1
|
Li X, Hegarty K, Lin F, Chang JL, Abdalla A, Dhanabalan K, Solomevich SO, Song W, Roder K, Yao C, Lu W, Carmeliet P, Choudhary G, Dennery PA, Yao H. Endothelial Cpt1a Inhibits Neonatal Hyperoxia-Induced Pulmonary Vascular Remodeling by Repressing Endothelial-Mesenchymal Transition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415824. [PMID: 39799584 PMCID: PMC11923872 DOI: 10.1002/advs.202415824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/15/2024] [Indexed: 01/15/2025]
Abstract
Pulmonary hypertension (PH) increases the mortality of preterm infants with bronchopulmonary dysplasia (BPD). There are no curative therapies for this disease. Lung endothelial carnitine palmitoyltransferase 1a (Cpt1a), the rate-limiting enzyme of the carnitine shuttle system, is reduced in a rodent model of BPD. It is unknown whether endothelial Cpt1a reduction causes pulmonary vascular (PV) remodeling. The latter can be the result of endothelial-mesenchymal transition (EndoMT). Here, endothelial cell (EC)-specific Cpt1a KO and WT mice (<12 h old) are exposed to hyperoxia (70% O2) for 14 days and allow them to recover in normoxia until postnatal day 28. Hyperoxia causes PH, which is aggravated in EC-specific Cpt1a KO mice. Upregulating endothelial Cpt1a expression inhibits hyperoxia-induced PV remodeling. Hyperoxia causes lung EndoMT, detected by immunofluorescence, scRNA-sequencing, and EC lineage tracing, which is further increased in EC-specific Cpt1a KO mice. Blocking EndoMT inhibits hyperoxia-induced PV remodeling. Male mice under the same high oxygen conditions develop a higher degree of PH than females, which is associated with reduced endothelial Cpt1a expression. Conclusively, neonatal hyperoxia causes PH by decreasing endothelial Cpt1a expression and upregulating EndoMT. This provides a valuable strategy for developing targeted therapies by upregulating endothelial Cpt1a levels or inhibiting EndoMT to treat BPD-associated PH.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
- College of PharmacyJinan UniversityGuangzhouGuangdong510632China
| | - Katy Hegarty
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
| | - Fanjie Lin
- State Key Laboratory of Respiratory DiseaseGuangdong Key Laboratory of Vascular DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510120China
| | - Jason L. Chang
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
| | - Amro Abdalla
- Providence VA Medical CenterProvidenceRI02908USA
| | - Karthik Dhanabalan
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Sergey O. Solomevich
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Wenliang Song
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Karim Roder
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Chenrui Yao
- College of Arts & SciencesBoston UniversityBostonMA02215USA
| | - Wenju Lu
- State Key Laboratory of Respiratory DiseaseGuangdong Key Laboratory of Vascular DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510120China
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular MetabolismDepartment of Oncology and Leuven Cancer InstituteKU LeuvenVIB Center for Cancer Biology, VIBLeuvenBrussels3000Belgium
- Center for BiotechnologyKhalifa UniversityAbu Dhabi127788UAE
| | - Gaurav Choudhary
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Phyllis A. Dennery
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Department of PediatricsWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Hongwei Yao
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| |
Collapse
|
2
|
Rodríguez-Rodríguez R, Baena M, Zagmutt S, Paraiso WK, Reguera AC, Fadó R, Casals N. International Union of Basic and Clinical Pharmacology: Fundamental insights and clinical relevance regarding the carnitine palmitoyltransferase family of enzymes. Pharmacol Rev 2025; 77:100051. [PMID: 40106976 DOI: 10.1016/j.pharmr.2025.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
The carnitine palmitoyltransferases (CPTs) play a key role in controlling the oxidation of long-chain fatty acids and are potential therapeutic targets for diseases with a strong metabolic component, such as obesity, diabetes, and cancer. Four distinct proteins are CPT1A, CPT1B, CPT1C, and CPT2, differing in tissue expression and catalytic activity. CPT1s are finely regulated by malonyl-CoA, a metabolite whose intracellular levels reflect the cell's nutritional state. Although CPT1C does not exhibit significant catalytic activity, it is capable of modulating the functioning of other neuronal proteins. Structurally, all CPTs share a Y-shaped catalytic tunnel that allows the entry of 2 substrates and accommodation of the acyl group in a hydrophobic pocket. Several molecules targeting these enzymes have been described, some showing potential in normalizing blood glucose levels in diabetic patients, and others that, through a central mechanism, are anorexigenic and enhance energy expenditure. However, given the critical roles that CPTs play in certain tissues, such as the heart, liver, and brain, it is essential to fully understand the differences between the various isoforms. We analyze in detail the structure of these proteins, their cellular and physiological functions, and their potential as therapeutic targets in diseases such as obesity, diabetes, and cancer. We also describe drugs identified to date as having inhibitory or activating capabilities for these proteins. This knowledge will support the design of new drugs specific to each isoform, and the development of nanomedicines that can selectively target particular tissues or cells. SIGNIFICANCE STATEMENT: Carnitine palmitoyltransferase (CPT) proteins, as gatekeepers of fatty acid oxidation, have great potential as pharmacological targets to treat metabolic diseases like obesity, diabetes, and cancer. In recent years, significant progress has been made in understanding the 3-dimensional structure of CPTs and their pathophysiological functions. A deeper understanding of the differences between the various CPT family members will enable the design of selective drugs and therapeutic approaches with fewer side effects.
Collapse
Affiliation(s)
- Rosalía Rodríguez-Rodríguez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel Baena
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Sebastián Zagmutt
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - West Kristian Paraiso
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Ana Cristina Reguera
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Rut Fadó
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Núria Casals
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
de la Serna-Soto M, Calleros L, Martos-Elvira M, Moreno-Piedra A, García-Villoria S, Griera M, Alcalde-Estévez E, Asenjo-Bueno A, Rodríguez-Puyol D, de Frutos S, Ruiz-Torres MP. Integrin-Linked Kinase (ILK) Promotes Mitochondrial Dysfunction by Decreasing CPT1A Expression in a Folic Acid-Based Model of Kidney Disease. Int J Mol Sci 2025; 26:1861. [PMID: 40076489 PMCID: PMC11899702 DOI: 10.3390/ijms26051861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Integrin-linked kinase (ILK) is a key scaffolding protein between extracellular matrix protein and the cytoskeleton and has been implicated previously in the pathogenesis of renal damage. However, its involvement in renal mitochondrial dysfunction remains to be elucidated. We studied the role of ILK and its downstream regulations in renal damage and mitochondria function both in vivo and vitro, using a folic acid (FA)-induced kidney disease model. Wild type (WT) and ILK conditional-knockdown (cKD-ILK) mice were injected with a single intraperitoneal dose of FA and studied after 15 days of chronic renal damage progression. Human Kidney tubular epithelial cells (HK2) were transfected with specific siRNAs targeting ILK, glycogen synthase kinase 3-β (GSK3β), or CCAAT/enhancer binding protein-β (C/EBPβ). The expressions and activities of renal ILK, GSK3β, C/EBPβ, mitochondrial oxidative phosphorylation enzymes, and mitochondrial membrane potential were assessed. Additionally, the expression of markers for fibrosis fibronectin (FN) and collagen 1 (COL1A1), for autophagy p62 and cytosolic light chain 3 (LC3B) isoforms II and I, and mitochondrial homeostasis marker carnitine palmitoyl-transferase 1A (CPT1A) were evaluated using immunoblotting, RT-qPCR, immunofluorescence, or colorimetric assays. FA upregulated ILK expression, leading to the decrease of GSK3β activity, increased tubular fibrosis, and produced mitochondrial dysfunction, both in vivo and vitro. These alterations were fully or partially reversed upon ILK depletion, mitigating FA-induced renal damage. The signaling axis composed by ILK, GSK3β, and C/EBPβ regulated CPT1A transcription as the limiting factor in the FA-based impaired mitochondrial activity. We highlight ILK as a potential therapeutical target for preserving mitochondrial function in kidney injury.
Collapse
Affiliation(s)
- Mariano de la Serna-Soto
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Laura Calleros
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - María Martos-Elvira
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Ariadna Moreno-Piedra
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Sergio García-Villoria
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Mercedes Griera
- Graphenano Medical Care S.L., Alcalá de Henares, 28871 Madrid, Spain;
| | - Elena Alcalde-Estévez
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Ana Asenjo-Bueno
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Diego Rodríguez-Puyol
- Department of Medicine, Universidad de Alcalá, Nephrology Service at Hospital Príncipe de Asturias, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain;
| | - Sergio de Frutos
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - María Piedad Ruiz-Torres
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| |
Collapse
|
4
|
Liao K, Liu K, Wang Z, Zhao K, Mei Y. TRIM2 promotes metabolic adaptation to glutamine deprivation via enhancement of CPT1A activity. FEBS J 2025; 292:275-293. [PMID: 38949993 DOI: 10.1111/febs.17218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 05/14/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Cancer cells undergo metabolic adaptation to promote their survival and growth under energy stress conditions, yet the underlying mechanisms remain largely unclear. Here, we report that tripartite motif-containing protein 2 (TRIM2) is upregulated in response to glutamine deprivation by the transcription factor cyclic AMP-dependent transcription factor (ATF4). TRIM2 is shown to specifically interact with carnitine O-palmitoyltransferase 1 (CPT1A), a rate-limiting enzyme of fatty acid oxidation. Via this interaction, TRIM2 enhances the enzymatic activity of CPT1A, thereby regulating intracellular lipid levels and protecting cells from glutamine deprivation-induced apoptosis. Furthermore, TRIM2 is able to promote both in vitro cell proliferation and in vivo xenograft tumor growth via CPT1A. Together, these findings establish TRIM2 as an important regulator of the metabolic adaptation of cancer cells to glutamine deprivation and implicate TRIM2 as a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Kaimin Liao
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiyue Liu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhongyu Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kailiang Zhao
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yide Mei
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
5
|
Wei J, Lv Q, Luan F, Zhang X, Guo D, Zhai B, Chen S, Zou J, Shi Y. Exploration of potential mechanism of Sanhua Jiangzhi granules for the treatment of hyperlipidemia based on network pharmacology and experimental verification. Fitoterapia 2024; 179:106271. [PMID: 39461567 DOI: 10.1016/j.fitote.2024.106271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Sanhua Jiangzhi Granules (SJG) is a traditional Chinese patent medicine known for regulating lipid metabolism. In this study, we utilized UPLC-TOF-MS to analyze the components of SJG and, in conjunction with network pharmacology, identified 125 core chemical constituents. These components were individually queried and intersected with targets related to hyperlipidemia, resulting in the identification of 312 core targets. KEGG and GO analyses suggested that the mechanism of SJG in treating hyperlipidemia may primarily involve the PPAR signaling pathway. To further validate the efficacy and underlying signaling mechanisms of SJG, we conducted experiments using 60 rats. The results indicated that SJG significantly reduced body weight, lowered serum levels of total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C), while increasing high-density lipoprotein cholesterol (HDLC) levels. Enzyme-linked immunosorbent assay (ELISA) results demonstrated that SJG decreased hepatic TC and TG levels and mitigated lipid accumulation in the liver. Hematoxylin and eosin (HE) staining indicated that SJG improved liver pathological morphology and reduced the risk of fatty liver disease. Western blot analyses showed that treatment with SJG down-regulated the expression of stearoyl-CoA desaturase (SCD), 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), phospholipid transfer protein (PLTP), and fatty acid-binding protein 1 (FABP1), while up-regulating the expression of cholesterol 7α-hydroxylase (CYP7A1), carnitine palmitoyltransferase 1 (CPT-1), and PPARα by activating the PPAR signaling pathway. In conclusion, this study demonstrated that SJG activates the PPAR signaling pathway, leading to decreased body weight, lowered blood lipid levels, reduced hepatic TC and TG, and improved liver pathology in rats.
Collapse
Affiliation(s)
- Junfei Wei
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Qian Lv
- Weinan Testing Institute, 714000, China
| | - Fei Luan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiaofei Zhang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Dongyan Guo
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Bingtao Zhai
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Shucun Chen
- Shangluo Hospital of Traditional Chinese Medicine, Shangluo 726000, China
| | - Junbo Zou
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| | - Yajun Shi
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| |
Collapse
|
6
|
Basumatary D, Das S, Bidyarani Devi M, Shalini Devi G, Sarma P, Mukherjee AK, Khan MR, Borah JC. Garcinol enriched fraction of Garcinia morella (Gaertn.) Desr. fruit rind improves gut health and reduces the risk of nonalcoholic fatty liver disease by regulating PCK1/ACC/SREBP1/FASn pathway in a mouse model. Food Res Int 2024; 197:115285. [PMID: 39577934 DOI: 10.1016/j.foodres.2024.115285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/07/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a fast-emerging global burden, is an umbrella term for several liver manifestations that result in excessive accumulation of fat in the liver. NAFLD leads to gut microbiome dysbiosis, loss in gut epithelia, increased gut permeability, etc. The limited availability of registered drugs for NAFLD highlights the urgent need to focus on understanding its pathogenesis and discovering new treatments, including the potential exploration of herbal therapies for managing the condition. In this study, we evaluated the bioactive potential of garcinol enriched fraction from Garcinia morella fruit rind in preventing NAFLD-associated increased gut permeability. Administration of garcinol-enriched fraction (GEF) significantly reduced body weight, serum lipids (triglyceride and total cholesterol) levels, and enzymes (alkaline phosphatase and aspartate aminotransferase) responsible for liver dysfunction in high-fat diet (HFD)-fed C57BL/6 mice. GEF treatment also regulated the alteration in signaling pathways of lipid metabolism in HFD-fed mice by inhibiting the overexpression of genes involved in de novo lipogenesis. Mice treated with GEF had increased gut microbial diversity, reduced pathogenic bacteria, and increased Lactococcus and Streptococcaceae genera. Additionally, GEF treatment could increase the expression of intestinal tight junction proteins, which were otherwise decreased in HFD-fed mice, stipulating its protective effect in maintaining gut barrier integrity. Our study demonstrated that GEF treatment reduces obesity in mice and improves gut health by keeping junctions tight and maintaining a healthy gut microbiome.
Collapse
Affiliation(s)
- Devi Basumatary
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Paschim Boragaon, Guwahati 35, Assam, India; Department of Biotechnology, Gauhati University, Guwahati 781014, Assam, India
| | - Santanu Das
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Paschim Boragaon, Guwahati 35, Assam, India
| | - M Bidyarani Devi
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Paschim Boragaon, Guwahati 35, Assam, India; Department of Biotechnology, Gauhati University, Guwahati 781014, Assam, India
| | - G Shalini Devi
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Paschim Boragaon, Guwahati 35, Assam, India; Department of Biotechnology, Gauhati University, Guwahati 781014, Assam, India
| | - Pranamika Sarma
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Paschim Boragaon, Guwahati 35, Assam, India
| | - Ashis K Mukherjee
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Paschim Boragaon, Guwahati 35, Assam, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, India
| | - Mojibur R Khan
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Paschim Boragaon, Guwahati 35, Assam, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, India.
| | - Jagat C Borah
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Paschim Boragaon, Guwahati 35, Assam, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, India; Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati 781101, Assam, India.
| |
Collapse
|
7
|
Sedghi M, Javanmard F, Amoozmehr A, Zamany S, Mohammadi I, Kim W, Choppa VSR. Lysophospholipid Supplementation in Broiler Breeders' Diet Benefits Offspring's Productive Performance, Blood Parameters, and Hepatic β-Oxidation Genes. Animals (Basel) 2024; 14:3066. [PMID: 39518789 PMCID: PMC11545463 DOI: 10.3390/ani14213066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
The present study aimed to investigate whether supplementation of modified lysophospholipids (LPLs) in the diet of broiler breeders can benefit their offspring. A total of 264 49-week-old breeders (Ross 308) were allocated and fed based on a 2 × 2 factorial arrangement with two levels of dietary energy (normal energy = 2800 kcal/kg and low energy = 2760 kcal/kg) and two LPL levels (0 and 0.5 g/kg) for periods of 8 and 12 weeks. The offspring were assessed for growth performance, serum parameters, hepatic antioxidative capability, and expression of genes involved in liver β-oxidation at 7 days old. The LPL inclusion improved (p < 0.01) average body weight (ABW), average daily gain (ADG), and feed conversion ratio (FCR). The offspring of 61-week-old breeders fed with LPL exhibited reduced serum triglyceride levels (p < 0.01) but an increase in hepatic glutathione peroxidase (p < 0.05). The LPL increased (p < 0.001) the mRNA expression of the PGC-1α gene in the liver. Supplementing LPL in low-energy diets resulted in higher FABP1 gene expression (p < 0.05) in the intestine. In conclusion, LPL supplementation in the breeders' diet improved offspring performance by enhancing fatty acid absorption, hepatic indices, and the expression of genes involved in liver β-oxidation.
Collapse
Affiliation(s)
- Mohammad Sedghi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran; (F.J.); (S.Z.); (I.M.)
| | - Fatemeh Javanmard
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran; (F.J.); (S.Z.); (I.M.)
| | - Anvar Amoozmehr
- Department of Animal and Poultry Nutrition, Faculty of Animal Science, Gorgan University of Agricultural Sciences and Natural Resources, Shahid Beheshti Ave, Gorgan 49138-15739, Iran;
| | - Saeid Zamany
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran; (F.J.); (S.Z.); (I.M.)
| | - Ishmael Mohammadi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran; (F.J.); (S.Z.); (I.M.)
| | - Woo Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (W.K.); (V.S.R.C.)
| | | |
Collapse
|
8
|
Tan H, Mi N, Tong F, Zhang R, Abudurexiti A, Lei Y, Zhong Y, Yan J, Yang J, Ma X. Lactucopicrin promotes fatty acid β-oxidation and attenuates lipid accumulation through adenosine monophosphate-activated protein kinase activation in free fatty acid-induced human hepatoblastoma cancer cells. Food Sci Nutr 2024; 12:5357-5372. [PMID: 39139977 PMCID: PMC11317671 DOI: 10.1002/fsn3.4176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 08/15/2024] Open
Abstract
With its annually increasing prevalence, non-alcoholic fatty liver disease (NAFLD) has become a serious threat to people's life and health. After a preliminary research, we found that Lactucopicrin has pharmacological effects, such as lowering blood lipids and protecting the liver. Further research showed its significant activation for fatty acid β-oxidase hydroxyacyl-coenzyme A (CoA) dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA), so we hypothesized that Lactucopicrin could ameliorate lipid accumulation in hepatocytes by promoting fatty acid β-oxidation. In this study, free fatty acid (FFA)-induced human hepatoblastoma cancer cells (HepG2) were used to establish an in vitro NAFLD model to investigate the molecular basis of Lactucopicrin in regulating lipid metabolism. Staining with Oil red O and measurements of triglyceride (TG) content, fatty acid β-oxidase (FaβO) activity, reactive oxygen species (ROS) content, mitochondrial membrane potential, and adenosine triphosphate (ATP) content were used to assess the extent to which Lactucopicrin ameliorates lipid accumulation and promotes fatty acid β-oxidation. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot methods were used to explore the regulatory effects of Lactucopicrin on factors related to fatty acid β-oxidation. Results showed that Lactucopicrin downregulated phosphorylated mammalian target of rapamycin (P-mTOR) by activating the adenosine monophosphate-activated protein kinase (AMPK) pathway and upregulated the messenger RNA (mRNA) and protein expression levels of coactivators (peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α)), transcription factors (peroxisome proliferator-activated receptor α (PPARα) and peroxisome proliferator-activated receptor γ (PPARγ)), and oxidative factors (carnitine palmitoyltransferase 1A (CPT1A) and HADHA). This phenomenon resulted in a significant increase in FaβO activity, ATP content, and JC-1 and a significant decrease in ROS level, TG content, and intracellular lipid droplets. With the addition of Dorsomorphin, all the effects of Lactucopicrin intervention were suppressed. In summary, Lactucopicrin promotes fatty acid β-oxidation by activating the AMPK pathway, thereby ameliorating FFA-induced intracellular lipid accumulation in HepG2 cells.
Collapse
Affiliation(s)
- Huiwen Tan
- College of PharmacyXinjiang Medical UniversityUrumqiXinjiangChina
- Affiliated Hospital of Chongqing Three Gorges Medical CollegeChongqingChina
| | - Na Mi
- The First Affiliated Hospital of Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Fenglian Tong
- College of PharmacyXinjiang Medical UniversityUrumqiXinjiangChina
| | - Rui Zhang
- College of PharmacyXinjiang Medical UniversityUrumqiXinjiangChina
| | | | - Yi Lei
- College of PharmacyXinjiang Medical UniversityUrumqiXinjiangChina
| | - Yewei Zhong
- College of PharmacyXinjiang Medical UniversityUrumqiXinjiangChina
| | - Junlin Yan
- College of PharmacyXinjiang Medical UniversityUrumqiXinjiangChina
| | - Jian Yang
- College of PharmacyXinjiang Medical UniversityUrumqiXinjiangChina
| | - Xiaoli Ma
- College of PharmacyXinjiang Medical UniversityUrumqiXinjiangChina
| |
Collapse
|
9
|
Smith L, Santiago EG, Eke C, Gu W, Wang W, Llivichuzhca-Loja D, Kehoe T, St Denis K, Strine M, Taylor S, Tseng G, Konnikova L. Human Milk Supports Robust Intestinal Organoid Growth, Differentiation, and Homeostatic Cytokine Production. GASTRO HEP ADVANCES 2024; 3:1030-1042. [PMID: 39529649 PMCID: PMC11550179 DOI: 10.1016/j.gastha.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/11/2024] [Indexed: 11/16/2024]
Abstract
Background and Aims Necrotizing enterocolitis is a severe gastrointestinal complication of prematurity. Using small intestinal organoids derived from fetal tissue of a gestational age similar to an extremely preterm infant, this study aims to assess the effect of diet on intestinal epithelial growth and differentiation to elucidate the role nutrition type plays in intestinal development and modifies the risk for necrotizing enterocolitis. Methods Organoids were cultured for 5 days in growth media and 5 days in differentiation media supplemented 1:40 with 4 different diets: parental milk, donor human milk, standard formula, or extensively hydrolyzed formula. Images were captured daily and organoids were quantified. Organoids were preserved for RNA sequencing and immunofluorescence staining with Ki67, cleaved caspase 3, and chromogranin-A. Media was saved for cytokine/chemokine and growth factor analysis. Results Human milk supplementation improved growth and differentiation of intestinal organoids generating larger organoids during the growth phase and organoids with longer and wider buds during differentiation compared to formula. Ki67 staining confirmed the proliferative nature of milk-supplemented organoids and chromogranin A staining proved that MM-supplemented organoids induced highest enteroendocrine differentiation. Human milk supplementation also upregulated genes involved in Wnt signaling and fatty acid metabolism pathways and promoted a homeostatic immune landscape, including via increased secretion of tumor necrosis factor-related apoptosis-inducing ligand among other cytokines. Conversely, organoids supplemented with formula had a downregulation of cell-cycle-promoting genes and a more inflammatory immune signature, including a reduced level of leukemia inhibitory factor. Conclusion Our results demonstrate that parental milk, and to a lesser extent donor human milk, support robust intestinal epithelial proliferation, differentiation, and homeostatic cytokine production, suggesting a critical role for factors enriched in human milk in intestinal epithelial health.
Collapse
Affiliation(s)
- Lauren Smith
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | | | - Chino Eke
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Weihong Gu
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Wenjia Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Tessa Kehoe
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Kerri St Denis
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Madison Strine
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Sarah Taylor
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Liza Konnikova
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut
- Program in Translational Biomedicine, Yale School of Medicine, New Haven, Connecticut
- Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
10
|
Parafati M, La Russa D, Lascala A, Crupi F, Riillo C, Fotschki B, Mollace V, Janda E. Dramatic Suppression of Lipogenesis and No Increase in Beta-Oxidation Gene Expression Are among the Key Effects of Bergamot Flavonoids in Fatty Liver Disease. Antioxidants (Basel) 2024; 13:766. [PMID: 39061835 PMCID: PMC11273501 DOI: 10.3390/antiox13070766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 07/28/2024] Open
Abstract
Bergamot flavonoids have been shown to prevent metabolic syndrome, non-alcoholic fatty liver disease (NAFLD) and stimulate autophagy in animal models and patients. To investigate further the mechanism of polyphenol-dependent effects, we performed a RT2-PCR array analysis on 168 metabolism, transport and autophagy-related genes expressed in rat livers exposed for 14 weeks to different diets: standard, cafeteria (CAF) and CAF diet supplemented with 50 mg/kg of bergamot polyphenol fraction (BPF). CAF diet caused a strong upregulation of gluconeogenesis pathway (Gck, Pck2) and a moderate (>1.7 fold) induction of genes regulating lipogenesis (Srebf1, Pparg, Xbp1), lipid and cholesterol transport or lipolysis (Fabp3, Apoa1, Lpl) and inflammation (Il6, Il10, Tnf). However, only one β-oxidation gene (Cpt1a) and a few autophagy genes were differentially expressed in CAF rats compared to controls. While most of these transcripts were significantly modulated by BPF, we observed a particularly potent effect on lipogenesis genes, like Acly, Acaca and Fasn, which were suppressed far below the mRNA levels of control livers as confirmed by alternative primers-based RT2-PCR analysis and western blotting. These effects were accompanied by downregulation of pro-inflammatory cytokines (Il6, Tnfa, and Il10) and diabetes-related genes. Few autophagy (Map1Lc3a, Dapk) and no β-oxidation gene expression changes were observed compared to CAF group. In conclusion, chronic BPF supplementation efficiently prevents NAFLD by modulating hepatic energy metabolism and inflammation gene expression programs, with no effect on β-oxidation, but profound suppression of de novo lipogenesis.
Collapse
Affiliation(s)
- Maddalena Parafati
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, 88100 Catanzaro, Italy; (M.P.); (F.C.); (C.R.); (V.M.)
| | - Daniele La Russa
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy;
| | - Antonella Lascala
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, 88100 Catanzaro, Italy; (M.P.); (F.C.); (C.R.); (V.M.)
| | - Francesco Crupi
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, 88100 Catanzaro, Italy; (M.P.); (F.C.); (C.R.); (V.M.)
| | - Concetta Riillo
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, 88100 Catanzaro, Italy; (M.P.); (F.C.); (C.R.); (V.M.)
| | - Bartosz Fotschki
- Department of Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland;
| | - Vincenzo Mollace
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, 88100 Catanzaro, Italy; (M.P.); (F.C.); (C.R.); (V.M.)
| | - Elzbieta Janda
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, 88100 Catanzaro, Italy; (M.P.); (F.C.); (C.R.); (V.M.)
| |
Collapse
|
11
|
Theys C, Vanderhaeghen T, Van Dijck E, Peleman C, Scheepers A, Ibrahim J, Mateiu L, Timmermans S, Vanden Berghe T, Francque SM, Van Hul W, Libert C, Vanden Berghe W. Loss of PPARα function promotes epigenetic dysregulation of lipid homeostasis driving ferroptosis and pyroptosis lipotoxicity in metabolic dysfunction associated Steatotic liver disease (MASLD). FRONTIERS IN MOLECULAR MEDICINE 2024; 3:1283170. [PMID: 39086681 PMCID: PMC11285560 DOI: 10.3389/fmmed.2023.1283170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/14/2023] [Indexed: 08/02/2024]
Abstract
Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD) is a growing epidemic with an estimated prevalence of 20%-30% in Europe and the most common cause of chronic liver disease worldwide. The onset and progression of MASLD are orchestrated by an interplay of the metabolic environment with genetic and epigenetic factors. Emerging evidence suggests altered DNA methylation pattern as a major determinant of MASLD pathogenesis coinciding with progressive DNA hypermethylation and gene silencing of the liver-specific nuclear receptor PPARα, a key regulator of lipid metabolism. To investigate how PPARα loss of function contributes to epigenetic dysregulation in MASLD pathology, we studied DNA methylation changes in liver biopsies of WT and hepatocyte-specific PPARα KO mice, following a 6-week CDAHFD (choline-deficient, L-amino acid-defined, high-fat diet) or chow diet. Interestingly, genetic loss of PPARα function in hepatocyte-specific KO mice could be phenocopied by a 6-week CDAHFD diet in WT mice which promotes epigenetic silencing of PPARα function via DNA hypermethylation, similar to MASLD pathology. Remarkably, genetic and lipid diet-induced loss of PPARα function triggers compensatory activation of multiple lipid sensing transcription factors and epigenetic writer-eraser-reader proteins, which promotes the epigenetic transition from lipid metabolic stress towards ferroptosis and pyroptosis lipid hepatoxicity pathways associated with advanced MASLD. In conclusion, we show that PPARα function is essential to support lipid homeostasis and to suppress the epigenetic progression of ferroptosis-pyroptosis lipid damage associated pathways towards MASLD fibrosis.
Collapse
Affiliation(s)
- Claudia Theys
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tineke Vanderhaeghen
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Cedric Peleman
- Laboratory of Experimental Medicine and Pediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anne Scheepers
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Joe Ibrahim
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Ligia Mateiu
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Steven Timmermans
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sven M. Francque
- Laboratory of Experimental Medicine and Pediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Wim Van Hul
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wim Vanden Berghe
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
12
|
Wu XF, Liu Y, Zhan JS, Huang QL, Li WY. A novel splice variant of goat CPT1a gene and their diverse mRNA expression profiles. Anim Biotechnol 2023; 34:2571-2581. [PMID: 36047452 DOI: 10.1080/10495398.2022.2106573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
The Alternative splicing (AS) of Carnitine palmitoyltransferase 1a (CPT1a) and their expression profiles had never been illuminated in goats until now. Herein, a novel splice transcript in the CPT1a gene that is predicted to result in the skipping of exons 6-19 (CPT1a-sv1) has been isolated in addition to the full-length transcript in goats. The result of RT-PCR showed that CPT1a-sv1 is 606 bp in length and consists of 6 exons. A novel exon 6 was consisted of partial exon 5 and partial exon 19, compared to that in CPT1a. RT-qPCR analysis showed that the expression patterns of CPT1a and CPT1a-sv1 are spatially different. In both kid and adult goats, the CPT1a transcript is strongly expressed in the liver, spleen, lung, kidney, and brain tissues. However, CPT1a-sv1 has a strong tissue-specific expression pattern, with moderate RNA levels in the liver and brain of kids, while highly expressed in the liver and minimally expressed in the brain of adults. We observed two transcripts to be involved in brain development. These findings improve our understanding of the function of the CPT1a gene in goats and provide information on the molecular mechanism of AS events.
Collapse
Affiliation(s)
- Xian-Feng Wu
- Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Yuan Liu
- Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Jin-Shun Zhan
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi, China
| | - Qin-Lou Huang
- Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Wen-Yang Li
- Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| |
Collapse
|
13
|
Drake RR, Louey S, Thornburg KL. Maturation of lipid metabolism in the fetal and newborn sheep heart. Am J Physiol Regul Integr Comp Physiol 2023; 325:R809-R819. [PMID: 37867472 PMCID: PMC11178298 DOI: 10.1152/ajpregu.00122.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023]
Abstract
At birth, the fetus experiences a dramatic change in environment that is accompanied by a shift in myocardial fuel preference from lactate and glucose in fetal life to fatty acid oxidation after birth. We hypothesized that fatty acid metabolic machinery would mature during fetal life in preparation for this extreme metabolic transformation at birth. We quantified the pre- (94-day and 135-day gestation, term ∼147 days) and postnatal (5 ± 4 days postnatal) gene expression and protein levels for fatty acid transporters and enzymes in hearts from a precocial species, the sheep. Gene expression of fatty acid translocase (CD36), acyl-CoA synthetase long-chain 1 (ACSL1), carnitine palmitoyltransferase 1 (CPT1), hydroxy-acyl dehydrogenase (HADH), acetyl-CoA acetyltransferase (ACAT1), isocitrate dehydrogenase (IDH), and glycerol phosphate acyltransferase (GPAT) progressively increased through the perinatal period, whereas several genes [fatty acid transport protein 6 (FATP6), acyl-CoA synthetase long chain 3 (ACSL3), long-chain acyl-CoA dehydrogenase (LCAD), very long-chain acyl-CoA dehydrogenase (VLCAD), pyruvate dehydrogenase kinase (PDK4), phosphatidic acid phosphatase (PAP), and diacylglycerol acyltransferase (DGAT)] were stable in fetal hearts and had high expression after birth. Protein expression of CD36 and ACSL1 progressively increased throughout the perinatal period, whereas protein expression of carnitine palmitoyltransferase 1a (fetal isoform) (CPT1a) decreased and carnitine palmitoyltransferase 1b (adult isoform) (CPT1b) remained constitutively expressed. Using fluorescent-tagged long-chain fatty acids (BODIPY-C12), we demonstrated that fetal (125 ± 1 days gestation) cardiomyocytes produce 59% larger lipid droplets (P < 0.05) compared with newborn (8 ± 1 day) cardiomyocytes. These results provide novel insights into the perinatal maturation of cardiac fatty acid metabolism in a precocial species.NEW & NOTEWORTHY This study characterized the previously unknown expression patterns of genes that regulate the metabolism of free fatty acids in the perinatal sheep myocardium. This study shows that the prenatal myocardium prepares for the dramatic switch from carbohydrate metabolism to near complete reliance on free fatty acids postnatally. Fetal and neonatal cardiomyocytes also demonstrate differing lipid storage mechanisms where fetal cardiomyocytes form larger lipid droplets compared with newborn cardiomyocytes.
Collapse
Affiliation(s)
- Rachel R Drake
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Samantha Louey
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Kent L Thornburg
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
14
|
Griffin JD, Buxton JM, Culver JA, Barnes R, Jordan EA, White AR, Flaherty SE, Bernardo B, Ross T, Bence KK, Birnbaum MJ. Hepatic Activin E mediates liver-adipose inter-organ communication, suppressing adipose lipolysis in response to elevated serum fatty acids. Mol Metab 2023; 78:101830. [PMID: 38787338 PMCID: PMC10656223 DOI: 10.1016/j.molmet.2023.101830] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/21/2023] [Indexed: 05/25/2024] Open
Abstract
OBJECTIVE The liver is a central regulator of energy metabolism exerting its influence both through intrinsic processing of substrates such as glucose and fatty acid as well as by secreting endocrine factors, known as hepatokines, which influence metabolism in peripheral tissues. Human genome wide association studies indicate that a predicted loss-of-function variant in the Inhibin βE gene (INHBE), encoding the putative hepatokine Activin E, is associated with reduced abdominal fat mass and cardiometabolic disease risk. However, the regulation of hepatic Activin E and the influence of Activin E on adiposity and metabolic disease are not well understood. Here, we examine the relationship between hepatic Activin E and adipose metabolism, testing the hypothesis that Activin E functions as part of a liver-adipose, inter-organ feedback loop to suppress adipose tissue lipolysis in response to elevated serum fatty acids and hepatic fatty acid exposure. METHODS The relationship between hepatic Activin E and non-esterified fatty acids (NEFA) released from adipose lipolysis was assessed in vivo using fasted CL 316,243 treated mice and in vitro using Huh7 hepatocytes treated with fatty acids. The influence of Activin E on adipose lipolysis was examined using a combination of Inhbe knockout mice, a mouse model of hepatocyte-specific overexpression of Activin E, and mouse brown adipocytes treated with Activin E enriched media. RESULTS Increasing hepatocyte NEFA exposure in vivo by inducing adipose lipolysis through fasting or CL 316,243 treatment increased hepatic Inhbe expression. Similarly, incubation of Huh7 human hepatocytes with fatty acids increased expression of INHBE. Genetic ablation of Inhbe in mice increased fasting circulating NEFA and hepatic triglyceride accumulation. Treatment of mouse brown adipocytes with Activin E conditioned media and overexpression of Activin E in mice suppressed adipose lipolysis and reduced serum FFA levels, respectively. The suppressive effects of Activin E on lipolysis were lost in CRISPR-mediated ALK7 deficient cells and ALK7 kinase deficient mice. Disruption of the Activin E-ALK7 signaling axis in Inhbe KO mice reduced adiposity upon HFD feeding, but caused hepatic steatosis and insulin resistance. CONCLUSIONS Taken together, our data suggest that Activin E functions as part of a liver-adipose feedback loop, such that in response to increased serum free fatty acids and elevated hepatic triglyceride, Activin E is released from hepatocytes and signals in adipose through ALK7 to suppress lipolysis, thereby reducing free fatty acid efflux to the liver and preventing excessive hepatic lipid accumulation. We find that disrupting this Activin E-ALK7 inter-organ communication network by ablation of Inhbe in mice increases lipolysis and reduces adiposity, but results in elevated hepatic triglyceride and impaired insulin sensitivity. These results highlight the liver-adipose, Activin E-ALK7 signaling axis as a critical regulator of metabolic homeostasis.
Collapse
Affiliation(s)
- John D Griffin
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA.
| | - Joanne M Buxton
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Jeffrey A Culver
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Robert Barnes
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Emily A Jordan
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Alexis R White
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Stephen E Flaherty
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Barbara Bernardo
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Trenton Ross
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Kendra K Bence
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| | - Morris J Birnbaum
- Internal Medicine Research Unit, Pfizer Inc.,1 Portland Street, Cambridge, MA 02139, USA
| |
Collapse
|
15
|
Tang Y, Zhang W, Wang Y, Li H, Zhang C, Wang Y, Lin Y, Shi H, Xiang H, Huang L, Zhu J. Expression Variation of CPT1A Induces Lipid Reconstruction in Goat Intramuscular Precursor Adipocytes. Int J Mol Sci 2023; 24:13415. [PMID: 37686221 PMCID: PMC10488119 DOI: 10.3390/ijms241713415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Intramuscular fat (IMF) deposition is one of the most important factors affecting meat quality and is closely associated with the expression of carnitine palmitoyl transferase 1A (CPT1A) which facilitates the transfer of long-chain fatty acids (LCFAs) into the mitochondria. However, the role of how CPT1A regulates the IMF formation remains unclear. Herein, we established the temporal expression profile of CPT1A during the differentiation of goat intramuscular precursor adipocytes. Functionally, the knockdown of CPT1A by siRNA treatment significantly increased the mRNA expression of adipogenic genes and promoted lipid deposition in goat intramuscular precursor adipocytes. Meanwhile, a CPT1A deficiency inhibited cell proliferation and promoted cell apoptosis significantly. CPT1A was then supported by the overexpression of CPT1A which significantly suppressed the cellular triglyceride deposition and promoted cell proliferation although the cell apoptosis also was increased. For RNA sequencing, a total of 167 differential expression genes (DEGs), including 125 upregulated DEGs and 42 downregulated DEGs, were observed after the RNA silencing of CPT1A compared to the control, and were predicted to enrich in the focal adhesion pathway, cell cycle, apoptosis and the MAPK signaling pathway by KEGG analysis. Specifically, blocking the MAPK signaling pathway by a specific inhibitor (PD169316) rescued the promotion of cell proliferation in CPT1A overexpression adipocytes. In conclusion, the expression variation of CPT1A may reconstruct the lipid distribution between cellular triglyceride deposition and cell proliferation in goat intramuscular precursor adipocyte. Furthermore, we demonstrate that CPT1A promotes the proliferation of goat adipocytes through the MAPK signaling pathway. This work widened the genetic regulator networks of IMF formation and delivered theoretical support for improving meat quality from the aspect of IMF deposition.
Collapse
Affiliation(s)
- Yinmei Tang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Wenyang Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| | - Yinggui Wang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Haiyang Li
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Changhui Zhang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Yong Wang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| | - Yaqiu Lin
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| | - Hengbo Shi
- College of Animal Science, Zhejiang University, Hangzhou 310058, China;
| | - Hua Xiang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| | - Lian Huang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Jiangjiang Zhu
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| |
Collapse
|
16
|
Su Y, Deng C, Liu X, Lian J. Epigenetic Histone Methylation of PPARγ and CPT1A Signaling Contributes to Betahistine Preventing Olanzapine-Induced Dyslipidemia. Int J Mol Sci 2023; 24:ijms24119143. [PMID: 37298094 DOI: 10.3390/ijms24119143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/08/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023] Open
Abstract
As a partial histamine H1 receptor agonist and H3 antagonist, betahistine has been reported to partially prevent olanzapine-induced dyslipidemia and obesity through a combination therapy, although the underlying epigenetic mechanisms are still not known. Recent studies have revealed that histone regulation of key genes for lipogenesis and adipogenesis in the liver is one of the crucial mechanisms for olanzapine-induced metabolic disorders. This study investigated the role of epigenetic histone regulation in betahistine co-treatment preventing dyslipidemia and fatty liver caused by chronic olanzapine treatment in a rat model. In addition to abnormal lipid metabolism, the upregulation of peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein (C/EBPα), as well as the downregulation of carnitine palmitoyltransferase 1A (CPT1A) in the liver induced by olanzapine, were significantly attenuated by betahistine co-treatment. In addition, betahistine co-treatment significantly enhanced the global expression of H3K4me and the enrichment of H3K4me binding on the promoter of Cpt1a gene as revealed by ChIP-qPCR, but inhibited the expression of one of its site-specific demethylases, lysine (K)-specific demethylase 1A (KDM1A). Betahistine co-treatment also significantly enhanced the global expression of H3K9me and the enrichment of H3K9me binding on the promoter of the Pparg gene, but inhibited the expression of two of its site-specific demethylases, lysine demethylase 4B (KDM4B) and PHD finger protein 2 (PHF2). These results suggest that betahistine attenuates abnormal adipogenesis and lipogenesis triggered by olanzapine through modulating hepatic histone methylation, and thus inhibiting the PPARγ pathway-mediated lipid storage, while at the same time promoting CP1A-mediated fatty acid oxidation.
Collapse
Affiliation(s)
- Yueqing Su
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynaecology and Paediatrics, Fujian Medical University, Fuzhou 350005, China
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Chao Deng
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Xuemei Liu
- School of Pharmaceutical Sciences, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Jiamei Lian
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
17
|
Zhang N, Tian X, Yan T, Wang H, Zhang D, Lin C, Liu Q, Jiang S. Insights into the role of nucleotide methylation in metabolic-associated fatty liver disease. Front Immunol 2023; 14:1148722. [PMID: 37020540 PMCID: PMC10067741 DOI: 10.3389/fimmu.2023.1148722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/22/2023] [Indexed: 04/07/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a chronic liver disease characterized by fatty infiltration of the liver. In recent years, the MAFLD incidence rate has risen and emerged as a serious public health concern. MAFLD typically progresses from the initial hepatocyte steatosis to steatohepatitis and then gradually advances to liver fibrosis, which may ultimately lead to cirrhosis and carcinogenesis. However, the potential evolutionary mechanisms still need to be clarified. Recent studies have shown that nucleotide methylation, which was directly associated with MAFLD's inflammatory grading, lipid synthesis, and oxidative stress, plays a crucial role in the occurrence and progression of MAFLD. In this review, we highlight the regulatory function and associated mechanisms of nucleotide methylation modification in the progress of MAFLD, with a particular emphasis on its regulatory role in the inflammation of MAFLD, including the regulation of inflammation-related immune and metabolic microenvironment. Additionally, we summarize the potential value of nucleotide methylation in the diagnosis and treatment of MAFLD, intending to provide references for the future investigation of MAFLD.
Collapse
Affiliation(s)
- Ni Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinchen Tian
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tinghao Yan
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haochen Wang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Dengtian Zhang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Cong Lin
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Qingbin Liu
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Qingbin Liu, ; Shulong Jiang,
| | - Shulong Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Qingbin Liu, ; Shulong Jiang,
| |
Collapse
|
18
|
Effect of green cardamom on the expression of genes implicated in obesity and diabetes among obese women with polycystic ovary syndrome: a double blind randomized controlled trial. GENES & NUTRITION 2022; 17:17. [PMID: 36522620 PMCID: PMC9753872 DOI: 10.1186/s12263-022-00719-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 11/22/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is an endocrine disease in which related to obesity, metabolic disorders and is considered as one of the main causes of infertility in women. This trial was investigated the effects of green cardamom on the expression of genes implicated in obesity and diabetes among obese women with PCOS. METHODS One hundred ninety-four PCOS women were randomly divided two groups: intervention (n = 99; 3 g/day green cardamom) and control groups (n = 95). All of them were given low calorie diet. Anthropometric, glycemic and androgen hormones were assessed before and after 16-week intervention. The reverse transcription-polymerase chain reaction (RT-PCR) method was used to measure fat mass and obesity-associated (FTO), peroxisome proliferative activating receptor- (PPAR-), carnitine palmitoyltransferase 1A (CPT1A), acetyl-CoA carboxylase beta (ACAB), leptin receptor (LEPR), ghrelin, and lamin A/C (LAMIN) genes expression in each group. RESULTS Anthropometric indices were significantly decreased after intervention in both two studied groups. Glycemic indices and androgen hormones were significantly improved in the intervention group compared to the control group. The expression levels of FTO, CPT1A, LEPR, and LAMIN were significantly downregulated compared to control group (P < 0.001), as well as, PPAR-y was significantly upregulated in the intervention group after intervention with green cardamom compared to control group (P < 0.001). CONCLUSION This current study showed that the administration of green cardamom is a beneficial approach for improving anthropometric, glycemic, and androgen hormones, as well as obesity and diabetes genes expression in PCOS women under the low-calorie diet. TRIAL REGISTRATION This trial was registered with the Iranian Clinical Trials Registry (registration number: IRCT20200608047697N1). 1 August, 2020; https://www.irct.ir/trial/48748.
Collapse
|
19
|
PPARα in the Epigenetic Driver Seat of NAFLD: New Therapeutic Opportunities for Epigenetic Drugs? Biomedicines 2022; 10:biomedicines10123041. [PMID: 36551797 PMCID: PMC9775974 DOI: 10.3390/biomedicines10123041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing epidemic and the most common cause of chronic liver disease worldwide. It consists of a spectrum of liver disorders ranging from simple steatosis to NASH which predisposes patients to further fibrosis, cirrhosis and even hepatocarcinoma. Despite much research, an approved treatment is still lacking. Finding new therapeutic targets has therefore been a main priority. Known as a main regulator of the lipid metabolism and highly expressed in the liver, the nuclear receptor peroxisome proliferator-activated receptor-α (PPARα) has been identified as an attractive therapeutic target. Since its expression is silenced by DNA hypermethylation in NAFLD patients, many research strategies have aimed to restore the expression of PPARα and its target genes involved in lipid metabolism. Although previously tested PPARα agonists did not ameliorate the disease, current research has shown that PPARα also interacts and regulates epigenetic DNMT1, JMJD3, TET and SIRT1 enzymes. Moreover, there is a growing body of evidence suggesting the orchestrating role of epigenetics in the development and progression of NAFLD. Therefore, current therapeutic strategies are shifting more towards epigenetic drugs. This review provides a concise overview of the epigenetic regulation of NAFLD with a focus on PPARα regulation and highlights recently identified epigenetic interaction partners of PPARα.
Collapse
|
20
|
Chang JL, Gong J, Rizal S, Peterson AL, Chang J, Yao C, Dennery PA, Yao H. Upregulating carnitine palmitoyltransferase 1 attenuates hyperoxia-induced endothelial cell dysfunction and persistent lung injury. Respir Res 2022; 23:205. [PMID: 35964084 PMCID: PMC9375342 DOI: 10.1186/s12931-022-02135-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a chronic lung disease in premature infants that may cause long-term lung dysfunction. Accumulating evidence supports the vascular hypothesis of BPD, in which lung endothelial cell dysfunction drives this disease. We recently reported that endothelial carnitine palmitoyltransferase 1a (Cpt1a) is reduced by hyperoxia, and that endothelial cell-specific Cpt1a knockout mice are more susceptible to developing hyperoxia-induced injury than wild type mice. Whether Cpt1a upregulation attenuates hyperoxia-induced endothelial cell dysfunction and lung injury remains unknown. We hypothesized that upregulation of Cpt1a by baicalin or L-carnitine ameliorates hyperoxia-induced endothelial cell dysfunction and persistent lung injury. METHODS Lung endothelial cells or newborn mice (< 12 h old) were treated with baicalin or L-carnitine after hyperoxia (50% and 95% O2) followed by air recovery. RESULTS We found that incubation with L-carnitine (40 and 80 mg/L) and baicalin (22.5 and 45 mg/L) reduced hyperoxia-induced apoptosis, impaired cell migration and angiogenesis in cultured lung endothelial cells. This was associated with increased Cpt1a gene expression. In mice, neonatal hyperoxia caused persistent alveolar and vascular simplification in a concentration-dependent manner. Treatment with L-carnitine (150 and 300 mg/kg) and baicalin (50 and 100 mg/kg) attenuated neonatal hyperoxia-induced alveolar and vascular simplification in adult mice. These effects were diminished in endothelial cell-specific Cpt1a knockout mice. CONCLUSIONS Upregulating Cpt1a by baicalin or L-carnitine ameliorates hyperoxia-induced lung endothelial cell dysfunction, and persistent alveolar and vascular simplification. These findings provide potential therapeutic avenues for using L-carnitine and baicalin as Cpt1a upregulators to prevent persistent lung injury in premature infants with BPD.
Collapse
Affiliation(s)
- Jason L Chang
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Jiannan Gong
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
- Department of Respiratory Medicine, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Salu Rizal
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Abigail L Peterson
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Julia Chang
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Chenrui Yao
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Phyllis A Dennery
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Hongwei Yao
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA.
| |
Collapse
|
21
|
Moody L, Xu GB, Pan YX, Chen H. Genome-wide cross-cancer analysis illustrates the critical role of bimodal miRNA in patient survival and drug responses to PI3K inhibitors. PLoS Comput Biol 2022; 18:e1010109. [PMID: 35639779 PMCID: PMC9187341 DOI: 10.1371/journal.pcbi.1010109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/10/2022] [Accepted: 04/15/2022] [Indexed: 11/24/2022] Open
Abstract
Heterogeneity of cancer means many tumorigenic genes are only aberrantly expressed in a subset of patients and thus follow a bimodal distribution, having two modes of expression within a single population. Traditional statistical techniques that compare sample means between cancer patients and healthy controls fail to detect bimodally expressed genes. We utilize a mixture modeling approach to identify bimodal microRNA (miRNA) across cancers, find consistent sources of heterogeneity, and identify potential oncogenic miRNA that may be used to guide personalized therapies. Pathway analysis was conducted using target genes of the bimodal miRNA to identify potential functional implications in cancer. In vivo overexpression experiments were conducted to elucidate the clinical importance of bimodal miRNA in chemotherapy treatments. In nine types of cancer, tumors consistently displayed greater bimodality than normal tissue. Specifically, in liver and lung cancers, high expression of miR-105 and miR-767 was indicative of poor prognosis. Functional pathway analysis identified target genes of miR-105 and miR-767 enriched in the phosphoinositide-3-kinase (PI3K) pathway, and analysis of over 200 cancer drugs in vitro showed that drugs targeting the same pathway had greater efficacy in cell lines with high miR-105 and miR-767 levels. Overexpression of the two miRNA facilitated response to PI3K inhibitor treatment. We demonstrate that while cancer is marked by considerable genetic heterogeneity, there is between-cancer concordance regarding the particular miRNA that are more variable. Bimodal miRNA are ideal biomarkers that can be used to stratify patients for prognosis and drug response in certain types of cancer. Bimodal genes can be defined as those having two modes of expression within the same population. A variety of statistical methodologies have been employed to assess bimodal gene expression, but current methods and their applications have been limited. Given the advances in next-generation sequencing as well as the extensive regulatory role of miRNA, assessing bimodality in miRNA-seq data can greatly broaden our understanding of factors underlying tumor progression. The goal of the current study was to utilize a novel mixture modeling approach to identify bimodal miRNA and then demonstrate their importance in cancer by evaluating their ability to predict overall survival and drug response. Our results showed that high levels of bimodal miRNA expression was characteristic of cancer. Additionally, several bimodal miRNA were common to multiple cancer types, suggesting that certain miRNA consistently account for tumor heterogeneity and may be involved in general oncogenic processes. Our study points to the potential of bimodal miRNA to facilitate precise prognostic evaluation and effective treatment strategies.
Collapse
Affiliation(s)
- Laura Moody
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Guanying Bianca Xu
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Hong Chen
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
22
|
Zhang G, Xu Y, Xia Y, Wang G, Zhao H. Transcriptomic Analysis of Hepatotoxicology of Adult Zebrafish (Danio rerio) Exposed to Environmentally Relevant Oxytetracycline. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2022; 82:539-550. [PMID: 35460351 DOI: 10.1007/s00244-022-00930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
The extensive use of the broad-spectrum antibiotics like oxytetracycline (OTC) has become a serious environmental issue globally. OTC has profound negative effects on aquatic organisms including fishes. In this study, RNA-Seq analysis was employed to examine the possible molecular mechanism of hepatotoxicology in zebrafish induced by OTC exposure. Adult male zebrafish was exposed to 0, 5, 90, and 450 μg/L OTC for 3 weeks. The results showed the decrease in body weight and tail length but the increase in total length of zebrafish under OTC exposure in a dose-dependent way. In addition, severe histopathological damages were featured by increasing tissue vacuolization in the livers of 450 μg/L OTC group. Moreover, RNA-Seq analysis revealed that molecular signaling and functional pathways in the liver were disrupted by OTC exposure. Furthermore, the down-regulation of gene expression after OTC exposure was found on both the genes related to fatty acid degradation and the genes related to lipid synthesis. The present study implied that OTC induced liver malfunction and fish health risks through growth retard, histopathological damages, molecular signaling disruption, genetic expression alteration, and lipid metabolism disturbance.
Collapse
Affiliation(s)
- Gaixia Zhang
- Collge of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, Shaanxi, China
| | - Yifan Xu
- AP Center, Changzhou Senior High School of Jiangsu Province, No. 8, Luohan Road, Tianning District, Changzhou, 213004, Jiangsu, China
| | - Youran Xia
- Changzhou No. 2 High School, No. 32, Xiheng Street, Zhonglou District, Changzhou, 213001, Jiangsu, China
| | - Gang Wang
- AP Center, Changzhou Senior High School of Jiangsu Province, No. 8, Luohan Road, Tianning District, Changzhou, 213004, Jiangsu, China
| | - Hongfeng Zhao
- Collge of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, Shaanxi, China.
| |
Collapse
|
23
|
Ando Y, Yamada H, Munetsuna E, Yamazaki M, Kageyama I, Teshigawara A, Nouchi Y, Fujii R, Mizuno G, Sadamoto N, Ishikawa H, Suzuki K, Hashimoto S, Ohashi K. Maternal High-Fructose Corn Syrup consumption causes insulin resistance and hyperlipidemia in offspring via DNA methylation of the Pparα promoter region. J Nutr Biochem 2022; 103:108951. [PMID: 35123000 DOI: 10.1016/j.jnutbio.2022.108951] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/16/2021] [Accepted: 01/04/2022] [Indexed: 01/21/2023]
Abstract
PURPOSE There are concerns about the negative effects of fructose intake during pregnancy on the next generation. We have previously reported that offspring from dams fed with fructose during gestation and lactation demonstrate abnormal lipid metabolism in the liver. In this study, we aimed to elucidate the molecular mechanism of the effects of maternal high-fructose corn syrup (HFCS) consumption on offspring. BASIC PROCEDURES Pregnant Sprague-Dawley rats were fed with 20% HFCS water solution during gestation and lactation. Offspring were put on a normal diet after weaning, and the serum parameters and gene expression patterns were studied at predetermined intervals. MAIN FINDINGS Offsprings from pregnant rats fed with 20% HFCS (HFCS group) developed insulin resistance and hyperlipidemia at 60 days of age. RNA-seq analysis demonstrated that peroxisome proliferator-activated receptor α (PPARα) expression is downregulated by maternal HFCS intake. Hepatic Pparα expression in the HFCS group appeared to be suppressed by the enhanced DNA methylation of its promoter region. PRINCIPAL CONCLUSIONS It is suggested that the development of insulin resistance and hyperlipidemia in the HFCS group may be attributable to aberrant Pparα methylation in the offspring liver. Pparα hypermethylation may be one of molecular mechanism underlying the toxicity of maternal fructose intake.
Collapse
Affiliation(s)
- Yoshitaka Ando
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Hiroya Yamada
- Department of Hygiene, Fujita Health University School of Medicine, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan.
| | - Eiji Munetsuna
- Department of Biochemistry, Fujita Health University School of Medicine, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Mirai Yamazaki
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, 281-1, Murechohara, Takamatsu, Kagawa 761-0123 Japan
| | - Itsuki Kageyama
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Atsushi Teshigawara
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University Hospital, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Yuki Nouchi
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Ryosuke Fujii
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Genki Mizuno
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan; Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University Hospital, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Nao Sadamoto
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Hiroaki Ishikawa
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Koji Suzuki
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Shuji Hashimoto
- Department of Hygiene, Fujita Health University School of Medicine, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| | - Koji Ohashi
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192 Japan
| |
Collapse
|
24
|
Mahmoud AM. An Overview of Epigenetics in Obesity: The Role of Lifestyle and Therapeutic Interventions. Int J Mol Sci 2022; 23:ijms23031341. [PMID: 35163268 PMCID: PMC8836029 DOI: 10.3390/ijms23031341] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity has become a global epidemic that has a negative impact on population health and the economy of nations. Genetic predispositions have been demonstrated to have a substantial role in the unbalanced energy metabolism seen in obesity. However, these genetic variations cannot entirely explain the massive growth in obesity over the last few decades. Accumulating evidence suggests that modern lifestyle characteristics such as the intake of energy-dense foods, adopting sedentary behavior, or exposure to environmental factors such as industrial endocrine disruptors all contribute to the rising obesity epidemic. Recent advances in the study of DNA and its alterations have considerably increased our understanding of the function of epigenetics in regulating energy metabolism and expenditure in obesity and metabolic diseases. These epigenetic modifications influence how DNA is transcribed without altering its sequence. They are dynamic, reflecting the interplay between the body and its surroundings. Notably, these epigenetic changes are reversible, making them appealing targets for therapeutic and corrective interventions. In this review, I discuss how these epigenetic modifications contribute to the disordered energy metabolism in obesity and to what degree lifestyle and weight reduction strategies and pharmacological drugs can restore energy balance by restoring normal epigenetic profiles.
Collapse
Affiliation(s)
- Abeer M Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
25
|
Role of macronutrient intake in the epigenetics of obesity. Biochem Soc Trans 2022; 50:487-497. [PMID: 34994392 DOI: 10.1042/bst20211069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022]
Abstract
Obesity is caused by a combination of hereditary and environmental factors. Despite extensive study, contemporary through diet, exercise, education, surgery, and pharmacological treatments, no effective long-term solution has been found to this epidemic. Over the last decade, there has been a tremendous advancement in understanding the science of epigenetics, as well as a rise in public interest in learning more about the influence of diet and lifestyle choices on the health of an individual. Without affecting the underlying DNA sequence, epigenetic alterations impact gene expression. Previous animal studies have shown a link between the type of diet and expression or suppression of obesity genes, but there are very few human studies that demonstrate the relationship between dietary intake and obesity gene expression. This review highlights the effects of carbohydrates, lipids, and protein intake from the diet on obesity-related genes.
Collapse
|
26
|
Porcuna J, Mínguez-Martínez J, Ricote M. The PPARα and PPARγ Epigenetic Landscape in Cancer and Immune and Metabolic Disorders. Int J Mol Sci 2021; 22:ijms221910573. [PMID: 34638914 PMCID: PMC8508752 DOI: 10.3390/ijms221910573] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 02/07/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-modulated nuclear receptors that play pivotal roles in nutrient sensing, metabolism, and lipid-related processes. Correct control of their target genes requires tight regulation of the expression of different PPAR isoforms in each tissue, and the dysregulation of PPAR-dependent transcriptional programs is linked to disorders, such as metabolic and immune diseases or cancer. Several PPAR regulators and PPAR-regulated factors are epigenetic effectors, including non-coding RNAs, epigenetic enzymes, histone modifiers, and DNA methyltransferases. In this review, we examine advances in PPARα and PPARγ-related epigenetic regulation in metabolic disorders, including obesity and diabetes, immune disorders, such as sclerosis and lupus, and a variety of cancers, providing new insights into the possible therapeutic exploitation of PPAR epigenetic modulation.
Collapse
|
27
|
Dessì A, Bosco A, Pintus R, Picari G, Mazza S, Fanos V. Epigenetics and Modulations of Early Flavor Experiences: Can Metabolomics Contribute to Prevention during Weaning? Nutrients 2021; 13:nu13103351. [PMID: 34684350 PMCID: PMC8539480 DOI: 10.3390/nu13103351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 11/29/2022] Open
Abstract
The significant increase in chronic non-communicable diseases has changed the global epidemiological landscape. Among these, obesity is the most relevant in the pediatric field. This has pushed the world of research towards a new paradigm: preventive and predictive medicine. Therefore, the window of extreme plasticity that characterizes the first stage of development cannot be underestimated. In this context, nutrition certainly plays a primary role, being one of the most important epigenetic modulators known to date. Weaning, therefore, has a crucial role that must be analyzed far beyond the simple achievement of nutritional needs. Furthermore, the taste experience and the family context are fundamental for future food choices and can no longer be underestimated. The use of metabolomics allows, through the recognition of early disease markers and food-specific metabolites, the planning of an individualized and precise diet. In addition, the possibility of identifying particular groups of subjects at risk and the careful monitoring of adherence to dietary therapy may represent the basis for this change.
Collapse
|
28
|
Kong Y, Li Y, Dai Z, Qin M, Fan H, Hao J, Zhang C, Zhong Q, Qi C, Wang P. Glycosaminoglycan from Ostrea rivularis attenuates hyperlipidemia and regulates gut microbiota in high-cholesterol diet-fed zebrafish. Food Sci Nutr 2021; 9:5198-5210. [PMID: 34532028 PMCID: PMC8441474 DOI: 10.1002/fsn3.2492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/01/2021] [Accepted: 07/11/2021] [Indexed: 12/22/2022] Open
Abstract
Hyperlipidemia an immense group of acquired or genetic metabolic disorders that is characterized by an excess of lipids in the bloodstream. Altogether, they have a high prevalence worldwide and constitute a major threat to human health. Glycosaminoglycans (GAG) are natural biomolecules that have hypolipidemic activity. The purpose of this study was to investigate the potential hypolipidemic effect of glycosaminoglycans extracted from Ostrea rivularis (OGAG) on hyperlipidemic zebrafish, as well as the possible underlying mechanism of such effect. Dietary supplementation with OGAG during 4 weeks significantly reduced the serum and hepatic lipid levels and the hepatosomatic index in hyperlipidemic zebrafish. In addition, histopathological showed that OGAG supplementation decreases the volume and number of lipid droplets in hepatocytes. Transcriptome and real-time quantitative polymerase chain reaction analysis revealed that the gene expression levels of PPARγ, SCD, HMGRA, ACAT2, HMGCS, and HMGCR were significantly downregulated by OGAG treatment in hepatocytes, whereas those of CD36, FABP2, FABP6, ABCG5, and CYP7A1 were significantly upregulated. This suggests that the hypolipidemic effect of OGAG relies on increasing the ketogenic metabolism of fatty acids, inhibiting cholesterol synthesis, and enhancing the transformation of cholesterol to bile acid. Furthermore, OGAG treatment improved gut microbiota imbalance by reducing the Firmicutes-to-Bacteroidetes ratio, increasing the relative abundance of beneficial bacteria (Bacteroidetes, Verrucomicrobia, Acidobacteria, and Sphingomonas), and reducing the relative abundance of harmful bacteria (Proteobacteria, Cohaesibacter, Vibrio, and Terrisporobacter). These findings highlight the potential benefit of implementing OGAG as a dietary supplement to prevent and treat hyperlipidemia.
Collapse
Affiliation(s)
- Yan Kong
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity ConservationBeibu Gulf UniversityQinzhouChina
- College of Light Industry and Food EngineeringGuangxi UniversityNanningChina
| | - Ying Li
- Qinzhou Key Laboratory of Food Flavor Analysis and ControlBeibu Gulf UniversityQinzhouChina
| | - Zi‐Ru Dai
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity ConservationBeibu Gulf UniversityQinzhouChina
- Qinzhou Key Laboratory of Food Flavor Analysis and ControlBeibu Gulf UniversityQinzhouChina
| | - Mei Qin
- Qinzhou Key Laboratory of Food Flavor Analysis and ControlBeibu Gulf UniversityQinzhouChina
| | - He‐Liang Fan
- College of Light Industry and Food EngineeringGuangxi UniversityNanningChina
- Qinzhou Key Laboratory of Food Flavor Analysis and ControlBeibu Gulf UniversityQinzhouChina
| | - Jun‐Guang Hao
- Qinzhou Key Laboratory of Food Flavor Analysis and ControlBeibu Gulf UniversityQinzhouChina
| | - Chen‐Xiao Zhang
- Qinzhou Key Laboratory of Food Flavor Analysis and ControlBeibu Gulf UniversityQinzhouChina
| | - Qiu‐Ping Zhong
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity ConservationBeibu Gulf UniversityQinzhouChina
| | - Cen Qi
- Qinzhou Key Laboratory of Food Flavor Analysis and ControlBeibu Gulf UniversityQinzhouChina
| | - Pei Wang
- Qinzhou Key Laboratory of Food Flavor Analysis and ControlBeibu Gulf UniversityQinzhouChina
| |
Collapse
|
29
|
Wang L, Li X, Zhang G, Zhao H. Transcriptomic analysis of lead-induced hepatoxicology in female Japanese quails (Coturnix japonica): Implications of triglyceride synthesis, degradation and transport disruption. Comp Biochem Physiol C Toxicol Pharmacol 2021; 244:109024. [PMID: 33631343 DOI: 10.1016/j.cbpc.2021.109024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
Lead (Pb) pollution poses great threats to mammals including human and it is also hazardous to bird life. In this study, RNA sequencing analysis was employed to examine the molecular responses to lead exposure in the liver of a toxicological model species Japanese quails (Coturnix japonica). Female birds were exposed to 0, 50, 500 and 1000 ppm waterborne Pb for 49 days. The results showed that hepatic microstructure was damaged under lead exposure featured by sinusoids dilation and irregularity as well as cell necrosis. Moreover, ultrastructural injury in the liver including mitochondrial swelling and vacuolization as well as nuclear deformation was induced by lead exposure. Lead exposure also caused the decrease of lipid droplets in the liver by oil red O staining. In addition, liver transcriptomic analysis revealed that molecular signaling and functional pathways were disrupted by lead exposure. Meanwhile, the expression of genes involved with hepatic glycerophospholipids metabolism of triglyceride synthesis and lipid transport of triglyceride transfer was significantly down-regulated by lead exposure. Moreover, the up-regulation of genes associated with fatty acid oxidation and the down-regulation of genes related with fatty acid synthesis were caused by lead exposure. The present study implied that lead induced liver malfunction and bird health risks through histopathological damages, molecular signaling disruption, genetic expression alteration and triglyceride metabolism disturbance.
Collapse
Affiliation(s)
- Ling Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119 No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi 710119, China
| | - Xuan Li
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119 No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi 710119, China
| | - Gaixia Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119 No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi 710119, China
| | - Hongfeng Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119 No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
30
|
Shrestha N, Vidimce J, Holland OJ, Cuffe JSM, Beck BR, Perkins AV, McAinch AJ, Hryciw DH. Maternal and Postnatal High Linoleic Acid Diet Impacts Lipid Metabolism in Adult Rat Offspring in a Sex-Specific Manner. Int J Mol Sci 2021; 22:ijms22062946. [PMID: 33799409 PMCID: PMC7999727 DOI: 10.3390/ijms22062946] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
Linoleic acid (LA), an n-6 polyunsaturated fatty acid (PUFA), is essential for fetal growth and development. We aimed to investigate the effect of maternal and postnatal high LA (HLA) diet on plasma FA composition, plasma and hepatic lipids and genes involved in lipid metabolism in the liver of adult offspring. Female rats were fed with low LA (LLA; 1.44% LA) or HLA (6.21% LA) diets for 10 weeks before pregnancy, and during gestation/lactation. Offspring were weaned at postnatal day 25 (PN25), fed either LLA or HLA diets and sacrificed at PN180. Postnatal HLA diet decreased circulating total n-3 PUFA and alpha-linolenic acid (ALA), while increased total n-6 PUFA, LA and arachidonic acid (AA) in both male and female offspring. Maternal HLA diet increased circulating leptin in female offspring, but not in males. Maternal HLA diet decreased circulating adiponectin in males. Postnatal HLA diet significantly decreased aspartate transaminase (AST) in females and downregulated total cholesterol, HDL-cholesterol and triglycerides in the plasma of males. Maternal HLA diet downregulated the hepatic mRNA expression of Hmgcr in both male and female offspring and decreased the hepatic mRNA expression of Cpt1a and Acox1 in females. Both maternal and postnatal HLA diet decreased hepatic mRNA expression of Cyp27a1 in females. Postnatal diet significantly altered circulating fatty acid concentrations, with sex-specific differences in genes that control lipid metabolism in the adult offspring following exposure to high LA diet in utero.
Collapse
Affiliation(s)
- Nirajan Shrestha
- School of Medical Science, Griffith University, Gold Coast, QLD 4222, Australia; (N.S.); (J.V.); (O.J.H.); (A.V.P.)
| | - Josif Vidimce
- School of Medical Science, Griffith University, Gold Coast, QLD 4222, Australia; (N.S.); (J.V.); (O.J.H.); (A.V.P.)
| | - Olivia J. Holland
- School of Medical Science, Griffith University, Gold Coast, QLD 4222, Australia; (N.S.); (J.V.); (O.J.H.); (A.V.P.)
- Institute of Health and Biomedical Innovation, Queensland University of Technology, South Brisbane, QLD 4001, Australia
| | - James S. M. Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Belinda R. Beck
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- School of Allied Health Sciences, Griffith University, Gold Coast, QLD 4222, Australia
| | - Anthony V. Perkins
- School of Medical Science, Griffith University, Gold Coast, QLD 4222, Australia; (N.S.); (J.V.); (O.J.H.); (A.V.P.)
| | - Andrew J. McAinch
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, VIC 3021, Australia
| | - Deanne H. Hryciw
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
- Environmental Futures Research Institute, Griffith University, Nathan, QLD 4111, Australia
- Correspondence:
| |
Collapse
|
31
|
Wang L, McFadden JW, Yang G, Zhu H, Lian H, Fu T, Sun Y, Gao T, Li M. Effect of melatonin on visceral fat deposition, lipid metabolism and hepatic lipo-metabolic gene expression in male rats. J Anim Physiol Anim Nutr (Berl) 2021; 105:787-796. [PMID: 33486831 DOI: 10.1111/jpn.13497] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/15/2020] [Accepted: 12/16/2020] [Indexed: 12/23/2022]
Abstract
Melatonin (MT) influences lipid metabolism in animals; however, the mechanistic effect of melatonin on liver fat and abdominal adipose deposition requires further clarity. In order to study the effects of melatonin on lipid metabolism, and hepatic fat and abdominal adipose deposition in animals, twenty Sprague-Dawley (SD) rats of 6 weeks of age with similar bodyweight were randomly divided into two groups: control (CTL) and MT-treated (10 mg/kg/day). During a 60-day experiment, food intake and bodyweight were measured daily and weekly respectively. At the end of treatment, blood samples were collected to collect plasma to quantify hormones and metabolic indicators of lipid metabolism. In addition, organ and abdominal adipose depots including liver, and omental, perirenal, and epididymal fat were weighed. Liver tissue was sampled for sectioning, long-chain fatty acid (LCFA) quantification, and gene chip and Real-time quantitative PCR (qPCR) analyses. The results showed that liver weight and index (ratio of liver weight to body weight) in MT group reduced by 20.69% and 9.63% respectively; omentum weight and index reduced by 59.88% and 54.93% respectively, and epididymal fat weight reduced by 45.34% (p = 0.049), relative to CTL. Plasma lipid indices, triglyceride (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL) and total cholesterol (TC) with MT treatment decreased significantly compared with the control. Fat and 8 LCFA content in liver in MT group also decreased. Gene chip and qPCR demonstrated that there were 289 genes up-regulated and 293 genes down-regulated by MT. Further analysis found that the mRNA expression of lipolysis-related genes increased, while the mRNA expression of lipogenesis-related enzymes decreased (p < 0.05) with MT. This study concluded that melatonin greatly affected fat deposition, and hepatic LCFA supply and the expression of genes associated with lipogenesis and lipolysis.
Collapse
Affiliation(s)
- Linfeng Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | | | - Gaiqing Yang
- Modern Experimental Technique and Management Centre, Henan Agricultural University, Zhengzhou, Henan, China
| | - Heshui Zhu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Hongxia Lian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Tong Fu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Yu Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Tengyun Gao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
32
|
Dykstra H, Fisk C, LaRose C, Waldhart A, Meng X, Zhao G, Wu N. Mouse long-chain acyl-CoA synthetase 1 is active as a monomer. Arch Biochem Biophys 2021; 700:108773. [PMID: 33485846 DOI: 10.1016/j.abb.2021.108773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 11/25/2022]
Abstract
Fatty acids are essential cellular building blocks and a major energy source. Regardless of their metabolic fate, fatty acids first need to be activated by forming a thioester with a coenzyme A group. This reaction is carried out by acyl-CoA synthetases (ACSs), of which ACSL1 (long-chain acyl-CoA synthetase 1) is an important member. Two bacterial homologues of ACSL1 crystal structures have been solved previously. One is a soluble dimeric protein, and the other is a monomeric peripheral membrane protein. The mammalian ACSL1 is a membrane protein with an N-terminal transmembrane helix. To characterize the mammalian ACSL1, we purified the full-length mouse ACSL1 and reconstituted it into lipid nanodiscs. Using enzymatic assays, mutational analysis, and cryo-electron microscopy, we show that mouse ACSL1 is active as a monomer.
Collapse
Affiliation(s)
| | - Chelsea Fisk
- Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Cassi LaRose
- Van Andel Institute, Grand Rapids, MI, 49503, USA
| | | | - Xing Meng
- Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Gongpu Zhao
- Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Ning Wu
- Van Andel Institute, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
33
|
Lai CQ, Parnell LD, Smith CE, Guo T, Sayols-Baixeras S, Aslibekyan S, Tiwari HK, Irvin MR, Bender C, Fei D, Hidalgo B, Hopkins PN, Absher DM, Province MA, Elosua R, Arnett DK, Ordovas JM. Carbohydrate and fat intake associated with risk of metabolic diseases through epigenetics of CPT1A. Am J Clin Nutr 2020; 112:1200-1211. [PMID: 32930325 PMCID: PMC7657341 DOI: 10.1093/ajcn/nqaa233] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Epigenome-wide association studies identified the cg00574958 DNA methylation site at the carnitine palmitoyltransferase-1A (CPT1A) gene to be associated with reduced risk of metabolic diseases (hypertriglyceridemia, obesity, type 2 diabetes, hypertension, metabolic syndrome), but the mechanism underlying these associations is unknown. OBJECTIVES We aimed to elucidate whether carbohydrate and fat intakes modulate cg00574958 methylation and the risk of metabolic diseases. METHODS We examined associations between carbohydrate (CHO) and fat (FAT) intake, as percentages of total diet energy, and the CHO/FAT ratio with CPT1A-cg00574958, and the risk of metabolic diseases in 3 populations (Genetics of Lipid Lowering Drugs and Diet Network, n = 978; Framingham Heart Study, n = 2331; and REgistre GIroní del COR study, n = 645) while adjusting for confounding factors. To understand possible causal effects of dietary intake on the risk of metabolic diseases, we performed meta-analysis, CPT1A transcription analysis, and mediation analysis with CHO and FAT intakes as exposures and cg00574958 methylation as the mediator. RESULTS We confirmed strong associations of cg00574958 methylation with metabolic phenotypes (BMI, triglyceride, glucose) and diseases in all 3 populations. Our results showed that CHO intake and CHO/FAT ratio were positively associated with cg00574958 methylation, whereas FAT intake was negatively correlated with cg00574958 methylation. Meta-analysis further confirmed this strong correlation, with β = 58.4 ± 7.27, P = 8.98 x 10-16 for CHO intake; β = -36.4 ± 5.95, P = 9.96 x 10-10 for FAT intake; and β = 3.30 ± 0.49, P = 1.48 x 10-11 for the CHO/FAT ratio. Furthermore, CPT1A mRNA expression was negatively associated with CHO intake, and positively associated with FAT intake, and metabolic phenotypes. Mediation analysis supports the hypothesis that CHO intake induces CPT1A methylation, hence reducing the risk of metabolic diseases, whereas FAT intake inhibits CPT1A methylation, thereby increasing the risk of metabolic diseases. CONCLUSIONS Our results suggest that the proportion of total energy supplied by CHO and FAT can have a causal effect on the risk of metabolic diseases via the epigenetic status of CPT1A.Study registration at https://www.clinicaltrials.gov/: the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN)-NCT01023750; and the Framingham Heart Study (FHS)-NCT00005121.
Collapse
Affiliation(s)
- Chao-Qiang Lai
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Laurence D Parnell
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Caren E Smith
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Tao Guo
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Sergi Sayols-Baixeras
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Catalonia, Spain
- CIBER Cardiovascular Diseases (CIBERCV), Barcelona, Catalonia, Spain
- Molecular Epidemiology, Department of Medical Sciences, Uppsala Universitet, Uppsala, Sweden
| | - Stella Aslibekyan
- Department of Epidemiology, School of Public Health, University of Alabama, Birmingham, AL, USA
| | - Hemant K Tiwari
- Department of Epidemiology, School of Public Health, University of Alabama, Birmingham, AL, USA
| | - Marguerite R Irvin
- Department of Epidemiology, School of Public Health, University of Alabama, Birmingham, AL, USA
| | - Carl Bender
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - David Fei
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama, Birmingham, AL, USA
| | - Paul N Hopkins
- Department of Cardiovascular Genetics, University of Utah, Salt Lake City, UT, USA
| | - Devin M Absher
- Hudson Alpha Institute for Biotechnology, Huntsville, AL, USA
| | - Michael A Province
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Catalonia, Spain
- CIBER Cardiovascular Diseases (CIBERCV), Barcelona, Catalonia, Spain
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
34
|
Dysregulation of metabolic pathways by carnitine palmitoyl-transferase 1 plays a key role in central nervous system disorders: experimental evidence based on animal models. Sci Rep 2020; 10:15583. [PMID: 32973137 PMCID: PMC7519132 DOI: 10.1038/s41598-020-72638-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
The etiology of CNS diseases including multiple sclerosis, Parkinson’s disease and amyotrophic lateral sclerosis remains elusive despite decades of research resulting in treatments with only symptomatic effects. In this study, we provide evidence that a metabolic shift from glucose to lipid is a key mechanism in neurodegeneration. We show that, by downregulating the metabolism of lipids through the key molecule carnitine palmitoyl transferase 1 (CPT1), it is possible to reverse or slowdown disease progression in experimental models of autoimmune encephalomyelitis-, SOD1G93A and rotenone models, mimicking these CNS diseases in humans. The effect was seen both when applying a CPT1 blocker or by using a Cpt1a P479L mutant mouse strain. Furthermore, we show that diet, epigenetics, and microbiota are key elements in this metabolic shift. Finally, we present a systemic model for understanding the complex etiology of neurodegeneration and how different regulatory systems are interconnected through a central metabolic pathway that becomes deregulated under specific conditions.
Collapse
|
35
|
Fernández-Ramos D, Lopitz-Otsoa F, Delacruz-Villar L, Bilbao J, Pagano M, Mosca L, Bizkarguenaga M, Serrano-Macia M, Azkargorta M, Iruarrizaga-Lejarreta M, Sot J, Tsvirkun D, van Liempd SM, Goni FM, Alonso C, Martínez-Chantar ML, Elortza F, Hayardeny L, Lu SC, Mato JM. Arachidyl amido cholanoic acid improves liver glucose and lipid homeostasis in nonalcoholic steatohepatitis via AMPK and mTOR regulation. World J Gastroenterol 2020; 26:5101-5117. [PMID: 32982112 PMCID: PMC7495035 DOI: 10.3748/wjg.v26.i34.5101] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/19/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Arachidyl amido cholanoic acid (Aramchol) is a potent downregulator of hepatic stearoyl-CoA desaturase 1 (SCD1) protein expression that reduces liver triglycerides and fibrosis in animal models of steatohepatitis. In a phase IIb clinical trial in patients with nonalcoholic steatohepatitis (NASH), 52 wk of treatment with Aramchol reduced blood levels of glycated hemoglobin A1c, an indicator of glycemic control. AIM To assess lipid and glucose metabolism in mouse hepatocytes and in a NASH mouse model [induced with a 0.1% methionine and choline deficient diet (0.1MCD)] after treatment with Aramchol. METHODS Isolated primary mouse hepatocytes were incubated with 20 μmol/L Aramchol or vehicle for 48 h. Subsequently, analyses were performed including Western blot, proteomics by mass spectrometry, and fluxomic analysis with 13C-uniformly labeled glucose. For the in vivo part of the study, male C57BL/6J mice were randomly fed a control or 0.1MCD for 4 wk and received 1 or 5 mg/kg/d Aramchol or vehicle by intragastric gavage for the last 2 wk. Liver metabolomics were assessed using ultra-high-performance liquid chromatography-time of flight-MS for the determination of glucose metabolism-related metabolites. RESULTS Combination of proteomics and Western blot analyses showed increased AMPK activity while the activity of nutrient sensor mTORC1 was decreased by Aramchol in hepatocytes. This translated into changes in the content of their downstream targets including proteins involved in fatty acid (FA) synthesis and oxidation [P-ACCα/β(S79), SCD1, CPT1A/B, HADHA, and HADHB], oxidative phosphorylation (NDUFA9, NDUFB11, NDUFS1, NDUFV1, ETFDH, and UQCRC2), tricarboxylic acid (TCA) cycle (MDH2, SUCLA2, and SUCLG2), and ribosome (P-p70S6K[T389] and P-S6[S235/S236]). Flux experiments with 13C-uniformely labeled glucose showed that TCA cycle cataplerosis was reduced by Aramchol in hepatocytes, as indicated by the increase in the number of rounds that malate remained in the TCA cycle. Finally, liver metabolomic analysis showed that glucose homeostasis was improved by Aramchol in 0.1MCD fed mice in a dose-dependent manner, showing normalization of glucose, G6P, F6P, UDP-glucose, and Rbl5P/Xyl5P. CONCLUSION Aramchol exerts its effect on glucose and lipid metabolism in NASH through activation of AMPK and inhibition of mTORC1, which in turn activate FA β-oxidation and oxidative phosphorylation.
Collapse
Affiliation(s)
- David Fernández-Ramos
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
- CIBERehd - Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Madrid 28029, Spain
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
| | - Laura Delacruz-Villar
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
| | - Jon Bilbao
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
| | - Martina Pagano
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Laura Mosca
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Maider Bizkarguenaga
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
| | - Marina Serrano-Macia
- Liver Disease Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Spain
| | - Mikel Azkargorta
- Proteomics Platform, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Spain
| | | | - Jesús Sot
- Instituto Biofisika (UPV/EHU, CSIC), Leioa 48940, Spain; Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, Leioa 48940, Spain
| | - Darya Tsvirkun
- Pre-clinical and Chemistry, Manufacturing and Controls, Galmed Pharmaceuticals, Tel Aviv 6578317, Israel
| | | | - Felix M Goni
- Instituto Biofisika (UPV/EHU, CSIC), Leioa 48940, Spain; Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, Leioa 48940, Spain
| | | | - María Luz Martínez-Chantar
- CIBERehd - Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Madrid 28029, Spain
- Liver Disease Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Spain
| | - Felix Elortza
- Proteomics Platform, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Spain
| | - Liat Hayardeny
- Pre-clinical and Chemistry, Manufacturing and Controls, Galmed Pharmaceuticals, Tel Aviv 6578317, Israel
| | - Shelly C Lu
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - José M Mato
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
- CIBERehd - Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Madrid 28029, Spain
| |
Collapse
|
36
|
Sawyer BT, Qamar L, Yamamoto TM, McMellen A, Watson ZL, Richer JK, Behbakht K, Schlaepfer IR, Bitler BG. Targeting Fatty Acid Oxidation to Promote Anoikis and Inhibit Ovarian Cancer Progression. Mol Cancer Res 2020; 18:1088-1098. [PMID: 32198139 PMCID: PMC7335321 DOI: 10.1158/1541-7786.mcr-19-1057] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/10/2020] [Accepted: 03/16/2020] [Indexed: 12/22/2022]
Abstract
Epithelial-derived high-grade serous ovarian cancer (HGSOC) is the deadliest gynecologic malignancy. Roughly 80% of patients are diagnosed with late-stage disease, which is defined by wide-spread cancer dissemination throughout the pelvic and peritoneal cavities. HGSOC dissemination is dependent on tumor cells acquiring the ability to resist anoikis (apoptosis triggered by cell detachment). Epithelial cell detachment from the underlying basement membrane or extracellular matrix leads to cellular stress, including nutrient deprivation. In this report, we examined the contribution of fatty acid oxidation (FAO) in supporting anoikis resistance. We examined expression Carnitine Palmitoyltransferase 1A (CPT1A) in a panel of HGSOC cell lines cultured in adherent and suspension conditions. With CPT1A knockdown cells, we evaluated anoikis by caspase 3/7 activity, cleaved caspase 3 immunofluorescence, flow cytometry, and colony formation. We assessed CPT1A-dependent mitochondrial activity and tested the effect of exogenous oleic acid on anoikis and mitochondrial activity. In a patient-derived xenograft model, we administered etomoxir, an FAO inhibitor, and/or platinum-based chemotherapy. CPT1A is overexpressed in HGSOC, correlates with poor overall survival, and is upregulated in HGSOC cells cultured in suspension. CPT1A knockdown promoted anoikis and reduced viability of cells cultured in suspension. HGSOC cells in suspension culture are dependent on CPT1A for mitochondrial activity. In a patient-derived xenograft model of HGSOC, etomoxir significantly inhibited tumor progression. IMPLICATIONS: Targeting FAO in HGSOC to promote anoikis and attenuate dissemination is a potential approach to promote a more durable antitumor response and improve patient outcomes.
Collapse
Affiliation(s)
- Brandon T Sawyer
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Lubna Qamar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Tomomi M Yamamoto
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Alexandra McMellen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Zachary L Watson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Kian Behbakht
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Isabel R Schlaepfer
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Benjamin G Bitler
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado.
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
37
|
Fukizawa S, Yamashita M, Wakabayashi KI, Fujisaka S, Tobe K, Nonaka Y, Murayama N. Anti-obesity effect of a hop-derived prenylflavonoid isoxanthohumol in a high-fat diet-induced obese mouse model. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2020; 39:175-182. [PMID: 32775137 PMCID: PMC7392919 DOI: 10.12938/bmfh.2019-040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/10/2020] [Indexed: 01/07/2023]
Abstract
We examined whether oral administration of a hop-derived prenylflavonoid isoxanthohumol (IX) would show anti-obesity activity and the underlying mechanism of the potential activity using a high-fat diet (HFD)-induced obese mouse model. Oral administration of 180 mg/kg IX for 8 weeks suppressed HFD-induced accumulation of visceral fat and body weight gain in mice. Simultaneously, IX changed the composition of the microbiome, as determined by a significant increase in the relative abundances of Akkermansia muciniphila, Blautia, and Escherichia coli. A. muciniphila accounted for 23% and 24% of the total microbiome in the HFD+60 mg/kg and 180 mg/kg IX groups, respectively, while it was undetectable in the normal diet (ND) and HFD groups. Similarly, Blautia accounted for 8% and 10% of the total microbiome in the HFD+60 mg/kg and 180 mg/kg IX groups, respectively, while it accounted for less than 1% in the ND and HFD groups. In contrast, a significant decrease in the relative abundance of Oscillospira was observed in the HFD+60 mg/kg and 180 mg/kg IX groups compared with the HFD group. We further examined the anti-obesity effect of IX using a germ-free (GF) mouse model to clarify the relationship between the microbiome and the effect of IX. IX showed no significant anti-obesity effect on fat accumulation and weight gain in GF mice. These results suggest that the anti-obesity effect of IX may involve microbial changes.
Collapse
Affiliation(s)
- Shinya Fukizawa
- Research Institute, Suntory Global Innovation Center Ltd., 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Mai Yamashita
- Research Institute, Suntory Global Innovation Center Ltd., 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Ken-Ichi Wakabayashi
- Research Institute, Suntory Global Innovation Center Ltd., 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Shiho Fujisaka
- 1st Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan
| | - Kazuyuki Tobe
- 1st Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan
| | - Yuji Nonaka
- Research Institute, Suntory Global Innovation Center Ltd., 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Norihito Murayama
- Research Institute, Suntory Global Innovation Center Ltd., 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| |
Collapse
|
38
|
Schlaepfer IR, Joshi M. CPT1A-mediated Fat Oxidation, Mechanisms, and Therapeutic Potential. Endocrinology 2020; 161:5695911. [PMID: 31900483 DOI: 10.1210/endocr/bqz046] [Citation(s) in RCA: 396] [Impact Index Per Article: 79.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/31/2019] [Indexed: 12/15/2022]
Abstract
Energy homeostasis during fasting or prolonged exercise depends on mitochondrial fatty acid oxidation (FAO). This pathway is crucial in many tissues with high energy demand and its disruption results in inborn FAO deficiencies. More than 15 FAO genetic defects have been currently described, and pathological variants described in circumpolar populations provide insights into its critical role in metabolism. The use of fatty acids as energy requires more than 2 dozen enzymes and transport proteins, which are involved in the activation and transport of fatty acids into the mitochondria. As the key rate-limiting enzyme of FAO, carnitine palmitoyltransferase I (CPT1) regulates FAO and facilitates adaptation to the environment, both in health and in disease, including cancer. The CPT1 family of proteins contains 3 isoforms: CPT1A, CPT1B, and CPT1C. This review focuses on CPT1A, the liver isoform that catalyzes the rate-limiting step of converting acyl-coenzyme As into acyl-carnitines, which can then cross membranes to get into the mitochondria. The regulation of CPT1A is complex and has several layers that involve genetic, epigenetic, physiological, and nutritional modulators. It is ubiquitously expressed in the body and associated with dire consequences linked with genetic mutations, metabolic disorders, and cancers. This makes CPT1A an attractive target for therapeutic interventions. This review discusses our current understanding of CPT1A expression, its role in heath and disease, and the potential for therapeutic opportunities targeting this enzyme.
Collapse
Affiliation(s)
- Isabel R Schlaepfer
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora
| | - Molishree Joshi
- University of Colorado School of Medicine, Department of Pharmacology, Aurora, Colorado
| |
Collapse
|
39
|
Kao YC, Wei WY, Tsai KJ, Wang LC. High Fat Diet Suppresses Peroxisome Proliferator-Activated Receptors and Reduces Dopaminergic Neurons in the Substantia Nigra. Int J Mol Sci 2019; 21:ijms21010207. [PMID: 31892244 PMCID: PMC6981702 DOI: 10.3390/ijms21010207] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
Although several epidemiologic and animal studies have revealed correlations between obesity and neurodegenerative disorders, such as Parkinson disease (PD), the underlying pathological mechanisms of obesity-induced PD remain unclear. Our study aimed to assess the effect of diet-induced obesity on the brain dopaminergic pathway. For five months, starting from weaning, we gave C57BL/6 mice a high-fat diet (HFD) to generate an obese mouse model and investigate whether the diet reprogrammed the midbrain dopaminergic system. Tyrosine hydroxylase staining showed that the HFD resulted in fewer dopaminergic neurons in the substantia nigra (SN), but not the striatum. It also induced neuroinflammation, with increased astrogliosis in the SN and striatum. Dendritic spine density in the SN of HFD-exposed mice decreased, which suggested that prolonged HFD altered dopaminergic neuroplasticity. All three peroxisome proliferator-activated receptor (PPAR) subtype (PPAR-α, PPAR-β/δ, PPAR-γ) levels were significantly reduced in the SN and the ventral tegmental area of HFD mice when compared to those in controls. This study showed that a prolonged HFD induced neuroinflammation, suppressed PPAR levels, caused degeneration of midbrain dopaminergic neurons, and resulted in symptoms reminiscent of human PD. To our knowledge, this is the first study documenting the effects of an HFD on PPARs in dopaminergic neurons.
Collapse
Affiliation(s)
- Yu-Chia Kao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-C.K.); (W.-Y.W.)
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan
| | - Wei-Yen Wei
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-C.K.); (W.-Y.W.)
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-C.K.); (W.-Y.W.)
- Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
- Correspondence: (K.-J.T.); (L.-C.W.); Tel.: +886-6-235-3535-4254 (K.-J.T.); +886-6-235-3535-7212 (L.-C.W.)
| | - Liang-Chao Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-C.K.); (W.-Y.W.)
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
- Correspondence: (K.-J.T.); (L.-C.W.); Tel.: +886-6-235-3535-4254 (K.-J.T.); +886-6-235-3535-7212 (L.-C.W.)
| |
Collapse
|
40
|
Chen X, Zhang J, Dai X. DNA methylation profiles capturing breast cancer heterogeneity. BMC Genomics 2019; 20:823. [PMID: 31699026 PMCID: PMC6839140 DOI: 10.1186/s12864-019-6142-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND As one of the most described epigenetic marks in human cancers, DNA methylation plays essential roles in gene expression regulation and has been implicated in the prognosis and therapeutics of many cancers. We are motivated in this study to explore DNA methylation profiles capturing breast cancer heterogeneity to improve breast cancer prognosis at the epigenetic level. RESULTS Through comparisons on differentially methylated CpG sites among breast cancer subtypes followed by a sequential validation and functional studies using computational approaches, we propose 313 CpG, corresponding to 191 genes, whose methylation pattern identifies the triple negative breast cancer subtype, and report cell migration as represented by extracellular matrix organization and cell proliferation as mediated via MAPK and Wnt signalings are the primary factors driving breast cancer subtyping. CONCLUSIONS Our study offers novel CpGs and gene methylation patterns with translational potential on triple negative breast cancer prognosis, as well as fresh insights from the epigenetic level on breast cancer heterogeneity.
Collapse
Affiliation(s)
- Xiao Chen
- School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianying Zhang
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
41
|
Weld KA, Erb SJ, White HM. Short communication: Effect of manipulating fatty acid profile on gluconeogenic gene expression in bovine primary hepatocytes. J Dairy Sci 2019; 102:7576-7582. [PMID: 31202663 DOI: 10.3168/jds.2018-16150] [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: 12/11/2018] [Accepted: 04/22/2019] [Indexed: 12/20/2022]
Abstract
During the peripartum period, dairy cows experience both an increase in circulating fatty acid (FA) profile and a change in circulating FA profile, which have been shown to alter regulation of gluconeogenic genes. The objective was to quantify gene expression of key enzymes involved in gluconeogenesis and FA transport into the mitochondria in primary hepatocytes in response to exposure to an FA mixture mimicking what is circulating in a transition dairy cow with or without enrichment of C16:0, C18:0, and C18:1. Primary hepatocytes were isolated from 4 Holstein bull calves 3 d of age (± standard deviation 2 d) and cultured. Twenty-four hours after plating, treatments were applied to the cells for 24-h incubation. Treatments consisted of (1) control (1% BSA), (2) 0.75 mM FA cocktail (3% C14:0, 27% C16:0, 23% C18:0, 31% C18:1, 8% C18:2, and 8% C18:3 to mimic the FA profile of dairy cattle at calving), (3) 0.90 mM FA cocktail, (4) 0.75 mM FA cocktail + 0.15 mM C16:0, (5) 0.75 mM FA cocktail + 0.15 mM C18:0, and (6) 0.75 mM FA cocktail + 0.15 mM C18:1. After harvest in Trizol (Life Technologies, Carlsbad, CA), samples were stored at -80°C until RNA extraction, purification, and reverse transcription. Abundance of mRNA was measured using quantitative real-time PCR. Expression of genes of interest [carnitine palmitoyltransferase 1A, pyruvate carboxylase, cytosolic phosphoenolpyruvate carboxykinase (PCK1), mitochondrial phosphoenolpyruvate carboxykinase, and glucose-6-phosphatase] was calculated relative to the average abundance of 2 reference genes (ribosomal protein L32 and glyceraldehyde 3-phosphate dehydrogenase), which were the most stable out of 3 tested. Data were analyzed using PROC MIXED (SAS version 9.4; SAS Institute, Cary, NC) with the fixed effect of treatment and calf in the random statement. The addition of FA compared with the 1% BSA treatment increased the expression of carnitine palmitoyltransferase 1A and cytosolic PCK1. Enrichment with individual FA did not further regulate pyruvate carboxylase or PCK1 beyond that achieved by the basal profile. These results suggest that shifts in circulating FA profile within a biological range, without a difference in the total FA concentration, have minimal effects on transcriptional regulation of hepatic gluconeogenic genes in primary bovine hepatocytes.
Collapse
Affiliation(s)
- K A Weld
- Department of Dairy Science, University of Wisconsin, Madison 53706
| | - S J Erb
- Department of Dairy Science, University of Wisconsin, Madison 53706
| | - H M White
- Department of Dairy Science, University of Wisconsin, Madison 53706.
| |
Collapse
|