1
|
Imierska M, Zabielski P, Roszczyc-Owsiejczuk K, Pogodzińska K, Błachnio-Zabielska A. Impact of reduced hepatic ceramide levels in high-fat diet mice on glucose metabolism. J Nutr Biochem 2024:109785. [PMID: 39427846 DOI: 10.1016/j.jnutbio.2024.109785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Dysregulation of insulin action in hepatocytes, common in obesity, significantly contributes to insulin resistance, type 2 diabetes, and metabolic syndrome. Previous research highlights ceramides' role in these conditions. This study explores the impact of ceramides by silencing the serine palmitoyltransferase (Sptlc2) gene, crucial for the initial ceramide biosynthesis, using hydrodynamic gene delivery. Male C57BL/6 mice were randomly divided into three groups: one on a low-fat diet (LFD) receiving scrambled shRNA plasmids, another on a high-fat diet (HFD) with scrambled shRNA plasmids, and a third on HFD with a plasmid targeting Sptlc2. Analyses included RT-PCR for gene expression, western blot for protein levels, and UHPLC/MS/MS for lipid profiling. Glucose metabolism was evaluated via oral glucose tolerance tests, homeostatic model assessment of insulin resistance, and glucose-6-phosphate analysis. Results showed that HFD induces insulin resistance by inhibiting insulin signaling and increasing active lipid levels in hepatocytes. Sptlc2 silencing reduced ceramide accumulation, improving insulin signaling and glucose metabolism. Notably, ceramide synthesis inhibition did not significantly affect other lipid levels, highlighting ceramide's critical role in hepatic insulin resistance.
Collapse
Affiliation(s)
- Monika Imierska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland.
| | - Piotr Zabielski
- Department of Medical Biology, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland.
| | - Kamila Roszczyc-Owsiejczuk
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland.
| | - Karolina Pogodzińska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland.
| | - Agnieszka Błachnio-Zabielska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland.
| |
Collapse
|
2
|
Bubak MP, Mann SN, Borowik AK, Pranay A, Batushansky A, Vieira de Sousa Neto I, Mondal SA, Doidge SM, Davidyan A, Szczygiel MM, Peelor FF, Rigsby S, Broomfield ME, Lacy CI, Rice HC, Stout MB, Miller BF. 17α-Estradiol alleviates high-fat diet-induced inflammatory and metabolic dysfunction in skeletal muscle of male and female mice. Am J Physiol Endocrinol Metab 2024; 326:E226-E244. [PMID: 38197793 PMCID: PMC11193529 DOI: 10.1152/ajpendo.00215.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/14/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
17α-estradiol (17α-E2) is a naturally occurring nonfeminizing diastereomer of 17β-estradiol that has life span-extending effects in rodent models. To date, studies of the systemic and tissue-specific benefits of 17α-E2 have largely focused on the liver, brain, and white adipose tissue with far less focus on skeletal muscle. Skeletal muscle has an important role in metabolic and age-related disease. Therefore, this study aimed to determine whether 17α-E2 treatment has positive, tissue-specific effects on skeletal muscle during a high-fat feeding. We hypothesized that male, but not female, mice, would benefit from 17α-E2 treatment during a high-fat diet (HFD) with changes in the mitochondrial proteome to support lipid oxidation and subsequent reductions in diacylglycerol (DAG) and ceramide content. To test this hypothesis, we used a multiomics approach to determine changes in lipotoxic lipid intermediates, metabolites, and proteins related to metabolic homeostasis. Unexpectedly, we found that 17α-E2 had marked, but different, beneficial effects within each sex. In male mice, we show that 17α-E2 alleviates HFD-induced metabolic detriments of skeletal muscle by reducing the accumulation of diacylglycerol (DAG), and inflammatory cytokine levels, and altered the abundance of most of the proteins related to lipolysis and β-oxidation. Similar to male mice, 17α-E2 treatment reduced fat mass while protecting muscle mass in female mice but had little muscle inflammatory cytokine levels. Although female mice were resistant to HFD-induced changes in DAGs, 17α-E2 treatment induced the upregulation of six DAG species. In female mice, 17α-E2 treatment changed the relative abundance of proteins involved in lipolysis, β-oxidation, as well as structural and contractile proteins but to a smaller extent than male mice. These data demonstrate the metabolic benefits of 17α-E2 in skeletal muscle of male and female mice and contribute to the growing literature of the use of 17α-E2 for multi tissue health span benefits.NEW & NOTEWORTHY Using a multiomics approach, we show that 17α-E2 alleviates HFD-induced metabolic detriments in skeletal muscle by altering bioactive lipid intermediates, inflammatory cytokines, and the abundance of proteins related to lipolysis and muscle contraction. The positive effects of 17α-E2 in skeletal muscle occur in both sexes but differ in their outcome.
Collapse
Affiliation(s)
- Matthew P Bubak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Shivani N Mann
- Department of Neuroscience, University of Arizona, Tucson, Arizona, United States
| | - Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheba, Israel
| | - Ivo Vieira de Sousa Neto
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Samim A Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Stephen M Doidge
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Arik Davidyan
- Department of Biological Sciences, California State University, Sacramento, California, United States
| | - Marcelina M Szczygiel
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Sandra Rigsby
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Matle E Broomfield
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Charles I Lacy
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, United States
| | - Heather C Rice
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, United States
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma, United States
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
3
|
Brown RDR, Green CD, Weigel C, Ni B, Celi FS, Proia RL, Spiegel S. Overexpression of ORMDL3 confers sexual dimorphism in diet-induced non-alcoholic steatohepatitis. Mol Metab 2024; 79:101851. [PMID: 38081412 PMCID: PMC10772294 DOI: 10.1016/j.molmet.2023.101851] [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: 09/24/2023] [Revised: 11/15/2023] [Accepted: 12/05/2023] [Indexed: 12/22/2023] Open
Abstract
OBJECTIVE The bioactive sphingolipid metabolites ceramide and sphingosine-1-phosphate (S1P) accumulate with overnutrition and have been implicated in non-alcoholic steatohepatitis (NASH) development. ORMDL3, a negative regulator of the rate-limiting step in ceramide biosynthesis, has been identified as an obesity-related gene. Therefore, we assessed the role of ORMDL3 in diet-induced obesity and development of NASH. METHODS Globally overexpressing Ormdl3-Flag transgenic mice (ORMDL3TG) were fed a western high-fat, carbohydrate and cholesterol enriched diet, with high fructose-glucose drinking water. Physiological, biochemical and sphingolipidomic analyses were employed to measure the effect of ORMDL3 overexpression on NASH development. RESULTS ORMDL3TG male but not female mice fed a western high-fat diet and sugar water had exacerbated adipocyte hypertrophy together with increased severity of white adipose inflammation and fibrosis. Hepatic steatosis, dyslipidemia, impaired glucose homeostasis, hyperinsulinemia, and insulin resistance were significantly more severe only in obese ORMDL3TG male mice that accompanied dramatic liver fibrosis, inflammation, and formation of hepatic crown-like structures, which are unique features of human and murine NASH. Obesogenic diet induces ORMDL expression in male mice but reduces it in females. Mechanistically, overexpression of Ormdl3 lowered the levels of S1P and ceramides only in obese female mice and antithetically increased them in tissues of obese males. ORMDL3TG male mice exhibited a much greater induction of the UPR, propagating ER stress that contributed to their early development of NASH. CONCLUSIONS This study uncovered a previously unrecognized role for ORMDL3 in sexual dimorphism important for the development and progression of NASH.
Collapse
Affiliation(s)
- Ryan D R Brown
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Christopher D Green
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Cynthia Weigel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Bin Ni
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Francesco S Celi
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Richard L Proia
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
4
|
Chella Krishnan K, El Hachem EJ, Keller MP, Patel SG, Carroll L, Vegas AD, Gerdes Gyuricza I, Light C, Cao Y, Pan C, Kaczor-Urbanowicz KE, Shravah V, Anum D, Pellegrini M, Lee CF, Seldin MM, Rosenthal NA, Churchill GA, Attie AD, Parker B, James DE, Lusis AJ. Genetic architecture of heart mitochondrial proteome influencing cardiac hypertrophy. eLife 2023; 12:e82619. [PMID: 37276142 PMCID: PMC10241513 DOI: 10.7554/elife.82619] [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: 08/11/2022] [Accepted: 05/18/2023] [Indexed: 06/07/2023] Open
Abstract
Mitochondria play an important role in both normal heart function and disease etiology. We report analysis of common genetic variations contributing to mitochondrial and heart functions using an integrative proteomics approach in a panel of inbred mouse strains called the Hybrid Mouse Diversity Panel (HMDP). We performed a whole heart proteome study in the HMDP (72 strains, n=2-3 mice) and retrieved 848 mitochondrial proteins (quantified in ≥50 strains). High-resolution association mapping on their relative abundance levels revealed three trans-acting genetic loci on chromosomes (chr) 7, 13 and 17 that regulate distinct classes of mitochondrial proteins as well as cardiac hypertrophy. DAVID enrichment analyses of genes regulated by each of the loci revealed that the chr13 locus was highly enriched for complex-I proteins (24 proteins, P=2.2E-61), the chr17 locus for mitochondrial ribonucleoprotein complex (17 proteins, P=3.1E-25) and the chr7 locus for ubiquinone biosynthesis (3 proteins, P=6.9E-05). Follow-up high resolution regional mapping identified NDUFS4, LRPPRC and COQ7 as the candidate genes for chr13, chr17 and chr7 loci, respectively, and both experimental and statistical analyses supported their causal roles. Furthermore, a large cohort of Diversity Outbred mice was used to corroborate Lrpprc gene as a driver of mitochondrial DNA (mtDNA)-encoded gene regulation, and to show that the chr17 locus is specific to heart. Variations in all three loci were associated with heart mass in at least one of two independent heart stress models, namely, isoproterenol-induced heart failure and diet-induced obesity. These findings suggest that common variations in certain mitochondrial proteins can act in trans to influence tissue-specific mitochondrial functions and contribute to heart hypertrophy, elucidating mechanisms that may underlie genetic susceptibility to heart failure in human populations.
Collapse
Affiliation(s)
- Karthickeyan Chella Krishnan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of MedicineCincinnatiUnited States
| | - Elie-Julien El Hachem
- Department of Integrative Biology and Physiology, Field Systems Biology, Sciences Sorbonne UniversitéParisFrance
| | - Mark P Keller
- Biochemistry Department, University of Wisconsin-MadisonMadisonUnited States
| | - Sanjeet G Patel
- Department of Surgery/Division of Cardiac Surgery, University of Southern California Keck School of MedicineLos AngelesUnited States
| | - Luke Carroll
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Alexis Diaz Vegas
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | | | - Christine Light
- Cardiovascular Biology Research Program, Oklahoma Medical Research FoundationOklahoma CityUnited States
| | - Yang Cao
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Division of Oral Biology and Medicine, UCLA School of DentistryLos AngelesUnited States
- UCLA Institute for Quantitative and Computational BiosciencesLos AngelesUnited States
| | - Varun Shravah
- Department of Chemistry, University of CaliforniaLos AngelesUnited States
| | - Diana Anum
- Department of Integrative Biology and Physiology, University of CaliforniaLos AngelesUnited States
| | - Matteo Pellegrini
- UCLA Institute for Quantitative and Computational BiosciencesLos AngelesUnited States
| | - Chi Fung Lee
- Cardiovascular Biology Research Program, Oklahoma Medical Research FoundationOklahoma CityUnited States
- Department of Physiology, University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Marcus M Seldin
- Center for Epigenetics and MetabolismIrvineUnited States
- Department of Biological Chemistry, University of CaliforniaIrvineUnited States
| | | | | | - Alan D Attie
- Biochemistry Department, University of Wisconsin-MadisonMadisonUnited States
| | - Benjamin Parker
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
| | - David E James
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, University of CaliforniaLos AngelesUnited States
- Department of Microbiology, Immunology and Molecular Genetics, University of CaliforniaLos AngelesUnited States
| |
Collapse
|
5
|
Jurrjens AW, Seldin MM, Giles C, Meikle PJ, Drew BG, Calkin AC. The potential of integrating human and mouse discovery platforms to advance our understanding of cardiometabolic diseases. eLife 2023; 12:e86139. [PMID: 37000167 PMCID: PMC10065800 DOI: 10.7554/elife.86139] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/15/2023] [Indexed: 04/01/2023] Open
Abstract
Cardiometabolic diseases encompass a range of interrelated conditions that arise from underlying metabolic perturbations precipitated by genetic, environmental, and lifestyle factors. While obesity, dyslipidaemia, smoking, and insulin resistance are major risk factors for cardiometabolic diseases, individuals still present in the absence of such traditional risk factors, making it difficult to determine those at greatest risk of disease. Thus, it is crucial to elucidate the genetic, environmental, and molecular underpinnings to better understand, diagnose, and treat cardiometabolic diseases. Much of this information can be garnered using systems genetics, which takes population-based approaches to investigate how genetic variance contributes to complex traits. Despite the important advances made by human genome-wide association studies (GWAS) in this space, corroboration of these findings has been hampered by limitations including the inability to control environmental influence, limited access to pertinent metabolic tissues, and often, poor classification of diseases or phenotypes. A complementary approach to human GWAS is the utilisation of model systems such as genetically diverse mouse panels to study natural genetic and phenotypic variation in a controlled environment. Here, we review mouse genetic reference panels and the opportunities they provide for the study of cardiometabolic diseases and related traits. We discuss how the post-GWAS era has prompted a shift in focus from discovery of novel genetic variants to understanding gene function. Finally, we highlight key advantages and challenges of integrating complementary genetic and multi-omics data from human and mouse populations to advance biological discovery.
Collapse
Affiliation(s)
- Aaron W Jurrjens
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, United States
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| |
Collapse
|
6
|
DeVeaux SA, Ogle ME, Vyshnya S, Chiappa NF, Leitmann B, Rudy R, Day A, Mortensen LJ, Kurtzberg J, Roy K, Botchwey EA. Characterizing human mesenchymal stromal cells' immune-modulatory potency using targeted lipidomic profiling of sphingolipids. Cytotherapy 2022; 24:608-618. [PMID: 35190267 PMCID: PMC10725732 DOI: 10.1016/j.jcyt.2021.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022]
Abstract
Cell therapies are expected to increase over the next decade owing to increasing demand for clinical applications. Mesenchymal stromal cells (MSCs) have been explored to treat a number of diseases, with some successes in early clinical trials. Despite early successes, poor MSC characterization results in lessened therapeutic capacity once in vivo. Here, we characterized MSCs derived from bone marrow (BM), adipose tissue and umbilical cord tissue for sphingolipids (SLs), a class of bioactive lipids, using liquid chromatography/tandem mass spectrometry. We found that ceramide levels differed based on the donor's sex in BM-MSCs. We detected fatty acyl chain variants in MSCs from all three sources. Linear discriminant analysis revealed that MSCs separated based on tissue source. Principal component analysis showed that interferon-γ-primed and unstimulated MSCs separated according to their SL signature. Lastly, we detected higher ceramide levels in low indoleamine 2,3-dioxygenase MSCs, indicating that sphingomyelinase or ceramidase enzymatic activity may be involved in their immune potency.
Collapse
Affiliation(s)
- S’Dravious A. DeVeaux
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Molly E. Ogle
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Sofiya Vyshnya
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Nathan F. Chiappa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Bobby Leitmann
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA
| | - Ryan Rudy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Abigail Day
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Luke J. Mortensen
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA
| | - Edward A. Botchwey
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
7
|
Hsieh J, Molusky MM, McCabe KM, Fotakis P, Xiao T, Tascau L, Zeana-Schliep L, DaSilva-Jardine P, Tall AR. TTC39B destabilizes retinoblastoma protein promoting hepatic lipogenesis in a sex-specific fashion. J Hepatol 2022; 76:383-393. [PMID: 34600974 PMCID: PMC8766887 DOI: 10.1016/j.jhep.2021.09.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND & AIMS Molecular mechanisms underlying the different susceptibility of men and women to non-alcoholic fatty liver disease (NAFLD) are poorly understood. The TTC39B locus encodes a scaffolding protein, associates with gynecological disorders and its deletion protects mice from diet-induced steatohepatitis. This study aimed to elucidate the molecular mechanisms linking TTC39B (T39) to the expression of lipogenic genes and to explore sex-specific effects. METHODS Co-expression in HEK293A cells validated the novel T39/pRb interaction predicted by a protein-protein interaction algorithm. T39 was knocked down using an antisense oligonucleotide (ASO) in mice with dietary NAFLD and a genetic deficiency of pRb or its downstream effector E2F1, as well as in primary human hepatocytes. RESULTS T39 interacts with pRb via its C-terminal TPR domain and promotes its proteasomal degradation. In female mice, T39 deficiency reduces the mRNA of lipogenic genes, especially Pnpla3, in a pRb- and E2F1-dependent manner. In contrast, in male mice, T39 deficiency results in a much smaller reduction in lipogenic gene expression that is independent of pRb/E2F1. T39 also interacts with VAPB via an N-terminal FFAT motif and stabilizes the interaction of VAPB with SCAP. Ovariectomy abolishes the effect of T39 knockdown on the hepatic pRb/E2F1/Pnpla3 axis. In both sexes T39 knockdown reduces SCAP independently of pRb. In primary human hepatocytes, T39 knockdown reduces expression of PNPLA3 and other lipogenic genes in women but not men. CONCLUSIONS We have uncovered a conserved sexual dimorphism in the regulation of hepatic lipogenic genes, with effects of T39 mediated through pRb/E2F1 in females and VAPB/SCAP in both sexes. T39 inhibition could be a novel strategy to downregulate PNPLA3 and treat NAFLD in women. LAY SUMMARY In females, the protein TTC39B degrades a tumor suppressor in the liver to promote the synthesis of new fat and the expression of a major genetic risk factor for non-alcoholic fatty liver disease. TTC39B is a potential therapeutic target for non-alcoholic fatty liver disease, especially in women.
Collapse
Affiliation(s)
- Joanne Hsieh
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA.
| | - Matthew M. Molusky
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Kristin M. McCabe
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Panagiotis Fotakis
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Tong Xiao
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Liana Tascau
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Lars Zeana-Schliep
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | | | - Alan R. Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
8
|
Chella Krishnan K, Vergnes L, Acín-Pérez R, Stiles L, Shum M, Ma L, Mouisel E, Pan C, Moore TM, Péterfy M, Romanoski CE, Reue K, Björkegren JLM, Laakso M, Liesa M, Lusis AJ. Sex-specific genetic regulation of adipose mitochondria and metabolic syndrome by Ndufv2. Nat Metab 2021; 3:1552-1568. [PMID: 34697471 PMCID: PMC8909918 DOI: 10.1038/s42255-021-00481-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/17/2021] [Indexed: 12/28/2022]
Abstract
We have previously suggested a central role for mitochondria in the observed sex differences in metabolic traits. However, the mechanisms by which sex differences affect adipose mitochondrial function and metabolic syndrome are unclear. Here we show that in both mice and humans, adipose mitochondrial functions are elevated in females and are strongly associated with adiposity, insulin resistance and plasma lipids. Using a panel of diverse inbred strains of mice, we identify a genetic locus on mouse chromosome 17 that controls mitochondrial mass and function in adipose tissue in a sex- and tissue-specific manner. This locus contains Ndufv2 and regulates the expression of at least 89 mitochondrial genes in females, including oxidative phosphorylation genes and those related to mitochondrial DNA content. Overexpression studies indicate that Ndufv2 mediates these effects by regulating supercomplex assembly and elevating mitochondrial reactive oxygen species production, which generates a signal that increases mitochondrial biogenesis.
Collapse
Affiliation(s)
- Karthickeyan Chella Krishnan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA.
| | - Laurent Vergnes
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rebeca Acín-Pérez
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael Shum
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular Medicine, Faculty of Medicine, Universite Laval, Quebec City, Quebec, Canada
| | - Lijiang Ma
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Etienne Mouisel
- INSERM, UMR1297, Institute of Metabolic and Cardiovascular Diseases, University of Toulouse, Paul Sabatier University, Toulouse, France
| | - Calvin Pan
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Timothy M Moore
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Miklós Péterfy
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Casey E Romanoski
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Karen Reue
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Marc Liesa
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA.
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Im YR, Hunter H, de Gracia Hahn D, Duret A, Cheah Q, Dong J, Fairey M, Hjalmarsson C, Li A, Lim HK, McKeown L, Mitrofan CG, Rao R, Utukuri M, Rowe IA, Mann JP. A Systematic Review of Animal Models of NAFLD Finds High-Fat, High-Fructose Diets Most Closely Resemble Human NAFLD. Hepatology 2021; 74:1884-1901. [PMID: 33973269 DOI: 10.1002/hep.31897] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Animal models of human disease are a key component of translational hepatology research, yet there is no consensus on which model is optimal for NAFLD. APPROACH AND RESULTS We generated a database of 3,920 rodent models of NAFLD. Study designs were highly heterogeneous, and therefore, few models had been cited more than once. Analysis of genetic models supported the current evidence for the role of adipose dysfunction and suggested a role for innate immunity in the progression of NAFLD. We identified that high-fat, high-fructose diets most closely recapitulate the human phenotype of NAFLD. There was substantial variability in the nomenclature of animal models: a consensus on terminology of specialist diets is needed. More broadly, this analysis demonstrates the variability in preclinical study design, which has wider implications for the reproducibility of in vivo experiments both in the field of hepatology and beyond. CONCLUSIONS This systematic analysis provides a framework for phenotypic assessment of NAFLD models and highlights the need for increased standardization and replication.
Collapse
Affiliation(s)
- Yu Ri Im
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Harriet Hunter
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Dana de Gracia Hahn
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Amedine Duret
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Qinrong Cheah
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Jiawen Dong
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Madison Fairey
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Alice Li
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Hong Kai Lim
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Lorcán McKeown
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Raunak Rao
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Mrudula Utukuri
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ian A Rowe
- Leeds Institute for Medical Research and Leeds Institute for Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Jake P Mann
- Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
10
|
Song Z, Yan W, Abulikemu M, Wang J, Xing Y, Zhou Q, Ma S, Chang C. Sphingolipid profiles and their relationship with inflammatory factors in asthmatic patients of different sexes. Chronic Dis Transl Med 2021; 7:199-205. [PMID: 34505020 PMCID: PMC8413120 DOI: 10.1016/j.cdtm.2021.04.002] [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: 11/29/2020] [Indexed: 11/26/2022] Open
Abstract
Background Asthma is a heterogeneous disease with distinct prevalence and manifestation between sexes. This study was to identify sex-specific features of asthma via metabolomic analysis of sphingolipids. Methods Forty-two asthma patients (27 women and 15 men) admitted to the Peking University Third Hospital from January 2015 to December 2015 were enrolled. Peripheral venous blood was collected for metabolomic analysis by targeted liquid chromatography-mass spectrometry. Sex hormones(estradiol, progesterone, testosterone, and androstenedione) and multiple inflammatory factors (periostin, leptin, IgE, IL-4, IL-5, IL-10, IL-13, IL-17A, and IFN-γ) were also assessed. The eosinophil percentage in induced sputum was also detected. All these data were applied to comparative analysis between sexes. Results Testosterone was negatively related to periostin (ρ = −0.420, P = 0.009) and IL-5 (ρ = −0.540, P = 0.012), while estradiol was positively related to the blood eosinophil percentage (ρ = 0.384, P = 0.025). Among the eighteen species of sphingolipids detected in the 42 patients, five ceramide (Cer) species (Cer16:0, Cer:20:0, Cer22:0, Cer24:0, and Cer26:0) and one sphingomyelin (SM) species (SM38:0) were significantly higher in male than in female patients. Further investigation found that the correlation between Cer20:0 and IL-5 was positive in males (ρ = 0.943, P = 0.005) but negative in females (ρ = −0.561, P = 0.030). Conclusions Testosterone was negatively correlated with eosinophil inflammatory factors, but estradiol was positively correlated. Male asthma patients had higher ceramide and sphingomyelin levels than female patients. Different sexes had opposite correlations with ceramide and IL-5, respectively, suggesting that therapeutic strategies targeting ceramide should be different between sexes.
Collapse
Affiliation(s)
- Zhu Song
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Wei Yan
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Mairipaiti Abulikemu
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Juan Wang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China
| | - Qingtao Zhou
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Shaohua Ma
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Chun Chang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
11
|
Bai X, Liao Y, Sun F, Xiao X, Fu S. Diurnal regulation of oxidative phosphorylation restricts hepatocyte proliferation and inflammation. Cell Rep 2021; 36:109659. [PMID: 34496251 DOI: 10.1016/j.celrep.2021.109659] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/14/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
The principles guiding the diurnal organization of biological pathways remain to be fully elucidated. Here, we perturb the hepatic transcriptome through nutrient regulators (high-fat diet and mTOR signaling components) to identify enduring properties of pathway organization. Temporal separation and counter-regulation between pathways of energy metabolism and inflammation/proliferation emerge as persistent transcriptome features across animal models, and network analysis identifies the G0s2 and Rgs16 genes as potential mediators at the metabolism-inflammation interface. Mechanistically, G0s2 and Rgs16 are sequentially induced during the light phase, promoting amino acid oxidation and suppressing overall mitochondrial respiration. In their absence, sphingolipids and diacylglycerides accumulate, accompanied by hepatic inflammation and hepatocyte proliferation. Notably, the expression of G0s2 and Rgs16 is further induced in obese mouse livers, and silencing of their expression accentuates hepatic fibrosis. Therefore, diurnal regulation of energy metabolism alleviates inflammatory and proliferative stresses under physiological and pathological conditions.
Collapse
Affiliation(s)
- Xiaojie Bai
- School of Life Sciences, Tsinghua University, Beijing, China 100084
| | - Yilie Liao
- School of Life Sciences, Tsinghua University, Beijing, China 100084
| | - Fangfang Sun
- School of Life Sciences, Tsinghua University, Beijing, China 100084
| | - Xia Xiao
- School of Life Sciences, Tsinghua University, Beijing, China 100084
| | - Suneng Fu
- School of Life Sciences, Tsinghua University, Beijing, China 100084; Department of Basic Research, Guangzhou Laboratory, Guangdong, China 510005.
| |
Collapse
|
12
|
Fineide F, Chen X, Bjellaas T, Vitelli V, Utheim TP, Jensen JL, Galtung HK. Characterization of Lipids in Saliva, Tears and Minor Salivary Glands of Sjögren's Syndrome Patients Using an HPLC/MS-Based Approach. Int J Mol Sci 2021; 22:ijms22168997. [PMID: 34445702 PMCID: PMC8396590 DOI: 10.3390/ijms22168997] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 01/11/2023] Open
Abstract
The diagnostic work-up of primary Sjögren’s syndrome (pSS) includes quantifying saliva and tear production, evaluation of autoantibodies in serum and histopathological analysis of minor salivary glands. Thus, the potential for further utilizing these fluids and tissues in the quest to find better diagnostic and therapeutic tools should be fully explored. Ten samples of saliva and tears from female patients diagnosed with pSS and ten samples of saliva and tears from healthy females were included for lipidomic analysis of tears and whole saliva using high-performance liquid chromatography coupled to time-of-flight mass spectrometry. In addition, lipidomic analysis was performed on minor salivary gland biopsies from three pSS and three non-SS females. We found significant differences in the lipidomic profiles of saliva and tears in pSS patients compared to healthy controls. Moreover, there were differences in individual lipid species in stimulated saliva that were comparable to those of glandular biopsies, representing an intriguing avenue for further research. We believe a comprehensive elucidation of the changes in lipid composition in saliva, tears and minor salivary glands in pSS patients may be the key to detecting pSS-related dry mouth and dry eyes at an early stage. The identified differences may illuminate the path towards future innovative diagnostic methodologies and treatment modalities for alleviating pSS-related sicca symptoms.
Collapse
Affiliation(s)
- Fredrik Fineide
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 1171 Oslo, Norway; (F.F.); (T.P.U.)
- The Norwegian Dry Eye Clinic, Ole Vigs Gate 32 E, 0366 Oslo, Norway
| | - Xiangjun Chen
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, 0317 Oslo, Norway; (X.C.); (J.L.J.)
| | | | - Valeria Vitelli
- Department of Biostatistics, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0316 Oslo, Norway;
| | - Tor Paaske Utheim
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 1171 Oslo, Norway; (F.F.); (T.P.U.)
- The Norwegian Dry Eye Clinic, Ole Vigs Gate 32 E, 0366 Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, 1171 Oslo, Norway
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, 0316 Oslo, Norway
| | - Janicke Liaaen Jensen
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, 0317 Oslo, Norway; (X.C.); (J.L.J.)
| | - Hilde Kanli Galtung
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, 0316 Oslo, Norway
- Correspondence:
| |
Collapse
|
13
|
Barron K, Ogretmen B, Krupenko N. Ceramide synthase 6 mediates sex-specific metabolic response to dietary folic acid in mice. J Nutr Biochem 2021; 98:108832. [PMID: 34358645 DOI: 10.1016/j.jnutbio.2021.108832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/20/2021] [Accepted: 07/01/2021] [Indexed: 11/26/2022]
Abstract
Folic acid-fortified foods and multi-vitamin supplements containing folic acid (FA) are widely used around the world, but the exact mechanisms/metabolic effects of FA are not precisely identified. We have demonstrated that Ceramide Synthase 6 (CerS6) and C16:0-ceramide mediate response to folate stress in cultured cells. Here we investigated the dietary FA effects on mouse liver metabolome, with a specific focus on sphingolipids, CerS6 and C16:0-ceramide. Wild-type and CerS6-/- mice were fed FA-deficient, control, or FA over-supplemented diets for 4 weeks. After dietary treatment, liver concentrations of ceramides, sphingomyelins and hexosylceramides were measured by LC-MS/MS and complemented by untargeted metabolomic characterization of mouse livers. Our study shows that alterations in dietary FA elicit multiple sphingolipid responses mediated by CerS6 in mouse livers. Folic acid-deficient diet elevated C14:0-, C18:0- and C20:0- but not C16:0-ceramide in WT male and female mice. Additionally, FA over-supplementation increased multiple sphingomyelin species, including total sphingomyelins, in both sexes. Of note, concentrations of C14:0- and C16:0-ceramides and hexosylceramides were significantly higher in female livers than in male. The latter were increased by FD diet, with no difference between sexes in total pools of these sphingolipid classes. Untargeted liver metabolomic analysis concurred with the targeted measurements and showed broad effects of dietary FA and CerS6 status on multiple lipid classes including sex-specific effects on phosphatidylethanolamines and diacylglycerols. Our study demonstrates that both dietary FA and CerS6 status exhibit pleiotropic and sex-dependent effects on liver metabolism, including hepatic sphingolipids, diacylglycerols, long chain fatty acids, and phospholipids.
Collapse
Affiliation(s)
- Keri Barron
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - Besim Ogretmen
- Department of Biochemistry & Molecular Biology, Hollings Cancer center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina
| | - Natalia Krupenko
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina,; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina,.
| |
Collapse
|
14
|
CD73 Maintains Hepatocyte Metabolic Integrity and Mouse Liver Homeostasis in a Sex-Dependent Manner. Cell Mol Gastroenterol Hepatol 2021; 12:141-157. [PMID: 33516905 PMCID: PMC8082562 DOI: 10.1016/j.jcmgh.2021.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Metabolic imbalance and inflammation are common features of chronic liver diseases. Molecular factors controlling these mechanisms represent potential therapeutic targets. CD73 is the major enzyme that dephosphorylates extracellular adenosine monophosphate (AMP) to form the anti-inflammatory adenosine. CD73 is expressed on pericentral hepatocytes, which are important for long-term liver homeostasis. We aimed to determine if CD73 has nonredundant hepatoprotective functions. METHODS Liver-specific CD73 knockout (CD73-LKO) mice were generated by targeting the Nt5e gene in hepatocytes. The CD73-LKO mice and hepatocytes were characterized using multiple approaches. RESULTS Deletion of hepatocyte Nt5e resulted in an approximately 70% reduction in total liver CD73 protein (P < .0001). Male and female CD73-LKO mice developed normally during the first 21 weeks without significant liver phenotypes. Between 21 and 42 weeks, the CD73-LKO mice developed spontaneous-onset liver disease, with significant severity in male mice. Middle-aged male CD73-LKO mice showed hepatocyte swelling and ballooning (P < .05), inflammation (P < .01), and variable steatosis. Female CD73-LKO mice had lower serum albumin levels (P < .05) and increased inflammatory genes (P < .01), but did not show the spectrum of histopathologic changes in male mice, potentially owing to compensatory induction of adenosine receptors. Serum analysis and proteomic profiling of hepatocytes from male CD73-LKO mice showed significant metabolic imbalance, with increased blood urea nitrogen (P < .0001) and impairments in major metabolic pathways, including oxidative phosphorylation and AMP-activated protein kinase (AMPK) signaling. There was significant hypophosphorylation of AMPK substrates in CD73-LKO livers (P < .0001), while in isolated hepatocytes treated with AMP, soluble CD73 induced AMPK activation (P < .001). CONCLUSIONS Hepatocyte CD73 supports long-term metabolic liver homeostasis through AMPK in a sex-dependent manner. These findings have implications for human liver diseases marked by CD73 dysregulation.
Collapse
|
15
|
Norheim F, Chella Krishnan K, Bjellaas T, Vergnes L, Pan C, Parks BW, Meng Y, Lang J, Ward JA, Reue K, Mehrabian M, Gundersen TE, Péterfy M, Dalen KT, Drevon CA, Hui ST, Lusis AJ, Seldin MM. Genetic regulation of liver lipids in a mouse model of insulin resistance and hepatic steatosis. Mol Syst Biol 2021; 17:e9684. [PMID: 33417276 PMCID: PMC7792507 DOI: 10.15252/msb.20209684] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/31/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
To elucidate the contributions of specific lipid species to metabolic traits, we integrated global hepatic lipid data with other omics measures and genetic data from a cohort of about 100 diverse inbred strains of mice fed a high-fat/high-sucrose diet for 8 weeks. Association mapping, correlation, structure analyses, and network modeling revealed pathways and genes underlying these interactions. In particular, our studies lead to the identification of Ifi203 and Map2k6 as regulators of hepatic phosphatidylcholine homeostasis and triacylglycerol accumulation, respectively. Our analyses highlight mechanisms for how genetic variation in hepatic lipidome can be linked to physiological and molecular phenotypes, such as microbiota composition.
Collapse
Affiliation(s)
- Frode Norheim
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
- Department of NutritionInstitute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
| | | | | | - Laurent Vergnes
- Department of Human GeneticsUniversity of California at Los AngelesLos AngelesCAUSA
| | - Calvin Pan
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - Brian W Parks
- Department of Nutritional SciencesUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Yonghong Meng
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - Jennifer Lang
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - James A Ward
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - Karen Reue
- Department of Human GeneticsUniversity of California at Los AngelesLos AngelesCAUSA
| | - Margarete Mehrabian
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | | | - Miklós Péterfy
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
- Depatrment of Basic Medical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Knut T Dalen
- Department of NutritionInstitute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
| | - Christian A Drevon
- Department of NutritionInstitute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
- Vitas ASOsloNorway
| | - Simon T Hui
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - Aldons J Lusis
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
- Department of Human GeneticsUniversity of California at Los AngelesLos AngelesCAUSA
| | - Marcus M Seldin
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
- Department of Biological Chemistry and Center for Epigenetics and MetabolismUniversity of California, IrvineIrvineCAUSA
| |
Collapse
|
16
|
Chella Krishnan K, Floyd RR, Sabir S, Jayasekera DW, Leon-Mimila PV, Jones AE, Cortez AA, Shravah V, Péterfy M, Stiles L, Canizales-Quinteros S, Divakaruni AS, Huertas-Vazquez A, Lusis AJ. Liver Pyruvate Kinase Promotes NAFLD/NASH in Both Mice and Humans in a Sex-Specific Manner. Cell Mol Gastroenterol Hepatol 2020; 11:389-406. [PMID: 32942044 PMCID: PMC7788245 DOI: 10.1016/j.jcmgh.2020.09.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The etiology of nonalcoholic fatty liver disease (NAFLD) is poorly understood, with males and certain populations exhibiting markedly increased susceptibility. Using a systems genetics approach involving multi-omic analysis of ∼100 diverse inbred strains of mice, we recently identified several candidate genes driving NAFLD. We investigated the role of one of these, liver pyruvate kinase (L-PK or Pklr), in NAFLD by using patient samples and mouse models. METHODS We examined L-PK expression in mice of both sexes and in a cohort of bariatric surgery patients. We used liver-specific loss- and gain-of-function strategies in independent animal models of diet-induced steatosis and fibrosis. After treatment, we measured several metabolic phenotypes including obesity, insulin resistance, dyslipidemia, liver steatosis, and fibrosis. Liver tissues were used for gene expression and immunoblotting, and liver mitochondria bioenergetics was characterized. RESULTS In both mice and humans, L-PK expression is up-regulated in males via testosterone and is strongly associated with NAFLD severity. In a steatosis model, L-PK silencing in male mice improved glucose tolerance, insulin sensitivity, and lactate/pyruvate tolerance compared with controls. Furthermore, these animals had reduced plasma cholesterol levels and intrahepatic triglyceride accumulation. Conversely, L-PK overexpression in male mice resulted in augmented disease phenotypes. In contrast, female mice overexpressing L-PK were unaffected. Mechanistically, L-PK altered mitochondrial pyruvate flux and its incorporation into citrate, and this, in turn, increased liver triglycerides via up-regulated de novo lipogenesis and increased PNPLA3 levels accompanied by mitochondrial dysfunction. Also, L-PK increased plasma cholesterol levels via increased PCSK9 levels. On the other hand, L-PK silencing reduced de novo lipogenesis and PNPLA3 and PCSK9 levels and improved mitochondrial function. Finally, in fibrosis model, we demonstrate that L-PK silencing in male mice reduced both liver steatosis and fibrosis, accompanied by reduced de novo lipogenesis and improved mitochondrial function. CONCLUSIONS L-PK acts in a male-specific manner in the development of liver steatosis and fibrosis. Because NAFLD/nonalcoholic steatohepatitis exhibit sexual dimorphism, our results have important implications for the development of personalized therapeutics.
Collapse
Affiliation(s)
- Karthickeyan Chella Krishnan
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, California,Correspondence Address correspondence to: Karthickeyan Chella Krishnan, PhD, UCLA Department of Medicine/Division of Cardiology, 650 Charles E. Young Drive South, Box 951679, Los Angeles, California 90095-1679. fax: (310) 794-7345, or
| | - Raquel R. Floyd
- Department of Biology, University of California, Los Angeles, California
| | - Simon Sabir
- Department of Psychology, University of California, Los Angeles, California
| | - Dulshan W. Jayasekera
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California
| | - Paola V. Leon-Mimila
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, California,Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Unidad de Genómica de Poblaciones Aplicada a la Salud, Mexico City, Mexico
| | - Anthony E. Jones
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | - Angel A. Cortez
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | - Varun Shravah
- Department of Chemistry, University of California, Los Angeles, California
| | - Miklós Péterfy
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, California,Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, California
| | - Linsey Stiles
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, California
| | - Samuel Canizales-Quinteros
- Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Unidad de Genómica de Poblaciones Aplicada a la Salud, Mexico City, Mexico
| | - Ajit S. Divakaruni
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | - Adriana Huertas-Vazquez
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, California
| | - Aldons J. Lusis
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, California,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California,Department of Human Genetics, University of California, Los Angeles, California,Aldons J. Lusis, PhD, UCLA Department of Medicine/Division of Cardiology, 650 Charles E. Young Drive South, Box 951679, Los Angeles, California 90095-1679.
| |
Collapse
|
17
|
Franco J, Rajwa B, Ferreira CR, Sundberg JP, HogenEsch H. Lipidomic Profiling of the Epidermis in a Mouse Model of Dermatitis Reveals Sexual Dimorphism and Changes in Lipid Composition before the Onset of Clinical Disease. Metabolites 2020; 10:metabo10070299. [PMID: 32708296 PMCID: PMC7408197 DOI: 10.3390/metabo10070299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 02/07/2023] Open
Abstract
Atopic dermatitis (AD) is a multifactorial disease associated with alterations in lipid composition and organization in the epidermis. Multiple variants of AD exist with different outcomes in response to therapies. The evaluation of disease progression and response to treatment are observational assessments with poor inter-observer agreement highlighting the need for molecular markers. SHARPIN-deficient mice (Sharpincpdm) spontaneously develop chronic proliferative dermatitis with features similar to AD in humans. To study the changes in the epidermal lipid-content during disease progression, we tested 72 epidermis samples from three groups (5-, 7-, and 10-weeks old) of cpdm mice and their WT littermates. An agnostic mass-spectrometry strategy for biomarker discovery termed multiple-reaction monitoring (MRM)-profiling was used to detect and monitor 1,030 lipid ions present in the epidermis samples. In order to select the most relevant ions, we utilized a two-tiered filter/wrapper feature-selection strategy. Lipid categories were compressed, and an elastic-net classifier was used to rank and identify the most predictive lipid categories for sex, phenotype, and disease stages of cpdm mice. The model accurately classified the samples based on phospholipids, cholesteryl esters, acylcarnitines, and sphingolipids, demonstrating that disease progression cannot be defined by one single lipid or lipid category.
Collapse
Affiliation(s)
- Jackeline Franco
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA;
| | - Bartek Rajwa
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: (B.R.); (H.H.)
| | - Christina R. Ferreira
- Metabolite Profiling Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA;
| | | | - Harm HogenEsch
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA;
- Purdue Institute of Inflammation, Immunology and Infectious Diseases, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: (B.R.); (H.H.)
| |
Collapse
|
18
|
Chella Krishnan K, Sabir S, Shum M, Meng Y, Acín-Pérez R, Lang JM, Floyd RR, Vergnes L, Seldin MM, Fuqua BK, Jayasekera DW, Nand SK, Anum DC, Pan C, Stiles L, Péterfy M, Reue K, Liesa M, Lusis AJ. Sex-specific metabolic functions of adipose Lipocalin-2. Mol Metab 2019; 30:30-47. [PMID: 31767179 PMCID: PMC6812340 DOI: 10.1016/j.molmet.2019.09.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/04/2019] [Accepted: 09/22/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Lipocalin-2 (LCN2) is a secreted protein involved in innate immunity and has also been associated with several cardiometabolic traits in both mouse and human studies. However, the causal relationship of LCN2 to these traits is unclear, and most studies have examined only males. METHODS Using adeno-associated viral vectors we expressed LCN2 in either adipose or liver in a tissue specific manner on the background of a whole-body Lcn2 knockout or wildtype mice. Metabolic phenotypes including body weight, body composition, plasma and liver lipids, glucose homeostasis, insulin resistance, mitochondrial phenotyping, and metabolic cage studies were monitored. RESULTS We studied the genetics of LCN2 expression and associated clinical traits in both males and females in a panel of 100 inbred strains of mice (HMDP). The natural variation in Lcn2 expression across the HMDP exhibits high heritability, and genetic mapping suggests that it is regulated in part by Lipin1 gene variation. The correlation analyses revealed striking tissue dependent sex differences in obesity, insulin resistance, hepatic steatosis, and dyslipidemia. To understand the causal relationships, we examined the effects of expression of LCN2 selectively in liver or adipose. On a Lcn2-null background, LCN2 expression in white adipose promoted metabolic disturbances in females but not males. It acted in an autocrine/paracrine manner, resulting in mitochondrial dysfunction and an upregulation of inflammatory and fibrotic genes. On the other hand, on a null background, expression of LCN2 in liver had no discernible impact on the traits examined despite increasing the levels of circulating LCN2 more than adipose LCN2 expression. The mechanisms underlying the sex-specific action of LCN2 are unclear, but our results indicate that adipose LCN2 negatively regulates its receptor, LRP2 (or megalin), and its repressor, ERα, in a female-specific manner and that the effects of LCN2 on metabolic traits are mediated in part by LRP2. CONCLUSIONS Following up on our population-based studies, we demonstrate that LCN2 acts in a highly sex- and tissue-specific manner in mice. Our results have important implications for human studies, emphasizing the importance of sex and the tissue source of LCN2.
Collapse
Affiliation(s)
| | - Simon Sabir
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Michaël Shum
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Yonghong Meng
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Rebeca Acín-Pérez
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Jennifer M Lang
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Raquel R Floyd
- Department of Biology, University of California, Los Angeles, CA, USA
| | - Laurent Vergnes
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Marcus M Seldin
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Brie K Fuqua
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Dulshan W Jayasekera
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Sereena K Nand
- Department of Biology, University of California, Los Angeles, CA, USA
| | - Diana C Anum
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Miklós Péterfy
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA; Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Karen Reue
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Marc Liesa
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Pujol-Lereis LM. Alteration of Sphingolipids in Biofluids: Implications for Neurodegenerative Diseases. Int J Mol Sci 2019; 20:ijms20143564. [PMID: 31330872 PMCID: PMC6678458 DOI: 10.3390/ijms20143564] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/14/2022] Open
Abstract
Sphingolipids (SL) modulate several cellular processes including cell death, proliferation and autophagy. The conversion of sphingomyelin (SM) to ceramide and the balance between ceramide and sphingosine-1-phosphate (S1P), also known as the SL rheostat, have been associated with oxidative stress and neurodegeneration. Research in the last decade has focused on the possibility of targeting the SL metabolism as a therapeutic option; and SL levels in biofluids, including serum, plasma, and cerebrospinal fluid (CSF), have been measured in several neurodegenerative diseases with the aim of finding a diagnostic or prognostic marker. Previous reviews focused on results from diseases such as Alzheimer's Disease (AD), evaluated total SL or species levels in human biofluids, post-mortem tissues and/or animal models. However, a comprehensive review of SL alterations comparing results from several neurodegenerative diseases is lacking. The present work compiles data from circulating sphingolipidomic studies and attempts to elucidate a possible connection between certain SL species and neurodegeneration processes. Furthermore, the effects of ceramide species according to their acyl-chain length in cellular pathways such as apoptosis and proliferation are discussed in order to understand the impact of the level alteration in specific species. Finally, enzymatic regulations and the possible influence of insulin resistance in the level alteration of SL are evaluated.
Collapse
Affiliation(s)
- Luciana M Pujol-Lereis
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas (CIDIE-CONICET), X5016DHK Córdoba, Argentina.
| |
Collapse
|
20
|
Identification of novel serum markers for the progression of coronary atherosclerosis in WHHLMI rabbits, an animal model of familial hypercholesterolemia. Atherosclerosis 2019; 284:18-23. [PMID: 30870703 DOI: 10.1016/j.atherosclerosis.2019.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIMS The development of serum markers specific for coronary lesions is important to prevent coronary events. However, analyses of serum markers in humans are affected by environmental factors and non-target diseases. Using an appropriate model animal can reduce these effects. To identify specific markers for coronary atherosclerosis, we comprehensively analyzed the serum of WHHLMI rabbits, which spontaneously develop coronary atherosclerosis. METHODS Female WHHLMI rabbits were fed standard chow. Serum and plasma were collected under fasting at intervals of 4 months from 4 months old, and a total of 313 lipid molecules, 59 metabolites, lipoprotein lipid levels, and various plasma biochemical parameters were analyzed. The severity of coronary lesions was evaluated with cross-sectional narrowing (CSN) corrected with a frequency of 75%-89% CSN and CSN> 90%. RESULTS There was a large variation in the severity of coronary lesions in WHHLMI rabbits despite almost no differences in plasma biochemical parameters and aortic lesion area between rabbits with severe and mild coronary lesions. The metabolites and lipid molecules selected as serum markers for coronary atherosclerosis were lysophosphatidylcholine (LPC) 22:4 and diacylglycerol 18:0-18:0 at 4 months old, LPC 20:4 (sn-2), ceramide d18:1-18:2, citric acid plus isocitric acid, and pyroglutamic acid at 8 months old, and phosphatidylethanolamine plasminogen 16:1p-22:2 at 16 months old. CONCLUSIONS These serum markers were coronary lesion-specific markers independent of cholesterol levels and aortic lesions and may be useful to detect patients who develop cardiovascular disease.
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Sex differences are pervasive in metabolic and cardiovascular traits, yet they have often been ignored in human and animal model research. Sex differences can arise from reversible hormonal effects, from irreversible organizational (developmental) processes, and from gene expression differences from the X and Y chromosomes. We briefly review our current understanding of the impact of these factors in metabolic traits and disorders, with an emphasis on the recent literature. RECENT FINDINGS Novel sex differences continue to be identified for metabolic and cardiovascular traits. For example, it is now clear that gut microbiota tend to differ between men and women, with potentially large implications for disease susceptibility. Also, tissue-specific gene regulation differs between men and women, contributing to differential metabolism. These new insights will open up personalized therapeutic avenues for cardiometabolic diseases. SUMMARY Sex differences in body fat distribution, glucose homeostasis, insulin signaling, ectopic fat accumulation, and lipid metabolism during normal growth and in response to hormonal or nutritional imbalance are mediated partly through sex hormones and the sex chromosome complement. Most of these differences are mediated in a tissue-specific manner. Important future goals are to better understand the interactions between genetic variation and sex differences, and to bring an understanding of sex differences into clinical practice.
Collapse
Affiliation(s)
| | | | - Aldons J. Lusis
- Department of Medicine/Division of Cardiology
- Department of Micro-biology, Immunology and Molecular Genetics
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|