Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Science ; 378(6618): eabj3510, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36302005

ABSTRACT

Spermidine (SPD) delays age-related pathologies in various organisms. SPD supplementation overcame the impaired immunotherapy against tumors in aged mice by increasing mitochondrial function and activating CD8+ T cells. Treatment of naïve CD8+ T cells with SPD acutely enhanced fatty acid oxidation. SPD conjugated to beads bound to the mitochondrial trifunctional protein (MTP). In the MTP complex, synthesized and purified from Escherichia coli, SPD bound to the α and ß subunits of MTP with strong affinity and allosterically enhanced their enzymatic activities. T cell-specific deletion of the MTP α subunit abolished enhancement of programmed cell death protein 1 (PD-1) blockade immunotherapy by SPD, indicating that MTP is required for SPD-dependent T cell activation.


Subject(s)
CD8-Positive T-Lymphocytes , Mitochondria , Mitochondrial Trifunctional Protein, alpha Subunit , Mitochondrial Trifunctional Protein, beta Subunit , Neoplasms , Spermidine , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Mitochondria/metabolism , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Spermidine/pharmacology , Spermidine/metabolism , Neoplasms/immunology
2.
Proteins ; 90(11): 2001-2005, 2022 11.
Article in English | MEDLINE | ID: mdl-35532281

ABSTRACT

The c subunits, which constitute the c-ring apparatus of the F1 FO -ATPase, could be the main components of the mitochondrial permeability transition pore (mPTP). The well-known modulator of the mPTP formation and opening is the cyclophilin D (CyPD), a peptidyl-prolyl cis-trans isomerase. On the loop, which connects the two hairpin α-helix of c subunit, is present the unique proline residue (Pro40 ) that could be a biological target of CyPD. Indeed, the proline cis-trans isomerization might provide the switch that interconverts the open/closed states of the pore by pulling out the c-ring lipid plug.


Subject(s)
Mitochondrial Permeability Transition Pore , Proline , Adenosine Triphosphatases/metabolism , Peptidyl-Prolyl Isomerase F , Ion Channels , Isomerism , Lipids , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Proline/chemistry , Protein Folding
3.
Pathology ; 54(3): 286-293, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34531036

ABSTRACT

In haematological malignancies, such as malignant lymphoma, reprogramming of fatty acid metabolism favours tumour cell survival and drug resistance. Hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA), an enzyme involved in fatty acid beta-oxidation (FAO), is overexpressed in high-grade lymphoma and is a predictor of poor prognosis in diffuse large B-cell lymphoma (DLBCL). HADHB forms a heterodimer with HADHA and functions as an FAO enzyme together with HADHA; however, the relevance of its expression in malignant lymphoma is unknown. In this study, we investigated the roles and antitumour effects of HADHB expression in malignant lymphoma. Immunohistochemical analysis showed that HADHB was frequently overexpressed in the high-grade lymphoma subtype. HADHB overexpression was observed in 68% (87/128) of DLBCL cases and was an independent predictor of poor prognosis (p=0.001). In vitro analysis demonstrated that HADHB knockdown suppressed cell proliferation in LCL-K and MD901 cells (p<0.05). Additionally, treatment with the FAO inhibitor, ranolazine, increased cell death in control cells compared with that in HADHB knockdown LCL-K and MD901 cells (p<0.01). Cell death was also suppressed by the ferroptosis inhibitor, ferrosatin-1, in LCL-K and MD901 cells (p<0.05). Collectively, these findings provide basic evidence for the development of new cell death-based therapies for refractory malignant lymphoma. We plan to perform prospective studies and preclinical studies using animal models to confirm these results.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Mitochondrial Trifunctional Protein, beta Subunit , Animals , Fatty Acids/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Prognosis , Prospective Studies
4.
Genes (Basel) ; 13(1)2021 12 28.
Article in English | MEDLINE | ID: mdl-35052415

ABSTRACT

Adequate protein nutrition is essential for good health. Effects of protein malnutrition in animals have been widely studied at the mRNA level with the development of DNA microarray technology. Although microRNAs (miRNAs) have attracted attention for their function in regulating gene expression and have been studied in several disciplines, fewer studies have clarified the effects of protein malnutrition on miRNA alterations. The present study aimed to elucidate the relationship between protein malnutrition and miRNAs. Six-week old Wistar male rats were fed a control diet (20% casein) or a low-protein diet (5% casein) for two weeks, and their livers were subjected to both DNA microarray and miRNA array analysis. miR-203 was downregulated and its putative target Hadhb (hydroxyacyl-CoA dehydrogenase ß subunit), known to regulate ß-oxidation of fatty acids, was upregulated by the low-protein diet. In an in vitro experiment, miR-203 or its inhibitor were transfected in HepG2 cells, and the pattern of Hadhb expression was opposite to that of miR-203 expression. In addition, to clarifying the hepatic miRNA profile in response to protein malnutrition, these results showed that a low-protein diet increased Hadhb expression through downregulation of miR-203 and induced ß-oxidation of fatty acids.


Subject(s)
Diet, Protein-Restricted , Gene Expression Regulation , Malnutrition/pathology , MicroRNAs/genetics , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Animals , Hep G2 Cells , Humans , Male , Malnutrition/genetics , Malnutrition/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Rats , Rats, Wistar
5.
Proc Natl Acad Sci U S A ; 117(25): 14365-14375, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513690

ABSTRACT

Proper resolution of inflammation is vital for repair and restoration of homeostasis after tissue damage, and its dysregulation underlies various noncommunicable diseases, such as cardiovascular and metabolic diseases. Macrophages play diverse roles throughout initial inflammation, its resolution, and tissue repair. Differential metabolic reprogramming is reportedly required for induction and support of the various macrophage activation states. Here we show that a long noncoding RNA (lncRNA), lncFAO, contributes to inflammation resolution and tissue repair in mice by promoting fatty acid oxidation (FAO) in macrophages. lncFAO is induced late after lipopolysaccharide (LPS) stimulation of cultured macrophages and in Ly6Chi monocyte-derived macrophages in damaged tissue during the resolution and reparative phases. We found that lncFAO directly interacts with the HADHB subunit of mitochondrial trifunctional protein and activates FAO. lncFAO deletion impairs resolution of inflammation related to endotoxic shock and delays resolution of inflammation and tissue repair in a skin wound. These results demonstrate that by tuning mitochondrial metabolism, lncFAO acts as a node of immunometabolic control in macrophages during the resolution and repair phases of inflammation.


Subject(s)
Fatty Acids/metabolism , Inflammation/immunology , Macrophages/immunology , Mitochondrial Trifunctional Protein, beta Subunit/genetics , RNA, Long Noncoding/metabolism , Animals , Disease Models, Animal , Gene Knockdown Techniques , Humans , Lipopolysaccharides/immunology , Macrophage Activation/genetics , Macrophages/metabolism , Male , Mice , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Oxidation-Reduction , Primary Cell Culture , RNA, Long Noncoding/genetics , Skin/immunology , Skin/injuries , Wound Healing/immunology
6.
J Cell Mol Med ; 24(12): 6833-6845, 2020 06.
Article in English | MEDLINE | ID: mdl-32340079

ABSTRACT

Ketone bodies (KBs) were known to suppress seizure. Untraditionally, neurons were recently reported to utilize fatty acids and produce KBs, but the effect of seizure on neuronal ketogenesis has not been researched. Zinc-α2-glycoprotein (ZAG) was reported to suppress seizure via unclear mechanism. Interestingly, ZAG was involved in fatty acid ß-oxidation and thus may exert anti-epileptic effect by promoting ketogenesis. However, this promotive effect of ZAG on neuronal ketogenesis has not been clarified. In this study, we performed immunoprecipitation and mass spectrometry to identify potential interaction partners with ZAG. The mechanisms of how ZAG translocated into mitochondria were determined by quantitative coimmunoprecipitation after treatment with apoptozole, a heat shock cognate protein 70 (HSC70) inhibitor. ZAG level was modulated by lentivirus in neurons or adeno-associated virus in rat brains. Seizure models were induced by magnesium (Mg2+ )-free artificial cerebrospinal fluid in neurons or intraperitoneal injection of pentylenetetrazole kindling in rats. Ketogenesis was determined by cyclic thio-NADH method in supernatant of neurons or brain homogenate. The effect of peroxisome proliferator-activated receptor γ (PPARγ) on ZAG expression was examined by Western blot, quantitative real-time polymerase chain reaction (qRT-PCR) and chromatin immunoprecipitation qRT-PCR. We found that seizure induced ketogenesis deficiency via a ZAG-dependent mechanism. ZAG entered mitochondria through a HSC70-dependent mechanism, promoted ketogenesis by binding to four ß-subunits of long-chain L-3-hydroxyacyl-CoA dehydrogenase (HADHB) and alleviated ketogenesis impairment in a neuronal seizure model and pentylenetetrazole-kindled epileptic rats. Additionally, PPARγ activation up-regulated ZAG expression by binding to promoter region of AZGP1 gene and promoted ketogenesis through a ZAG-dependent mechanism.


Subject(s)
Ketone Bodies/metabolism , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , Seizures/pathology , Seminal Plasma Proteins/metabolism , Animals , Cells, Cultured , HSC70 Heat-Shock Proteins/metabolism , Male , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , Rats, Sprague-Dawley , Zn-Alpha-2-Glycoprotein
7.
J Biol Chem ; 295(9): 2839-2849, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31980458

ABSTRACT

Understanding the mechanisms by which viruses evade host cell immune defenses is important for developing improved antiviral therapies. In an unusual twist, human cytomegalovirus co-opts the antiviral radical SAM enzyme viperin (virus-inhibitory protein, endoplasmic reticulum-associated, interferon-inducible) to enhance viral infectivity. This process involves translocation of viperin to the mitochondrion, where it binds the ß-subunit (HADHB) of the mitochondrial trifunctional enzyme complex that catalyzes thiolysis of ß-ketoacyl-CoA esters as part of fatty acid ß-oxidation. Here we investigated how the interaction between these two enzymes alters their activities and affects cellular ATP levels. Experiments with purified enzymes indicated that viperin inhibits the thiolase activity of HADHB, but, unexpectedly, HADHB activates viperin, leading to synthesis of the antiviral nucleotide 3'-deoxy-3',4'-didehydro-CTP. Measurements of enzyme activities in lysates prepared from transfected HEK293T cells expressing these enzymes mirrored the findings obtained with purified enzymes. Thus, localizing viperin to mitochondria decreased thiolase activity, and coexpression of HADHB significantly increased viperin activity. Furthermore, targeting viperin to mitochondria also increased the rate at which HADHB is retrotranslocated out of mitochondria and degraded, providing an additional mechanism by which viperin reduces HADHB activity. Targeting viperin to mitochondria decreased cellular ATP levels by more than 50%, consistent with the enzyme disrupting fatty acid catabolism. These results provide biochemical insight into the mechanism by which human cytomegalovirus subverts viperin; they also provide a biochemical rationale for viperin's recently discovered role in regulating thermogenesis in adipose tissues.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Mitochondria/metabolism , Proteins/metabolism , Adenosine Triphosphate/metabolism , Cytomegalovirus/physiology , HEK293 Cells , Humans , Immune Evasion , Mitochondrial Trifunctional Protein, beta Subunit/antagonists & inhibitors , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/physiology , Oxidoreductases Acting on CH-CH Group Donors
8.
Mitochondrion ; 49: 200-205, 2019 11.
Article in English | MEDLINE | ID: mdl-31521624

ABSTRACT

Mitochondrial trifunctional protein (MTP) deficiency is a rare autosomal recessive disorder with several phenotypes. Neuromyopathic form of MTP deficiency is characterized by infantile or juvenile-onset, progressive peripheral neuropathy and rhabdomyolysis. To date, only one Chinese patient harboring homozygous c. 739C>T (p.R247C) in HADHB has been reported. Here, using whole exome sequencing (WES), we identified a compound heterozygote of c.407T>C (p.M136T) and c.421G>A (p.A141T) within HADHB in a Chinese MTP deficiency patient of neuromyopathic form. In vitro cell functional studies were performed to evaluate the effect of mutations on MTP complex expression and subcellular location, which revealed that p.M136T and p.A141T mutations compromised MTP complex stability but not altered subcellular localization, resulting in lower protein level at 37 °C but higher at 30 °C. These results indicated that both mutations were pathogenic through a loss-of-function mechanism and temperature-sensitive leading to their correlation with the mild phenotype. The current study broadens the genetic spectrum of HADHB and highlights the importance of screening fatty acid oxidation deficiency-related gene mutations among patients with intermittent rhabdomyolysis, as in the patient reported here, although extremely rare.


Subject(s)
Genetic Diseases, Inborn/genetics , Loss of Function Mutation , Mitochondrial Trifunctional Protein, beta Subunit/deficiency , Mutation, Missense , Rhabdomyolysis/genetics , Adult , Amino Acid Substitution , Female , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Rhabdomyolysis/metabolism , Rhabdomyolysis/pathology
9.
Br Poult Sci ; 60(2): 115-124, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30698464

ABSTRACT

1. Limiting the growth of adipose tissue in chickens is a major issue in the poultry industry. In chickens, de novo synthesis of lipids occurs primarily in the liver. Thus, it is necessary to understand how fatty acid accumulation in the liver is controlled. The miR-33 is an intronic microRNA (miRNA) of the chicken sterol regulatory element binding transcription factor 2 (SREBF2), which is a master switch in activating many genes involved in the uptake and synthesis of cholesterol, triglycerides, fatty acids and phospholipids. 2. In the current study, the genes CROT and HADHB known to encode enzymes critical for fatty acid oxidation were predicted to be potential targets of miR-33 in chickens via the miRNA target prediction programs 'miRanda' and 'TargetScan'. Co-transfection and dual-luciferase reporter assays showed that the expression of luciferase reporter gene linked to the 3'-untranslated region (3'UTR) of the chicken CROT and HADHB mRNA was down-regulated by overexpression of the chicken miR-33 (P < 0.05). This down-regulation was completely abolished when the predicted miR-33 target sites in the CROT and HADHB 3'UTR were mutated. 3. Transfecting miR-33 mimics into the LMH cells led to a decrease in the mRNA expression of CROT and HADHB (P < 0.01), and this transfection had a similar effect on the proteins (P < 0.05). In contrast, the expression of CROT in primary chicken hepatocytes was up-regulated after transfection with the miR-33 inhibitor LNA-anti-miR-33 (P < 0.05). 4. Using quantitative RT-PCR, it was shown that the expression of miR-33 was increased in the chicken liver from day 0 to day 49 of age, whereas the CROT and HADHB mRNA levels decreased during the same period. 5. These findings support the conclusion that miR-33 might play an important role in lipid metabolism in the chicken liver by negatively regulating the expression of the CROT and HADHB genes, which encode enzymes critical for lipid oxidation.


Subject(s)
Avian Proteins/genetics , Chickens/genetics , Gene Expression Regulation , MicroRNAs/genetics , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Animals , Avian Proteins/metabolism , Base Sequence , Chickens/metabolism , Introns , Liver/metabolism , MicroRNAs/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Plasmids/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
10.
Clin Epigenetics ; 10: 30, 2018.
Article in English | MEDLINE | ID: mdl-29507648

ABSTRACT

Background: DNA methylation is an important epigenetic modification, associated with gene expression. 5-Methylcytosine and 5-hydroxymethylcytosine are two epigenetic hallmarks that maintain the equilibrium of epigenetic reprogramming. Disequilibrium in genomic methylation leads to carcinogenesis. The purpose of this study was to elucidate the epigenetic mechanisms of DNA methylation and hydroxymethylation in the carcinogenesis of colorectal cancer. Methods: Genome-wide patterns of DNA methylation and hydroxymethylation in six paired colorectal tumor tissues and corresponding normal tissues were determined using immunoprecipitation and sequencing. Transcriptional expression was determined by RNA sequencing (RNA-Seq). Groupwise differential methylation regions (DMR), differential hydroxymethylation regions (DhMR), and differentially expressed gene (DEG) regions were identified. Epigenetic biomarkers were screened by integrating DMR, DhMR, and DEGs and confirmed using functional analysis. Results: We identified a genome-wide distinct hydroxymethylation pattern that could be used as an epigenetic biomarker for clearly differentiating colorectal tumor tissues from normal tissues. We identified 59,249 DMRs, 187,172 DhMRs, and 948 DEGs by comparing between tumors and normal tissues. After cross-matching genes containing DMRs or DhMRs with DEGs, we screened seven genes that were aberrantly regulated by DNA methylation in tumors. Furthermore, hypermethylation of the HADHB gene was persistently found to be correlated with downregulation of its transcription in colorectal cancer (CRC). These findings were confirmed in other patients of colorectal cancer. Tumor functional analysis indicated that HADHB reduced cancer cell migration and invasiveness. These findings suggested its possible role as a tumor suppressor gene (TSG). Conclusion: This study reveals the global patterns of methylation and hydroxymethylation in CRC. Several CRC-associated genes were screened with multi-omic analysis. Aberrant methylation and hydroxymethylation were found to be in the carcinogenesis of CRC.


Subject(s)
Colorectal Neoplasms/genetics , Epigenomics/methods , Gene Expression Profiling/methods , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/analysis , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/metabolism , DNA Methylation , Down-Regulation , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Male , Sequence Analysis, DNA , Sequence Analysis, RNA
11.
Mol Cell ; 69(3): 480-492.e7, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395065

ABSTRACT

Fatty acid oxidation (FAO) is crucial for cells to overcome metabolic stress by providing ATP and NADPH. However, the mechanism by which FAO is regulated in tumors remains elusive. Here we show that Nur77 is required for the metabolic adaptation of melanoma cells by protecting FAO. Glucose deprivation activates ERK2 to phosphorylate and induce Nur77 translocation to the mitochondria, where Nur77 binds to TPß, a rate-limiting enzyme in FAO. Although TPß activity is normally inhibited by oxidation under glucose deprivation, the Nur77-TPß association results in Nur77 self-sacrifice to protect TPß from oxidation. FAO is therefore able to maintain NADPH and ATP levels and prevent ROS increase and cell death. The Nur77-TPß interaction further promotes melanoma metastasis by facilitating circulating melanoma cell survival. This study demonstrates a novel regulatory function of Nur77 with linkage of the FAO-NADPH-ROS pathway during metabolic stress, suggesting Nur77 as a potential therapeutic target in melanoma.


Subject(s)
Melanoma/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Animals , Cell Survival/physiology , Fatty Acids/metabolism , Glucose/metabolism , HEK293 Cells , Humans , Lipid Metabolism , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
12.
Cell Rep ; 21(5): 1317-1330, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29091769

ABSTRACT

As an important regulator of macrophage cholesterol efflux and HDL biogenesis, miR-33 is a promising target for treatment of atherosclerosis, and numerous studies demonstrate that inhibition of miR-33 increases HDL levels and reduces plaque burden. However, important questions remain about how miR-33 impacts atherogenesis, including whether this protection is primarily due to direct effects on plaque macrophages or regulation of lipid metabolism in the liver. We demonstrate that miR-33 deficiency in Ldlr-/- mice promotes obesity, insulin resistance, and hyperlipidemia but does not impact plaque development. We further assess how loss of miR-33 or addition of miR-33b in macrophages and other hematopoietic cells impact atherogenesis. Macrophage-specific loss of miR-33 decreases lipid accumulation and inflammation under hyperlipidemic conditions, leading to reduced plaque burden. Therefore, the pro-atherogenic effects observed in miR-33-deficient mice are likely counterbalanced by protective effects in macrophages, which may be the primary mechanism through which anti-miR-33 therapies reduce atherosclerosis.


Subject(s)
Atherosclerosis/pathology , MicroRNAs/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Aorta/pathology , Atherosclerosis/metabolism , Atherosclerosis/veterinary , Blood Glucose/analysis , Cells, Cultured , Cholesterol/metabolism , Cholesterol, HDL/blood , Disease Progression , Gene Regulatory Networks , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Myocardium/metabolism , Myocardium/pathology , Receptors, LDL/deficiency , Receptors, LDL/genetics
13.
J Hum Genet ; 62(9): 809-814, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28515471

ABSTRACT

Mitochondrial trifunctional protein (TFP) deficiency is an inherited metabolic disorder of mitochondrial fatty-acid oxidation. Isolated long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency is often reported in Caucasian countries due to a common mutation. However, the molecular and clinical basis of complete TFP deficiency has not been extensively reported. In this study, 14 Japanese cases (13 families) with complete TFP deficiency, including 9 previously reported cases, were analyzed to clarify the clinical and molecular characteristics of TFP deficiency. The clinical types of the 14 patients were as follows: 12 cases of neonatal (n=7) or myopathic (n=5) types and 2 cases of intermediate type. Peripheral neuropathy was found in four cases and hypocalcemia due to hypoparathyroidism, which is rarely reported in Caucasian patients, had developed in four cases. Maternal hemolysis, elevated liver enzymes and low platelet count syndrome and acute fatty liver of pregnancy were noted in two and one mothers, respectively. Fourteen mutations were identified in 26 alleles in Japanese patients, including two novel mutations (HADHA: c.361C>T, and HADHA-HADHB: g.26233880_ 26248855del), although no common mutations were found. This study suggests that the molecular and clinical aspects of Japanese patients with TFP deficiencies differ from those of Caucasian patients.


Subject(s)
Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/genetics , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/diagnosis , Nervous System Diseases/genetics , Rhabdomyolysis/diagnosis , Rhabdomyolysis/genetics , Adolescent , Asian People/genetics , Child , Child, Preschool , Enzyme Activation , Family , Female , Genetic Testing , Genotype , Humans , Infant , Infant, Newborn , Male , Mitochondrial Trifunctional Protein/genetics , Mitochondrial Trifunctional Protein, alpha Subunit/genetics , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Mutation , White People/genetics
14.
Am J Physiol Heart Circ Physiol ; 311(1): H64-75, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27199118

ABSTRACT

Myocardial fatty acid ß-oxidation is critical for the maintenance of energy homeostasis and contractile function in the heart, but its regulation is still not fully understood. While thioredoxin-interacting protein (TXNIP) has recently been implicated in cardiac metabolism and mitochondrial function, its effects on ß-oxidation have remained unexplored. Using a new cardiomyocyte-specific TXNIP knockout mouse and working heart perfusion studies, as well as loss- and gain-of-function experiments in rat H9C2 and human AC16 cardiomyocytes, we discovered that TXNIP deficiency promotes myocardial ß-oxidation via signaling through a specific microRNA, miR-33a. TXNIP deficiency leads to increased binding of nuclear factor Y (NFYA) to the sterol regulatory element binding protein 2 (SREBP2) promoter, resulting in transcriptional inhibition of SREBP2 and its intronic miR-33a. This allows for increased translation of the miR-33a target genes and ß-oxidation-promoting enzymes, carnitine octanoyl transferase (CROT), carnitine palmitoyl transferase 1 (CPT1), hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase-ß (HADHB), and AMPKα and is associated with an increase in phospho-AMPKα and phosphorylation/inactivation of acetyl-CoA-carboxylase. Thus, we have identified a novel TXNIP-NFYA-SREBP2/miR-33a-AMPKα/CROT/CPT1/HADHB pathway that is conserved in mouse, rat, and human cardiomyocytes and regulates myocardial ß-oxidation.


Subject(s)
Carrier Proteins/metabolism , Energy Metabolism , Fatty Acids/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Thioredoxins/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line , Gene Expression Regulation, Enzymologic , Genotype , Humans , Isolated Heart Preparation , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Myocytes, Cardiac/enzymology , Oxidation-Reduction , Phenotype , RNA Interference , Rats , Signal Transduction , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Thioredoxins/genetics , Transfection
15.
Arterioscler Thromb Vasc Biol ; 35(12): 2536-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26427794

ABSTRACT

OBJECTIVE: miR-33 has emerged as an important regulator of lipid homeostasis. Inhibition of miR-33 has been demonstrated as protective against atherosclerosis; however, recent studies in mice suggest that miR-33 inhibition may have adverse effects on lipid and insulin metabolism. Given the therapeutic interest in miR-33 inhibitors for treating atherosclerosis, we sought to test whether pharmacologically inhibiting miR-33 at atheroprotective doses affected metabolic parameters in a mouse model of diet-induced obesity. APPROACH AND RESULTS: High-fat diet (HFD) feeding in conjunction with treatment of male mice with 10 mg/kg control anti-miR or anti-miR33 inhibitors for 20 weeks promoted equivalent weight gain in all groups. miR-33 inhibitors increased plasma total cholesterol and decreased serum triglycerides compared with control anti-miR, but not compared with PBS-treated mice. Metrics of insulin resistance were not altered in anti-miR33-treated mice compared with controls; however, respiratory exchange ratio was decreased in anti-miR33-treated mice. Hepatic expression of miR-33 targets Abca1 and Hadhb were derepressed on miR-33 inhibition. In contrast, protein levels of putative miR-33 target gene SREBP-1 or its downstream targets genes Fasn and Acc were not altered in anti-miR33-treated mice, and hepatic lipid accumulation did not differ between groups. In the adipose tissue, anti-miR33 treatment increased Ampk gene expression and markers of M2 macrophage polarization. CONCLUSIONS: We demonstrate in a mouse model of diet-induced obesity that therapeutic silencing of miR-33 may promote whole-body oxidative metabolism but does not affect metabolic dysregulation. This suggests that pharmacological inhibition of miR-33 at doses known to reduce atherosclerosis may be a safe future therapeutic.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Liver/metabolism , MicroRNAs/metabolism , Obesity/therapy , Oligonucleotides, Antisense/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cholesterol/blood , Disease Models, Animal , Insulin/blood , Insulin Resistance , Macrophages/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Obesity/genetics , Obesity/metabolism , Oligonucleotides, Antisense/genetics , Oxidation-Reduction , Phenotype , Time Factors , Triglycerides/blood , Weight Gain
16.
Int J Mol Sci ; 16(6): 12737-52, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26057744

ABSTRACT

BACKGROUND: miRNAs are single-stranded, small RNA molecules with a length of 18-25 nucleotides. They bind to the 3' untranslated regions of mRNA transcripts to reduce the translation of these transcripts or to cause their degradation. The roles of these molecules differ in biological processes, such as cell differentiation, proliferation, apoptosis and tumor genesis. miRNA-33 is encoded by the gene introns of proteins that bind sterol-regulatory elements. This molecule cooperates with these proteins to control cholesterol homeostasis, fatty acid levels and the genes that are related to the expression of fat metabolism. The examination of miR-33 expression and its target genes can promote the in-depth study of the miRNA regulation mechanism in the formation process of goose fatty liver and can lay a foundation for research into human fatty liver. METHODOLOGY/PRINCIPAL FINDINGS: (1) Through real-time fluorescent quantitative polymerase chain reaction (TaqMan MicroRNA Assay), we detected the expression of miR-33 during the feeding of Landes geese. The expression level of miR-33 increases significantly in the liver after 19 days in comparison with the control group; (2) By using the bioinformatics software programs TargetScan, miRDB and miRCosm to predict the target genes of miR-33 according to laboratory prophase transcriptome results and references, we screen nine target genes: adenosine triphosphate binding cassette transporters A1, adenosine triphosphate binding cassette transporters G1, Neimann Pick C, carnitine O-octanoyltransferase (CROT), cyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase, beta subunit (HADHB), AMP-activated protein kinase, alpha subunit 1 (AMPKα1), insulin receptor substrate 2, glutamic pyruvate transaminase and adipose differentiation-related protein. The dual luciferase reporter gene system in the CHO cell line verifies that CROT, HADHB and NPC1 are the target genes of miR-33 in geese. The inhibition rate of CROT is highest and reaches 70%; (3) The seed sequence (5' 2-8 bases) is the acting site of miR-33. The two predicted target sites of CROT are the target sites of miR-33. Moreover, the predicted target site of HADHB and NPC1 is the target site of miR-33. CONCLUSIONS/SIGNIFICANCE: (1) After 19 days of overfeeding, the expression level of miR-33 increases significantly in the livers of geese; (2) CROT, HADHB and NPC1 are the target genes of miR-33 in geese. These genes determine the combined target site.


Subject(s)
Fatty Liver/metabolism , MicroRNAs/genetics , Transcriptome , Animals , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Geese , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mitochondrial Trifunctional Protein, beta Subunit/metabolism
17.
PLoS Pathog ; 11(3): e1004750, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25816318

ABSTRACT

Infection with Japanese encephalitis virus (JEV) can induce the expression of pro-inflammatory cytokines and cause acute encephalitis in humans. ß-oxidation breaks down fatty acids for ATP production in mitochondria, and impaired ß-oxidation can induce pro-inflammatory cytokine expression. To address the role of fatty-acid ß-oxidation in JEV infection, we measured the oxygen consumption rate of mock- and JEV-infected cells cultured with or without long chain fatty acid (LCFA) palmitate. Cells with JEV infection showed impaired LCFA ß-oxidation and increased interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) expression. JEV nonstructural protein 5 (NS5) interacted with hydroxyacyl-CoA dehydrogenase α and ß subunits, two components of the mitochondrial trifunctional protein (MTP) involved in LCFA ß-oxidation, and NS5 proteins were detected in mitochondria and co-localized with MTP. LCFA ß-oxidation was impaired and higher cytokines were induced in cells overexpressing NS5 protein as compared with control cells. Deletion and mutation studies showed that the N-terminus of NS5 was involved in the MTP association, and a single point mutation of NS5 residue 19 from methionine to alanine (NS5-M19A) reduced its binding ability with MTP. The recombinant JEV with NS5-M19A mutation (JEV-NS5-M19A) was less able to block LCFA ß-oxidation and induced lower levels of IL-6 and TNF-α than wild-type JEV. Moreover, mice challenged with JEV-NS5-M19A showed less neurovirulence and neuroinvasiveness. We identified a novel function of JEV NS5 in viral pathogenesis by impairing LCFA ß-oxidation and inducing cytokine expression by association with MTP.


Subject(s)
Encephalitis Virus, Japanese/metabolism , Encephalitis, Japanese/metabolism , Fatty Acids/metabolism , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Encephalitis Virus, Japanese/genetics , Encephalitis, Japanese/genetics , Fatty Acids/genetics , HEK293 Cells , Humans , Mice , Mitochondrial Trifunctional Protein, alpha Subunit/genetics , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Oxidation-Reduction , Point Mutation , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...