1
|
Liepinsh E, Gukalova B, Krims‐Davis K, Kuka J, Leduskrasta A, Korzh S, Vilskersts R, Makrecka‐Kuka M, Konrade I, Dambrova M. EPA and DHA acylcarnitines are less cardiotoxic than are saturated and monounsaturated long-chain acylcarnitines. Biofactors 2025; 51:e70014. [PMID: 40197855 PMCID: PMC11976691 DOI: 10.1002/biof.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Elevated levels of fatty acid-derived long-chain acylcarnitines are detrimental to cardiac health, primarily because of their adverse effects on mitochondrial function and key metabolic pathways in the heart. While trans-fatty acids are considered harmful and omega-3 polyunsaturated fatty acids (PUFAs) are considered beneficial, the specific properties of acylcarnitines derived from these types of fatty acids are not characterized. This study aimed to compare the effects of saturated palmitoylcarnitine (PC), monounsaturated cis-oleoylcarnitine (cis-OC), trans-elaidoylcarnitine (trans-EC), and polyunsaturated eicosapentaenoylcarnitine (EPAC) and docosahexaenoylcarnitine (DHAC) on heart function, cardiac cell viability, mitochondrial functionality, and insulin signaling pathways. Saturated and monounsaturated acylcarnitines, particularly trans-EC, significantly reduced cardiac contractility at concentrations of 8-12 μM, and trans-EC was identified as the most cardiotoxic acylcarnitine. Conversely, the presence of EPAC and DHAC in the perfusion buffer did not impair heart functionality. Saturated and monounsaturated acylcarnitines also drastically reduced H9C2 cell viability and suppressed mitochondrial OXPHOS by up to 70% at 25 μM, whereas PUFA-derived acylcarnitines caused only a 20%-25% reduction in OXPHOS and did not decrease cell viability. Furthermore, PC, cis-OC, and trans-EC significantly inhibited Akt phosphorylation, whereas EPAC and DHAC had a much weaker effect on insulin signaling. In conclusion, saturated and monounsaturated acylcarnitines, particularly trans-EC, exert significant cardiotoxic effects, primarily through the impairment of cardiac mitochondrial function. The omega-3 PUFA-derived acylcarnitines EPAC and DHAC are safe and less likely to damage cardiac mitochondria, cardiac cells, and the heart than other acylcarnitines. PUFA intake might be safer than other long-chain fatty acid-containing lipid sources in patients with FAODs and cardiometabolic diseases.
Collapse
Affiliation(s)
- Edgars Liepinsh
- Latvian Institute of Organic SynthesisRigaLatvia
- Riga Stradins UniversityRigaLatvia
| | - Baiba Gukalova
- Latvian Institute of Organic SynthesisRigaLatvia
- Riga Stradins UniversityRigaLatvia
| | | | - Janis Kuka
- Latvian Institute of Organic SynthesisRigaLatvia
| | | | | | - Reinis Vilskersts
- Latvian Institute of Organic SynthesisRigaLatvia
- Riga Stradins UniversityRigaLatvia
| | | | - Ilze Konrade
- Riga Stradins UniversityRigaLatvia
- Riga East Clinical HospitalRigaLatvia
| | - Maija Dambrova
- Latvian Institute of Organic SynthesisRigaLatvia
- Riga Stradins UniversityRigaLatvia
| |
Collapse
|
2
|
Rostad KO, Trognitz T, Frøyset AK, Bifulco E, Fladmark KE. Accelerated Sarcopenia Phenotype in the DJ-1/ Park7-Knockout Zebrafish. Antioxidants (Basel) 2024; 13:1509. [PMID: 39765837 PMCID: PMC11673048 DOI: 10.3390/antiox13121509] [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: 11/05/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Age-dependent loss of muscle mass and function is associated with oxidative stress. DJ-1/Park7 acts as an antioxidant through multiple signalling pathways. DJ-1-knockout zebrafish show a decline in swimming performance and loss of weight gain between 6 and 9 months of age. Here, we address the degree to which this is associated with muscle degeneration and identify molecular changes preceding dysregulation of muscle performance. Loss of DJ-1 reduced the skeletal muscle fibre cross-section area. The highly mitochondrial-dependent red slow muscle was more affected than the white muscle, and degeneration of sub-sarcolemma red muscle mitochondria was observed. Using TandemMassTag-based quantitative proteomics, we identified a total of 3721 proteins in the multiplex sample of 4 and 12-month-old muscles. A total of 68 proteins, mainly associated with inflammation and mitochondrial function, were dysregulated in the young DJ-1-null adults, with Annexin A3, Sphingomyelin phosphodiesterase acid-like 3B, Complement C3a, and 2,4-dienoyl CoA reductase 1 being the most affected. The loss of DJ-1 also accelerated molecular features associated with sarcopenia, such as a decrease in the NAD+/NADH ratio and a reduction in Prostaglandin reductase 2 and Cytosolic glycerol-3-phosphate dehydrogenase levels. In view of the experimental power of zebrafish, the DJ-1-null zebrafish makes a valuable model for understanding the connection between oxidative stress and age-dependent muscle loss and function.
Collapse
Affiliation(s)
| | | | | | | | - Kari E. Fladmark
- Department of Biological Sciences, University of Bergen, 5020 Bergen, Norway (T.T.); (A.K.F.); (E.B.)
| |
Collapse
|
3
|
Kohlmaier B, Skok K, Lackner C, Haselrieder G, Müller T, Sailer S, Zschocke J, Keller MA, Knisely AS, Janecke AR. Steatotic liver disease associated with 2,4-dienoyl-CoA reductase 1 deficiency. Int J Obes (Lond) 2024; 48:1818-1821. [PMID: 39277655 PMCID: PMC11584395 DOI: 10.1038/s41366-024-01634-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is considered multifactorial with a number of predisposing gene polymorphisms known. METHODS The occurrence of MASLD in 7 and 10 year old siblings, one without classical risk factors and one with type 2 diabetes suggested a monogenic etiology and prompted next-generation sequencing. Exome sequencing was performed in the proband, both parents and both siblings. The impact of a likely disease-causing DNA variant was assessed on the transcript and protein level. RESULTS Two siblings have hepatomegaly, elevated serum transaminase activity, and steatosis and harbor a homozygous DECR1 splice-site variant, c.330+3A>T. The variant caused DECR1 transcript decay. Immunostaining demonstrated lack of DECR1 in patient liver. CONCLUSIONS These patients may represent the first individuals with DECR1 deficiency, then defining within MASLD an autosomal-recessive entity, well corresponding to the reported steatotic liver disease in Decr1 knockout mice. DECR1 may need to be considered in the genetic work-up of MASLD.
Collapse
Affiliation(s)
- Benno Kohlmaier
- Department of General Paediatrics, Medical University of Graz, 8010, Graz, Austria
| | - Kristijan Skok
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010, Graz, Austria
| | - Carolin Lackner
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010, Graz, Austria
| | - Greta Haselrieder
- Department of Paediatrics I, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Sabrina Sailer
- Institute of Human Genetics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Johannes Zschocke
- Institute of Human Genetics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - A S Knisely
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010, Graz, Austria.
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, 6020, Innsbruck, Austria.
- Institute of Human Genetics, Medical University of Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
4
|
Duan Z, Huang Z, Lei W, Zhang K, Xie W, Jin H, Wu M, Wang N, Li X, Xu A, Zhou H, Wu F, Li Y, Lin Z. Bone Morphogenetic Protein 9 Protects Against Myocardial Infarction by Improving Lymphatic Drainage Function and Triggering DECR1-Mediated Mitochondrial Bioenergetics. Circulation 2024; 150:1684-1701. [PMID: 39315433 DOI: 10.1161/circulationaha.123.065935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/01/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND BMP9 (bone morphogenetic protein 9) is a member of the TGF-β (transforming growth factor β) family of cytokines with pleiotropic effects on glucose metabolism, fibrosis, and lymphatic development. However, the role of BMP9 in myocardial infarction (MI) remains elusive. METHODS The expressional profiles of BMP9 in cardiac tissues and plasma samples of subjects with MI were determined by immunoassay or immunoblot. The role of BMP9 in MI was determined by evaluating the impact of BMP9 deficiency and replenishment with adeno-associated virus-mediated BMP9 expression or recombinant human BMP9 protein in mice. RESULTS We show that circulating BMP9 and its cardiac levels are markedly increased in humans and mice with MI and are negatively associated with cardiac function. It is important to note that BMP9 deficiency exacerbates left ventricular dysfunction, increases infarct size, and augments cardiac fibrosis in mice with MI. In contrast, replenishment of BMP9 significantly attenuates these adverse effects. We further demonstrate that BMP9 improves lymphatic drainage function, thereby leading to a decrease of cardiac edema. In addition, BMP9 increases the expression of mitochondrial DECR1 (2,4-dienoyl-CoA [coenzyme A] reductase 1), a rate-limiting enzyme involved in β-oxidation, which, in turn, promotes cardiac mitochondrial bioenergetics and mitigates MI-induced cardiomyocyte injury. Moreover, DECR1 deficiency exacerbates MI-induced cardiac damage in mice, whereas this adverse effect is restored by the treatment of adeno-associated virus-mediated DECR1. Consistently, DECR1 deletion abrogates the beneficial effect of BMP9 against MI-induced cardiomyopathy and cardiac damage in mice. CONCLUSIONS These results suggest that BMP9 protects against MI by fine-tuning the multiorgan cross-talk among the liver, lymph, and the heart.
Collapse
Affiliation(s)
- Zikun Duan
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
| | - Zhouqing Huang
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Wei Lei
- Precision Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China (W.L.)
| | - Ke Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Wei Xie
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Hua Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Maolan Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Ningrui Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, China (A.X.)
| | - Hao Zhou
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Fan Wu
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Innovation Center of Cardiometabolic Disease (F.W., Z.L.), Guangdong Medical University, Dongguan, China
| | - Yulin Li
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Anzhen Hospital of Capital Medical University, China (Y.L.)
| | - Zhuofeng Lin
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Innovation Center of Cardiometabolic Disease (F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| |
Collapse
|
5
|
Jurado-Aguilar J, Barroso E, Bernard M, Zhang M, Peyman M, Rada P, Valverde ÁM, Wahli W, Palomer X, Vázquez-Carrera M. GDF15 activates AMPK and inhibits gluconeogenesis and fibrosis in the liver by attenuating the TGF-β1/SMAD3 pathway. Metabolism 2024; 152:155772. [PMID: 38176644 DOI: 10.1016/j.metabol.2023.155772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
INTRODUCTION The levels of the cellular energy sensor AMP-activated protein kinase (AMPK) have been reported to be decreased via unknown mechanisms in the liver of mice deficient in growth differentiation factor 15 (GDF15). This stress response cytokine regulates energy metabolism mainly by reducing food intake through its hindbrain receptor GFRAL. OBJECTIVE To examine how GDF15 regulates AMPK. METHODS Wild-type and Gdf15-/- mice, mouse primary hepatocytes and the human hepatic cell line Huh-7 were used. RESULTS Gdf15-/- mice showed glucose intolerance, reduced hepatic phosphorylated AMPK levels, increased levels of phosphorylated mothers against decapentaplegic homolog 3 (SMAD3; a mediator of the fibrotic response), elevated serum levels of transforming growth factor (TGF)-β1, as well as upregulated gluconeogenesis and fibrosis. In line with these observations, recombinant (r)GDF15 promoted AMPK activation and reduced the levels of phosphorylated SMAD3 and the markers of gluconeogenesis and fibrosis in the liver of mice and in mouse primary hepatocytes, suggesting that these effects may be independent of GFRAL. Pharmacological inhibition of SMAD3 phosphorylation in Gdf15-/- mice prevented glucose intolerance, the deactivation of AMPK and the increase in the levels of proteins involved in gluconeogenesis and fibrosis, suggesting that overactivation of the TGF-β1/SMAD3 pathway is responsible for the metabolic alterations in Gdf15-/- mice. CONCLUSIONS Overall, these findings indicate that GDF15 activates AMPK and inhibits gluconeogenesis and fibrosis by lowering the activity of the TGF-β1/SMAD3 pathway.
Collapse
Affiliation(s)
- Javier Jurado-Aguilar
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Maribel Bernard
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Meijian Zhang
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Mona Peyman
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Patricia Rada
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Madrid, Spain
| | - Ángela M Valverde
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Madrid, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland; Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232; ToxAlim (Research Center in Food Toxicology), INRAE, UMR1331, F-31300 Toulouse Cedex, France
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain.
| |
Collapse
|
6
|
Fujinuma S, Nakatsumi H, Shimizu H, Sugiyama S, Harada A, Goya T, Tanaka M, Kohjima M, Takahashi M, Izumi Y, Yagi M, Kang D, Kaneko M, Shigeta M, Bamba T, Ohkawa Y, Nakayama KI. FOXK1 promotes nonalcoholic fatty liver disease by mediating mTORC1-dependent inhibition of hepatic fatty acid oxidation. Cell Rep 2023; 42:112530. [PMID: 37209098 DOI: 10.1016/j.celrep.2023.112530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/14/2023] [Accepted: 05/02/2023] [Indexed: 05/22/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic metabolic disorder caused by overnutrition and can lead to nonalcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC). The transcription factor Forkhead box K1 (FOXK1) is implicated in regulation of lipid metabolism downstream of mechanistic target of rapamycin complex 1 (mTORC1), but its role in NAFLD-NASH pathogenesis is understudied. Here, we show that FOXK1 mediates nutrient-dependent suppression of lipid catabolism in the liver. Hepatocyte-specific deletion of Foxk1 in mice fed a NASH-inducing diet ameliorates not only hepatic steatosis but also associated inflammation, fibrosis, and tumorigenesis, resulting in improved survival. Genome-wide transcriptomic and chromatin immunoprecipitation analyses identify several lipid metabolism-related genes, including Ppara, as direct targets of FOXK1 in the liver. Our results suggest that FOXK1 plays a key role in the regulation of hepatic lipid metabolism and that its inhibition is a promising therapeutic strategy for NAFLD-NASH, as well as for HCC.
Collapse
Affiliation(s)
- Shun Fujinuma
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hirokazu Nakatsumi
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hideyuki Shimizu
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Akihito Harada
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takeshi Goya
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatake Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Motoyuki Kohjima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Mikako Yagi
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan; Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Mayo Shigeta
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
7
|
Ranea-Robles P, Houten SM. The biochemistry and physiology of long-chain dicarboxylic acid metabolism. Biochem J 2023; 480:607-627. [PMID: 37140888 PMCID: PMC10214252 DOI: 10.1042/bcj20230041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/05/2023]
Abstract
Mitochondrial β-oxidation is the most prominent pathway for fatty acid oxidation but alternative oxidative metabolism exists. Fatty acid ω-oxidation is one of these pathways and forms dicarboxylic acids as products. These dicarboxylic acids are metabolized through peroxisomal β-oxidation representing an alternative pathway, which could potentially limit the toxic effects of fatty acid accumulation. Although dicarboxylic acid metabolism is highly active in liver and kidney, its role in physiology has not been explored in depth. In this review, we summarize the biochemical mechanism of the formation and degradation of dicarboxylic acids through ω- and β-oxidation, respectively. We will discuss the role of dicarboxylic acids in different (patho)physiological states with a particular focus on the role of the intermediates and products generated through peroxisomal β-oxidation. This review is expected to increase the understanding of dicarboxylic acid metabolism and spark future research.
Collapse
Affiliation(s)
- Pablo Ranea-Robles
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Sander M Houten
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| |
Collapse
|
8
|
Zhang R, Zhang K. Mitochondrial NAD kinase in health and disease. Redox Biol 2023; 60:102613. [PMID: 36689815 PMCID: PMC9873681 DOI: 10.1016/j.redox.2023.102613] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADP), a co-enzyme and an electron carrier, plays crucial roles in numerous biological functions, including cellular metabolism and antioxidation. Because NADP is subcellular-membrane impermeable, eukaryotes compartmentalize NAD kinases (NADKs), the NADP biosynthetic enzymes. Mitochondria are fundamental organelles for energy production through oxidative phosphorylation. Ten years after the discovery of the mitochondrial NADK (known as MNADK or NADK2), a significant amount of knowledge has been obtained regarding its functions, mechanism of action, human biology, mouse models, crystal structures, and post-translation modifications. NADK2 phosphorylates NAD(H) to generate mitochondrial NADP(H). NADK2-deficient patients suffered from hyperlysinemia, elevated plasma C10:2-carnitine (due to the inactivity of relevant NADP-dependent enzymes), and neuronal development defects. Nadk2-deficient mice recapitulate key features of NADK2-deficient patients, including metabolic and neuronal abnormalities. Crystal structures of human NADK2 show a dimer, with the NADP+-binding site located at the dimer interface. NADK2 activity is highly regulated by post-translational modifications, including S188 phosphorylation, K76 and K304 acetylation, and C193 S-nitrosylation; mutations in each site affect NADK2 activity and function. In mice, hepatic Nadk2 functions as a major metabolic regulator upon increased energy demands by regulating sirtuin 3 activity and fatty acid oxidation. Hopefully, future research on NADK2 will not only elucidate its functional roles in health and disease but will also pave the way for novel therapeutics for both rare and common diseases, including NADK2 deficiency and metabolic syndrome.
Collapse
Affiliation(s)
- Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
9
|
Murray GC, Bais P, Hatton CL, Tadenev ALD, Hoffmann BR, Stodola TJ, Morelli KH, Pratt SL, Schroeder D, Doty R, Fiehn O, John SWM, Bult CJ, Cox GA, Burgess RW. Mouse models of NADK2 deficiency analyzed for metabolic and gene expression changes to elucidate pathophysiology. Hum Mol Genet 2022; 31:4055-4074. [PMID: 35796562 PMCID: PMC9703942 DOI: 10.1093/hmg/ddac151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 06/17/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
NADK2 encodes the mitochondrial form of nicotinamide adenine dinucleotide (NAD) kinase, which phosphorylates NAD. Rare recessive mutations in human NADK2 are associated with a syndromic neurological mitochondrial disease that includes metabolic changes, such as hyperlysinemia and 2,4 dienoyl CoA reductase (DECR) deficiency. However, the full pathophysiology resulting from NADK2 deficiency is not known. Here, we describe two chemically induced mouse mutations in Nadk2-S326L and S330P-which cause severe neuromuscular disease and shorten lifespan. The S330P allele was characterized in detail and shown to have marked denervation of neuromuscular junctions by 5 weeks of age and muscle atrophy by 11 weeks of age. Cerebellar Purkinje cells also showed progressive degeneration in this model. Transcriptome profiling on brain and muscle was performed at early and late disease stages. In addition, metabolomic profiling was performed on the brain, muscle, liver and spinal cord at the same ages and on plasma at 5 weeks. Combined transcriptomic and metabolomic analyses identified hyperlysinemia, DECR deficiency and generalized metabolic dysfunction in Nadk2 mutant mice, indicating relevance to the human disease. We compared findings from the Nadk model to equivalent RNA sequencing and metabolomic datasets from a mouse model of infantile neuroaxonal dystrophy, caused by recessive mutations in Pla2g6. This enabled us to identify disrupted biological processes that are common between these mouse models of neurological disease, as well as those processes that are gene-specific. These findings improve our understanding of the pathophysiology of neuromuscular diseases and describe mouse models that will be useful for future preclinical studies.
Collapse
Affiliation(s)
- G C Murray
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - P Bais
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - C L Hatton
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - A L D Tadenev
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - B R Hoffmann
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - T J Stodola
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - K H Morelli
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - S L Pratt
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - D Schroeder
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - R Doty
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - O Fiehn
- West Coast Metabolomics Center, University of California Davis, 451 Health Science Dr., Davis, CA 95618, USA
| | - S W M John
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10032, USA
| | - C J Bult
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - G A Cox
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - R W Burgess
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
10
|
Dave A, Park EJ, Kumar A, Parande F, Beyoğlu D, Idle JR, Pezzuto JM. Consumption of Grapes Modulates Gene Expression, Reduces Non-Alcoholic Fatty Liver Disease, and Extends Longevity in Female C57BL/6J Mice Provided with a High-Fat Western-Pattern Diet. Foods 2022; 11:1984. [PMID: 35804799 PMCID: PMC9265568 DOI: 10.3390/foods11131984] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 02/05/2023] Open
Abstract
A key objective of this study was to explore the potential of dietary grape consumption to modulate adverse effects caused by a high-fat (western-pattern) diet. Female C57BL/6J mice were purchased at six-weeks-of-age and placed on a standard (semi-synthetic) diet (STD). At 11 weeks-of-age, the mice were continued on the STD or placed on the STD supplemented with 5% standardized grape powder (STD5GP), a high-fat diet (HFD), or an HFD supplemented with 5% standardized grape powder (HFD5GP). After being provided with the respective diets for 13 additional weeks, the mice were euthanized, and liver was collected for biomarker analysis, determination of genetic expression (RNA-Seq), and histopathological examination. All four dietary groups demonstrated unique genetic expression patterns. Using pathway analysis tools (GO, KEGG and Reactome), relative to the STD group, differentially expressed genes of the STD5GP group were significantly enriched in RNA, mitochondria, and protein translation related pathways, as well as drug metabolism, glutathione, detoxification, and oxidative stress associated pathways. The expression of Gstp1 was confirmed to be upregulated by about five-fold (RT-qPCR), and, based on RNA-Seq data, the expression of additional genes associated with the reduction of oxidative stress and detoxification (Gpx4 and 8, Gss, Gpx7, Sod1) were enhanced by dietary grape supplementation. Cluster analysis of genetic expression patterns revealed the greatest divergence between the HFD5GP and HFD groups. In the HFD5GP group, relative to the HFD group, 14 genes responsible for the metabolism, transportation, hydrolysis, and sequestration of fatty acids were upregulated. Conversely, genes responsible for lipid content and cholesterol synthesis (Plin4, Acaa1b, Slc27a1) were downregulated. The two top classifications emerging as enriched in the HFD5GP group vs. the HFD group (KEGG pathway analysis) were Alzheimer's disease and nonalcoholic fatty liver disease (NAFLD), both of which have been reported in the literature to bear a causal relationship. In the current study, nonalcoholic steatohepatitis was indicated by histological observations that revealed archetype markers of fatty liver induced by the HFD. The adverse response was diminished by grape intervention. In addition to these studies, life-long survival was assessed with C57BL/6J mice. C57BL/6J mice were received at four-weeks-of-age and placed on the STD. At 14-weeks-of-age, the mice were divided into two groups (100 per group) and provided with the HFD or the HFD5GP. Relative to the HFD group, the survival time of the HFD5GP group was enhanced (log-rank test, p = 0.036). The respective hazard ratios were 0.715 (HFD5GP) and 1.397 (HFD). Greater body weight positively correlated with longevity; the highest body weight of the HFD5GP group was attained later in life than the HFD group (p = 0.141). These results suggest the potential of dietary grapes to modulate hepatic gene expression, prevent oxidative damage, induce fatty acid metabolism, ameliorate NAFLD, and increase longevity when co-administered with a high-fat diet.
Collapse
Affiliation(s)
- Asim Dave
- Division of Pharmaceutical Sciences, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (A.D.); (E.-J.P.); (A.K.); (F.P.)
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eun-Jung Park
- Division of Pharmaceutical Sciences, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (A.D.); (E.-J.P.); (A.K.); (F.P.)
| | - Avinash Kumar
- Division of Pharmaceutical Sciences, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (A.D.); (E.-J.P.); (A.K.); (F.P.)
| | - Falguni Parande
- Division of Pharmaceutical Sciences, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (A.D.); (E.-J.P.); (A.K.); (F.P.)
- Artus Therapeutics, Harvard Life Lab, Allston, MA 02134, USA
| | - Diren Beyoğlu
- Arthur G. Zupko’s Institute of Systems Pharmacology and Pharmacogenomics, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (D.B.); (J.R.I.)
| | - Jeffrey R. Idle
- Arthur G. Zupko’s Institute of Systems Pharmacology and Pharmacogenomics, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (D.B.); (J.R.I.)
| | - John M. Pezzuto
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| |
Collapse
|
11
|
Dambrova M, Makrecka-Kuka M, Kuka J, Vilskersts R, Nordberg D, Attwood MM, Smesny S, Sen ZD, Guo AC, Oler E, Tian S, Zheng J, Wishart DS, Liepinsh E, Schiöth HB. Acylcarnitines: Nomenclature, Biomarkers, Therapeutic Potential, Drug Targets, and Clinical Trials. Pharmacol Rev 2022; 74:506-551. [PMID: 35710135 DOI: 10.1124/pharmrev.121.000408] [Citation(s) in RCA: 236] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acylcarnitines are fatty acid metabolites that play important roles in many cellular energy metabolism pathways. They have historically been used as important diagnostic markers for inborn errors of fatty acid oxidation and are being intensively studied as markers of energy metabolism, deficits in mitochondrial and peroxisomal β -oxidation activity, insulin resistance, and physical activity. Acylcarnitines are increasingly being identified as important indicators in metabolic studies of many diseases, including metabolic disorders, cardiovascular diseases, diabetes, depression, neurologic disorders, and certain cancers. The US Food and Drug Administration-approved drug L-carnitine, along with short-chain acylcarnitines (acetylcarnitine and propionylcarnitine), is now widely used as a dietary supplement. In light of their growing importance, we have undertaken an extensive review of acylcarnitines and provided a detailed description of their identity, nomenclature, classification, biochemistry, pathophysiology, supplementary use, potential drug targets, and clinical trials. We also summarize these updates in the Human Metabolome Database, which now includes information on the structures, chemical formulae, chemical/spectral properties, descriptions, and pathways for 1240 acylcarnitines. This work lays a solid foundation for identifying, characterizing, and understanding acylcarnitines in human biosamples. We also discuss the emerging opportunities for using acylcarnitines as biomarkers and as dietary interventions or supplements for many wide-ranging indications. The opportunity to identify new drug targets involved in controlling acylcarnitine levels is also discussed. SIGNIFICANCE STATEMENT: This review provides a comprehensive overview of acylcarnitines, including their nomenclature, structure and biochemistry, and use as disease biomarkers and pharmaceutical agents. We present updated information contained in the Human Metabolome Database website as well as substantial mapping of the known biochemical pathways associated with acylcarnitines, thereby providing a strong foundation for further clarification of their physiological roles.
Collapse
Affiliation(s)
- Maija Dambrova
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Marina Makrecka-Kuka
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Janis Kuka
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Reinis Vilskersts
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Didi Nordberg
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Misty M Attwood
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Stefan Smesny
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Zumrut Duygu Sen
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - An Chi Guo
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Eponine Oler
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Siyang Tian
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Jiamin Zheng
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - David S Wishart
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Edgars Liepinsh
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| | - Helgi B Schiöth
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia (M.D., M.M.-K., J.K., R.V., E.L.); Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden, (D.N., M.M.A., H.B.S.); Department of Psychiatry, Jena University Hospital, Jena, Germany (S.S., Z.D.S.); and Department of Biological Sciences, University of Alberta, Edmonton, Canada (A.C.G., E.O., S.T., J.Z., D.S.W.)
| |
Collapse
|
12
|
Abstract
Regulatory RNAs like microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) control vascular and immune cells' phenotype and thus play a crucial role in atherosclerosis. Moreover, the mutual interactions between miRNAs and lncRNAs link both types of regulatory RNAs in a functional network that affects lesion formation. In this review, we deduce novel concepts of atherosclerosis from the analysis of the current data on regulatory RNAs' role in endothelial cells (ECs) and macrophages. In contrast to arterial ECs, which adopt a stable phenotype by adaptation to high shear stress, macrophages are highly plastic and quickly change their activation status. At predilection sites of atherosclerosis, such as arterial bifurcations, ECs are exposed to disturbed laminar flow, which generates a dysadaptive stress response mediated by miRNAs. Whereas the highly abundant miR-126-5p promotes regenerative proliferation of dysadapted ECs, miR-103-3p stimulates inflammatory activation and impairs endothelial regeneration by aberrant proliferation and micronuclei formation. In macrophages, miRNAs are essential in regulating energy and lipid metabolism, which affects inflammatory activation and foam cell formation.Moreover, lipopolysaccharide-induced miR-155 and miR-146 shape inflammatory macrophage activation through their oppositional effects on NF-kB. Most lncRNAs are not conserved between species, except a small group of very long lncRNAs, such as MALAT1, which blocks numerous miRNAs by providing non-functional binding sites. In summary, regulatory RNAs' roles are highly context-dependent, and therapeutic approaches that target specific functional interactions of miRNAs appear promising against cardiovascular diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany.
| | - Saffiyeh Saboor Maleki
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Maliheh Nazari-Jahantigh
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
13
|
Shen Z, Tang Y, Song Y, Shen W, Zou C. Differences of DNA methylation patterns in the placenta of large for gestational age infant. Medicine (Baltimore) 2020; 99:e22389. [PMID: 32991460 PMCID: PMC7523834 DOI: 10.1097/md.0000000000022389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
To investigate the molecular mechanisms of later metabolic health changes in large for gestational age (LGA) newborns by analyzing deoxyribonucleic acid (DNA) methylation patterns in the placenta of LGA and appropriate for gestational age (AGA) newborns.A total of 6 placentas of LGA and 6 placentas of AGA newborns were enrolled as LGA group and AGA group. DNA methylation was analyzed using the Illumina Infinium Human MethylationEPIC BeadChip microarrays and verified via pyrosequencing and reverse transcription-quantitative real-time polymerase chain reaction. Functional enrichment analysis were constructed by gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis based on the differentially methylated regions between LGA and AGA groups.Clinical investigation showed that LGA newborns had significantly lower hemoglobin and blood glucose compared to AGA newborns. Birth weight was negatively correlated to hemoglobin and blood glucose. Genome-wide DNA methylation analysis identified 17 244 methylation variable positions achieving genome-wide significance (adjusted P < .05). 34% methylation variable positions were located in the gene promoter region. A total of 117 differentially methylated regions were revealed by bump hunting analysis, which mapped to 107 genes. Function analysis showed 13 genes enriched in "adhesion and infection process, endocrine and other factor-regulated calcium reabsorption, calcium signaling pathway and transmembrane transport". Four genes linked to type II diabetes mellitus. Among the 13 genes, we selected GNAS and calcium voltage-gated channel subunit alpha1 G for independent verification of pyrosequencing, and the messenger ribonucleic acid levels of guanine nucleotide binding protein, calcium voltage-gated channel subunit alpha1 G, DECR1, and FK506 binding protein 11 were verified by reverse transcription-quantitative real-time polymerase chain reaction.DNA methylation variation and gene expression differences in placental samples were associated with LGA newborns, which linking the effect of intrauterine environment to regulation of the offspring's gene expression. Furthermore, pathway analysis suggested that intrauterine environment affecting fetal growth might had a functional impact on multiple signaling pathways involved in fetal growth, metabolism, and inflammation. Further studies were required to understand the differences of methylation patterns.
Collapse
Affiliation(s)
- Zheng Shen
- Department of Clinical laboratory, Zhejiang University School of Medicine Children's Hospital
- National Clinical Research Center for Child Health
| | - Yanfei Tang
- Department of Endocrinology, Zhejiang University School of Medicine Children's Hospital
- Second Hospital of Jiaxing
| | - Yemei Song
- Department of Endocrinology, Zhejiang University School of Medicine Children's Hospital
- Huzhou Central Hospital
| | - Wenxia Shen
- Department of Endocrinology, Zhejiang University School of Medicine Children's Hospital
- Women and Children's Hospital of Shaoxin
| | - Chaochun Zou
- National Clinical Research Center for Child Health
- Department of Endocrinology, Zhejiang University School of Medicine Children's Hospital
| |
Collapse
|
14
|
de Sousa IF, Pedroso AP, de Andrade IS, Boldarine VT, Tashima AK, Oyama LM, Lionetti L, Ribeiro EB. High-fat but not normal-fat intake of extra virgin olive oil modulates the liver proteome of mice. Eur J Nutr 2020; 60:1375-1388. [PMID: 32712699 DOI: 10.1007/s00394-020-02323-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/01/2020] [Indexed: 11/27/2022]
Abstract
PURPOSE The metabolic benefits of the Mediterranean diet have been largely attributed to its olive oil content. Whether the ingested fat amount is relevant to these effects is not clear. We thus compared the effects of high-fat and normal-fat intake of extra-virgin olive oil (EVOO) on the liver proteome. METHODS Three groups of mice were fed for 12 weeks with either normal-fat diets containing either soybean oil (control, C) or EVOO (NO) or a high-fat EVOO diet (HO). Body weight and food intake were measured weekly and serum parameters were analyzed. The liver was processed for data-independent acquisition mass spectrometry-based proteomics. The differentially expressed proteins among the groups were submitted to pathway enrichment analysis. RESULTS The consumption of HO diet reduced food intake and serum triglycerides, while it preserved body weight gain, adiposity, and glycemia. However, it increased serum cholesterol and liver mass. The proteomic analysis showed 98 altered proteins, which were allocated in 27 significantly enriched pathways. The pathway analysis suggested stimulation of mitochondrial and peroxissomal β-oxidation, and inhibition of lipid synthesis and gluconeogenesis in the HO group. Although the NO group failed to show significant liver proteome alterations, it presented reduced body fat, body weight gain, and serum triglycerides and glucose levels. CONCLUSION The data indicate that the intake of the HO diet induced hepatic adjustments, which were partially successful in counteracting the detrimental outcomes of a high-fat feeding. Contrastingly, the NO diet had beneficial effects which were not accompanied by significant modifications on hepatic proteome.
Collapse
Affiliation(s)
- Isy F de Sousa
- Departamento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, Rua Botucatu 862, Vila Clementino, São Paulo, SP, 04023-062, Brazil
- Dipartimento Di Chimica E Biologia "Adolfo Zambelli", Università Degli Studi Di Salerno, Salerno, Italy
| | - Amanda P Pedroso
- Departamento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, Rua Botucatu 862, Vila Clementino, São Paulo, SP, 04023-062, Brazil
| | - Iracema S de Andrade
- Departamento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, Rua Botucatu 862, Vila Clementino, São Paulo, SP, 04023-062, Brazil
| | - Valter T Boldarine
- Departamento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, Rua Botucatu 862, Vila Clementino, São Paulo, SP, 04023-062, Brazil
| | - Alexandre K Tashima
- Departamento de Bioquímica, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Lila M Oyama
- Departamento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, Rua Botucatu 862, Vila Clementino, São Paulo, SP, 04023-062, Brazil
| | - Lillà Lionetti
- Dipartimento Di Chimica E Biologia "Adolfo Zambelli", Università Degli Studi Di Salerno, Salerno, Italy
| | - Eliane B Ribeiro
- Departamento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, Rua Botucatu 862, Vila Clementino, São Paulo, SP, 04023-062, Brazil.
| |
Collapse
|
15
|
Nassar ZD, Mah CY, Dehairs J, Burvenich IJG, Irani S, Centenera MM, Helm M, Shrestha RK, Moldovan M, Don AS, Holst J, Scott AM, Horvath LG, Lynn DJ, Selth LA, Hoy AJ, Swinnen JV, Butler LM. Human DECR1 is an androgen-repressed survival factor that regulates PUFA oxidation to protect prostate tumor cells from ferroptosis. eLife 2020; 9:e54166. [PMID: 32686647 PMCID: PMC7386908 DOI: 10.7554/elife.54166] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 07/16/2020] [Indexed: 12/27/2022] Open
Abstract
Fatty acid β-oxidation (FAO) is the main bioenergetic pathway in human prostate cancer (PCa) and a promising novel therapeutic vulnerability. Here we demonstrate therapeutic efficacy of targeting FAO in clinical prostate tumors cultured ex vivo, and identify DECR1, encoding the rate-limiting enzyme for oxidation of polyunsaturated fatty acids (PUFAs), as robustly overexpressed in PCa tissues and associated with shorter relapse-free survival. DECR1 is a negatively-regulated androgen receptor (AR) target gene and, therefore, may promote PCa cell survival and resistance to AR targeting therapeutics. DECR1 knockdown selectively inhibited β-oxidation of PUFAs, inhibited proliferation and migration of PCa cells, including treatment resistant lines, and suppressed tumor cell proliferation and metastasis in mouse xenograft models. Mechanistically, targeting of DECR1 caused cellular accumulation of PUFAs, enhanced mitochondrial oxidative stress and lipid peroxidation, and induced ferroptosis. These findings implicate PUFA oxidation via DECR1 as an unexplored facet of FAO that promotes survival of PCa cells.
Collapse
Grants
- Early Career Fellowship,1138648 National Health and Medical Research Council
- Project Grants C16/15/073 and C32/17/052 KU Leuven
- Future Fellowship,FT130101004 Australian Research Council
- Beat Cancer Fellowship,PRF1117 Cancer Council South Australia
- Revolutionary Team Award,MRTA3 Movember Foundation
- Project Grant,1121057 National Health and Medical Research Council
- Project Grant,1100626 National Health and Medical Research Council
- Fellowship,1084178 National Health and Medical Research Council
- Young Investigator Award,YI 1417 Prostate Cancer Foundation of Australia
- Project Grant,1164798 Cure Cancer Australia Foundation
- Group Leader Award EMBL Australia
- Robinson Fellowship University of Sydney
- Project Grants G.0841.15 and G.0C22.19N Fonds Wetenschappelijk Onderzoek
- 1138648 National Health and Medical Research Council
- 1121057 National Health and Medical Research Council
- 1100626 National Health and Medical Research Council
- 1084178 National Health and Medical Research Council
- YI 1417 Prostate Cancer Foundation of Australia
- 1164798 Cure Cancer Australia Foundation
- FT130101004 Australian Research Council
- PRF1117 Cancer Council South Australia
- MRTA3 Movember Foundation
- Freemasons Foundation Centre for Men's Health, University of Adelaide
Collapse
Affiliation(s)
- Zeyad D Nassar
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Chui Yan Mah
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Jonas Dehairs
- KU Leuven- University of Leuven, LKI- Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and CancerLeuvenBelgium
| | - Ingrid JG Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe UniversityMelbourneAustralia
| | - Swati Irani
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Margaret M Centenera
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Madison Helm
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Raj K Shrestha
- Dame Roma Mitchell Cancer Research Laboratories, University of AdelaideAdelaideAustralia
| | - Max Moldovan
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Anthony S Don
- NHMRC Clinical Trials Centre, and Centenary Institute, The University of SydneyCamperdownAustralia
| | - Jeff Holst
- Translational Cancer Metabolism Laboratory, School of Medical Sciences and Prince of Wales Clinical School, UNSW SydneySydneyAustralia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe UniversityMelbourneAustralia
| | - Lisa G Horvath
- Garvan Institute of Medical Research, NSW 2010; University of Sydney, NSW 2006; and University of New South WalesDarlinghurstAustralia
| | - David J Lynn
- South Australian Health and Medical Research InstituteAdelaideAustralia
- College of Medicine and Public Health, Flinders UniversityBedford ParkAustralia
| | - Luke A Selth
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- Dame Roma Mitchell Cancer Research Laboratories, University of AdelaideAdelaideAustralia
- College of Medicine and Public Health, Flinders UniversityBedford ParkAustralia
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of SydneyCamperdownAustralia
| | - Johannes V Swinnen
- KU Leuven- University of Leuven, LKI- Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and CancerLeuvenBelgium
| | - Lisa M Butler
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| |
Collapse
|
16
|
Leishmania Encodes a Bacterium-like 2,4-Dienoyl-Coenzyme A Reductase That Is Required for Fatty Acid β-Oxidation and Intracellular Parasite Survival. mBio 2020; 11:mBio.01057-20. [PMID: 32487758 PMCID: PMC7267886 DOI: 10.1128/mbio.01057-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Leishmania spp. are protozoan parasites that cause a spectrum of important diseases in humans. These parasites develop as extracellular promastigotes in the digestive tract of their insect vectors and as obligate intracellular amastigotes that infect macrophages and other phagocytic cells in their vertebrate hosts. Promastigote-to-amastigote differentiation is associated with marked changes in metabolism, including the upregulation of enzymes involved in fatty acid β-oxidation, which may reflect adaptation to the intracellular niche. Here, we have investigated the function of one of these enzymes, a putative 2,4-dienoyl-coenzyme A (CoA) reductase (DECR), which is specifically required for the β-oxidation of polyunsaturated fatty acids. The Leishmania DECR shows close homology to bacterial DECR proteins, suggesting that it was acquired by lateral gene transfer. It is present in other trypanosomatids that have obligate intracellular stages (i.e., Trypanosoma cruzi and Angomonas) but is absent from dixenous parasites with an exclusively extracellular lifestyle (i.e., Trypanosoma brucei). A DECR-green fluorescent protein (GFP) fusion protein was localized to the mitochondrion in both promastigote and amastigote stages, and the levels of expression increased in the latter stages. A Leishmania major Δdecr null mutant was unable to catabolize unsaturated fatty acids and accumulated the intermediate 2,4-decadienoyl-CoA, confirming DECR's role in β-oxidation. Strikingly, the L. major Δdecr mutant was unable to survive in macrophages and was avirulent in BALB/c mice. These findings suggest that β-oxidation of polyunsaturated fatty acids is essential for intracellular parasite survival and that the bacterial origin of key enzymes in this pathway could be exploited in developing new therapies.IMPORTANCE The Trypanosomatidae are protozoan parasites that infect insects, plants, and animals and have evolved complex monoxenous (single host) and dixenous (two hosts) lifestyles. A number of species of Trypanosomatidae, including Leishmania spp., have evolved the capacity to survive within intracellular niches in vertebrate hosts. The adaptations, metabolic and other, that are associated with development of intracellular lifestyles remain poorly defined. We show that genomes of Leishmania and Trypanosomatidae that can survive intracellularly encode a 2,4-dienoyl-CoA reductase that is involved in catabolism of a subclass of fatty acids. The trypanosomatid enzyme shows closest similarity to the corresponding bacterial enzymes and is located in the mitochondrion and essential for intracellular growth of Leishmania The findings suggest that acquisition of this gene by lateral gene transfer from bacteria by ancestral monoxenous Trypanosomatidae likely contributed to the development of a dixenous lifestyle of these parasites.
Collapse
|
17
|
Blomme A, Ford CA, Mui E, Patel R, Ntala C, Jamieson LE, Planque M, McGregor GH, Peixoto P, Hervouet E, Nixon C, Salji M, Gaughan L, Markert E, Repiscak P, Sumpton D, Blanco GR, Lilla S, Kamphorst JJ, Graham D, Faulds K, MacKay GM, Fendt SM, Zanivan S, Leung HY. 2,4-dienoyl-CoA reductase regulates lipid homeostasis in treatment-resistant prostate cancer. Nat Commun 2020; 11:2508. [PMID: 32427840 PMCID: PMC7237503 DOI: 10.1038/s41467-020-16126-7] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 03/24/2020] [Indexed: 12/21/2022] Open
Abstract
Despite the clinical success of Androgen Receptor (AR)-targeted therapies, reactivation of AR signalling remains the main driver of castration-resistant prostate cancer (CRPC) progression. In this study, we perform a comprehensive unbiased characterisation of LNCaP cells chronically exposed to multiple AR inhibitors (ARI). Combined proteomics and metabolomics analyses implicate an acquired metabolic phenotype common in ARI-resistant cells and associated with perturbed glucose and lipid metabolism. To exploit this phenotype, we delineate a subset of proteins consistently associated with ARI resistance and highlight mitochondrial 2,4-dienoyl-CoA reductase (DECR1), an auxiliary enzyme of beta-oxidation, as a clinically relevant biomarker for CRPC. Mechanistically, DECR1 participates in redox homeostasis by controlling the balance between saturated and unsaturated phospholipids. DECR1 knockout induces ER stress and sensitises CRPC cells to ferroptosis. In vivo, DECR1 deletion impairs lipid metabolism and reduces CRPC tumour growth, emphasizing the importance of DECR1 in the development of treatment resistance.
Collapse
Affiliation(s)
- Arnaud Blomme
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Catriona A Ford
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Ernest Mui
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Rachana Patel
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Chara Ntala
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Lauren E Jamieson
- Centre for Molecular Nanometrology, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium
| | - Grace H McGregor
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Paul Peixoto
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, 25000, Besançon, France
- EPIGENExp (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
- DIMACELL Dispositif Interrégional d'Imagerie Cellulaire, Dijon, France
| | - Eric Hervouet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, 25000, Besançon, France
- EPIGENExp (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
- DIMACELL Dispositif Interrégional d'Imagerie Cellulaire, Dijon, France
| | - Colin Nixon
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Mark Salji
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Luke Gaughan
- Northern Institute for Cancer Research, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Elke Markert
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Peter Repiscak
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - David Sumpton
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | | | - Sergio Lilla
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Jurre J Kamphorst
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Duncan Graham
- Centre for Molecular Nanometrology, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Karen Faulds
- Centre for Molecular Nanometrology, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Gillian M MacKay
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium
| | - Sara Zanivan
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Hing Y Leung
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
| |
Collapse
|
18
|
Karshovska E, Wei Y, Subramanian P, Mohibullah R, Geißler C, Baatsch I, Popal A, Corbalán Campos J, Exner N, Schober A. HIF-1α (Hypoxia-Inducible Factor-1α) Promotes Macrophage Necroptosis by Regulating miR-210 and miR-383. Arterioscler Thromb Vasc Biol 2020; 40:583-596. [PMID: 31996026 DOI: 10.1161/atvbaha.119.313290] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Inflammatory activation changes the mitochondrial function of macrophages from oxidative phosphorylation to reactive oxygen species production, which may promote necrotic core formation in atherosclerotic lesions. In hypoxic and cancer cells, HIF-1α (hypoxia-inducible factor) promotes oxygen-independent energy production by microRNAs. Therefore, we studied the role of HIF-1α in the regulation of macrophage energy metabolism in the context of atherosclerosis. Approach and Results: Myeloid cell-specific deletion of Hif1a reduced atherosclerosis and necrotic core formation by limiting macrophage necroptosis in apolipoprotein E-deficient mice. In inflammatory bone marrow-derived macrophages, deletion of Hif1a increased oxidative phosphorylation, ATP levels, and the expression of genes encoding mitochondrial proteins and reduced reactive oxygen species production and necroptosis. microRNA expression profiling showed that HIF-1α upregulates miR-210 and downregulates miR-383 levels in lesional macrophages and inflammatory bone marrow-derived macrophages. In contrast to miR-210, which inhibited oxidative phosphorylation and enhanced mitochondrial reactive oxygen species production, miR-383 increased ATP levels and inhibited necroptosis. The effect of miR-210 was due to targeting 2,4-dienoyl-CoA reductase, which is essential in the β oxidation of unsaturated fatty acids. miR-383 affected the DNA damage repair pathway in bone marrow-derived macrophages by targeting poly(ADP-ribose)-glycohydrolase (Parg), which reduced energy consumption and increased cell survival. Blocking the targeting of Parg by miR-383 prevented the protective effect of Hif1a deletion in macrophages on atherosclerosis and necrotic core formation in mice. CONCLUSIONS Our findings unveil a new mechanism by which activation of HIF-1α in inflammatory macrophages increases necroptosis through microRNA-mediated ATP depletion, thus increasing atherosclerosis by necrotic core formation.
Collapse
Affiliation(s)
- Ela Karshovska
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.).,DZHK, German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (E.K., Y.W., A.S.)
| | - Yuanyuan Wei
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.).,DZHK, German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (E.K., Y.W., A.S.)
| | - Pallavi Subramanian
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.)
| | - Rokia Mohibullah
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.)
| | - Claudia Geißler
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.)
| | - Isabelle Baatsch
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.)
| | - Aamoun Popal
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.)
| | - Judit Corbalán Campos
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.)
| | - Nicole Exner
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany (N.E.).,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany (N.E.)
| | - Andreas Schober
- From Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany (E.K., Y.W., P.S., R.M., C.G., I.B., A.P., J.C.C., A.S.).,DZHK, German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (E.K., Y.W., A.S.)
| |
Collapse
|
19
|
Mäkelä AM, Hohtola E, Miinalainen IJ, Autio JA, Schmitz W, Niemi KJ, Hiltunen JK, Autio KJ. Mitochondrial 2,4-dienoyl-CoA reductase (Decr) deficiency and impairment of thermogenesis in mouse brown adipose tissue. Sci Rep 2019; 9:12038. [PMID: 31427678 PMCID: PMC6700156 DOI: 10.1038/s41598-019-48562-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/07/2019] [Indexed: 12/17/2022] Open
Abstract
A large number of studies have demonstrated significance of polyunsaturated fatty acids (PUFAs) for human health. However, many aspects on signals translating PUFA-sensing into body homeostasis have remained enigmatic. To shed light on PUFA physiology, we have generated a mouse line defective in mitochondrial dienoyl-CoA reductase (Decr), which is a key enzyme required for β-oxidation of PUFAs. Previously, we have shown that these mice, whose oxidation of saturated fatty acid is intact but break-down of unsaturated fatty acids is blunted, develop severe hypoglycemia during metabolic stresses and fatal hypothermia upon acute cold challenge. In the current work, indirect calorimetry and thermography suggested that cold intolerance of Decr−/− mice is due to failure in maintaining appropriate heat production at least partly due to failure of brown adipose tissue (BAT) thermogenesis. Magnetic resonance imaging, electron microscopy, mass spectrometry and biochemical analysis showed attenuation in activation of lipolysis despite of functional NE-signaling and inappropriate expression of genes contributing to thermogenesis in iBAT when the Decr−/− mice were exposed to cold. We hypothesize that the failure in turning on BAT thermogenesis occurs due to accumulation of unsaturated long-chain fatty acids or their metabolites in Decr−/− mice BAT suppressing down-stream propagation of NE-signaling.
Collapse
Affiliation(s)
- Anne M Mäkelä
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Esa Hohtola
- Department of Ecology and Genetics, University of Oulu, Oulu, Finland
| | | | - Joonas A Autio
- Center for Biosystems Dynamics Research, RIKEN, Kobe, Japan.,Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | | | - Kalle J Niemi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - J Kalervo Hiltunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Kaija J Autio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
20
|
Roussel E, Drolet MC, Lavigne AM, Arsenault M, Couet J. Multiple short-chain dehydrogenases/reductases are regulated in pathological cardiac hypertrophy. FEBS Open Bio 2018; 8:1624-1635. [PMID: 30338214 PMCID: PMC6168690 DOI: 10.1002/2211-5463.12506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/04/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022] Open
Abstract
Cardiac hypertrophy (CH) is an important and independent predictor of morbidity and mortality. Through expression profiling, we recently identified a subset of genes (Dhrs7c, Decr, Dhrs11, Dhrs4, Hsd11b1, Hsd17b10, Hsd17b8, Blvrb, Pecr), all of which are members of the short‐chain dehydrogenase/reductase (SDR) superfamily and are highly expressed in the heart, that were significantly dysregulated in a rat model of CH caused by severe aortic valve regurgitation (AR). Here, we studied their expression in various models of CH, as well as factors influencing their regulation. Among the nine SDR genes studied, all but Hsd11b1 were down‐regulated in CH models (AR rats or mice infused with either isoproterenol or angiotensin II). This regulation showed a clear sex dimorphism, being more evident in males than in females irrespective of CH levels. In neonatal rat cardiomyocytes, we observed that treatment with the α1‐adrenergic receptor agonist phenylephrine mostly reproduced the observations made in CH animals models. Retinoic acid, on the other hand, stimulated the expression of most of the SDR genes studied, suggesting that their expression may be related to cardiomyocyte differentiation. Indeed, levels of expression were found to be higher in the hearts of adult animals than in neonatal cardiomyocytes. In conclusion, we identified a group of genes modulated in animal models of CH and mostly in males. This could be related to the activation of the fetal gene expression program in pathological CH situations, in which these highly expressed genes are down‐regulated in the adult heart.
Collapse
Affiliation(s)
- Elise Roussel
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| | - Marie-Claude Drolet
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| | - Anne-Marie Lavigne
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| | - Marie Arsenault
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| | - Jacques Couet
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| |
Collapse
|
21
|
Pomerantz DJ, Ferdinandusse S, Cogan J, Cooper DN, Reimschisel T, Robertson A, Bican A, McGregor T, Gauthier J, Millington DS, Andrae JLW, Tschannen MR, Helbling DC, Demos WM, Denis S, Wanders RJA, Newman JN, Hamid R, Phillips JA. Clinical heterogeneity of mitochondrial NAD kinase deficiency caused by a NADK2 start loss variant. Am J Med Genet A 2018; 176:692-698. [PMID: 29388319 PMCID: PMC6185736 DOI: 10.1002/ajmg.a.38602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 12/13/2017] [Accepted: 12/16/2017] [Indexed: 11/12/2022]
Abstract
Mitochondrial NAD kinase deficiency (NADK2D, OMIM #615787) is a rare autosomal recessive disorder of NADPH biosynthesis that can cause hyperlysinemia and dienoyl-CoA reductase deficiency (DECRD, OMIM #616034). NADK2 deficiency has been reported in only three unrelated patients. Two had severe, unremitting disease; one died at 4 months and the other at 5 years of age. The third was a 10 year old female with CNS anomalies, ataxia, and incoordination. In two cases mutations in NADK2 have been demonstrated. Here, we report the fourth known case, a 15 year old female with normal intelligence and a mild clinical and biochemical phenotype presumably without DECRD. Her clinical symptoms, which are now stable, became evident at the age of 9 with the onset of decreased visual acuity, bilateral optic atrophy, nystagmus, episodic lower extremity weakness, peripheral neuropathy, and gait abnormalities. Plasma amino acid levels were within normal limits except for mean lysine and proline levels that were 3.7 and 2.5 times the upper limits of normal. Whole exome sequencing (WES) revealed homozygosity for a g.36241900 A>G p. Met1Val start loss mutation in the primary NADK2 transcript (NM_001085411.1) encoding the 442 amino acid isoform. This presumed hypomorphic mutation has not been previously reported and is absent from the v1000GP, EVS, and ExAC databases. Our patient's normal intelligence and stable disease expands the clinical heterogeneity and the prognosis associated with NADK2 deficiency. Our findings also clarify the mechanism underlying NADK2 deficiency and suggest that this disease should be ruled out in cases of hyperlysinemia, especially those with visual loss, and neurological phenotypes.
Collapse
Affiliation(s)
- Daniel J. Pomerantz
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Joy Cogan
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David N. Cooper
- Institute of Medical Genetics, School of Medicine, Heath Park, Cardiff University, Cardiff, United Kingdom
| | - Tyler Reimschisel
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Amy Robertson
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna Bican
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tracy McGregor
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jackie Gauthier
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David S. Millington
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | | | | | | | | | - Simone Denis
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Ronald J. A. Wanders
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - John N. Newman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rizwan Hamid
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John A. Phillips
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | | |
Collapse
|
22
|
Zhang K, Kim H, Fu Z, Qiu Y, Yang Z, Wang J, Zhang D, Tong X, Yin L, Li J, Wu J, Qi NR, Houten SM, Zhang R. Deficiency of the Mitochondrial NAD Kinase Causes Stress-Induced Hepatic Steatosis in Mice. Gastroenterology 2018; 154:224-237. [PMID: 28923496 PMCID: PMC5742027 DOI: 10.1053/j.gastro.2017.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 08/31/2017] [Accepted: 09/10/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The mitochondrial nicotinamide adenine dinucleotide (NAD) kinase (NADK2, also called MNADK) catalyzes phosphorylation of NAD to yield NADP. Little is known about the functions of mitochondrial NADP and MNADK in liver physiology and pathology. We investigated the effects of reduced mitochondrial NADP by deleting MNADK in mice. METHODS We generated MNADK knockout (KO) mice on a C57BL/6NTac background; mice with a wild-type Mnadk gene were used as controls. Some mice were placed on an atherogenic high-fat diet (16% fat, 41% carbohydrate, and 1.25% cholesterol supplemented with 0.5% sodium cholate) or given methotrexate intraperitoneally. We measured rates of fatty acid oxidation in primary hepatocytes using radiolabeled palmitate and in mice using indirect calorimetry. We measured levels of reactive oxygen species in mouse livers and primary hepatocytes. Metabolomic analyses were used to quantify serum metabolites, such as amino acids and acylcarnitines. RESULTS The KO mice had metabolic features of MNADK-deficient patients, such as increased serum concentrations of lysine and C10:2 carnitine. When placed on the atherogenic high-fat diet, the KO mice developed features of nonalcoholic fatty liver disease and had increased levels of reactive oxygen species in livers and primary hepatocytes, compared with control mice. During fasting, the KO mice had a defect in fatty acid oxidation. MNADK deficiency reduced the activation of cAMP-responsive element binding protein-hepatocyte specific and peroxisome proliferator-activated receptor alpha, which are transcriptional activators that mediate the fasting response. The activity of mitochondrial sirtuins was reduced in livers of the KO mice. Methotrexate inhibited the catalytic activity of MNADK in hepatocytes and in livers in mice with methotrexate injection. In mice given injections of methotrexate, supplementation of a diet with nicotinamide riboside, an NAD precursor, replenished hepatic NADP and protected the mice from hepatotoxicity, based on markers such as increased level of serum alanine aminotransferase. CONCLUSION MNADK facilitates fatty acid oxidation, counteracts oxidative damage, maintains mitochondrial sirtuin activity, and prevents metabolic stress-induced non-alcoholic fatty liver disease in mice.
Collapse
Affiliation(s)
- Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan; Department of Microbiology, Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan.
| | - Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhiyao Fu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yining Qiu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhao Yang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jiemei Wang
- College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Deqiang Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Xin Tong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lei Yin
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jing Li
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Jianmei Wu
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Nathan R. Qi
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sander M. Houten
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan.
| |
Collapse
|
23
|
Wiśniewska A, Olszanecki R, Totoń-Żurańska J, Kuś K, Stachowicz A, Suski M, Gębska A, Gajda M, Jawień J, Korbut R. Anti-Atherosclerotic Action of Agmatine in ApoE-Knockout Mice. Int J Mol Sci 2017; 18:ijms18081706. [PMID: 28777310 PMCID: PMC5578096 DOI: 10.3390/ijms18081706] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is an inflammatory disease in which dysfunction of mitochondria play an important role, and disorders of lipid management intensify this process. Agmatine, an endogenous polyamine formed by decarboxylation of arginine, exerts a protective effect on mitochondria and modulates fatty acid metabolism. We investigated the effect of exogenous agmatine on the development of atherosclerosis and changes in lipid profile in apolipoprotein E knockout (apoE-/-) mice. Agmatine caused an approximate 40% decrease of atherosclerotic lesions, as estimated by en face and cross-section methods with an influence on macrophage but not on smooth muscle content in the plaques. Agmatine treatment did not changed gelatinase activity within the plaque area. What is more, the action of agmatine was associated with an increase in the number of high density lipoproteins (HDL) in blood. Real-Time PCR analysis showed that agmatine modulates liver mRNA levels of many factors involved in oxidation of fatty acid and cholesterol biosynthesis. Two-dimensional electrophoresis coupled with mass spectrometry identified 27 differentially expressed mitochondrial proteins upon agmatine treatment in the liver of apoE-/- mice, mostly proteins related to metabolism and apoptosis. In conclusion, prolonged administration of agmatine inhibits atherosclerosis in apoE-/- mice; however, the exact mechanisms linking observed changes and elevations of HDL plasma require further investigation.
Collapse
Affiliation(s)
- Anna Wiśniewska
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Rafał Olszanecki
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Justyna Totoń-Żurańska
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Katarzyna Kuś
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Aneta Stachowicz
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Maciej Suski
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Anna Gębska
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Mariusz Gajda
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland.
| | - Jacek Jawień
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Ryszard Korbut
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| |
Collapse
|
24
|
The genomic landscape of evolutionary convergence in mammals, birds and reptiles. Nat Ecol Evol 2017; 1:41. [PMID: 28812724 DOI: 10.1038/s41559-016-0041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 11/23/2016] [Indexed: 01/11/2023]
Abstract
Many lineage-defining (nodal) mutations possess high functionality. However, differentiating adaptive nodal mutations from those that are functionally compensated remains challenging. To address this challenge, we identified functional nodal mutations (fNMs) in ~3,400 nuclear DNA (nDNA) and 4 mitochondrial DNA (mtDNA) protein structures from 91 and 1,003 species, respectively, representing the entire mammalian, bird and reptile phylogeny. A screen for candidate compensatory mutations among co-occurring amino acid changes in close structural proximity revealed that such compensated fNMs encompass 37% and 27% of the mtDNA and nDNA datasets, respectively. Analysis of the remaining (non-compensated) mutations, which are enriched for adaptive mutations, showed that birds and mammals share most such recurrent fNMs (N = 51). Among the latter, we discovered mutations in thermoregulation-related genes. These represent the best candidates to explain the molecular basis of convergent body thermoregulation in birds and mammals. Our analysis reveals the landscape of possible mutational compensation and convergence in amniote phylogeny.
Collapse
|
25
|
Roifman M, Choufani S, Turinsky AL, Drewlo S, Keating S, Brudno M, Kingdom J, Weksberg R. Genome-wide placental DNA methylation analysis of severely growth-discordant monochorionic twins reveals novel epigenetic targets for intrauterine growth restriction. Clin Epigenetics 2016; 8:70. [PMID: 27330572 PMCID: PMC4915063 DOI: 10.1186/s13148-016-0238-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/12/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR), which refers to reduced fetal growth in the context of placental insufficiency, is etiologically heterogeneous. IUGR is associated not only with perinatal morbidity and mortality but also with adult-onset disorders, such as cardiovascular disease and diabetes, posing a major health burden. Placental epigenetic dysregulation has been proposed as one mechanism that causes IUGR; however, the spectrum of epigenetic pathophysiological mechanisms leading to IUGR remains to be elucidated. Monozygotic monochorionic twins are particularly affected by IUGR, in the setting of severe discordant growth. Because monozygotic twins have the same genotype at conception and a shared maternal environment, they provide an ideal model system for studying epigenetic dysregulation of the placenta. RESULTS We compared genome-wide placental DNA methylation patterns of severely growth-discordant twins to identify novel candidate genes for IUGR. Snap-frozen placental samples for eight severely growth-discordant monozygotic monochorionic twin pairs were obtained at delivery from each twin. A high-resolution DNA methylation array platform was used to identify methylation differences between IUGR and normal twins. Our analysis revealed differentially methylated regions in the promoters of eight genes: DECR1, ZNF300, DNAJA4, CCL28, LEPR, HSPA1A/L, GSTO1, and GNE. The largest methylation differences between the two groups were in the promoters of DECR1 and ZNF300. The significance of these group differences was independently validated by bisulfite pyrosequencing, implicating aberrations in fatty acid beta oxidation and transcriptional regulation, respectively. Further analysis of the array data identified methylation changes most prominently affecting the Wnt and cadherin pathways in the IUGR cohort. CONCLUSIONS Our results suggest that IUGR in monozygotic twins is associated with impairments in lipid metabolism and transcriptional regulation as well as cadherin and Wnt signaling. We show that monozygotic monochorionic twins discordant for growth provide a useful model to study one type of the epigenetic placental dysregulation that drives IUGR.
Collapse
Affiliation(s)
- Maian Roifman
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario Canada ; Department of Paediatrics, University of Toronto, Toronto, Ontario Canada ; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada ; The Prenatal and Medical Genetics Program, Department of Obstetrics and Gynaecology, Mount Sinai Hospital, Toronto, Ontario Canada
| | - Sanaa Choufani
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Andrei L Turinsky
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada ; Centre for Computational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Sascha Drewlo
- C.S. Mott Center for Human Growth and Development, Wayne State School of Medicine, Wayne State University, Detroit, MI USA
| | - Sarah Keating
- Maternal-Fetal Medicine Division, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Toronto, Ontario Canada ; Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario Canada
| | - Michael Brudno
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada ; Centre for Computational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada ; Department of Computer Science, University of Toronto, Toronto, Ontario Canada
| | - John Kingdom
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario Canada ; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario Canada
| | - Rosanna Weksberg
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario Canada ; Department of Paediatrics, University of Toronto, Toronto, Ontario Canada ; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada ; Institute of Medical Science, University of Toronto, Toronto, Ontario Canada
| |
Collapse
|
26
|
A Metabolic Signature of Mitochondrial Dysfunction Revealed through a Monogenic Form of Leigh Syndrome. Cell Rep 2015; 13:981-9. [PMID: 26565911 PMCID: PMC4644511 DOI: 10.1016/j.celrep.2015.09.054] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 07/13/2015] [Accepted: 09/18/2015] [Indexed: 11/20/2022] Open
Abstract
A decline in mitochondrial respiration represents the root cause of a large number of inborn errors of metabolism. It is also associated with common age-associated diseases and the aging process. To gain insight into the systemic, biochemical consequences of respiratory chain dysfunction, we performed a case-control, prospective metabolic profiling study in a genetically homogenous cohort of patients with Leigh syndrome French Canadian variant, a mitochondrial respiratory chain disease due to loss-of-function mutations in LRPPRC. We discovered 45 plasma and urinary analytes discriminating patients from controls, including classic markers of mitochondrial metabolic dysfunction (lactate and acylcarnitines), as well as unexpected markers of cardiometabolic risk (insulin and adiponectin), amino acid catabolism linked to NADH status (α-hydroxybutyrate), and NAD+ biosynthesis (kynurenine and 3-hydroxyanthranilic acid). Our study identifies systemic, metabolic pathway derangements that can lie downstream of primary mitochondrial lesions, with implications for understanding how the organelle contributes to rare and common diseases.
Collapse
|
27
|
Houten SM, Violante S, Ventura FV, Wanders RJA. The Biochemistry and Physiology of Mitochondrial Fatty Acid β-Oxidation and Its Genetic Disorders. Annu Rev Physiol 2015; 78:23-44. [PMID: 26474213 DOI: 10.1146/annurev-physiol-021115-105045] [Citation(s) in RCA: 558] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondrial fatty acid β-oxidation (FAO) is the major pathway for the degradation of fatty acids and is essential for maintaining energy homeostasis in the human body. Fatty acids are a crucial energy source in the postabsorptive and fasted states when glucose supply is limiting. But even when glucose is abundantly available, FAO is a main energy source for the heart, skeletal muscle, and kidney. A series of enzymes, transporters, and other facilitating proteins are involved in FAO. Recessively inherited defects are known for most of the genes encoding these proteins. The clinical presentation of these disorders may include hypoketotic hypoglycemia, (cardio)myopathy, arrhythmia, and rhabdomyolysis and illustrates the importance of FAO during fasting and in hepatic and (cardio)muscular function. In this review, we present the current state of knowledge on the biochemistry and physiological functions of FAO and discuss the pathophysiological processes associated with FAO disorders.
Collapse
Affiliation(s)
- Sander M Houten
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; ,
| | - Sara Violante
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; ,
| | - Fatima V Ventura
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences, iMed.ULisboa, 1649-003 Lisboa, Portugal; .,Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisboa, Portugal
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, University of Amsterdam, 1100 DE Amsterdam, The Netherlands; .,Department of Pediatrics, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
28
|
Vähätalo LH, Ruohonen ST, Mäkelä S, Kovalainen M, Huotari A, Mäkelä KA, Määttä JA, Miinalainen I, Gilsbach R, Hein L, Ailanen L, Mattila M, Eerola K, Röyttä M, Ruohonen S, Herzig KH, Savontaus E. Neuropeptide Y in the noradrenergic neurones induces obesity and inhibits sympathetic tone in mice. Acta Physiol (Oxf) 2015; 213:902-19. [PMID: 25482272 DOI: 10.1111/apha.12436] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/21/2014] [Accepted: 11/30/2014] [Indexed: 12/21/2022]
Abstract
AIM Neuropeptide Y (NPY) co-localized with noradrenaline in central and sympathetic nervous systems seems to play a role in the control of energy metabolism. In this study, the aim was to elucidate the effects and pathophysiological mechanisms of increased NPY in catecholaminergic neurones on accumulation of body adiposity. METHODS Transgenic mice overexpressing NPY under the dopamine-beta-hydroxylase promoter (OE-NPY(DβH) ) and wild-type control mice were followed for body weight gain and body fat content. Food intake, energy expenditure, physical activity, body temperature, serum lipid content and markers of glucose homoeostasis were monitored. Thermogenic and lipolytic responses in adipose tissues, and urine catecholamine and tissue catecholamine synthesizing enzyme levels were analysed as indices of sympathetic tone. RESULTS Homozygous OE-NPY(DβH) mice showed significant obesity accompanied with impaired glucose tolerance and insulin resistance. Increased adiposity was explained by neither increased food intake or fat absorption nor by decreased total energy expenditure or physical activity. Adipocyte hypertrophy and decreased circulating lipid levels suggested decreased lipolysis and increased lipid uptake. Brown adipose tissue thermogenic capacity was decreased and brown adipocytes filled with lipids. Enhanced response to adrenergic stimuli, downregulation of catecholamine synthesizing enzyme expressions in the brainstem and lower adrenaline excretion supported the notion of low basal catecholaminergic activity. CONCLUSION Increased NPY in catecholaminergic neurones induces obesity that seems to be a result of preferential fat storage. These results support the role of NPY as a direct effector in peripheral tissues and an inhibitor of sympathetic activity in the pathogenesis of obesity.
Collapse
Affiliation(s)
- L. H. Vähätalo
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Drug Research Doctoral Program; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - S. T. Ruohonen
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - S. Mäkelä
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - M. Kovalainen
- Faculty of Health Sciences; School of Pharmacy; Pharmaceutical Technology; University of Eastern Finland; Kuopio Finland
- Institute of Biomedicine and Biocenter of Oulu; University of Oulu; Oulu Finland
| | - A. Huotari
- Faculty of Health Sciences; School of Pharmacy; Pharmaceutical Technology; University of Eastern Finland; Kuopio Finland
- Institute of Biomedicine and Biocenter of Oulu; University of Oulu; Oulu Finland
| | - K. A. Mäkelä
- Institute of Biomedicine and Biocenter of Oulu; University of Oulu; Oulu Finland
| | - J. A. Määttä
- Turku Center for Disease Modeling; University of Turku; Turku Finland
- Department of Cell Biology and Anatomy; Institute of Biomedicine; University of Turku; Turku Finland
| | - I. Miinalainen
- Biocenter Oulu Electron Microscopy Core Facility; University of Oulu; Oulu Finland
| | - R. Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology and BIOSS Centre for Biological Signalling Studies; University of Freiburg; Freiburg Germany
| | - L. Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology and BIOSS Centre for Biological Signalling Studies; University of Freiburg; Freiburg Germany
| | - L. Ailanen
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Drug Research Doctoral Program; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - M. Mattila
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Drug Research Doctoral Program; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - K. Eerola
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - M. Röyttä
- Department of Pathology; University of Turku; Turku Finland
| | - S. Ruohonen
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku; Turku Finland
| | - K. -H. Herzig
- Institute of Biomedicine and Biocenter of Oulu; University of Oulu; Oulu Finland
- Medical Research Center Oulu and Oulu University Hospital; Oulu Finland
| | - E. Savontaus
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
- Unit of Clinical Pharmacology; Turku University Hospital; Turku Finland
| |
Collapse
|
29
|
Fat mass- and obesity-associated gene Fto affects the dietary response in mouse white adipose tissue. Sci Rep 2015; 5:9233. [PMID: 25782772 PMCID: PMC4363842 DOI: 10.1038/srep09233] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/25/2015] [Indexed: 12/18/2022] Open
Abstract
Common variants of human fat mass- and obesity-associated gene Fto have been linked with higher body mass index, but the biological explanation for the link has remained obscure. Recent findings suggest that these variants affect the homeobox protein IRX3. Here we report that FTO has a role in white adipose tissue which modifies its response to high-fat feeding. Wild type and Fto-deficient mice were exposed to standard or high-fat diet for 16 weeks after which metabolism, behavior and white adipose tissue morphology were analyzed together with adipokine levels and relative expression of genes regulating white adipose tissue adipogenesis and Irx3. Our results indicate that Fto deficiency increases the expression of genes related to adipogenesis preventing adipocytes from becoming hypertrophic after high-fat diet. In addition, we report a novel finding of increased Irx3 expression in Fto-deficient mice after high-fat feeding indicating a complex link between FTO, IRX3 and fat metabolism.
Collapse
|
30
|
Onwukwe GU, Kursula P, Koski MK, Schmitz W, Wierenga RK. Human Δ3,Δ2-enoyl-CoA isomerase, type 2: a structural enzymology study on the catalytic role of its ACBP domain and helix-10. FEBS J 2015; 282:746-68. [DOI: 10.1111/febs.13179] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Goodluck U. Onwukwe
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine; University of Oulu; Finland
| | - Petri Kursula
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine; University of Oulu; Finland
- Department of Biomedicine; University of Bergen; Norway
| | - M. Kristian Koski
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine; University of Oulu; Finland
| | - Werner Schmitz
- Theodor Boveri Institute of Biosciences (Biocenter); University of Würzburg; Germany
| | - Rik K. Wierenga
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine; University of Oulu; Finland
| |
Collapse
|
31
|
Putri M, Syamsunarno MRAA, Iso T, Yamaguchi A, Hanaoka H, Sunaga H, Koitabashi N, Matsui H, Yamazaki C, Kameo S, Tsushima Y, Yokoyama T, Koyama H, Abumrad NA, Kurabayashi M. CD36 is indispensable for thermogenesis under conditions of fasting and cold stress. Biochem Biophys Res Commun 2015; 457:520-5. [PMID: 25596128 DOI: 10.1016/j.bbrc.2014.12.124] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 11/25/2022]
Abstract
Hypothermia can occur during fasting when thermoregulatory mechanisms, involving fatty acid (FA) utilization, are disturbed. CD36/FA translocase is a membrane protein which facilitates membrane transport of long-chain FA in the FA consuming heart, skeletal muscle (SkM) and adipose tissues. It also accelerates uptake of triglyceride-rich lipoprotein by brown adipose tissue (BAT) in a cold environment. In mice deficient for CD36 (CD36(-/-) mice), FA uptake is markedly reduced with a compensatory increase in glucose uptake in the heart and SkM, resulting in lower levels of blood glucose especially during fasting. However, the role of CD36 in thermogenic activity during fasting remains to be determined. In fasted CD36(-/-) mice, body temperature drastically decreased shortly after cold exposure. The hypothermia was accompanied by a marked reduction in blood glucose and in stores of triacylglycerols in BAT and of glycogen in glycolytic SkM. Biodistribution analysis using the FA analogue (125)I-BMIPP and the glucose analogue (18)F-FDG revealed that uptake of FA and glucose was severely impaired in BAT and glycolytic SkM in cold-exposed CD36(-/-) mice. Further, induction of the genes of thermogenesis in BAT was blunted in fasted CD36(-/-) mice after cold exposure. These findings strongly suggest that CD36(-/-) mice exhibit pronounced hypothermia after fasting due to depletion of energy storage in BAT and glycolytic SkM and to reduced supply of energy substrates to these tissues. Our study underscores the importance of CD36 for nutrient homeostasis to survive potentially life-threatening challenges, such as cold and starvation.
Collapse
Affiliation(s)
- Mirasari Putri
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Public Health, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Mas Rizky A A Syamsunarno
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Biochemistry, Universitas Padjadjaran, Jl. Raya Bandung Sumedang KM 21, Jatinangor, West Java 45363, Indonesia
| | - Tatsuya Iso
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| | - Aiko Yamaguchi
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hirofumi Hanaoka
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hiroaki Sunaga
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Norimichi Koitabashi
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Chiho Yamazaki
- Department of Public Health, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Satomi Kameo
- Department of Public Health, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Tomoyuki Yokoyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hiroshi Koyama
- Department of Public Health, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Nada A Abumrad
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Masahiko Kurabayashi
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
32
|
Justus J, Weigand E. The effect of a moderate zinc deficiency and dietary fat source on the activity and expression of the Δ(3)Δ (2)-enoyl-CoA isomerase in the liver of growing rats. Biol Trace Elem Res 2014; 158:365-75. [PMID: 24682920 DOI: 10.1007/s12011-014-9940-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/09/2014] [Indexed: 10/25/2022]
Abstract
Auxiliary enzymes participate in β-oxidation of unsaturated fatty acids. The objective of the study was to investigate the impact of a moderate zinc deficiency and a high intake of polyunsaturated fat on Δ(3)Δ(2)-enoyl-CoA isomerase (ECI) in the liver and other tissues. Five groups of eight weanling rats each were fed moderately zinc-deficient (ZD) or zinc-adequate (ZA) semisynthetic diets (7 or 50 mg Zn/kg) enriched with 22 % cocoa butter (CB) or 22 % safflower oil (SO) for 4 weeks: (1) ZD-CB, fed free choice; (2) ZA-CBR, ZA-CB diet fed in equivalent amounts consumed by the ZD-CB group; (3) ZD-SO, fed free choice; (4) ZA-SOR, ZA-SO diet fed in equivalent amounts consumed by the ZD-SO group; and (5) ZA-SO, fed free choice. Growth and Zn status markers were markedly reduced in the ZD groups. ECI activity in the liver of the animals fed the ZD- and ZA-SO diets were significantly higher (approximately 2- and 3-fold, respectively) as compared with the CB-fed animals, whereas activities in extrahepatic tissues (kidneys, heart, skeletal muscle, testes, adipose tissue) were not altered by dietary treatments. Transcript levels of the mitochondrial Eci gene in the liver did not significantly differ between ZD and ZA rats, but were 1.6-fold higher in the ZA-SO- than in the ZD-CB-fed animals (P < 0.05). It is concluded that diets enriched with safflower oil as a source high in linoleic acid induce markedly increased hepatic ECI activities and that a moderate Zn deficiency does not affect transcription of the mitochondrial Eci gene in the liver.
Collapse
Affiliation(s)
- Jennifer Justus
- Dussmann Service Deutschland GmbH, Frankfurt am Main, Germany
| | | |
Collapse
|
33
|
Houten SM, Denis S, Te Brinke H, Jongejan A, van Kampen AHC, Bradley EJ, Baas F, Hennekam RCM, Millington DS, Young SP, Frazier DM, Gucsavas-Calikoglu M, Wanders RJA. Mitochondrial NADP(H) deficiency due to a mutation in NADK2 causes dienoyl-CoA reductase deficiency with hyperlysinemia. Hum Mol Genet 2014; 23:5009-16. [PMID: 24847004 DOI: 10.1093/hmg/ddu218] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dienoyl-CoA reductase (DECR) deficiency with hyperlysinemia is a rare disorder affecting the metabolism of polyunsaturated fatty acids and lysine. The molecular basis of this condition is currently unknown. We describe a new case with failure to thrive, developmental delay, lactic acidosis and severe encephalopathy suggestive of a mitochondrial disorder. Exome sequencing revealed a causal mutation in NADK2. NADK2 encodes the mitochondrial NAD kinase, which is crucial for NADP biosynthesis evidenced by decreased mitochondrial NADP(H) levels in patient fibroblasts. DECR and also the first step in lysine degradation are performed by NADP-dependent oxidoreductases explaining their in vivo deficiency. DECR activity was also deficient in lysates of patient fibroblasts and could only be rescued by transfecting patient cells with functional NADK2. Thus NADPH is not only crucial as a cosubstrate, but can also act as a molecular chaperone that activates and stabilizes enzymes. In addition to polyunsaturated fatty acid oxidation and lysine degradation, NADPH also plays a role in various other mitochondrial processes. We found decreased oxygen consumption and increased extracellular acidification in patient fibroblasts, which may explain why the disease course is consistent with clinical criteria for a mitochondrial disorder. We conclude that DECR deficiency with hyperlysinemia is caused by mitochondrial NADP(H) deficiency due to a mutation in NADK2.
Collapse
Affiliation(s)
- Sander M Houten
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Department of Pediatrics, Emma Children's Hospital, Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY 10029, USA
| | - Simone Denis
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases
| | - Heleen Te Brinke
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases
| | - Aldo Jongejan
- Bioinformatics Laboratory, Clinical Epidemiology, Biostatistics and Bioinformatics and
| | - Antoine H C van Kampen
- Bioinformatics Laboratory, Clinical Epidemiology, Biostatistics and Bioinformatics and Biosystems Data Analysis Group, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Edward J Bradley
- Department of Genome Analysis, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Frank Baas
- Department of Genome Analysis, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | | | - David S Millington
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Sarah P Young
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Dianne M Frazier
- Division of Genetics and Metabolism, Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| | - Muge Gucsavas-Calikoglu
- Division of Genetics and Metabolism, Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| | - Ronald J A Wanders
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Department of Pediatrics, Emma Children's Hospital
| |
Collapse
|
34
|
Syamsunarno MRAA, Iso T, Yamaguchi A, Hanaoka H, Putri M, Obokata M, Sunaga H, Koitabashi N, Matsui H, Maeda K, Endo K, Tsushima Y, Yokoyama T, Kurabayashi M. Fatty acid binding protein 4 and 5 play a crucial role in thermogenesis under the conditions of fasting and cold stress. PLoS One 2014; 9:e90825. [PMID: 24603714 PMCID: PMC3946242 DOI: 10.1371/journal.pone.0090825] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/04/2014] [Indexed: 12/30/2022] Open
Abstract
Hypothermia is rapidly induced during cold exposure when thermoregulatory mechanisms, including fatty acid (FA) utilization, are disturbed. FA binding protein 4 (FABP4) and FABP5, which are abundantly expressed in adipose tissues and macrophages, have been identified as key molecules in the pathogenesis of overnutrition-related diseases, such as insulin resistance and atherosclerosis. We have recently shown that FABP4/5 are prominently expressed in capillary endothelial cells in the heart and skeletal muscle and play a crucial role in FA utilization in these tissues. However, the role of FABP4/5 in thermogenesis remains to be determined. In this study, we showed that thermogenesis is severely impaired in mice lacking both FABP4 and FABP5 (DKO mice), as manifested shortly after cold exposure during fasting. In DKO mice, the storage of both triacylglycerol in brown adipose tissue (BAT) and glycogen in skeletal muscle (SkM) was nearly depleted after fasting, and a biodistribution analysis using 125I-BMIPP revealed that non-esterified FAs (NEFAs) are not efficiently taken up by BAT despite the robustly elevated levels of serum NEFAs. In addition to the severe hypoglycemia observed in DKO mice during fasting, cold exposure did not induce the uptake of glucose analogue 18F-FDG by BAT. These findings strongly suggest that DKO mice exhibit pronounced hypothermia after fasting due to the depletion of energy storage in BAT and SkM and the reduced supply of energy substrates to these tissues. In conclusion, FABP4/5 play an indispensable role in thermogenesis in BAT and SkM. Our study underscores the importance of FABP4/5 for overcoming life-threatening environments, such as cold and starvation.
Collapse
Affiliation(s)
| | - Tatsuya Iso
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
- Education and Research Support Center, Gunma University Graduate School of Medicine, Gunma, Japan
- * E-mail:
| | - Aiko Yamaguchi
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hirofumi Hanaoka
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Mirasari Putri
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
- Department of Public Health, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Masaru Obokata
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroaki Sunaga
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Gunma, Japan
| | - Norimichi Koitabashi
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Gunma, Japan
| | - Kazuhisa Maeda
- Department of Complementary and Alternative Medicine, Graduate School of Medicine, Osaka University Hospital, Osaka, Japan
| | - Keigo Endo
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Tomoyuki Yokoyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Gunma, Japan
| | - Masahiko Kurabayashi
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
- Education and Research Support Center, Gunma University Graduate School of Medicine, Gunma, Japan
| |
Collapse
|
35
|
O’Neil D, Mendez-Figueroa H, Mistretta TA, Su C, Lane RH, Aagaard KM. Dysregulation of Npas2 leads to altered metabolic pathways in a murine knockout model. Mol Genet Metab 2013; 110:378-87. [PMID: 24067359 PMCID: PMC3874417 DOI: 10.1016/j.ymgme.2013.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 08/27/2013] [Indexed: 12/15/2022]
Abstract
In our primate model of maternal high fat diet exposure, we have described that fetal epigenomic modifications to the peripheral circadian Npas2 are associated with persistent alterations in fetal hepatic metabolism and non-alcoholic fatty liver. As the interaction of circadian response with metabolism is not well understood, we employed a murine knockout model to characterize the molecular mechanisms with which Npas2 reprograms the fetal hepatic metabolic response. cDNA was generated from Npas2-/- and +/+ (wild type) livers at day 2 (newborn) and at 25 weeks (adult) of life. Newborn samples were analyzed by exon array (n = 3/cohort). Independent pathway analysis software determined that the primary dysregulated pathway(s) in the Npas2-/- animals uniformly converged on lipid metabolism. Of particular interest, Ppargc1a, which integrates circadian and metabolism pathways, was significantly (p < .01) over expressed in newborn (1.7 fold) and adult (1.8 fold) Npas2-/- animals. These findings are consistent with an essential role for Npas2 in programming the peripheral circadian response and hepatic metabolism, which has not been previously described.
Collapse
Affiliation(s)
- Derek O’Neil
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine; Baylor College of Medicine; Houston, TX, 77030; USA
- Translational Biology and Molecular Medicine Program; Baylor College of Medicine; Houston, TX, 77030; USA
| | - Hector Mendez-Figueroa
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine; Baylor College of Medicine; Houston, TX, 77030; USA
| | - Toni-Ann Mistretta
- Department of Pathology; Texas Children’s Hospital, Baylor College of Medicine; Houston, TX, 77030; USA
| | - Chunliu Su
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine; Baylor College of Medicine; Houston, TX, 77030; USA
| | - Robert H. Lane
- Department of Pediatrics; University of Utah; Salt Lake City, UT, 84112; USA
| | - Kjersti M. Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine; Baylor College of Medicine; Houston, TX, 77030; USA
- Translational Biology and Molecular Medicine Program; Baylor College of Medicine; Houston, TX, 77030; USA
| |
Collapse
|
36
|
Strand E, Pedersen ER, Svingen GFT, Schartum-Hansen H, Rebnord EW, Bjørndal B, Seifert R, Bohov P, Meyer K, Hiltunen JK, Nordrehaug JE, Nilsen DWT, Berge RK, Nygård O. Dietary intake of n-3 long-chain polyunsaturated fatty acids and risk of myocardial infarction in coronary artery disease patients with or without diabetes mellitus: a prospective cohort study. BMC Med 2013; 11:216. [PMID: 24103380 PMCID: PMC3853070 DOI: 10.1186/1741-7015-11-216] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 09/12/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A beneficial effect of a high n-3 long-chain polyunsaturated fatty acid (LCPUFA) intake has been observed in heart failure patients, who are frequently insulin resistant. We investigated the potential influence of impaired glucose metabolism on the relation between dietary intake of n-3 LCPUFAs and risk of acute myocardial infarction (AMI) in patients with coronary artery disease. METHODS This prospective cohort study was based on the Western Norway B-Vitamin Intervention Trial and included 2,378 patients with coronary artery disease with available baseline glycosylated hemoglobin (HbA1c) and dietary data. Patients were sub-grouped as having no diabetes (HbA1c <5.7%), pre-diabetes (HbA1c ≥5.7%), or diabetes (previous diabetes, fasting baseline serum glucose ≥7.0, or non-fasting glucose ≥11.1 mmol/L). AMI risk was evaluated by Cox regression (age and sex adjusted), comparing the upper versus lower tertile of daily dietary n-3 LCPUFA intake. RESULTS The participants (80% males) had a mean age of 62 and follow-up of 4.8 years. A high n-3 LCPUFA intake was associated with reduced risk of AMI (hazard ratio 0.38, 95%CI 0.18, 0.80) in diabetes patients (median HbA1c = 7.2%), whereas no association was observed in pre-diabetes patients. In patients without diabetes a high intake tended to be associated with an increased risk (hazard ratio1.45, 95%CI 0.84, 2.53), which was significant for fatal AMI (hazard ratio 4.79, 95%CI 1.05, 21.90) and associated with lower HbA1c (mean ± standard deviation 4.55 ±0.68 versus 4.92 ±0.60, P = 0.02). No such differences in HbA1c were observed in those with pre-diabetes or diabetes. CONCLUSIONS A high intake of n-3 LCPUFAs was associated with a reduced risk of AMI, independent of HbA1c, in diabetic patients, but with an increased risk of fatal AMI and lower HbA1c among patients without impaired glucose metabolism. Further studies should investigate whether patients with diabetes may benefit from having a high intake of n-3 LCPUFAs and whether patients with normal glucose tolerance should be careful with a very high intake of these fatty acids. TRIAL REGISTRATION This trial is registered at clinicaltrials.gov as NCT00354081.
Collapse
Affiliation(s)
- Elin Strand
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fu Z, Wang M, Everett A, Lakatta E, Van Eyk J. Can proteomics yield insight into aging aorta? Proteomics Clin Appl 2013; 7:477-89. [PMID: 23788441 DOI: 10.1002/prca.201200138] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 06/13/2013] [Accepted: 06/14/2013] [Indexed: 12/16/2022]
Abstract
The aging aorta exhibits structural and physiological changes that are reflected in the proteome of its component cells types. The advance in proteomic technologies has made it possible to analyze the quantity of proteins associated with the natural history of aortic aging. These alterations reflect the molecular and cellular mechanisms of aging and could provide an opportunity to predict vascular health. This paper focuses on whether discoveries stemming from the application of proteomic approaches of the intact aging aorta or vascular smooth muscle cells can provide useful insights. Although there have been limited studies to date, a number of interesting proteins have been identified that are closely associated with aging in the rat aorta. Such proteins, including milk fat globule-EGF factor 8, matrix metalloproteinase type-2, and vitronectin, could be used as indicators of vascular health, or even explored as therapeutic targets for aging-related vascular diseases.
Collapse
Affiliation(s)
- Zongming Fu
- Department of Pediatrics, The Johns Hopkins University, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
38
|
Ramírez O, Quintanilla R, Varona L, Gallardo D, Díaz I, Pena R, Amills M. DECR1
and ME1
genotypes are associated with lipid composition traits in Duroc pigs. J Anim Breed Genet 2013; 131:46-52. [DOI: 10.1111/jbg.12035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 02/27/2013] [Indexed: 11/29/2022]
Affiliation(s)
- O. Ramírez
- Departament de Ciència Animal i dels Aliments; Facultat de Veterinària; Universitat Autònoma de Barcelona; Bellaterra Spain
| | | | - L. Varona
- Genètica i Millora Animal; IRTA; Lleida Spain
| | - D. Gallardo
- Departament de Ciència Animal i dels Aliments; Facultat de Veterinària; Universitat Autònoma de Barcelona; Bellaterra Spain
| | - I. Díaz
- Centre de Tecnologia de la Carn (IRTA); Monells Spain
| | - R.N. Pena
- Genètica i Millora Animal; IRTA; Lleida Spain
| | - M. Amills
- Departament de Ciència Animal i dels Aliments; Facultat de Veterinària; Universitat Autònoma de Barcelona; Bellaterra Spain
- Center for Research in Agricultural Genomics (CSIC-IRTA-UAB-UB); Universitat Autònoma de Barcelona; Bellaterra Spain
| |
Collapse
|
39
|
Ruotsalainen H, Risteli M, Wang C, Wang Y, Karppinen M, Bergmann U, Kvist AP, Pospiech H, Herzig KH, Myllylä R. The activities of lysyl hydroxylase 3 (LH3) regulate the amount and oligomerization status of adiponectin. PLoS One 2012; 7:e50045. [PMID: 23209641 PMCID: PMC3510199 DOI: 10.1371/journal.pone.0050045] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 10/15/2012] [Indexed: 01/04/2023] Open
Abstract
Lysyl hydroxylase 3 (LH3) has lysyl hydroxylase, galactosyltransferase, and glucosyltransferase activities, which are sequentially required for the formation of glucosylgalactosyl hydroxylysines in collagens. Here we demonstrate for the first time that LH3 also modifies the lysine residues in the collagenous domain of adiponectin, which has important roles in glucose and lipid metabolism and inflammation. Hydroxylation and, especially, glycosylation of the lysine residues of adiponectin have been shown to be essential for the formation of the more active high molecular weight adiponectin oligomers and thus for its function. In cells that totally lack LH3 enzyme, the galactosylhydroxylysine residues of adiponectin were not glucosylated to glucosylgalactosylhydroxylysine residues and the formation of high and middle molecular weight adiponectin oligomers was impaired. Circulating adiponectin levels in mutant mice lacking the lysyl hydroxylase activity of LH3 were significantly reduced, which indicates that LH3 is required for complete modification of lysine residues in adiponectin and the loss of some of the glycosylated hydroxylysine residues severely affects the secretion of adiponectin. LH mutant mice with reduced adiponectin level showed a high fat diet-induced increase in glucose, triglyceride, and LDL-cholesterol levels, hallmarks of the metabolic syndrome in humans. Our results reveal the first indication that LH3 is an important regulator of adiponectin biosynthesis, secretion and activity and thus might be a potential candidate for therapeutic applications in diseases associated with obesity and insulin resistance.
Collapse
|
40
|
Weeghel M, Brinke HT, Lenthe H, Kulik W, Minkler PE, Stoll MSK, Sass JO, Janssen U, Stoffel W, Schwab KO, Wanders RJA, Hoppel CL, Houten SM. Functional redundancy of mitochondrial enoyl‐CoA isomerases in the oxidation of unsaturated fatty acids. FASEB J 2012; 26:4316-26. [DOI: 10.1096/fj.12-206326] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Michel Weeghel
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Heleen te Brinke
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Henk Lenthe
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Wim Kulik
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Paul E. Minkler
- Department of PharmacologyCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Maria S. K. Stoll
- Department of PharmacologyCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Jörn Oliver Sass
- Center for Children's Hospital in FreiburgFreiburgGermany
- Department of Clinical Chemistry and BiochemistryUniversity Children's Hospital ZürichZürichSwitzerland
| | - Uwe Janssen
- Miltenyi Biotec GmbHBergisch GladbachGermany
| | - Wilhelm Stoffel
- Institute of BiochemistryCenter of Molecular Medicine Cologne, Cluster of Excellence, Cellular Stress Response in Aging Related Diseases (CECAD)University of CologneGermany
| | | | - Ronald J. A. Wanders
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
- Department of PediatricsEmma Children's HospitalAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Charles L. Hoppel
- Department of PharmacologyCase Western Reserve University School of MedicineClevelandOhioUSA
- Department of MedicineCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Sander M. Houten
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
- Department of PediatricsEmma Children's HospitalAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
41
|
Goetzman ES. Modeling Disorders of Fatty Acid Metabolism in the Mouse. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 100:389-417. [DOI: 10.1016/b978-0-12-384878-9.00010-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
42
|
Houten SM, Wanders RJA. A general introduction to the biochemistry of mitochondrial fatty acid β-oxidation. J Inherit Metab Dis 2010; 33:469-77. [PMID: 20195903 PMCID: PMC2950079 DOI: 10.1007/s10545-010-9061-2] [Citation(s) in RCA: 657] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Revised: 10/14/2009] [Accepted: 01/28/2010] [Indexed: 12/30/2022]
Abstract
Over the years, the mitochondrial fatty acid β-oxidation (FAO) pathway has been characterised at the biochemical level as well as the molecular biological level. FAO plays a pivotal role in energy homoeostasis, but it competes with glucose as the primary oxidative substrate. The mechanisms behind this so-called glucose-fatty acid cycle operate at the hormonal, transcriptional and biochemical levels. Inherited defects for most of the FAO enzymes have been identified and characterised and are currently included in neonatal screening programmes. Symptoms range from hypoketotic hypoglycaemia to skeletal and cardiac myopathies. The pathophysiology of these diseases is still not completely understood, hampering optimal treatment. Studies of patients and mouse models will contribute to our understanding of the pathogenesis and will ultimately lead to better treatment.
Collapse
Affiliation(s)
- Sander Michel Houten
- Department of Clinical Chemistry, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
43
|
Leonardi R, Rehg JE, Rock CO, Jackowski S. Pantothenate kinase 1 is required to support the metabolic transition from the fed to the fasted state. PLoS One 2010; 5:e11107. [PMID: 20559429 PMCID: PMC2885419 DOI: 10.1371/journal.pone.0011107] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 05/24/2010] [Indexed: 12/25/2022] Open
Abstract
Coenzyme A (CoA) biosynthesis is regulated by the pantothenate kinases (PanK), of which there are four active isoforms. The PanK1 isoform is selectively expressed in liver and accounted for 40% of the total PanK activity in this organ. CoA synthesis was limited using a Pank1(-/-) knockout mouse model to determine whether the regulation of CoA levels was critical to liver function. The elimination of PanK1 reduced hepatic CoA levels, and fasting triggered a substantial increase in total hepatic CoA in both Pank1(-/-) and wild-type mice. The increase in hepatic CoA during fasting was blunted in the Pank1(-/-) mouse, and resulted in reduced fatty acid oxidation as evidenced by abnormally high accumulation of long-chain acyl-CoAs, acyl-carnitines, and triglycerides in the form of lipid droplets. The Pank1(-/-) mice became hypoglycemic during a fast due to impaired gluconeogenesis, although ketogenesis was normal. These data illustrate the importance of PanK1 and elevated liver CoA levels during fasting to support the metabolic transition from glucose utilization and fatty acid synthesis to gluconeogenesis and fatty acid oxidation. The findings also suggest that PanK1 may be a suitable target for therapeutic intervention in metabolic disorders that feature hyperglycemia and hypertriglyceridemia.
Collapse
Affiliation(s)
- Roberta Leonardi
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Jerold E. Rehg
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Charles O. Rock
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Suzanne Jackowski
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|