitive discrimination in the active place avoidance task. Next, we performed synaptic transmission studies in brain slices obtained from older mice. We found anomalies in synaptic plasticity at the intracortical layer II/III lateral entorhinal cortex synapse but not in the hippocampal synapses in KO mice. These synaptic abnormalities in KO brain slices were rescued with extracellular SM supplementation, but not by supplementation with phosphatidylcholine. Taken together, these studies identify a role of ABCA7 in brain SM metabolism and the importance of SM in synaptic plasticity and cognition, as well as provide a possible explanation for the association between ABCA7 and LOAD.
Collapse
Affiliation(s)
- Jahangir Iqbal
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Al Ahsa, Saudi Arabia
| | - Manuel D Suarez
- Departments of Neurology and Physiology/Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, and Kings County Hospital, Brooklyn, NY
| | - Pradeep K Yadav
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY
| | - Meghan T Walsh
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Yimeng Li
- Institute of Mental Health, the Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, 325007, China
| | - Yiyang Wu
- Institute of Mental Health, the Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, 325007, China
| | - Zhengwei Huang
- Institute of Mental Health, the Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, 325007, China
| | | | - Victor Escobar
- Departments of Neurology and Physiology/Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, and Kings County Hospital, Brooklyn, NY
| | - Ashwag Mokbe
- Departments of Neurology and Physiology/Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, and Kings County Hospital, Brooklyn, NY
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's disease and the Aging Brain and Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - José A Luchsinger
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, NY
| | - Kezhi Dai
- Institute of Mental Health, the Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, 325007, China; School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Herman Moreno
- Departments of Neurology and Physiology/Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, and Kings County Hospital, Brooklyn, NY.
| | - M Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY.
| |
Collapse
|
47
|
Cheng ZQ, Liu TM, Ren PF, Chen C, Wang YL, Dai Y, Zhang X. Duodenal-jejunal bypass reduces serum ceramides via inhibiting intestinal bile acid-farnesoid X receptor pathway. World J Gastroenterol 2022; 28:4328-4337. [PMID: 36159007 PMCID: PMC9453759 DOI: 10.3748/wjg.v28.i31.4328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/12/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bile acids play an important role in the amelioration of type 2 diabetes following duodenal-jejunal bypass (DJB). Serum bile acids are elevated postoperatively. However, the clinical relevance is not known. Bile acids in the peripheral circulation reflect the amount of bile acids in the gut. Therefore, a further investigation of luminal bile acids following DJB is of great significance.
AIM To investigate changes of luminal bile acids following DJB.
METHODS Salicylhydroxamic acid (SHAM), DJB, and DJB with oral chenodeoxycholic acid (CDCA) supplementation were performed in a high-fat-diet/streptozotocin-induced diabetic rat model. Body weight, energy intake, oral glucose tolerance test, luminal bile acids, serum ceramides and intestinal ceramide synthesis were analyzed at week 12 postoperatively.
RESULTS Compared to SHAM, DJB achieved rapid and durable improvement in glucose tolerance and led to increased total luminal bile acid concentrations with preferentially increased proportion of farnesoid X receptor (FXR) - inhibitory bile acids within the common limb. Intestinal ceramide synthesis was repressed with decreased serum ceramides, and this phenomenon could be partially antagonized by luminal supplementation of FXR activating bile acid CDCA.
CONCLUSION DJB significantly changes luminal bile acid composition with increased proportion FXR-inhibitory bile acids and reduces serum ceramide levels. There observations suggest a novel mechanism of bile acids in metabolic regulation after DJB.
Collapse
Affiliation(s)
- Zhi-Qiang Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Tong-Ming Liu
- Department of Colorectal and Anal Surgery, Feicheng Hospital Affiliated to Shandong First Medical University, Feicheng 271600, Shandong Province, China
| | - Peng-Fei Ren
- Department of General Surgery, Lincheng People’s Hospital, Dezhou 253500, Shandong Province, China
| | - Chang Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yan-Lei Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yong Dai
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
48
|
Aftermath of AGE-RAGE Cascade in the pathophysiology of cardiovascular ailments. Life Sci 2022; 307:120860. [DOI: 10.1016/j.lfs.2022.120860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/21/2022]
|
49
|
Ke JT, Zhang H, Bu YH, Gan PR, Chen FY, Dong XT, Wang Y, Wu H. Metabonomic analysis of abnormal sphingolipid metabolism in rheumatoid arthritis synovial fibroblasts in hypoxia microenvironment and intervention of geniposide. Front Pharmacol 2022; 13:969408. [PMID: 35935818 PMCID: PMC9353937 DOI: 10.3389/fphar.2022.969408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by a joint hypoxia microenvironment. Our previous untargeted metabolomics study found that sphingolipid (SPL) metabolism was abnormal in the joint synovial fluid samples from adjuvant arthritis (AA) rats. Geniposide (GE), an iridoid glycoside component of the dried fruit of Gardenia jasminoides Ellis, is commonly used for RA treatment in many Asian countries. At present, the mechanism of GE in the treatment of RA, especially in the joint hypoxia microenvironment, is not entirely clear from the perspective of SPL metabolism. The purpose of this research was to explore the potential mechanism of abnormal SPL metabolism in RA joint hypoxia microenvironment and the intervention effect of GE, through the untargeted metabolic analysis based on the ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). Arthritis index, foot swelling and histopathology were used to assess whether the AA rat model was successfully established. The SPLs extracts collected from AA rats’ synovial tissue, serum and rheumatoid arthritis synovial fibroblasts (RASFs, MH7A cells, hypoxia/normoxia culture) were analyzed by metabolomics and lipdomics approach based on UPLC-Q-TOF/MS, to identify potential biomarkers associated with disorders of GE regulated RA sphingolipid metabolism. As a result, 11 sphingolipid metabolites related to RA were screened and identified. Except for galactosylceramide (d18:1/20:0), GE could recover the change levels of the above 10 sphingolipid biomarkers in varying degrees. Western blotting results showed that the changes in ceramide (Cer) level regulated by GE were related to the down-regulation of acid-sphingomyelinase (A-SMase) expression in synovial tissue of AA rats. To sum up, this research examined the mechanism of GE in the treatment of RA from the perspective of SPL metabolism and provided a new strategy for the screening of biomarkers for clinical diagnosis of RA.
Collapse
Affiliation(s)
- Jiang-Tao Ke
- Key Laboratory of Xin’an Medicine, Ministry of Education, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Heng Zhang
- Key Laboratory of Xin’an Medicine, Ministry of Education, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Yan-Hong Bu
- Key Laboratory of Xin’an Medicine, Ministry of Education, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Pei-Rong Gan
- Key Laboratory of Xin’an Medicine, Ministry of Education, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Fang-Yuan Chen
- Key Laboratory of Xin’an Medicine, Ministry of Education, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Xin-Tong Dong
- Key Laboratory of Xin’an Medicine, Ministry of Education, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Yan Wang
- Key Laboratory of Xin’an Medicine, Ministry of Education, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- *Correspondence: Yan Wang, ; Hong Wu,
| | - Hong Wu
- Key Laboratory of Xin’an Medicine, Ministry of Education, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- *Correspondence: Yan Wang, ; Hong Wu,
| |
Collapse
|
50
|
Contribution of specific ceramides to obesity-associated metabolic diseases. Cell Mol Life Sci 2022; 79:395. [PMID: 35789435 PMCID: PMC9252958 DOI: 10.1007/s00018-022-04401-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/04/2022]
Abstract
Ceramides are a heterogeneous group of bioactive membrane sphingolipids that play specialized regulatory roles in cellular metabolism depending on their characteristic fatty acyl chain lengths and subcellular distribution. As obesity progresses, certain ceramide molecular species accumulate in metabolic tissues and cause cell-type-specific lipotoxic reactions that disrupt metabolic homeostasis and lead to the development of cardiometabolic diseases. Several mechanisms for ceramide action have been inferred from studies in vitro, but only recently have we begun to better understand the acyl chain length specificity of ceramide-mediated signaling in the context of physiology and disease in vivo. New discoveries show that specific ceramides affect various metabolic pathways and that global or tissue-specific reduction in selected ceramide pools in obese rodents is sufficient to improve metabolic health. Here, we review the tissue-specific regulation and functions of ceramides in obesity, thus highlighting the emerging concept of selectively inhibiting production or action of ceramides with specific acyl chain lengths as novel therapeutic strategies to ameliorate obesity-associated diseases.
Collapse
|