Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Mol Brain ; 14(1): 132, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34479615

ABSTRACT

The medium-chain fatty acids octanoic acid (C8) and decanoic acid (C10) are gaining attention as beneficial brain fuels in several neurological disorders. The protective effects of C8 and C10 have been proposed to be driven by hepatic production of ketone bodies. However, plasma ketone levels correlates poorly with the cerebral effects of C8 and C10, suggesting that additional mechanism are in place. Here we investigated cellular C8 and C10 metabolism in the brain and explored how the protective effects of C8 and C10 may be linked to cellular metabolism. Using dynamic isotope labeling, with [U-13C]C8 and [U-13C]C10 as metabolic substrates, we show that both C8 and C10 are oxidatively metabolized in mouse brain slices. The 13C enrichment from metabolism of [U-13C]C8 and [U-13C]C10 was particularly prominent in glutamine, suggesting that C8 and C10 metabolism primarily occurs in astrocytes. This finding was corroborated in cultured astrocytes in which C8 increased the respiration linked to ATP production, whereas C10 elevated the mitochondrial proton leak. When C8 and C10 were provided together as metabolic substrates in brain slices, metabolism of C10 was predominant over that of C8. Furthermore, metabolism of both [U-13C]C8 and [U-13C]C10 was unaffected by etomoxir indicating that it is independent of carnitine palmitoyltransferase I (CPT-1). Finally, we show that inhibition of glutamine synthesis selectively reduced 13C accumulation in GABA from [U-13C]C8 and [U-13C]C10 metabolism in brain slices, demonstrating that the glutamine generated from astrocyte C8 and C10 metabolism is utilized for neuronal GABA synthesis. Collectively, the results show that cerebral C8 and C10 metabolism is linked to the metabolic coupling of neurons and astrocytes, which may serve as a protective metabolic mechanism of C8 and C10 supplementation in neurological disorders.


Subject(s)
Astrocytes/metabolism , Caprylates/metabolism , Cerebral Cortex/metabolism , Decanoic Acids/metabolism , Glutamine/metabolism , Neurons/metabolism , gamma-Aminobutyric Acid/biosynthesis , Animals , Animals, Outbred Strains , Carnitine O-Palmitoyltransferase/physiology , Cells, Cultured , Cerebral Cortex/cytology , Epoxy Compounds/pharmacology , Glucose/metabolism , Male , Mice , Mitochondria/metabolism , Oxygen Consumption , Specific Pathogen-Free Organisms
2.
Oncogene ; 40(4): 833-847, 2021 01.
Article in English | MEDLINE | ID: mdl-33273726

ABSTRACT

Trastuzumab resistance has been becoming a major obstacle for treatment of HER-2-positive breast cancer patients. Increasing evidence suggests that mesenchymal stem cells (MSCs) play critical roles during the formation of drug resistance, however, the underlying mechanism is not well known. In this study, mass spectrometry, RNA pulldown and RNA immunoprecipitation assays were performed to verify the direct interactions among AGAP2-AS1 and other associated targets, such as human antigen R (HuR), miR-15a-5p, and carnitine palmitoyl transferase 1 (CPT1). In vitro and in vivo experimental assays were done to clarify the functional role of AGAP2-AS1 in trastuzumab resistance, stemness, and fatty acid oxidation (FAO). The results showed that MSC co-culture induced trastuzumab resistance. AGAP2-AS1 was upregulated in MSC-cultured cells, and knockdown of AGAP2-AS1 reversed the MSC-mediated trastuzumab resistance. Furthermore, MSC culture-induced AGAP2-AS1 regulates stemness and trastuzumab resistance via activating FAO. Mechanistically, AGAP2-AS1 is associated with HuR, and the AGAP2-AS1-HuR complex could directly bind to the CPT1, increasing its expression via improving RNA stability. In addition, AGAP2-AS1 could serve as ceRNA via sponging miR-15a-5p and releasing CPT1 mRNA. Clinically, increased expression of serum AGAP2-AS1 predicts poor response to trastuzumab treatment in breast cancer patients. In conclusion, MSC culture-induced AGAP2-AS1 caused stemness and trastuzumab resistance via promoting CPT1 expression and inducing FAO. Our results provide new insight of the role of MSCs in trastuzumab resistance and AGAP2-AS1 could be promising predictive biomarker and therapeutic target for HER-2+ breast cancer patients.


Subject(s)
Breast Neoplasms/drug therapy , Carnitine O-Palmitoyltransferase/genetics , Fatty Acids/metabolism , Mesenchymal Stem Cells/physiology , RNA, Long Noncoding/physiology , Trastuzumab/therapeutic use , Breast Neoplasms/metabolism , Carnitine O-Palmitoyltransferase/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm , ELAV-Like Protein 1/physiology , Female , Humans , MicroRNAs/physiology , Oxidation-Reduction
3.
J Biomed Mater Res A ; 109(5): 766-778, 2021 05.
Article in English | MEDLINE | ID: mdl-32681806

ABSTRACT

Lipid overload of the adipose tissue, which can be caused by overnutrition, underlies metabolic disease. We hypothesized that increasing the energy demand of adipose tissue is a promising strategy to combat excessive lipid accumulation. Resveratrol, a natural polyphenol, activates lipid catabolism in fat tissue; however, its clinical success is hindered by poor bioavailability. Here, we implanted resveratrol releasing poly(lactide-co-glycolide) scaffolds into epididymal fat to overcome its poor bioavailability with the goal of enhancing local lipid catabolism. In lean mice, resveratrol scaffolds decreased adipocyte size relative to scaffolds with no drug, a response that correlated with AMP kinase activation. Immunohistochemistry indicated that macrophages and multinucleated giant cells within the scaffold expressed carnitine palmitoyltransferase 1 (CPT1) at higher levels than other cells in the adipose tissue. Furthermore, resveratrol increased CPT1 levels in cultured macrophages. Taken together, we propose that resveratrol scaffolds decrease adipocyte size because resveratrol increases lipid utilization in scaffold-infiltrating immune cells, possibly through elevating CPT1 levels or activity. In a follow-up study, mice that received resveratrol scaffolds 28-day prior to a high-fat diet exhibited decreased weight gain, adipose tissue expansion, and adipocyte hypertrophy compared to mice with control scaffolds. Notably, this scaffold-based strategy required a single resveratrol administration compared to the daily regiment generally needed for oral administration. These results indicate that localized delivery of metabolism modulating agents to the adipose tissue may overcome issues with bioavailability and that the role of biomaterials should be further investigated in this therapeutic strategy for metabolic disease.


Subject(s)
Adipocytes/drug effects , Epididymis/drug effects , Resveratrol/pharmacology , Tissue Scaffolds , Adenylate Kinase/metabolism , Animals , Carnitine O-Palmitoyltransferase/physiology , Cell Size/drug effects , Diet, High-Fat , Drug Liberation , Epididymis/ultrastructure , Implants, Experimental , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Polylactic Acid-Polyglycolic Acid Copolymer , RAW 264.7 Cells , Resveratrol/administration & dosage , Weight Gain/drug effects
4.
Molecules ; 25(18)2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32916992

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) is involved in lipid metabolism; however, the role of LRRK2 in lipid metabolism to affect non-alcoholic fatty liver disease (NAFLD) is still unclear. In the mouse model of NAFLD induced by a high-fat diet, we observed that LRRK2 was decreased in livers. In HepG2 cells, exposure to palmitic acid (PA) down-regulated LRRK2. Overexpression and knockdown of LRRK2 in HepG2 cells were performed to further investigate the roles of LRRK2 in lipid metabolism. Our results showed that ß-oxidation in HepG2 cells was promoted by LRRK2 overexpression, whereas LRRK2 knockdown inhibited ß-oxidation. The critical enzyme of ß-oxidation, carnitine palmitoyltransferase 1A (CPT1A), was positively regulated by LRRK2. Our data suggested that the regulation of CPT1A by LRRK2 may be via the activation of AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor α (PPARα). The overexpression of LRRK2 reduced the concentration of a pro-inflammatory cytokine, tumor necrosis factor α (TNFα), induced by PA. The increase in ß-oxidation may promote lipid catabolism to suppress inflammation induced by PA. These results indicated that LRRK2 participated in the regulation of ß-oxidation and suggested that the decreased LRRK2 may promote inflammation by suppressing ß-oxidation in the liver.


Subject(s)
Carnitine O-Palmitoyltransferase/physiology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/physiology , Oxygen/metabolism , Animals , Cell Nucleus/metabolism , Cytokines/metabolism , Diet, High-Fat , Hep G2 Cells , Humans , Inflammation , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Oxidation-Reduction , PPAR alpha/metabolism , Palmitic Acid/pharmacology , Tumor Necrosis Factor-alpha/metabolism
5.
Sci Rep ; 10(1): 13523, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32782332

ABSTRACT

Autophagy, an integral part of the waste recycling process, plays an important role in cellular physiology and pathophysiology. Impaired autophagic flux causes ectopic lipid deposition, which is defined as the accumulation of lipids in non-adipose tissue. Ectopic lipid accumulation is observed in patients with cardiometabolic syndrome, including obesity, diabetes, insulin resistance, and cardiovascular complications. Metformin is the first line of treatment for type 2 diabetes, and one of the underlying mechanisms for the anti-diabetic effect of metformin is mediated by the stimulation of AMP-activated protein kinase (AMPK). Because the activation of AMPK is crucial for the initiation of autophagy, we hypothesize that metformin reduces the accumulation of lipid droplets by increasing autophagic flux in vascular endothelial cells. Incubation of vascular endothelial cells with saturated fatty acid (SFA) increased the accumulation of lipid droplets and impaired autophagic flux. We observed that the accumulation of lipid droplets was reduced, and the autophagic flux was enhanced by treatment with metformin. The knock-down of AMPKα by using siRNA blunted the effect of metformin. Furthermore, treatment with SFA or inhibition of autophagy increased leukocyte adhesion, whereas treatment with metformin decreased the SFA-induced leukocyte adhesion. The results suggest a novel mechanism by which metformin protects vascular endothelium from SFA-induced ectopic lipid accumulation and pro-inflammatory responses. In conclusion, improving autophagic flux may be a therapeutic strategy to protect endothelial function from dyslipidemia and diabetic complications.


Subject(s)
Autophagy , Carnitine O-Palmitoyltransferase/physiology , Endothelium, Vascular/drug effects , Fatty Acids/toxicity , Hypoglycemic Agents/pharmacology , Inflammation/drug therapy , Metformin/pharmacology , AMP-Activated Protein Kinases , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Am J Physiol Endocrinol Metab ; 319(2): E265-E275, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32459525

ABSTRACT

Saturated fatty acids (SFAs) are implicated in muscle inflammation/cell stress and insulin resistance, but the catalog of factors involved is incomplete. SFA derivatives that accumulate with mismatched FA availability and FA oxidation (FAO) are likely involved, and evidence has emerged that select acylcarnitines should be considered. To understand if excessive long-chain acylcarnitine accumulation and limited FAO associate with lipotoxicity, carnitine palmitoyltransferase 2 knockout C2C12 cells were generated (CPT2 KO). CPT2 KO was confirmed by Western blot, increased palmitoylcarnitine accumulation, and loss of FAO capacity. There was no effect of CPT2 KO on palmitic acid (PA) concentration-dependent increases in media IL-6 or adenylate kinase. PA at 200 and 500 µM did not trigger cell stress responses (phospho-Erk, -JNK, or -p38) above that of vehicle in WT or CPT2 KO cells. In contrast, loss of CPT2 exacerbated PA-induced insulin resistance (acute phospho-Akt; 10 or 100 nM insulin) by as much as ~50-96% compared with WT. Growing cells in carnitine-free media abolished differences between WT and CPT2 KO, but this did not fully rescue PA-induced insulin resistance. The results suggest that PA-induced insulin resistance stems in part from palmitoylcarnitine accumulation, further supporting the hypothesis that select acylcarnitines participate in cell signaling and, when in excess, can compromise cell function. Since carnitine-free conditions could not fully rescue insulin signaling, and CPT2 KO did not alter cell stress responses, the majority of PA-induced "lipotoxicity" in C2C12 myotubes cannot be attributed to palmitoylcarnitine alone.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/physiology , Gene Knockout Techniques , Insulin Resistance/physiology , Muscle Fibers, Skeletal/physiology , Palmitic Acid/pharmacology , Animals , Cell Line , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Palmitoylcarnitine/metabolism , Signal Transduction/physiology
7.
Biomed Pharmacother ; 127: 110127, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32325349

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a commonly-encountered chronic liver disease which lacks verified pharmacological interventions. Gan-Jiang-Ling-Zhu decoction (GJLZ) is a classic formula utilized in clinical practice. In this study, we aimed to evaluate the therapeutic effect of GJLZ in NAFLD and explore the possible underlying mechanisms. METHODS: Twenty-four rats were randomly divided into three groups: normal group, fed with chow diet for 8 weeks; model group, fed with high fat diet for 8 weeks; and GJLZ group, initially fed HFD for 4 weeks, and then administered the GJLZ decoction for 4 weeks by oral gavage while continuously feeding HFD. Rats were sacrificed after the intervention, and liver tissues and blood samples were harvested. Liver steatosis was detected by HE and Oil Red O staining. Body weight and liver index were analyzed. Liver triglyceride (TG), total cholesterol (TC), and low-density lipoprotein (LDL), serum almandine aminotransferase (ALT), aspartate aminotransferase (AST), and nonesterified fatty acid (NEFA) were assayed using commercial kits. Differentially expressed genes were identified by RNA-sequencing and verified using real-time PCR (RT-PCR) and western blotting. Whole miRNAs were detected by RNA-sequence analysis, and mRNA-targeted miRNAs were verified by RT-PCR. The miRNA-mRNA regulation pattern was confirmed using the dual-luciferase reporter assay. RESULTS: Treatment with GJLZ significantly improved hepatic steatosis and inflammation, reduced liver index and liver TG content, and also significantly reduced serum ALT and AST levels. Based on the results of RNA-sequence analysis, five differentially expressed genes (DEGs) in the peroxisome proliferator-activated receptor (PPAR) signaling pathway were recognized. RT-PCR confirmed that carnitine palmitoyltransferase 1b (CPT1B) expression was significantly regulated by GJLZ treatment. GJLZ decoction intervention also increased significantly hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA) expression. Next, miRNA profiling and screening were performed based on CPT1B alteration. Rno-miR-138-5p likely responded to GJLZ intervention, and rno-miR-138-5p inhibitor increased CPT1B expression while rno-miR-138-5p mimic reduced CPT1B expression. When CPT1B mutated, miR-138-5p mimic and inhibitor could not regulate the luciferase activity of CPT1B. CONCLUSIONS: GJLZ is an effective formula for NAFLD management, and its possible mechanism of action involves the regulation of CPT1B expression via rno-miR-138-5p.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Drugs, Chinese Herbal/therapeutic use , Medicine, Chinese Traditional , MicroRNAs/physiology , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Carnitine O-Palmitoyltransferase/physiology , Drugs, Chinese Herbal/pharmacology , Fatty Acids/metabolism , Gene Expression Regulation/drug effects , Liver/metabolism , Male , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisome Proliferator-Activated Receptors/physiology , Rats , Rats, Wistar , Triglycerides/metabolism
8.
Lipids Health Dis ; 18(1): 215, 2019 Dec 10.
Article in English | MEDLINE | ID: mdl-31823799

ABSTRACT

BACKGROUND: Macrophage are specialized cells that contributes to the removal of detrimental contents via phagocytosis. Lipid accumulation in macrophages, whether from phagocytosis of dying cells or from circulating oxidized low-density lipoproteins, alters macrophage biology and functionality. It is known that carnitine palmitoyl transferase 1-a (CPT1a) gene encodes an enzyme involved in fatty acid oxidation and, therefore, lipid content. However, the potential of CPT1a to activate macrophage phagocytic function have not been elucidated. METHODS: Using a murine macrophage cell line, RAW264.7, we determine if intracellular accumulation of 7-ketocholesterol (7-KC) modulates macrophage phagocytic function through CPT1a gene expression. In addition, the effects of CPT1a genetic modification on macrophage phenotype and phagocytosis has been studied. RESULTS: Our results revealed that CPT1a gene expression decreased by the accumulation of 7-KC at the higher dose of 7-KC. This was concomitant with an impair ability to phagocytize bioparticles and an inflammatory phenotype. GW3965 treatment, which have shown to facilitate the efflux of cholesterol, eliminated the intracellular lipid droplets of 7-KC-laden macrophages, increased the gene expression of CPT1a, diminished the gene expression of the inflammatory marker iNOS and restored macrophage phagocytosis. Furthermore, CPT1a Knockdown per se was detrimental for macrophage phagocytosis whereas transcriptional activation of CPT1a heightened the uptake of bioparticles. CONCLUSIONS: Altogether, our findings indicate that downregulation of CPT1a by lipid content modulates macrophage phagocytosis and inflammatory phenotype.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Gene Expression/physiology , Inflammation , Ketocholesterols/physiology , Macrophages/physiology , Phagocytosis/physiology , Animals , Carnitine O-Palmitoyltransferase/physiology , Down-Regulation , Gene Knockdown Techniques , Ketocholesterols/pharmacology , Macrophage Activation/physiology , Mice , RAW 264.7 Cells , Transfection
9.
Cells ; 8(10)2019 09 20.
Article in English | MEDLINE | ID: mdl-31547059

ABSTRACT

Prostate cancer (PCa) is the most common cancer in men, and the global burden of the disease is rising. The majority of PCa deaths are due to metastasis that are highly resistant to current hormonal treatments; this state is called castration-resistant prostate cancer (CRPC). In this study, we focused on the role of the lipid catabolism enzyme CPT1A in supporting CRPC growth in an androgen-dependent manner. We found that androgen withdrawal promoted the growth of CPT1A over-expressing (OE) tumors while it decreased the growth of CPT1A under-expressing (KD) tumors, increasing their sensitivity to enzalutamide. Mechanistically, we found that CPT1A-OE cells burned more lipid and showed increased histone acetylation changes that were partially reversed with a p300 specific inhibitor. Conversely, CPT1A-KD cells showed less histone acetylation when grown in androgen-deprived conditions. Our results suggest that CPT1A supports CRPC by supplying acetyl groups for histone acetylation, promoting growth and antiandrogen resistance.


Subject(s)
Androgen Antagonists/therapeutic use , Androgens/deficiency , Carnitine O-Palmitoyltransferase/physiology , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Prostatic Neoplasms, Castration-Resistant , Acetylation , Androgen Antagonists/pharmacology , Androgens/pharmacology , Benzamides , Carnitine O-Palmitoyltransferase/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Histone Acetyltransferases/metabolism , Histones/metabolism , Humans , Male , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Processing, Post-Translational/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
10.
J Therm Biol ; 80: 113-118, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30784474

ABSTRACT

The naked neck gene was introduced by crossbreeding into Egyptian breeds to improve body weight. Expression levels of HSP70 and CPT-1 were used to assess the heat tolerance of three Egyptian local breeds (Fayoumi, Dandarawi and Sinai) with and without the naked neck gene and under normal and heat stress conditions. There were two genotypes from each breed that had the same genetic origin (the naked neck and normal plumage genotypes). For each genotype, chicks were divided into two groups, a control group and a treated group. Chicks in the treated group were subjected to heat stress (40 °C) for four hours when they were between 3 and 5 days old. This treatment was associated with a highly significant increase in HSP70 and CPT-1 gene expression for the Dandarawi breed compared to the levels in the Fayoumi and Sinai breeds. Moreover, the introduction of the naked neck gene into these local breeds caused marked increases in CPT-1 gene expression, but these increases did not significantly differ among different naked neck genotypes. Therefore, it could be concluded that the Dandarawi breed exhibited the best heat tolerance, followed by the Sinai breed, whereas the Fayoumi breed was inferior in this respect. Furthermore, the naked neck gene improved heat tolerance by increasing HSP70 gene expression rather than only by reducing feather cover. The results obtained recommended using the Sinia naked neck chicken as a male line in commercial parent stock to produce broiler chicks adapted to the hot and warm climates.


Subject(s)
Avian Proteins/physiology , Carnitine O-Palmitoyltransferase/physiology , Chickens/physiology , HSP70 Heat-Shock Proteins/physiology , Hot Temperature , Thermotolerance/physiology , Animals , Gene Expression , Genotype , Male
11.
FASEB J ; 33(3): 4355-4364, 2019 03.
Article in English | MEDLINE | ID: mdl-30540494

ABSTRACT

Peroxisomes are essential organelles for the specialized oxidation of a wide variety of fatty acids, but they are also able to degrade fatty acids that are typically handled by mitochondria. Using a combination of pharmacological inhibition and clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated protein 9 genome editing technology to simultaneously manipulate peroxisomal and mitochondrial fatty acid ß-oxidation (FAO) in HEK-293 cells, we identified essential players in the metabolic crosstalk between these organelles. Depletion of carnitine palmitoyltransferase (CPT)2 activity through pharmacological inhibition or knockout (KO) uncovered a significant residual peroxisomal oxidation of lauric and palmitic acid, leading to the production of peroxisomal acylcarnitine intermediates. Generation and analysis of additional single- and double-KO cell lines revealed that the D-bifunctional protein (HSD17B4) and the peroxisomal ABC transporter ABCD3 are essential in peroxisomal oxidation of lauric and palmitic acid. Our results indicate that peroxisomes not only accept acyl-CoAs but can also oxidize acylcarnitines in a similar biochemical pathway. By using an Hsd17b4 KO mouse model, we demonstrated that peroxisomes contribute to the plasma acylcarnitine profile after acute inhibition of CPT2, proving in vivo relevance of this pathway. We summarize that peroxisomal FAO is important when mitochondrial FAO is defective or overloaded.-Violante, S., Achetib, N., van Roermund, C. W. T., Hagen, J., Dodatko, T., Vaz, F. M., Waterham, H. R., Chen, H., Baes, M., Yu, C., Argmann, C. A., Houten, S. M. Peroxisomes can oxidize medium- and long-chain fatty acids through a pathway involving ABCD3 and HSD17B4.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Fatty Acids/metabolism , Peroxisomal Multifunctional Protein-2/physiology , Peroxisomes/enzymology , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Animals , CRISPR-Cas Systems , Carnitine/analogs & derivatives , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/physiology , HEK293 Cells , Humans , Lauric Acids/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondria/enzymology , Oxidation-Reduction , Palmitic Acid/metabolism , Peroxisomal Bifunctional Enzyme/deficiency , Peroxisomal Multifunctional Protein-2/deficiency , Peroxisomal Multifunctional Protein-2/genetics , Recombinant Proteins/metabolism
12.
Cell Metab ; 28(3): 504-515.e7, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30043753

ABSTRACT

T cell subsets including effector (Teff), regulatory (Treg), and memory (Tmem) cells are characterized by distinct metabolic profiles that influence their differentiation and function. Previous research suggests that engagement of long-chain fatty acid oxidation (LC-FAO) supports Foxp3+ Treg cell and Tmem cell survival. However, evidence for this is mostly based on inhibition of Cpt1a, the rate-limiting enzyme for LC-FAO, with the drug etomoxir. Using genetic models to target Cpt1a specifically in T cells, we dissected the role of LC-FAO in primary, memory, and regulatory T cell responses. Here we show that the ACC2/Cpt1a axis is largely dispensable for Teff, Tmem, or Treg cell formation, and that the effects of etomoxir on T cell differentiation and function are independent of Cpt1a expression. Together our data argue that metabolic pathways other than LC-FAO fuel Tmem or Treg differentiation and suggest alternative mechanisms for the effects of etomoxir that involve mitochondrial respiration.


Subject(s)
Acetyl-CoA Carboxylase/physiology , CD8-Positive T-Lymphocytes/metabolism , Carnitine O-Palmitoyltransferase/physiology , Epoxy Compounds/pharmacology , Fatty Acids/metabolism , Immunologic Memory/drug effects , Mitochondria/metabolism , T-Lymphocytes, Regulatory/drug effects , Acetyl-CoA Carboxylase/genetics , Animals , Carnitine O-Palmitoyltransferase/genetics , Cell Differentiation/drug effects , Cells, Cultured , Child , Child, Preschool , Female , Gene Knockout Techniques , Humans , Lymphocyte Activation/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction/drug effects , Oxidative Phosphorylation/drug effects , T-Lymphocytes, Regulatory/metabolism
13.
PLoS Biol ; 16(3): e2003782, 2018 03.
Article in English | MEDLINE | ID: mdl-29596410

ABSTRACT

It has been suggested that some cancer cells rely upon fatty acid oxidation (FAO) for energy. Here we show that when FAO was reduced approximately 90% by pharmacological inhibition of carnitine palmitoyltransferase I (CPT1) with low concentrations of etomoxir, the proliferation rate of various cancer cells was unaffected. Efforts to pharmacologically inhibit FAO more than 90% revealed that high concentrations of etomoxir (200 µM) have an off-target effect of inhibiting complex I of the electron transport chain. Surprisingly, however, when FAO was reduced further by genetic knockdown of CPT1, the proliferation rate of these same cells decreased nearly 2-fold and could not be restored by acetate or octanoic acid supplementation. Moreover, CPT1 knockdowns had altered mitochondrial morphology and impaired mitochondrial coupling, whereas cells in which CPT1 had been approximately 90% inhibited by etomoxir did not. Lipidomic profiling of mitochondria isolated from CPT1 knockdowns showed depleted concentrations of complex structural and signaling lipids. Additionally, expression of a catalytically dead CPT1 in CPT1 knockdowns did not restore mitochondrial coupling. Taken together, these results suggest that transport of at least some long-chain fatty acids into the mitochondria by CPT1 may be required for anabolic processes that support healthy mitochondrial function and cancer cell proliferation independent of FAO.


Subject(s)
Carnitine O-Palmitoyltransferase/physiology , Cell Proliferation/physiology , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacology , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/metabolism , Cell Line, Tumor , Electron Transport/drug effects , Fatty Acids/metabolism , Gene Knockdown Techniques , Humans , Mitochondria/drug effects , Mitochondria/physiology , Oxidation-Reduction/drug effects , Oxygen Consumption , RNA Interference
14.
Mol Cell ; 69(4): 689-698.e7, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29429925

ABSTRACT

Endothelial-to-mesenchymal transition (EndoMT) is a cellular process often initiated by the transforming growth factor ß (TGF-ß) family of ligands. Although required for normal heart valve development, deregulated EndoMT is linked to a wide range of pathological conditions. Here, we demonstrate that endothelial fatty acid oxidation (FAO) is a critical in vitro and in vivo regulator of EndoMT. We further show that this FAO-dependent metabolic regulation of EndoMT occurs through alterations in intracellular acetyl-CoA levels. Disruption of FAO via conditional deletion of endothelial carnitine palmitoyltransferase II (Cpt2E-KO) augments the magnitude of embryonic EndoMT, resulting in thickening of cardiac valves. Consistent with the known pathological effects of EndoMT, adult Cpt2E-KO mice demonstrate increased permeability in multiple vascular beds. Taken together, these results demonstrate that endothelial FAO is required to maintain endothelial cell fate and that therapeutic manipulation of endothelial metabolism could provide the basis for treating a growing number of EndoMT-linked pathological conditions.


Subject(s)
Carnitine O-Palmitoyltransferase/physiology , Endothelium, Vascular/metabolism , Epithelial-Mesenchymal Transition , Fatty Acids/chemistry , 3-Hydroxyacyl CoA Dehydrogenases , Acetyl Coenzyme A/metabolism , Acetyl-CoA C-Acyltransferase , Animals , Carbon-Carbon Double Bond Isomerases , Cells, Cultured , Endothelium, Vascular/cytology , Enoyl-CoA Hydratase , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Racemases and Epimerases , Signal Transduction , Transforming Growth Factor beta/metabolism
15.
Cell Syst ; 6(1): 90-102.e4, 2018 Jan 24.
Article in English | MEDLINE | ID: mdl-29199021

ABSTRACT

Identifying genetic and environmental factors that impact complex traits and common diseases is a high biomedical priority. Here, we developed, validated, and implemented a series of multi-layered systems approaches, including (expression-based) phenome-wide association, transcriptome-/proteome-wide association, and (reverse-) mediation analysis, in an open-access web server (systems-genetics.org) to expedite the systems dissection of gene function. We applied these approaches to multi-omics datasets from the BXD mouse genetic reference population, and identified and validated associations between genes and clinical and molecular phenotypes, including previously unreported links between Rpl26 and body weight, and Cpt1a and lipid metabolism. Furthermore, through mediation and reverse-mediation analysis we established regulatory relations between genes, such as the co-regulation of BCKDHA and BCKDHB protein levels, and identified targets of transcription factors E2F6, ZFP277, and ZKSCAN1. Our multifaceted toolkit enabled the identification of gene-gene and gene-phenotype links that are robust and that translate well across populations and species, and can be universally applied to any populations with multi-omics datasets.


Subject(s)
Gene Expression Profiling/methods , Genomics/methods , Proteomics/methods , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/physiology , Databases, Genetic , Genome-Wide Association Study , Genotype , Mice , Mice, Inbred Strains/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci , Ribosomal Proteins/genetics , Ribosomal Proteins/physiology , Systems Biology/methods , Transcriptome
16.
Endocrinology ; 158(7): 2226-2238, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28472467

ABSTRACT

Carnitine palmitoyltransferase (CPT) 1C, a brain-specific protein localized in the endoplasmic reticulum of neurons, is expressed in almost all brain regions. Based on global knockout (KO) models, CPT1C has demonstrated relevance in hippocampus-dependent spatial learning and in hypothalamic regulation of energy balance. Specifically, it has been shown that CPT1C is protective against high-fat diet-induced obesity (DIO), and that CPT1C KO mice show reduced peripheral fatty acid oxidation (FAO) during both fasting and DIO. However, the mechanisms mediating CPT1C-dependent regulation of energy homeostasis remain unclear. Here, we focus on the mechanistic understanding of hypothalamic CPT1C on the regulation of fuel selection in liver and muscle of male mice during energy deprivation situations, such as fasting. In CPT1C-deficient mice, modulation of the main hypothalamic energy sensors (5' adenosine monophosphate-activated protein kinase, Sirtuin 1, and mammalian target of rapamycin) was impaired and plasma catecholamine levels were decreased. Consequently, CPT1C-deficient mice presented defective fasting-induced FAO in liver, leading to higher triacylglycerol accumulation and lower glycogen levels. Moreover, muscle pyruvate dehydrogenase activity was increased, which was indicative of glycolysis enhancement. The respiratory quotient did not decrease in CPT1C KO mice after 48 hours of fasting, confirming a defective switch on fuel substrate selection under hypoglycemia. Phenotype reversion studies identified the mediobasal hypothalamus (MBH) as the main area mediating CPT1C effects on fuel selection. Overall, our data demonstrate that CPT1C in the MBH is necessary for proper hypothalamic sensing of a negative energy balance and fuel partitioning in liver and muscle.


Subject(s)
Carnitine O-Palmitoyltransferase/physiology , Energy Metabolism/genetics , Hypothalamus/physiology , Liver/metabolism , Muscles/metabolism , Animals , Brain/metabolism , Carnitine O-Palmitoyltransferase/genetics , Homeostasis , Hypothalamus/metabolism , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/genetics
17.
Biochim Biophys Acta ; 1857(4): 415-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26772728

ABSTRACT

Sea urchin sperm have only one mitochondrion, that in addition to being the main source of energy, may modulate intracellular Ca(2+) concentration ([Ca(2+)]i) to regulate their motility and possibly the acrosome reaction. Speract is a decapeptide from the outer jelly layer of the Strongylocentrotus purpuratus egg that upon binding to its receptor in the sperm, stimulates sperm motility, respiration and ion fluxes, among other physiological events. Altering the sea urchin sperm mitochondrial function with specific inhibitors of this organelle, increases [Ca(2+)]i in an external Ca(2+) concentration ([Ca(2+)]ext)-dependent manner (Ardón, et al., 2009. BBActa 1787: 15), suggesting that the mitochondrion is involved in sperm [Ca(2+)]i homeostasis. To further understand the interrelationship between the mitochondrion and the speract responses, we measured mitochondrial membrane potential (ΔΨ) and NADH levels. We found that the stimulation of sperm with speract depolarizes the mitochondrion and increases the levels of NADH. Surprisingly, these responses are independent of external Ca(2+) and are due to the increase in intracellular pH (pHi) induced by speract. Our findings indicate that speract, by regulating pHi, in addition to [Ca(2+)]i, may finely modulate mitochondrial metabolism to control motility and ensure that sperm reach the egg and fertilize it.


Subject(s)
Mitochondria/drug effects , Oligopeptides/pharmacology , Sperm Motility/drug effects , Spermatozoa/metabolism , Animals , Calcium/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Carnitine O-Palmitoyltransferase/physiology , Hydrogen-Ion Concentration , Male , Mitochondria/metabolism , NAD/analysis , Proton-Translocating ATPases/metabolism
18.
Clin Epigenetics ; 8: 6, 2016.
Article in English | MEDLINE | ID: mdl-26798409

ABSTRACT

BACKGROUND: There is growing interest in the hypertriglyceridemic waist (HTGW) phenotype, defined as high waist circumference (≥95 cm in males and ≥80 cm in females) combined with high serum triglyceride concentration (≥2.0 mmol/L in males and ≥1.5 mmol/L in females) as a marker of type 2 diabetes (T2D) and cardiovascular disease. However, the prevalence of this phenotype in high-risk populations, its association with T2D, and the genetic or epigenetic influences on HTGW are not well explored. Using data from large, extended families of Mexican Americans (a high-risk minority population in the USA) we aimed to: (1) estimate the prevalence of this phenotype, (2) test its association with T2D and related traits, and (3) dissect out the genetic and epigenetic associations with this phenotype using genome-wide and epigenome-wide studies, respectively. RESULTS: Data for this study was from 850 Mexican American participants (representing 39 families) recruited under the ongoing San Antonio Family Heart Study, 26 % of these individuals had HTGW. This phenotype was significantly heritable (h (2) r = 0.52, p = 1.1 × 10(-5)) and independently associated with T2D as well as fasting glucose levels and insulin resistance. We conducted genome-wide association analyses using 759,809 single nucleotide polymorphisms (SNPs) and epigenome-wide association analyses using 457,331 CpG sites. There was no evidence of any SNP associated with HTGW at the genome-wide level but two CpG sites (cg00574958 and cg17058475) in CPT1A and one CpG site (cg06500161) in ABCG1 were significantly associated with HTGW and remained significant after adjusting for the closely related components of metabolic syndrome. CPT1A holds a cardinal position in the metabolism of long-chain fatty acids while ABCG1 plays a role in triglyceride metabolism. CONCLUSIONS: Our results reemphasize the value of HTGW as a marker of T2D. This phenotype shows association with DNA methylation within CPT1A and ABCG1, genes involved in fatty acid and triglyceride metabolism. Our results underscore the importance of epigenetics in a clinically informative phenotype.


Subject(s)
Epigenesis, Genetic , Hypertriglyceridemia/genetics , Mexican Americans/genetics , Waist Circumference/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/physiology , Diabetes Mellitus, Type 2/genetics , Epigenomics , Family , Female , Genetic Markers/genetics , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
19.
PLoS One ; 11(1): e0145789, 2016.
Article in English | MEDLINE | ID: mdl-26808626

ABSTRACT

In this study, we conducted an epigenome-wide association study of metabolic syndrome (MetS) among 846 participants of European descent in the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN). DNA was isolated from CD4+ T cells and methylation at ~470,000 cytosine-phosphate-guanine dinucleotide (CpG) pairs was assayed using the Illumina Infinium HumanMethylation450 BeadChip. We modeled the percentage methylation at individual CpGs as a function of MetS using linear mixed models. A Bonferroni-corrected P-value of 1.1 x 10(-7) was considered significant. Methylation at two CpG sites in CPT1A on chromosome 11 was significantly associated with MetS (P for cg00574958 = 2.6x10(-14) and P for cg17058475 = 1.2x10(-9)). Significant associations were replicated in both European and African ancestry participants of the Bogalusa Heart Study. Our findings suggest that methylation in CPT1A is a promising epigenetic marker for MetS risk which could become useful as a treatment target in the future.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , CpG Islands/genetics , DNA Methylation , Metabolic Syndrome/genetics , Aged , Anthropometry , Black People/genetics , Blood Pressure , Carnitine O-Palmitoyltransferase/physiology , Chromosomes, Human, Pair 11/genetics , Cohort Studies , Female , Humans , Male , Metabolic Syndrome/ethnology , Middle Aged , Minnesota/epidemiology , Oligonucleotide Array Sequence Analysis , Risk Factors , Triglycerides/blood , Utah/epidemiology , White People/genetics
20.
Anim Sci J ; 87(1): 61-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26031853

ABSTRACT

Liver is the most important target organ for investigation of lipid metabolism in domestic fowls. However, little is known about the regulatory mechanism of fatty acid oxidation in chicken liver. In mammals, proliferator-activated receptor alpha (PPARα), a transcription factor, plays an essential role in the regulation of hepatic fatty acid oxidation. The aim of the present study was to investigate the regulatory mechanisms of PPARα-induced gene expression involved in hepatic fatty acid oxidation in chickens in vivo and in vitro. WY14643, a PPARα agonist, significantly increased the messenger RNA (mRNA) levels of carnitine palmitoyltransferase 1a (CPT1a) and acyl-coenzyme A oxidase (ACO), but not long-, middle- and short-chain acyl-coenzyme A dehydrogenase (LCAD, MCAD and SCAD, respectively), hydroxyacyl-coenzyme A dehydrogenase (HAD), and PPARα itself in chicken hepatoma cells. In contrast, WY14643 significantly increased the mRNA levels of CPT1a, ACO, MCAD, SCAD, HAD and PPARα in human hepatoma cells. The mRNA levels of CPT1a and ACO in the liver were significantly increased by 6 h of fasting in chickens, whereas the mRNA levels of LCAD, MCAD, SCAD and HAD were unchanged. These results suggest that, unlike in mammals, CPT1a and ACO might play an important role in PPARα-induced fatty acid oxidation in the liver of chickens.


Subject(s)
Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/physiology , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/physiology , Fatty Acids/metabolism , Gene Expression/genetics , Liver/metabolism , PPAR alpha/physiology , Animals , Chickens , Fasting/metabolism , Hep G2 Cells , Humans , Male , Oxidation-Reduction , PPAR alpha/agonists , Pyrimidines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL