Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
2.
Cardiovasc Toxicol ; 21(8): 619-629, 2021 08.
Article in English | MEDLINE | ID: mdl-33929718

ABSTRACT

Fatty acid-binding protein 5 (FABP5) is an important member of the FABP family and plays a vital role in the metabolism of fatty acids. However, few studies have examined the role of FABP5 in pathological cardiac remodeling and heart failure. The aim of this study was to explore the role of FABP5 in transverse aortic constriction (TAC)-induced pathological cardiac remodeling and dysfunction in mice. Quantitative RT-PCR (qRT-PCR) and western blotting (WB) analysis showed that the levels of FABP5 mRNA and protein, respectively, were upregulated in hearts of the TAC model. Ten weeks after TAC in FABP5 knockout and wild type control mice, echocardiography, histopathology, qRT-PCR, and WB demonstrated that FABP5 deficiency aggravated cardiac injury (both cardiac hypertrophy and fibrosis) and dysfunction. In addition, transmission electron microscopy, ATP detection, and WB revealed that TAC caused severe impairment to mitochondria in the hearts of FABP5-deficient mice compared with that in control mice. When FABP5 was downregulated by siRNA in primary mouse cardiac fibroblasts, FABP5 silencing increased oxidative stress, reduced mitochondrial respiration, and increased the expression of myofibroblast activation marker genes in response to treatment with transforming growth factor-ß. Our findings demonstrate that FABP5 deficiency aggravates cardiac pathological remodeling and dysfunction by damaging cardiac mitochondrial function.


Subject(s)
Fatty Acid-Binding Proteins/deficiency , Fibroblasts/metabolism , Heart Failure/metabolism , Hypertrophy, Left Ventricular/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Neoplasm Proteins/deficiency , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Ventricular Remodeling , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Fatty Acid-Binding Proteins/genetics , Fibroblasts/ultrastructure , Fibrosis , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/ultrastructure , Neoplasm Proteins/genetics , Oxidative Stress , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
3.
Physiol Rep ; 9(3): e14733, 2021 02.
Article in English | MEDLINE | ID: mdl-33527741

ABSTRACT

Bile acids are natural detergents that aid in the absorption of dietary lipids. Fatty acid binding protein 6 (Fabp6) is a component of the bile acid recovery system that operates in the small intestine. The aim of this study was to determine if Fabp6 deficiency causes dietary fat malabsorption. Wild-type and Fabp6-deficient mice were fed a Western-style diet (WSD) or a reference low-fat diet (LFD) for 10 weeks. The body weight gain, bile acid excretion, fat excretion, energy metabolism, and major gut microbial phyla of the mice were assessed at the end of the controlled diet period. Fabp6-/- mice exhibited enhanced excretion of both bile acids and fat on the WSD but not on the LFD diet. Paradoxically, male Fabp6-/- mice, but not female Fabp6-/- mice, had greater adiposity despite increased fat excretion. Analysis of energy intake and of expenditure by indirect calorimetry revealed sex differences in physical activity level and respiratory quotient, but these did not account for the enhanced adiposity displayed by male Fabp6-/- mice. Analysis of stool DNA showed sex-specific changes in the abundance of major phyla of bacteria in response to Fabp6 deficiency and WSD feeding. The results obtained indicate that the malabsorption of bile acids that occurs in Fabp6-/- mice is associated with dietary fat malabsorption on the high-fat diet but not on the low-fat diet. The WSD induced a sexually dimorphic increase in adiposity displayed by Fabp6-/- mice and sexually distinct pattern of change in gut microbiota composition.


Subject(s)
Adiposity , Diet, Western/adverse effects , Fatty Acid-Binding Proteins/deficiency , Gastrointestinal Microbiome , Intestinal Absorption , Lipid Metabolism , Malabsorption Syndromes/metabolism , Animals , Bile Acids and Salts/metabolism , Dysbiosis , Energy Metabolism , Fatty Acid-Binding Proteins/genetics , Female , Genotype , Malabsorption Syndromes/genetics , Malabsorption Syndromes/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Sex Characteristics , Sex Factors , Weight Gain
4.
Histochem Cell Biol ; 155(4): 439-449, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33398436

ABSTRACT

In our previous study, fatty acid-binding protein 5 (FABP5) was expressed in septoclasts with long processes which are considered to resorb uncalcified matrix of the growth plate (GP) cartilage, and no apparent abnormalities were detected in the histo-architecture of the GP of FABP5-deficient (FABP5-/-) mice. Those finding lead us to hypothesize that another FABP can compensate the deletion of FABP5 in septoclasts of its gene-mutant mice. Based on the hypothesis, the present study examined the expression levels of several other FABPs in septoclasts and their morphology in FABP5-/- mouse tibiae. Processes of FABP5-/- septoclasts tend to be shorter than wild septoclasts. FABP4-positive septoclasts in FABP5-/- mice were more numerous than those cells in wild mice.Peroxisome proliferator-activated receptor (PPAR) γ was expressed in FABP4-positive septoclasts of FABP5-/- mice as well as mice administered with GW1929, a PPARγ agonist, suggesting that the occurrence of PPARγ induces an increase of FABP4-positive septoclasts. The present finding suggests that the functional exertion of FABP5 in septoclasts is supplemented by FABP4 in normal and FABP5-/- mice, and that the expression of FABP4 is up-regulated in accompany with PPARγ in FABP5-/- for maintenance of resorptive activity in the GP.


Subject(s)
Chondrocytes/metabolism , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acid-Binding Proteins/metabolism , Growth Plate/metabolism , Neoplasm Proteins/metabolism , Tibia/metabolism , Animals , Cartilage/metabolism , Fatty Acid-Binding Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/deficiency , Phenotype
5.
Neuropharmacology ; 183: 108398, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33181146

ABSTRACT

Substance use disorders for cocaine are major public health concerns with few effective treatment options. Therefore, identification of novel pharmacotherapeutic targets is critical for future therapeutic development. Evolution has ensured that genes are expressed largely only where they are needed. Therefore, examining the gene expression landscape of the nucleus accumbens shell (NAcSh), a brain region important for reward related behaviors, may lead to the identification of novel targets for cocaine use disorder. In this study, we conducted a novel two-step topographic transcriptomic analysis using five seed transcripts with enhanced expression in the NAcSh to identify transcripts with similarly enhanced expression utilizing the correlation feature to search the more than 20,000 in situ hybridization experiments of the Allen Mouse Brain Atlas. Transcripts that correlated with at least three seed transcripts were analyzed with Ingenuity Pathway Analysis (IPA). We identified 7-fold more NAcSh-enhanced transcripts than our previous analysis using single voxels in the NAcSh as the seed. Analysis of the resulting transcripts with IPA identified many previously identified signaling pathways such as retinoic acid signaling as well as novel pathways. Manipulation of the retinoic acid pathway specifically in the NAcSh of male rats via viral vector-mediated RNA interference targeting fatty acid binding protein 5 (FABP5) decreased cocaine self-administration and modulates excitability of medium spiny neurons in the NAcSh. These results not only validate the prospective strategy of conducting a topographic transcriptomic analysis, but also further validate retinoic acid signaling as a promising pathway for pharmacotherapeutic development against cocaine use disorder.


Subject(s)
Cocaine-Related Disorders/metabolism , Eye Proteins/physiology , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/physiology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/physiology , Nucleus Accumbens/metabolism , Transcriptome , Action Potentials/drug effects , Animals , Cocaine/pharmacology , Gene Expression , Male , Nucleus Accumbens/physiology , Rats , Rats, Sprague-Dawley , Self Administration , Tretinoin/metabolism
6.
Development ; 147(12)2020 06 19.
Article in English | MEDLINE | ID: mdl-32467236

ABSTRACT

The zebrafish retina grows for a lifetime. Whether embryonic and postembryonic retinogenesis conform to the same developmental program is an outstanding question that remains under debate. Using single-cell RNA sequencing of ∼20,000 cells of the developing zebrafish retina at four different stages, we identified seven distinct developmental states. Each state explicitly expresses a gene set. Disruption of individual state-specific marker genes results in various defects ranging from small eyes to the loss of distinct retinal cell types. Using a similar approach, we further characterized the developmental states of postembryonic retinal stem cells (RSCs) and their progeny in the ciliary marginal zone. Expression pattern analysis of state-specific marker genes showed that the developmental states of postembryonic RSCs largely recapitulated those of their embryonic counterparts, except for some differences in rod photoreceptor genesis. Thus, our findings reveal the unifying developmental program used by the embryonic and postembryonic retinogenesis in zebrafish.


Subject(s)
Embryo, Nonmammalian/metabolism , Neurogenesis/genetics , Retina/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Development , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Retina/cytology , Retina/growth & development , Sequence Analysis, RNA , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/growth & development , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
7.
Am J Physiol Gastrointest Liver Physiol ; 318(3): G518-G530, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31905021

ABSTRACT

Intestinal-fatty acid binding protein (IFABP; FABP2) is a 15-kDa intracellular protein abundantly present in the cytosol of the small intestinal (SI) enterocyte. High-fat (HF) feeding of IFABP-/- mice resulted in reduced weight gain and fat mass relative to wild-type (WT) mice. Here, we examined intestinal properties that may underlie the observed lean phenotype of high fat-fed IFABP-/- mice. No alterations in fecal lipid content were found, suggesting that the IFABP-/- mice are not malabsorbing dietary fat. However, the total excreted fecal mass, normalized to food intake, was increased for the IFABP-/- mice relative to WT mice. Moreover, intestinal transit time was more rapid in the IFABP-/- mice. IFABP-/- mice displayed a shortened average villus length, a thinner muscularis layer, reduced goblet cell density, and reduced Paneth cell abundance. The number of proliferating cells in the crypts of IFABP-/- mice did not differ from that of WT mice, suggesting that the blunt villi phenotype is not due to alterations in proliferation. IFABP-/- mice were observed to have altered expression of genes and proteins related to intestinal structure, while immunohistochemical analyses revealed increased staining for markers of inflammation. Taken together, these studies indicate that the ablation of IFABP, coupled with high-fat feeding, leads to changes in gut motility and morphology, which likely contribute to the relatively leaner phenotype occurring at the whole-body level. Thus, IFABP is likely involved in dietary lipid sensing and signaling, influencing intestinal motility, intestinal structure, and nutrient absorption, thereby impacting systemic energy metabolism.NEW & NOTEWORTHY Intestinal fatty acid binding protein (IFABP) is thought to be essential for the efficient uptake and trafficking of dietary fatty acids. In this study, we demonstrate that high-fat-fed IFABP-/- mice have an increased fecal output and are likely malabsorbing other nutrients in addition to lipid. Furthermore, we observe that the ablation of IFABP leads to marked alterations in intestinal morphology and secretory cell abundance.


Subject(s)
Adiposity , Diet, High-Fat , Fatty Acid-Binding Proteins/deficiency , Gastrointestinal Motility , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Weight Gain , Animals , Cell Death , Defecation , Energy Metabolism , Enterocytes/metabolism , Enterocytes/pathology , Fatty Acid-Binding Proteins/genetics , Feces/chemistry , Gene Deletion , Genotype , Intestinal Absorption , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Intestine, Small/pathology , Intestine, Small/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Time Factors
8.
Mod Pathol ; 33(4): 665-675, 2020 04.
Article in English | MEDLINE | ID: mdl-31570768

ABSTRACT

The molecular classification of hepatocellular adenomas highlights a distinctive genotype-phenotype correlation. Malignant transformation is an exceptionally rare complication of hepatocyte nuclear factor 1α (HNF1A)-inactivated hepatocellular adenomas. This subtype is characterized by loss of liver fatty acid binding protein immunoexpression. In this study, we characterized the histopathologic spectrum of 13 liver fatty acid binding protein-deficient hepatocellular adenoma cases showing malignant transformation from multiple centers. Clinicopathologic characteristics of these patients were evaluated. Stains for reticulin, liver fatty acid binding protein, beta-catenin and glutamine synthetase were applied to these lesions. Moreover, the findings were compared to patients with ß-catenin mutated hepatocellular adenoma. Liver fatty acid binding protein-deficient hepatocellular adenomas with borderline features/carcinoma were seen predominantly in females (77%) with an average age of 46 ± 18 years and multiple lesions (77%; five patients with adenomatosis). Meanwhile, ß-catenin mutated hepatocellular adenoma patients with malignant transformation were predominantly male (67%, p = 0.018) with single lesion (86%, p = 0.0009). The largest liver fatty acid binding protein-deficient hepatocellular adenoma nodule in each patient ranged from 4 to 15.5 cm. Loss of liver fatty acid binding protein by immunohistochemistry was noted in all adenoma and borderline/carcinoma components. Features of malignant transformation were pseudoglandular architecture (85%), cytologic atypia (85%), architectural atypia (100%) and lack of steatosis (100%). Other findings included myxoid change (39%), peliosis (46%) and sinusoidal dilatation (46%). Molecular studies confirmed somatic inactivation of HNF1A in 3 cases and absence of TERT promotor and exon 3 CTNNB1 mutations in five cases. To summarize, liver fatty acid binding protein-deficient hepatocellular adenoma with malignant transformation is most frequently seen in female patients with multiple lesions. Most of these lesions demonstrate pseudoglandular architecture, cytologic and architectural atypia, with lack of steatosis. The natural history of these lesions is relatively benign with the exception of disease recurrence in 1 patient.


Subject(s)
Adenoma, Liver Cell/chemistry , Biomarkers, Tumor/deficiency , Cell Transformation, Neoplastic/chemistry , Fatty Acid-Binding Proteins/deficiency , Liver Neoplasms/chemistry , Adenoma, Liver Cell/genetics , Adenoma, Liver Cell/pathology , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chromogranins/genetics , Europe , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Silencing , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Mutation , Prognosis , Retrospective Studies , Sex Factors , Telomerase/genetics , United States , Young Adult , beta Catenin/genetics
9.
Oncogene ; 39(7): 1445-1465, 2020 02.
Article in English | MEDLINE | ID: mdl-31659255

ABSTRACT

Aneuploidy can instigate tumorigenesis. However, mutations in genes that control chromosome segregation are rare in human tumors as these mutations reduce cell fitness. Screening experiments indicate that the knockdown of multiple classes of genes that are not directly involved in chromosome segregation can lead to aneuploidy induction. The possible contribution of these genes to cancer formation remains yet to be defined. Here we identified gene knockdowns that lead to an increase in aneuploidy in checkpoint-deficient human cancer cells. Computational analysis revealed that the identified genes overlap with recurrent mutations in human cancers. The knockdown of the three strongest selected candidate genes (ORP3, GJB3, and RXFP1) enhances the malignant transformation of human fibroblasts in culture. Furthermore, the knockout of Orp3 results in an aberrant expansion of lymphoid progenitor cells and a high penetrance formation of chromosomal instable, pauci-clonal B-cell lymphoma in aging mice. At pre-tumorous stages, lymphoid cells from the animals exhibit deregulated phospholipid metabolism and an aberrant induction of proliferation regulating pathways associating with increased aneuploidy in hematopoietic progenitor cells. Together, these results support the concept that aneuploidy-inducing gene deficiencies contribute to cellular transformation and carcinogenesis involving the deregulation of various molecular processes such as lipid metabolism, proliferation, and cell survival.


Subject(s)
Aneuploidy , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Gene Knockdown Techniques , Lymphoma, B-Cell/genetics , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Fibroblasts/pathology , Humans , Lymphoma, B-Cell/pathology , Mice
10.
Lipids ; 54(10): 583-601, 2019 10.
Article in English | MEDLINE | ID: mdl-31487051

ABSTRACT

Brain endocannabinoids (EC) such as arachidonoylethanolamine (AEA) and 2-arachidonoylglycerol (2-AG) primarily originate from serum arachidonic acid (ARA), whose level is regulated in part by a cytosolic ARA-binding protein, that is, liver fatty acid binding protein-1 (FABP1), not expressed in the brain. Ablation of the Fabp1 gene (LKO) increases brain AEA and 2-AG by decreasing hepatic uptake of ARA to increase serum ARA, thereby increasing ARA availability for uptake by the brain. The brain also expresses sterol carrier protein-2 (SCP-2), which is also a cytosolic ARA-binding protein. To further resolve the role of SCP-2 independent of FABP1, mice ablated in the Scp-2/Scp-x gene (DKO) were crossed with mice ablated in the Fabp1 gene (LKO) mice to generate triple knock out (TKO) mice. TKO impaired the ability of LKO to increase brain AEA and 2-AG. While a high-fat diet (HFD) alone increased brain AEA, TKO impaired this effect. Overall, these TKO-induced blocks were not attributable to altered expression of brain proteins in ARA uptake, AEA/2-AG synthesis, or AEA/2-AG degrading enzymes. Instead, TKO reduced serum levels of free ARA and/or total ARA and thereby decreased ARA availability for uptake to the brain and downstream synthesis of AEA and 2-AG therein. In summary, Scp-2/Scp-x gene ablation in Fabp1 null (LKO) mice antagonized the impact of LKO and HFD on brain ARA and, subsequently, EC levels. Thus, both FABP1 and SCP-2 participate in regulating the EC system in the brain.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Diet, High-Fat , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/metabolism , Animals , Carrier Proteins/genetics , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(7): 985-1004, 2019 07.
Article in English | MEDLINE | ID: mdl-30910689

ABSTRACT

Liver fatty acid binding protein (L-FABP) is the major fatty acid binding/"chaperone" protein in hepatic cytosol. Although fatty acids can be derived from the breakdown of dietary fat and glucose, relatively little is known regarding the impact of L-FABP on phenotype in the context of high dietary glucose. Potential impact was examined in wild-type (WT) and Lfabp gene ablated (LKO) female mice fed either a control or pair-fed high glucose diet (HGD). WT mice fed HGD alone exhibited decreased whole body weight gain and weight gain/kcal food consumed-both as reduced lean tissue mass (LTM) and fat tissue mass (FTM). Conversely, LKO alone increased weight gain, lean tissue mass, and fat tissue mass while decreasing serum ß-hydroxybutyrate (indicative of hepatic fatty acid oxidation)-regardless of diet. Both LKO alone and HGD alone significantly altered the serum lipoprotein profile and increased triacylglycerol (TG), but in HGD mice the LKO did not further exacerbate serum TG content. HGD had little effect on hepatic lipid composition in WT mice, but prevented the LKO-induced selective increase in hepatic phospholipid, free-cholesterol and cholesteryl-ester. Taken together, these findings suggest that high glucose diet diminished the effects of LKO on the whole body and lipid phenotype of these mice.


Subject(s)
Fatty Acid-Binding Proteins/deficiency , Glucose/pharmacology , Lipid Metabolism , Animals , Cholesterol/metabolism , Diet , Fatty Acid-Binding Proteins/genetics , Female , Liver/metabolism , Mice , Phospholipids/metabolism , Triglycerides/metabolism , Weight Gain
12.
FASEB J ; 33(3): 4610-4625, 2019 03.
Article in English | MEDLINE | ID: mdl-30576225

ABSTRACT

Liver fatty acid binding protein (L-Fabp) modulates lipid trafficking in enterocytes, hepatocytes, and hepatic stellate cells (HSCs). We examined hepatocyte vs. HSC L-Fabp deletion in hepatic metabolic adaptation and fibrotic injury. Floxed L-Fabp mice were bred to different transgenic Cre mice or injected with adeno-associated virus type 8 (AAV8) Cre and fed diets to promote steatosis and fibrosis or were subjected to either bile duct ligation or CCl4 injury. Albumin-Cre-mediated L-Fabp deletion revealed recombination in hepatocytes and HSCs; these findings were confirmed with 2 other floxed alleles. Glial fibrillary acid protein-Cre and platelet-derived growth factor receptor ß-Cre-mediated L-Fabp deletion demonstrated recombination only in HSCs. Mice with albumin promoter-driven Cre recombinase (Alb-Cre)-mediated or AAV8-mediated L-Fabp deletion were protected against food withdrawal-induced steatosis. Mice with Alb-Cre-mediated L-Fabp deletion were protected against high saturated fat-induced steatosis and fibrosis, phenocopying germline L-Fabp-/- mice. Mice with HSC-specific L-Fabp deletion exhibited retinyl ester depletion yet demonstrated no alterations in fibrosis. On the other hand, fibrogenic resolution after CCl4 administration was impaired in mice with Alb-Cre-mediated L-Fabp deletion. These findings suggest cell type-specific roles for L-Fabp in mitigating hepatic steatosis and in modulating fibrogenic injury and reversal.-Newberry, E. P., Xie, Y., Lodeiro, C., Solis, R., Moritz, W., Kennedy, S., Barron, L., Onufer, E., Alpini, G., Zhou, T., Blaner, W. S., Chen, A., Davidson, N. O. Hepatocyte and stellate cell deletion of liver fatty acid binding protein reveal distinct roles in fibrogenic injury.


Subject(s)
Carbon Tetrachloride Poisoning/metabolism , Fatty Acid-Binding Proteins/physiology , Fatty Liver/metabolism , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , Albumins/genetics , Animals , Bile Ducts , Carbon Tetrachloride Poisoning/pathology , Crosses, Genetic , Dependovirus/genetics , Dietary Fats/toxicity , Fatty Acid-Binding Proteins/deficiency , Fatty Acids/toxicity , Fatty Liver/etiology , Fatty Liver/pathology , Female , Fibrosis , Food Deprivation , Gene Deletion , Genes, Synthetic , Hepatic Stellate Cells/pathology , Hepatocytes/pathology , Integrases , Ligation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Promoter Regions, Genetic
13.
Cardiovasc Res ; 114(8): 1132-1144, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29554241

ABSTRACT

Aims: The metabolism of the failing heart is characterized by an increase in glucose uptake with reduced fatty acid (FA) oxidation. We previously found that the genetic deletion of FA-binding protein-4 and -5 [double knockout (DKO)] induces an increased myocardial reliance on glucose with decreased FA uptake in mice. However, whether this fuel switch confers functional benefit during the hypertrophic response remains open to debate. To address this question, we investigated the contractile function and metabolic profile of DKO hearts subjected to pressure overload. Methods and results: Transverse aortic constriction (TAC) significantly reduced cardiac contraction in DKO mice (DKO-TAC), although an increase in cardiac mass and interstitial fibrosis was comparable with wild-type TAC (WT-TAC). DKO-TAC hearts exhibited enhanced glucose uptake by 8-fold compared with WT-TAC. Metabolic profiling and isotopomer analysis revealed that the pool size in the TCA cycle and the level of phosphocreatine were significantly reduced in DKO-TAC hearts, despite a marked increase in glycolytic flux. The ingestion of a diet enriched in medium-chain FAs restored cardiac contractile dysfunction in DKO-TAC hearts. The de novo synthesis of amino acids as well as FA from glycolytic flux was unlikely to be suppressed, despite a reduction in each precursor. The pentose phosphate pathway was also facilitated, which led to the increased production of a coenzyme for lipogenesis and a precursor for nucleotide synthesis. These findings suggest that reduced FA utilization is not sufficiently compensated by a robust increase in glucose uptake when the energy demand is elevated. Glucose utilization for sustained biomass synthesis further enhances diminishment of the pool size in the TCA cycle. Conclusions: Our data suggest that glucose is preferentially utilized for biomass synthesis rather than ATP production during pressure-overload-induced cardiac hypertrophy and that the efficient supplementation of energy substrates may restore cardiac dysfunction caused by energy insufficiency.


Subject(s)
Cardiomegaly/metabolism , Energy Metabolism , Fatty Acid-Binding Proteins/deficiency , Glucose/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Neoplasm Proteins/deficiency , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Citric Acid Cycle , Disease Models, Animal , Fatty Acid-Binding Proteins/genetics , Fatty Acids/metabolism , Genotype , Glycolysis , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocardium/pathology , Neoplasm Proteins/genetics , Oxidation-Reduction , Phenotype , Time Factors
14.
Behav Pharmacol ; 29(6): 503-508, 2018 09.
Article in English | MEDLINE | ID: mdl-29570114

ABSTRACT

Inhibition and genetic deletion of fatty acid-binding proteins (FABPs) 5 and 7 have been shown to increase the levels of the endocannabinoid anandamide as well as the related N-acylethanolamine's palmitoylethanolamide and oleoylethanolamide. This study examined the role of these FABPs on forced-swim (FS) behavior and on sucrose consumption in two experiments: (experiment 1) using wild-type (WT) mice treated with the FABP inhibitor SBFI26 or vehicle and (experiment 2) using WT and FABP5/7 deficient mice. Results from experiment 1 showed that acute treatment with SBFI26 did not have any effect on sucrose intake or FS behavior in mice. In experiment 2, male and female FABP5/7 deficient mice showed significant increases in sucrose consumption (25 and 21%, respectively) compared with their WT counterparts. In addition, immobility time during the FS was decreased by 27% in both male and female FABP5/7 knockout mice compared with their WT counterparts. The fact that such differences were seen between the acute pharmacological approach and the genetic approach (gene deletion) of FABP needs to be further investigated. The function of FABPs and their specific effects on endocannabinoid anandamide, oleoylethanolamide, and palmitoylethanolamide may play an important role in the development of reward and mood behaviors and could provide opportunities for potential therapeutic targets.


Subject(s)
Fatty Acid-Binding Protein 7/deficiency , Fatty Acid-Binding Proteins/deficiency , Food Preferences/psychology , Freezing Reaction, Cataleptic/physiology , Gene Deletion , Neoplasm Proteins/deficiency , Sucrose/metabolism , Analysis of Variance , Animals , Arachidonic Acids/metabolism , Cyclobutanes/pharmacology , Dicarboxylic Acids/pharmacology , Endocannabinoids/metabolism , Exploratory Behavior/physiology , Fatty Acid-Binding Protein 7/genetics , Fatty Acid-Binding Proteins/genetics , Female , Food Preferences/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Oleic Acids/metabolism , Polyunsaturated Alkamides/metabolism , Sex Factors , Sucrose/administration & dosage , Swimming/psychology
15.
Synapse ; 72(6): e22031, 2018 06.
Article in English | MEDLINE | ID: mdl-29457656

ABSTRACT

Genetic and pharmacological manipulation of endocannabinoid (eCB) signaling has previously been shown to have an important role on the rewarding properties of drugs of abuse, including cocaine. Recently, fatty acid binding proteins (FABPs) have been proposed as intracellular transporters of the endocannabinoid anandamide (AEA) as well as other bioactive lipids to their catabolic enzyme, fatty acid amide hydrolase (FAAH). The role of these transporters in modulating the brains reward system has yet to be investigated. This study examined the effects of genetic deletion of FABP 5/7 on cocaine preference, as assessed by the Conditioned Place Preference (CPP) paradigm. Male and female wild type (WT) and FABP 5/7 KO mice showed similar acquisition of cocaine CPP, with no differences found in overall locomotor activity. In addition, while male and female WT mice showed stress-induced CPP for cocaine, male and female FABP 5/7 KO mice failed to show a stress-induced preference for the cocaine-paired chamber. Additionally, serum corticosterone levels were analyzed to explore any potential differences in stress response that may be responsible for the lack of stress-induced preference for cocaine. Serum samples were obtained in animals under basal conditions as well as following a 30-min tube restraint stress. Male and female FABP 5/7 KO mice showed reduced corticosterone levels under stress compared to their WT counterparts. The reduction in corticosterone response under stress may mediate that lack of a stress-induced preference for cocaine in the FABP 5/7 KO mice. Thus, the role of FABPs may play an important role in drug-seeking behavior under stressful conditions.


Subject(s)
Cocaine , Corticosterone/blood , Drug-Seeking Behavior/physiology , Fatty Acid-Binding Protein 7/deficiency , Fatty Acid-Binding Proteins/deficiency , Neoplasm Proteins/deficiency , Stress, Psychological/blood , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Drug-Seeking Behavior/drug effects , Extinction, Psychological/physiology , Fatty Acid-Binding Protein 7/genetics , Fatty Acid-Binding Proteins/genetics , Female , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Reward , Stress, Psychological/genetics
16.
J Lipid Res ; 59(4): 646-657, 2018 04.
Article in English | MEDLINE | ID: mdl-29414765

ABSTRACT

Phytocannabinoids, such as Δ9-tetrahydrocannabinol (THC), bind and activate cannabinoid (CB) receptors, thereby "piggy-backing" on the same pathway's endogenous endocannabinoids (ECs). The recent discovery that liver fatty acid binding protein-1 (FABP1) is the major cytosolic "chaperone" protein with high affinity for both Δ9-THC and ECs suggests that Δ9-THC may alter hepatic EC levels. Therefore, the impact of Δ9-THC or EC treatment on the levels of endogenous ECs, such as N-arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG), was examined in cultured primary mouse hepatocytes from WT and Fabp1 gene-ablated (LKO) mice. Δ9-THC alone or 2-AG alone significantly increased AEA and especially 2-AG levels in WT hepatocytes. LKO alone markedly increased AEA and 2-AG levels. However, LKO blocked/diminished the ability of Δ9-THC to further increase both AEA and 2-AG. In contrast, LKO potentiated the ability of exogenous 2-AG to increase the hepatocyte level of AEA and 2-AG. These and other data suggest that Δ9-THC increases hepatocyte EC levels, at least in part, by upregulating endogenous AEA and 2-AG levels. This may arise from Δ9-THC competing with AEA and 2-AG binding to FABP1, thereby decreasing targeting of bound AEA and 2-AG to the degradative enzymes, fatty acid amide hydrolase and monoacylglyceride lipase, to decrease hydrolysis within hepatocytes.


Subject(s)
Dronabinol/adverse effects , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Animals , Dronabinol/pharmacology , Fatty Acid-Binding Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
J Lipid Res ; 59(3): 416-428, 2018 03.
Article in English | MEDLINE | ID: mdl-29317465

ABSTRACT

Hepatic stellate cell (HSC) activation occurs along with decreased Perilipin5 (Plin5) and liver fatty acid-binding protein (L-Fabp) expression and coincident lipid droplet (LD) depletion. Conversely, the activated phenotype is reversible in WT HSCs upon forced expression of Plin5. Here, we asked if L-Fabp expression is required for Plin5-mediated rescue of the quiescent phenotype. Lentiviral Plin5 transduction of passaged L-Fabp-/- HSCs failed to reverse activation markers or restore lipogenic gene expression and LD formation. However, adenoviral L-Fabp infection of lentiviral Plin5 transduced L-Fabp-/- HSCs restored both the quiescent phenotype and LD formation, an effect also mediated by adenoviral intestine-Fabp or adipocyte-Fabp. Expression of exogenous Plin5 in activated WT HSCs induced a transcriptional program of lipogenic gene expression including endogenous L-Fabp, but none of the other FABPs. We further demonstrated that selective, small molecule inhibition of endogenous L-Fabp also eliminated the ability of exogenous Plin5 to rescue LD formation and reverse activation of WT HSCs. This functional coordination of L-Fabp with Plin5 was 5'-AMP-activated protein kinase (AMPK)-dependent and was eliminated by AMPK inhibition. Taken together, our results indicate that L-Fabp is required for Plin5 to activate a transcriptional program that restores LD formation and reverses HSC activation.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Perilipin-5/metabolism , Animals , Cells, Cultured , Fatty Acid-Binding Proteins/deficiency , Female , Lipid Droplets/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Perilipin-5/antagonists & inhibitors , Perilipin-5/genetics , Small Molecule Libraries/pharmacology
18.
J Lipid Res ; 58(11): 2114-2126, 2017 11.
Article in English | MEDLINE | ID: mdl-28972119

ABSTRACT

Upregulation of the hepatic endocannabinoid (EC) receptor [cannabinoid receptor-1 (CB1)] and arachidonoylethanolamide (AEA) is associated with nonalcoholic fatty liver disease (NAFLD). Male mice fed high-fat diet (HFD) ad libitum also exhibit NAFLD, increased hepatic AEA, and obesity. But, preference for HFD complicates interpretation and almost nothing is known about these effects in females. These issues were addressed by pair-feeding HFD. Similarly to ad libitum-fed HFD, pair-fed HFD also increased WT male and female mouse fat tissue mass (FTM), but preferentially at the expense of lean tissue mass. In contrast, pair-fed HFD did not elicit NAFLD in WT mice regardless of sex. Concomitantly, pair-fed HFD oppositely impacted hepatic AEA, 2-arachidonoyl glycerol, and/or CB1 in WT males versus females. In pair-fed HFD mice, liver FA binding protein-1 (Fabp1) gene ablation (LKO): i) exacerbated FTM in both sexes; ii) did not elicit liver neutral lipid accumulation in males and only slightly in females; iii) increased liver AEA in males, but decreased it in females; and iv) decreased CB1 only in males. Thus, pair-fed HFD selectively impacted hepatic ECs more in females, but did not elicit NAFLD in either sex. These effects were modified by LKO consistent with FABP1's ability to impact EC and FA metabolism.


Subject(s)
Diet, High-Fat/adverse effects , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Gene Knockout Techniques , Liver/drug effects , Liver/metabolism , Animals , Biological Transport/drug effects , Biological Transport/genetics , Biomarkers/blood , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytosol/drug effects , Cytosol/metabolism , Fatty Acids/metabolism , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Organ Size/genetics , Phenotype , Sterol Regulatory Element Binding Protein 1/genetics
19.
J Am Heart Assoc ; 6(9)2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28903937

ABSTRACT

BACKGROUND: Fatty acid-binding protein 4 (FABP4) is expressed in adipocytes, macrophages, and endothelial cells of capillaries but not arteries. FABP4 is secreted from adipocytes in association with lipolysis, and an elevated circulating FABP4 level is associated with obesity, insulin resistance, and atherosclerosis. However, little is known about the link between FABP4 and endovascular injury. We investigated the involvement of ectopic FABP4 expression in endothelial cells in neointima hyperplasia after vascular injury. METHODS AND RESULTS: Femoral arteries of 8-week-old male mice were subjected to wire-induced vascular injury. After 4 weeks, immunofluorescence staining showed that FABP4 was ectopically expressed in endothelial cells of the hyperplastic neointima. Neointima formation determined by intima area and intima to media ratio was significantly decreased in FABP4-defficient mice compared with that in wild-type mice. Adenovirus-mediated overexpression of FABP4 in human coronary artery endothelial cells (HCAECs) in vitro increased inflammatory cytokines and decreased phosphorylation of nitric oxide synthase 3. Furthermore, FABP4 was secreted from HCAECs. Treatment of human coronary smooth muscle cells or HCAECs with the conditioned medium of Fabp4-overexpressed HCAECs or recombinant FABP4 significantly increased gene expression of inflammatory cytokines and proliferation- and adhesion-related molecules in cells, promoted cell proliferation and migration of human coronary smooth muscle cells, and decreased phosphorylation of nitric oxide synthase 3 in HCAECs, which were attenuated in the presence of an anti-FABP4 antibody. CONCLUSIONS: Ectopic expression and secretion of FABP4 in vascular endothelial cells contribute to neointima formation after vascular injury. Suppression of ectopic FABP4 in the vascular endothelium would be a novel strategy against post-angioplasty vascular restenosis.


Subject(s)
Endothelial Cells/metabolism , Fatty Acid-Binding Proteins/metabolism , Femoral Artery/metabolism , Neointima , Vascular Remodeling , Vascular System Injuries/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Culture Media, Conditioned/metabolism , Cytokines/metabolism , Endothelial Cells/pathology , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Femoral Artery/injuries , Femoral Artery/pathology , Genotype , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Phenotype , Phosphorylation , Signal Transduction , Time Factors , Transfection , Vascular System Injuries/genetics , Vascular System Injuries/pathology
20.
Metabolism ; 77: 47-57, 2017 12.
Article in English | MEDLINE | ID: mdl-28941596

ABSTRACT

BACKGROUND: Myocardial contractile dysfunction in sepsis has been attributed mainly to increased inflammatory cytokines, insulin resistance, and impaired oxidative phosphorylation of fatty acids (FAs). However, precise molecular mechanisms underlying the cardiac dysfunction in sepsis remain to be determined. We previously reported major shift in myocardial energy substrates from FAs to glucose, and increased hepatic ketogenesis in mice lacking fatty acid-binding protein 4 (FABP4) and FABP5 (DKO). PURPOSE: We sought to determine whether a shift of energy substrates from FAs to glucose and increased availability of ketone bodies are beneficial or detrimental to cardiac function under the septic condition. METHODS: Lipopolysaccharide (LPS, 10mg/kg) was intraperitoneally injected into wild-type (WT) and DKO mice. Twelve hours after injection, cardiac function was assessed by echocardiography and serum and hearts were collected for further analyses. RESULTS: Cardiac contractile function was more deteriorated by LPS injection in DKO mice than WT mice despite comparable changes in pro-inflammatory cytokine production. LPS injection reduced myocardial uptake of FA tracer by 30% in both types of mice, while uptake of the glucose tracer did not significantly change in either group of mice in sepsis. Storage of glycogen and triacylglycerol in hearts was remarkably increased by LPS injection in both mice. Metabolome analysis revealed that LPS-induced suppression of pool size in the TCA cycle was more enhanced in DKO hearts. A tracing study with 13C6-glucose further revealed that LPS injection substantially reduced glucose-derived metabolites in the TCA cycle and related amino acids in DKO hearts. Consistent with these findings, glucose oxidation in vitro was similarly and markedly reduced in both mice. Serum concentration of ß-hydroxybutyrate and cardiac expression of genes associated with ketolysis were reduced in septic mice. CONCLUSIONS: Our study demonstrated that LPS-induced cardiac contractile dysfunction is associated with the robust suppression of catabolism of energy substrates including FAs, glucose and ketone bodies and accumulation of glycogen and triacylglycerol in the heart. Thus, a fuel shift from FAs to glucose and/or ketone bodies may be detrimental rather than protective under septic conditions.


Subject(s)
Energy Metabolism , Myocardium/metabolism , Sepsis/physiopathology , Animals , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Fatty Acids/metabolism , Glucose/metabolism , Glycogen/metabolism , Heart/physiopathology , Ketone Bodies/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Sepsis/chemically induced , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...