1
|
Rivera CN, Hinkle JS, Watne RM, Macgowan TC, Wommack AJ, Vaughan RA. PPAR β/ δ Agonism with GW501516 Increases Myotube PGC-1 α Content and Reduces BCAA Media Content Independent of Changes in BCAA Catabolic Enzyme Expression. PPAR Res 2023; 2023:4779199. [PMID: 37325367 PMCID: PMC10264138 DOI: 10.1155/2023/4779199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/01/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
Background Type 2 diabetes is characterized by reduced insulin sensitivity, elevated blood metabolites, and reduced mitochondrial metabolism with reduced expression of genes governing metabolism such as peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). PGC-1α regulates the expression of branched-chain amino acid (BCAA) metabolism, and thus, increased circulating BCAA in diabetics may be partially explained by reduced PGC-1α expression. PGC-1α functions in-part through interactions with peroxisome proliferator-activated receptor β/δ (PPARβ/δ). The present report examined the effects of the PPARβ/δ agonism on cell metabolism and related gene/protein expression of cultured myotubes, with a primary emphasis on determining the effects of GW on BCAA disposal and catabolic enzyme expression. Methods C2C12 myotubes were treated with GW501516 (GW) for up to 24 hours. Mitochondrial and glycolytic metabolism were measured via oxygen consumption and extracellular acidification rate, respectively. Metabolic gene and protein expression were assessed via quantitative real-time polymerase chain reaction (qRT-PCR) and western blot, respectively. Media BCAA content was assessed via liquid chromatography-mass spectrometry (LC/MS). Results GW significantly increased PGC-1α protein expression, mitochondrial content, and mitochondrial function. GW also significantly reduced BCAA content within culture media following 24-hour treatment; however, expression of BCAA catabolic enzymes/transporter was unchanged. Conclusion These data confirm the ability of GW to increase muscle PGC-1α content and decrease BCAA media content without affecting BCAA catabolic enzymes/transporter. These findings suggest heightened BCAA uptake (and possibly metabolism) may occur without substantial changes in the protein levels of related cell machinery.
Collapse
Affiliation(s)
- Caroline N. Rivera
- Department of Exercise Science, High Point University, High Point, NC, USA
| | - Jason S. Hinkle
- Department of Exercise Science, High Point University, High Point, NC, USA
| | - Rachel M. Watne
- Department of Chemistry, High Point University, High Point, NC, USA
| | | | | | - Roger A. Vaughan
- Department of Exercise Science, High Point University, High Point, NC, USA
| |
Collapse
|
2
|
Zhang X, Halberstam AA, Zhu W, Leitner BP, Thakral D, Bosenberg MW, Perry RJ. Isotope tracing reveals distinct substrate preference in murine melanoma subtypes with differing anti-tumor immunity. Cancer Metab 2022; 10:21. [PMID: 36457136 PMCID: PMC9714036 DOI: 10.1186/s40170-022-00296-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Research about tumor "metabolic flexibility"-the ability of cells to toggle between preferred nutrients depending on the metabolic context-has largely focused on obesity-associated cancers. However, increasing evidence for a key role for nutrient competition in the tumor microenvironment, as well as for substrate regulation of immune function, suggests that substrate metabolism deserves reconsideration in immunogenic tumors that are not strongly associated with obesity. METHODS We compare two murine models: immunologically cold YUMM1.7 and immunologically-hot YUMMER1.7. We utilize stable isotope and radioisotope tracer-based metabolic flux studies as well as gas and liquid chromatography-based metabolomics analyses to comprehensively probe substrate preference in YUMM1.7 and YUMMER1.7 cells, with a subset of studies on the impact of available metabolites across a panel of five additional melanoma cell lines. We analyze bulk RNA-seq data and identify increased expression of amino acid and glucose metabolism genes in YUMMER1.7. Finally, we analyze melanoma patient RNA-seq data to identify potential prognostic predictors rooted in metabolism. RESULTS We demonstrate using stable isotope tracer-based metabolic flux studies as well as gas and liquid chromatography-based metabolomics that immunologically-hot melanoma utilizes more glutamine than immunologically-cold melanoma in vivo and in vitro. Analyses of human melanoma RNA-seq data demonstrate that glutamine transporter and other anaplerotic gene expression positively correlates with lymphocyte infiltration and function. CONCLUSIONS Here, we highlight the importance of understanding metabolism in non-obesity-associated cancers, such as melanoma. This work advances the understanding of the correlation between metabolism and immunogenicity in the tumor microenvironment and provides evidence supporting metabolic gene expression as potential prognostic factors of melanoma progression and may inform investigations of adjunctive metabolic therapy in melanoma. TRIAL REGISTRATION Deidentified data from The Cancer Genome Atlas were analyzed.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, P.O. Box 208026, 333 Cedar St., SHM BE36-B, New Haven, CT, 06520-8026, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Alexandra A Halberstam
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, P.O. Box 208026, 333 Cedar St., SHM BE36-B, New Haven, CT, 06520-8026, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Wanling Zhu
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, P.O. Box 208026, 333 Cedar St., SHM BE36-B, New Haven, CT, 06520-8026, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Brooks P Leitner
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, P.O. Box 208026, 333 Cedar St., SHM BE36-B, New Haven, CT, 06520-8026, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Durga Thakral
- Department of Pathology, Yale School of Medicine, New Haven, USA
| | - Marcus W Bosenberg
- Department of Pathology, Yale School of Medicine, New Haven, USA
- Department of Dermatology, Yale School of Medicine, New Haven, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Yale Cancer Center, New Haven, CT, USA
- Yale Center for Immuno-Oncology, New Haven, CT, USA
| | - Rachel J Perry
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, P.O. Box 208026, 333 Cedar St., SHM BE36-B, New Haven, CT, 06520-8026, USA.
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA.
| |
Collapse
|
3
|
Urso C, Zhou H. Palmitic acid-induced defects in cell cycle progression and cytokinesis in Neuro-2a cells. Cell Cycle 2022; 21:1048-1057. [PMID: 35171079 PMCID: PMC9037450 DOI: 10.1080/15384101.2022.2040769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Obesity is associated with elevated levels of free fatty acids (FFAs). Excessive saturated fatty acids (SFAs) exhibit significant deleterious cytotoxic effects in many types of cells. However, the effects of palmitic acid (PA), the most common circulating SFA, on cell cycle progression in neuronal cells have not been well-examined. The aim of this study was to examine whether PA affects the proliferation and cell cycle progression in mouse neuroblastoma Neuro-2a (N2a) cells. Our studies found that 200 µM PA significantly decreased DNA synthesis and mitotic index in N2a cells as early as 4 h following treatment. 24 h treatment with 200 µM PA significantly decreased the percentage of diploid (2 N) cells while dramatically increasing the percentage of tetraploid (4 N) cells as compared to the BSA control. Moreover, our studies found that 24 h treatment with 200 µM PA increased the percentage of binucleate cells as compared to the BSA control. Our studies also found that unsaturated fatty acids (UFAs), including linoleic acid, oleic acid, α-linolenic acid, and docosahexaenoic acid, were able to abolish PA-induced decrease of 2 N cells, increase of 4 N cells, and accumulation of binucleate cells. Taken together, these results suggest that PA may affect multiple aspects of the cell cycle progression in N2a cells, including decreased DNA synthesis, G2/M arrest, and cytokinetic failure, which could be abolished by UFAs.Abbreviations: 4-PBA, 4-Phenylbutyric Acid; ALA, α-linolenic acid; BrdU, 5-bromo-2'-deoxyuridine; DAPI, 4',6-diamidino-2-phenylindole; ER, endoplasmic reticulum; FFA, free fatty acids; FITC, fluorescein isothiocyanate; LA, linoleic acid; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; N2a, Neuro-2a; NAC, N-acetyl cysteine; OA, oleic acid; PA, palmitic acid; pHH3, Phosphorylation of histone H3; PI, propidium iodide; SFA, saturated fatty acids; PUFA, polyunsaturated fatty acids; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; UFA, unsaturated fatty acids.
Collapse
Affiliation(s)
- C.J. Urso
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA
| | - Heping Zhou
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA,CONTACT Heping Zhou Department of Biological Sciences, Seton Hall University, South Orange, NJ07079, USA
| |
Collapse
|
4
|
Vishnyakova KS, Popov KV, Pan X, Jasko MV, Yegorov YE. Long-Chain Free Fatty Acids Influence Lipid Accumulation, Lysosome Activation and Glycolytic Shift in Various Cells In Vitro. Mol Biol 2021. [DOI: 10.1134/s0026893321030146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
5
|
Urso C, Zhou H. Palmitic Acid Lipotoxicity in Microglia Cells Is Ameliorated by Unsaturated Fatty Acids. Int J Mol Sci 2021; 22:ijms22169093. [PMID: 34445796 PMCID: PMC8396597 DOI: 10.3390/ijms22169093] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 01/04/2023] Open
Abstract
Obesity and metabolic syndrome are associated with cognitive decline and dementia. Palmitic acid (PA) is increased in the cerebrospinal fluid of obese patients with cognitive impairment. This study was therefore designed to examine fatty acid (FA) lipotoxicity in BV2 microglia cells. We found that PA induced time- and dose-dependent decrease in cell viability and increase in cell death without affecting the cell cycle profile and that PA lipotoxicity did not depend on cell surface free fatty acid receptors but rather on FA uptake. Treatment with sulfosuccinimidyl oleate (SSO), an irreversible inhibitor of fatty acid translocase CD36, significantly inhibited FA uptake in BSA- and PA-treated cells and blocked PA-induced decrease in cell viability. Inhibition of ER stress or treatment with N-acetylcysteine was not able to rescue PA lipotoxicity. Our study also showed that unsaturated fatty acids (UFAs), such as linoleic acid (LA), oleic acid (OA), α-linolenic acid (ALA), and docosahexaenoic acid (DHA), were not lipotoxic but instead protected microglia against PA-induced decrease in cell viability. Co-treatment of PA with LA, OA, and DHA significantly inhibited FA uptake in PA-treated cells. All UFAs tested induced the incorporation of FAs into and the amount of neutral lipids, while PA did not significantly affect the amount of neutral lipids compared with BSA control.
Collapse
|
6
|
Maternal high-fat-diet exposure is associated with elevated blood pressure and sustained increased leptin levels through epigenetic memory in offspring. Sci Rep 2021; 11:316. [PMID: 33431976 PMCID: PMC7801715 DOI: 10.1038/s41598-020-79604-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
Maternal metabolism dysregulation during pregnancy predisposes offspring to major diseases, including hypertension, in later life, but the mechanism involved remains to be fully elucidated. A high-fat-diet (HFD) pregnant rat model was used to investigate whether excessive intrauterine lipid exposure was associated with elevated blood pressure in offspring and increased levels of leptin, an important biomarker and mediator of vascular dysfunction and hypertension. We found that gestational hyperlipidemia predisposed offspring to blood pressure elevation and sustained increases in leptin levels with no difference in body weight in the rat model. Increased leptin expression and leptin promoter hypomethylation were found in adipose tissues of HFD-exposed offspring. The treatment of mesenchymal stem cells with free fatty acids during adipogenic differentiation resulted in increased leptin expression, accompanied by leptin promoter hypomethylation. In addition, we also followed up 121 children to evaluate the association between maternal triglyceride levels and offspring blood pressure. Consistent with the animal study results, we observed elevated serum leptin levels and blood pressure in the offspring born to women with gestational hypertriglyceridemia. Our findings provide new insights that maternal hyperlipidemia is associated with elevated blood pressure in offspring and is associated with increases in leptin levels through epigenetic memory.
Collapse
|
7
|
Pavlović K, Lalić N. Cell models for studying muscle insulin resistance. MEDICINSKI PODMLADAK 2021. [DOI: 10.5937/mp72-31381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Type 2 diabetes is one of the most prevalent chronic diseases in the world today. Insulin resistance - a reduced responsiveness of tissues to insulin - is a hallmark of type 2 diabetes pathology. Skeletal muscle plays a pivotal role in glucose homeostasis - it is responsible for the majority of insulin-mediated glucose disposal and thus is one of the tissues most affected by insulin resistance. To study the molecular mechanisms of a disease, researchers often turn to cell models - they are inexpensive, easy to use, and exist in a controlled environment with few unknown variables. Cell models for exploring muscle insulin resistance are constructed using primary cell cultures or immortalised cell lines and treating them with fatty acids, high insulin or high glucose concentrations. The choice of cell culture, concentration and duration of the treatment and the methods for measuring insulin sensitivity, in order to confirm the model, are rarely discussed. Choosing an appropriate and physiologically relevant model for a particular topic of interest is required in order for the results to be reproducible, relevant, comparable and translatable to more complex biological systems. Cell models enable research that would otherwise be inaccessible but, especially when studying human disease, they do not serve a purpose if they are not in line with the biological reality. This review aims to summarise and critically evaluate the most commonly used cell models of muscle insulin resistance: the rationale for choosing these exact treatments and conditions, the protocols for constructing the models and the measurable outcomes used for confirming insulin resistance in the cells.
Collapse
|
8
|
Oleic acid up-regulates myosin heavy chain (MyHC) 1 expression and increases mitochondrial mass and maximum respiration in C2C12 myoblasts. Biochem Biophys Res Commun 2020; 525:406-411. [PMID: 32093891 DOI: 10.1016/j.bbrc.2020.02.099] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/15/2020] [Indexed: 11/20/2022]
Abstract
Skeletal muscle is divided into type 1 and type 2 fibers. Type 1 fibers are rich in mitochondria, have high oxidative metabolism, and are resistant to fatigue. Muscle-specific overexpression of peroxisome proliferator-activated receptor (PPAR)δ drastically increases the number of type 1 fibers. We focused on oleic acid, an omega-9 monounsaturated fatty acid, as a factor that activates PPARδ. In this study, we examined the effects of oleic acid on the muscle fiber type of C2C12 myotubes and its relationship with PPARδ. Our results showed that oleic acid treatment increased the levels of myosin heavy chain (MyHC)1, a known type 1 fiber marker, as well as mitochondrial mass and maximum respiration in C2C12 cells. To confirm the relationship between PPARδ activation and oleic acid-induced MyHC1 increase, we examined the effects of oleic acid in PPARδ knockdown C2C12 myoblasts. We found that oleic acid supplementation increased the mRNA expression of MyHC1 in PPARδ-knockdown C2C12 cells. Our data suggest that oleic acid increases type 1 fiber levels in C2C12 myotubes in a PPARδ-independent manner.
Collapse
|
9
|
Miklas JW, Clark E, Levy S, Detraux D, Leonard A, Beussman K, Showalter MR, Smith AT, Hofsteen P, Yang X, Macadangdang J, Manninen T, Raftery D, Madan A, Suomalainen A, Kim DH, Murry CE, Fiehn O, Sniadecki NJ, Wang Y, Ruohola-Baker H. TFPa/HADHA is required for fatty acid beta-oxidation and cardiolipin re-modeling in human cardiomyocytes. Nat Commun 2019; 10:4671. [PMID: 31604922 PMCID: PMC6789043 DOI: 10.1038/s41467-019-12482-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial trifunctional protein deficiency, due to mutations in hydratase subunit A (HADHA), results in sudden infant death syndrome with no cure. To reveal the disease etiology, we generated stem cell-derived cardiomyocytes from HADHA-deficient hiPSCs and accelerated their maturation via an engineered microRNA maturation cocktail that upregulated the epigenetic regulator, HOPX. Here we report, matured HADHA mutant cardiomyocytes treated with an endogenous mixture of fatty acids manifest the disease phenotype: defective calcium dynamics and repolarization kinetics which results in a pro-arrhythmic state. Single cell RNA-seq reveals a cardiomyocyte developmental intermediate, based on metabolic gene expression. This intermediate gives rise to mature-like cardiomyocytes in control cells but, mutant cells transition to a pathological state with reduced fatty acid beta-oxidation, reduced mitochondrial proton gradient, disrupted cristae structure and defective cardiolipin remodeling. This study reveals that HADHA (tri-functional protein alpha), a monolysocardiolipin acyltransferase-like enzyme, is required for fatty acid beta-oxidation and cardiolipin remodeling, essential for functional mitochondria in human cardiomyocytes.
Collapse
Affiliation(s)
- Jason W Miklas
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Elisa Clark
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Shiri Levy
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA
| | - Damien Detraux
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA
| | - Andrea Leonard
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Kevin Beussman
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Megan R Showalter
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, 95616, USA
| | - Alec T Smith
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Peter Hofsteen
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Jesse Macadangdang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Tuula Manninen
- Helsinki University Hospital, 00290, Helsinki, Finland
- Research Programs Unit, Stem Cells and Metabolism, University of Helsinki, 00290, Helsinki, Finland
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Anup Madan
- Covance Genomics Laboratory, Redmond, WA, 98052, USA
| | - Anu Suomalainen
- Helsinki University Hospital, 00290, Helsinki, Finland
- Research Programs Unit, Stem Cells and Metabolism, University of Helsinki, 00290, Helsinki, Finland
- Neuroscience Center, University of Helsinki, 00290, Helsinki, Finland
| | - Deok-Ho Kim
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, 98109, USA
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, 95616, USA
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nathan J Sniadecki
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Hannele Ruohola-Baker
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA.
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
10
|
Analysis of the PPARD Gene Expression Level Changes in Football Players in Response to the Training Cycle. Balkan J Med Genet 2018; 21:19-25. [PMID: 30425906 PMCID: PMC6231314 DOI: 10.2478/bjmg-2018-0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The PPARD gene codes protein that belongs to the peroxisome proliferator-activated receptor (PPAR) family engaged in a variety of biological processes, including lipid metabolism in muscle cells. In this study, we assess the relationship between PPARD gene expression lipid metabolism parameters and the variation of the PPARD gene expression before (T1) and after 12 hours of training (T2) sessions in a group of football players. Peripheral blood lymphocytes were obtained from 22 football players (17.5±0.7 years, 178±0.7 cm, 68.05±9.18 kg). The PPARD gene expression, analyzed by quantitative polymerase chain reaction (qPCR), was significantly higher after T2 (p = 0.0006). Moreover, at the end of the training cycle, there was a significant decrease in relative fat tissue (FAT) (%) (p = 0.01) and absolute FAT (kg) (p = 0.01). A negative correlation was observed between absolute FAT (kg) and PPARD gene expression level in T2 (p = 0.03). The levels of cholesterol and triglyceride (TG) fractions were not significantly different (p >0.05) before and after training. No significant relationship between PPARD expression and cholesterol or TG levels was found. We found that physical training affects PPARD expression. Moreover, the negative correlation between PPARD expression and absolute FAT (kg) level may be indicative of the contribution of PPARD in metabolic adaptation to increased lipid uptake that can be used to control the body composition of athletes.
Collapse
|
11
|
Manuel CR, Charron MJ, Ashby CR, Reznik SE. Saturated and unsaturated fatty acids differentially regulate in vitro and ex vivo placental antioxidant capacity. Am J Reprod Immunol 2018; 80:e12868. [PMID: 29736947 DOI: 10.1111/aji.12868] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/05/2018] [Indexed: 12/22/2022] Open
Abstract
PROBLEM Complications from prematurity are the leading cause of death among children under 5 years of age. Although clinical studies have shown a positive correlation between maternal high-fat diet (HFD) and preterm birth (PTB), the underlying mechanisms remain to be elucidated. Furthermore, it remains unclear how fatty acid type influences the effects of bacterial endotoxins. METHOD OF STUDY HTR-8/SVneo trophoblasts were cultured in either 0.5 mmol L-1 palmitic acid (PA) or linoleic acid (LA) in the absence or presence of 100 μg mL-1 of lipopolysaccharide (LPS) or lipoteichoic acid (LTA). Murine placental explants were cultured in either 2 mmol L-1 PA or LA, and cell viability, total antioxidant capacity (TAC), lipid peroxidation, H2 O2 , heme oxygenase-1 (HO-1), and nuclear erythroid 2-related factor 2 (Nrf-2) and nuclear factor-kappa light-chain enhancer of activated B cells (NF-κB) transcription factor activity assays were assessed. RESULTS Palmitic acid significantly (i) increased cell death, (ii) decreased TAC, and (iii) increased lipid peroxidation; but did not significantly increase HO-1. In contrast, LA maintained cell viability and significantly increased TAC and HO-1. In addition, incubating placental explants with PA significantly increased NF-κB activity. Co-incubating cells with PA and LPS or LTA significantly potentiated H2 O2 production and increased lipid peroxidation. Co-incubating cells with PA and LTA synergistically impaired TAC, and LTA decreased TAC more so than LPS. Co-incubation with PA/LA and LPS/LTA decreased HO-1 levels compared to treatment with either fatty acid alone. CONCLUSION Our findings suggest that saturated and unsaturated fats differentially regulate placental viability, antioxidant capacity, and inflammation and the actions of gram-positive and gram-negative endotoxins.
Collapse
Affiliation(s)
- Clarence R Manuel
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | - Maureen J Charron
- Departments of Biochemistry, Obstetrics & Gynecology and Women's Health and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA.,Departments of Pathology and Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
12
|
Yang C, Lim W, Bazer FW, Song G. Down-regulation of stearoyl-CoA desaturase-1 increases susceptibility to palmitic-acid-induced lipotoxicity in human trophoblast cells. J Nutr Biochem 2017; 54:35-47. [PMID: 29242171 DOI: 10.1016/j.jnutbio.2017.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/12/2017] [Accepted: 11/11/2017] [Indexed: 01/22/2023]
Abstract
In early pregnancy, adequate dietary factors are important for the growth of human trophoblast cells, followed by placental development. Although stearoyl-CoA desaturase 1 (SCD1) is expected to relieve palmitic acid (PA)-induced lipotoxicity by regulating diacylglycerol and ceramide, its function is unclear in human trophoblast cells. The aim was to investigate inhibitory effects of SCD1 activity on PA-induced trophoblast cell death. PA induces cell death and inhibits the invasion of human trophoblast cells (HTR8/SVneo). In addition, we demonstrate that SCD1 has a protective role against PA in human trophoblast cells by regulating AKT-mediated signaling pathway and mitochondrial membrane potential. The knockdown of SCD1 enhances the proapoptotic activity of PA in HTR8/SVneo cells. Lastly, we investigated microRNA expression predicted to target SCD1 and diacylglycerol O-acyltransferase 1 (DGAT1) by PA. Collectively, the results suggest potential roles of SCD1 and DGAT1 in alleviating the toxicity of PA and maintaining lipid homeostasis for normal placentation.
Collapse
Affiliation(s)
- Changwon Yang
- Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea; Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Whasun Lim
- Department of Biomedical Sciences, Catholic Kwandong University, Gangneung, 25601, Republic of Korea
| | - Fuller W Bazer
- Center for Animal Biotechnology and Genomics and Department of Animal Science, Texas A&M University, College Station, 77843-2471, Texas, USA
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea; Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
13
|
Mao Y, Wang J, Yu F, Li Z, Li H, Guo C, Fan X. Ghrelin protects against palmitic acid or lipopolysaccharide-induced hepatocyte apoptosis through inhibition of MAPKs/iNOS and restoration of Akt/eNOS pathways. Biomed Pharmacother 2016; 84:305-313. [DOI: 10.1016/j.biopha.2016.09.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 01/24/2023] Open
|
14
|
Echeverría F, Ortiz M, Valenzuela R, Videla LA. Long-chain polyunsaturated fatty acids regulation of PPARs, signaling: Relationship to tissue development and aging. Prostaglandins Leukot Essent Fatty Acids 2016; 114:28-34. [PMID: 27926461 DOI: 10.1016/j.plefa.2016.10.001] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/04/2016] [Accepted: 10/05/2016] [Indexed: 12/31/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that function as ligand-dependent transcription factors that can be activated by different types of fatty acids (FAs). Three isoforms of PPARs have been identify, namely, PPARα, PPARβ/δ, and PPARγ, which are able to bind long-chain polyunsaturated FAs (LCPUFAs), n-3 LCPUFAs being bound with greater affinity to achieve activation. FA binding induces a conformational change of the nuclear receptors, triggering the transcription of specific genes including those encoding for various metabolic and cellular processes such as FA β-oxidation and adipogenesis, thus representing key mediators of lipid homeostasis. In addition, PPARs have important roles during placental, embryonal, and fetal development, and in the regulation of processes related to aging comprising oxidative stress, inflammation, and neuroprotection. The aim of this review was to assess the role of FAs as PPARs ligands, in terms of their main functions associated with FA metabolism and their relevance in the prevention and treatment of related pathologies during human life span.
Collapse
Affiliation(s)
| | - Macarena Ortiz
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|