Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Soil Environ Health ; 1(2)2023 Jun.
Article in English | MEDLINE | ID: mdl-37830053

ABSTRACT

The use of nanoparticles in agrichemical formula and food products as additives has increased their chances of accumulation in humans via oral intake. Due to their potential toxicity, it is critical to understand their fate and distribution following oral intake. Cerium oxide nanoparticle (CeO2NP) is commonly used in agriculture and is highly stable in the environment. As such, it has been used as a model chemical to investigate nanoparticle's distribution and clearance. Based on their estimated human exposure levels, 0.15-0.75 mg/kg body weight/day of CeO2NPs with different sizes and surface charges (30-50 nm with negative charge and <25 nm with positive charge) were gavaged into C57BL/6 female mice daily. After 10-d, 50% of mice in each treatment were terminated, with the remaining being gavaged with 0.2 mL of deionized water daily for 7-d. Mouse organ tissues, blood, feces, and urine were collected at termination. At the tested levels, CeO2NPs displayed minimal overt toxicity to the mice, with their accumulation in various organs being negligible. Fecal discharge as the predominant clearance pathway took less than 7-d regardless of charges. Single particle inductively coupled plasma mass spectrometry analysis demonstrated minimal aggregation of CeO2NPs in the gastrointestinal tract. These findings suggest that nanoparticle additives >25 nm are unlikely to accumulate in mouse organ after oral intake, indicating limited impacts on human health.

2.
Nat Commun ; 14(1): 4342, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37468468

ABSTRACT

Although the role of the Wnt pathway in colon carcinogenesis has been described previously, it has been recently demonstrated that Wnt signaling originates from highly dynamic nano-assemblies at the plasma membrane. However, little is known regarding the role of oncogenic APC in reshaping Wnt nanodomains. This is noteworthy, because oncogenic APC does not act autonomously and requires activation of Wnt effectors upstream of APC to drive aberrant Wnt signaling. Here, we demonstrate the role of oncogenic APC in increasing plasma membrane free cholesterol and rigidity, thereby modulating Wnt signaling hubs. This results in an overactivation of Wnt signaling in the colon. Finally, using the Drosophila sterol auxotroph model, we demonstrate the unique ability of exogenous free cholesterol to disrupt plasma membrane homeostasis and drive Wnt signaling in a wildtype APC background. Collectively, these findings provide a link between oncogenic APC, loss of plasma membrane homeostasis and CRC development.


Subject(s)
Wnt Signaling Pathway , beta Catenin , Animals , beta Catenin/genetics , beta Catenin/metabolism , Carcinogenesis/genetics , Cell Membrane/metabolism , Colon/metabolism , Drosophila/metabolism , Wnt Signaling Pathway/genetics
3.
Behav Brain Res ; 440: 114256, 2023 02 25.
Article in English | MEDLINE | ID: mdl-36528169

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a key regulator in the microbiome-gut-brain axis, and AhR-active microbial metabolites modulate multiple neuronal responses. We recently demonstrated that 3,3'-diindolylmethane (DIM) and 1,4-dihydroxy-2-naphthoic acid (DHNA), two selective AhR modulators (SAhRMs), act as antidepressants in female mice. Thus, to examine the role of intestinal AhR in depression, anxiety, and spatial learning, this study employed transgenic mice in which the AhR was knockout only in the intestinal epithelium (AhRΔIEC). Additionally, this study examined whether the antidepressant effects of dietary DIM and DHNA is mediated by intestinal AhR. AhRΔIEC and WT female mice were fed daily with vehicle, 20 mg/kg DIM or DHNA for three weeks prior to four weeks of unpredictable chronic mild stress (UCMS). Mice were examined for weight gain, anhedonia-like behavior (sucrose preference test), anxiety levels (open field, light/dark, elevated plus maze, novelty-induced hypophagia, and marble burying tests), and spatial learning (Morris water maze). UCMS reduced weight gain in AhRΔIECs, but not WTs. Moreover, UCMS initially reduced sucrose preference in both AhRΔIECs and WTs, but over 4 weeks of UCMS, AhRΔIECs develop resilience to UCMS-induced anhedonia. Additionally, AhRΔIECs exhibit slightly reduced anxiety in certain tests and faster spatial learning. DIM and DHNA acted as antidepressants in both AhRΔIECs and WTs. Thus, this study suggests that intestinal AhR plays differential roles, mitigating stress effects on weight gain, and increasing stress effects on mood. However, the site of antidepressant action of SAhRMs, such as DIM and DHNA, is not dependent on the expression of intestinal AhR.


Subject(s)
Depression , Receptors, Aryl Hydrocarbon , Animals , Female , Mice , Anhedonia , Antidepressive Agents/pharmacology , Depression/drug therapy , Mice, Transgenic , Sucrose , Weight Gain
4.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G93-G106, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34755534

ABSTRACT

IL22 signaling plays an important role in maintaining gastrointestinal epithelial barrier function, cell proliferation, and protection of intestinal stem cells from genotoxicants. Emerging studies indicate that the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, promotes production of IL22 in gut immune cells. However, it remains to be determined if AhR signaling can also affect the responsiveness of colonic epithelial cells to IL22. Here, we show that IL22 treatment induces the phosphorylation of STAT3, inhibits colonic organoid growth, and promotes colonic cell proliferation in vivo. Notably, intestinal cell-specific AhR knockout (KO) reduces responsiveness to IL22 and compromises DNA damage response after exposure to carcinogen, in part due to the enhancement of suppressor of cytokine signaling 3 (SOCS3) expression. Deletion of SOCS3 increases levels of pSTAT3 in AhR KO organoids, and phenocopies the effects of IL22 treatment on wild-type (WT) organoid growth. In addition, pSTAT3 levels are inversely associated with increased azoxymethane/dextran sulfate sodium (AOM/DSS)-induced colon tumorigenesis in AhR KO mice. These findings indicate that AhR function is required for optimal IL22 signaling in colonic epithelial cells and provide rationale for targeting AhR as a means of reducing colon cancer risk.NEW & NOTEWORTHY AhR is a key transcription factor controlling expression of IL22 in gut immune cells. In this study, we show for the first time that AhR signaling also regulates IL22 response in colonic epithelial cells by modulating SOCS3 expression.


Subject(s)
Colon/drug effects , Colonic Neoplasms/drug therapy , Interleukins/pharmacology , Receptors, Aryl Hydrocarbon/drug effects , STAT3 Transcription Factor/drug effects , Animals , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Colon/metabolism , Colonic Neoplasms/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Mice, Knockout , Organoids/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein/drug effects , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transcriptional Activation/physiology , Interleukin-22
5.
Mol Nutr Food Res ; 65(20): e2100539, 2021 10.
Article in English | MEDLINE | ID: mdl-34406707

ABSTRACT

SCOPE: This study investigates the mechanism of action and functional effects of coffee extracts in colonic cells, on intestinal stem cell growth, and inhibition of dextran sodium sulfate (DSS)-induced intestinal barrier damage in mice. METHODS AND RESULTS: Aqueous coffee extracts induced Ah receptor (AhR) -responsive CYP1A1, CYP1B1, and UGT1A1 gene expression in colon-derived Caco2 and YAMC cells. Tissue-specific AhR knockout (AhRf/f x Lgr5-GFP-CreERT2 x Villin-Cre), wild-type (Lgr5-CreERT2 x Villin-Cre) mice are sources of stem cell enriched organoids and both coffee extracts and norharman, an AhR-active component of these extracts inhibited stem cell growth. Coffee extracts also inhibit DSS-induced damage to intestinal barrier function and DSS-induced mucosal inflammatory genes such as IL-6 and TGF-ß1 in wild-type (AhR+/+ ) but not AhR-/- mice. In contrast, coffee does not exhibit protective effects in intestinal-specific AhR knockout mice. Coffee extracts also enhanced overall formation of AhR-active microbial metabolites. CONCLUSIONS: In colon-derived cells and in the mouse intestine, coffee induced several AhR-dependent responses including gene expression, inhibition of intestinal stem cell-enriched organoid growth, and inhibition of DSS-induced intestinal barrier damage. We conclude that the anti-inflammatory effects of coffee in the intestine are due, in part, to activation of AhR signaling.


Subject(s)
Coffee , Colon/drug effects , Plant Extracts/pharmacology , Receptors, Aryl Hydrocarbon/physiology , Animals , Caco-2 Cells , Colon/metabolism , Cytochrome P-450 CYP1A1/physiology , Cytochrome P-450 CYP1B1/physiology , Dextran Sulfate/toxicity , Female , Humans , Male , Mice
6.
Microb Cell Fact ; 19(1): 219, 2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33256731

ABSTRACT

BACKGROUND: Diet, loss of aryl hydrocarbon receptor (AhR) expression and their modification of the gut microbiota community composition and its metabolites affect the development of colorectal cancer (CRC). However, the concordance between fecal microbiota composition and the fecal metabolome is poorly understood. Mice with specific AhR deletion (AhRKO) in intestinal epithelial cell and their wild-type littermates were fed a low-fat diet or a high-fat diet. Shifts in the fecal microbiome and metabolome associated with diet and loss of AhR expression were assessed. Microbiome and metabolome data were integrated to identify specific microbial taxa that contributed to the observed metabolite shifts. RESULTS: Our analysis shows that diet has a more pronounced effect on mouse fecal microbiota composition than the impact of the loss of AhR. In contrast, metabolomic analysis showed that the loss of AhR in intestinal epithelial cells had a more pronounced effect on metabolite profile compared to diet. Integration analysis of microbiome and metabolome identified unclassified Clostridiales, unclassified Desulfovibrionaceae, and Akkermansia as key contributors to the synthesis and/or utilization of tryptophan metabolites. CONCLUSIONS: Akkermansia are likely to contribute to the synthesis and/or degradation of tryptophan metabolites. Our study highlights the use of multi-omic analysis to investigate the relationship between the microbiome and metabolome and identifies possible taxa that can be targeted to manipulate the microbiome for CRC treatment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Diet , Feces/microbiology , Metabolome , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Akkermansia/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Colonic Neoplasms/microbiology , DNA, Bacterial , Female , Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , RNA, Ribosomal, 16S , Receptors, Aryl Hydrocarbon/genetics
7.
EMBO J ; 39(19): e104319, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32915464

ABSTRACT

The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that senses xenobiotics, diet, and gut microbial-derived metabolites, is increasingly recognized as a key regulator of intestinal biology. However, its effects on the function of colonic stem and progenitor cells remain largely unexplored. Here, we observed that inducible deletion of AhR in Lgr5+ stem cells increases the percentage of colonic stem cells and enhances organoid initiating capacity and growth of sorted stem and progenitor cells, while AhR activation has the opposite effect. Moreover, intestinal-specific AhR knockout increases basal stem cell and crypt injury-induced cell proliferation and promotes colon tumorigenesis in a preclinical colitis-associated tumor model by upregulating FoxM1 signaling. Mechanistically, AhR transcriptionally suppresses FoxM1 expression. Activation of AhR in human organoids recapitulates phenotypes observed in mice, such as reduction in the percentage of colonic stem cells, promotion of stem cell differentiation, and attenuation of FoxM1 signaling. These findings indicate that the AhR-FoxM1 axis, at least in part, mediates colonic stem/progenitor cell behavior.


Subject(s)
Colon/metabolism , Forkhead Box Protein M1/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Signal Transduction , Stem Cells/metabolism , Animals , Female , Forkhead Box Protein M1/genetics , Gene Knockout Techniques , Humans , Male , Mice , Mice, Transgenic , Receptors, Aryl Hydrocarbon/metabolism
8.
Lipids ; 55(4): 309-327, 2020 07.
Article in English | MEDLINE | ID: mdl-32314395

ABSTRACT

Ad libitum-fed diets high in fat and carbohydrate (especially fructose) induce weight gain, obesity, and nonalcoholic fatty liver disease (NAFLD) in humans and animal models. However, interpretation is complicated since ad libitum feeding of such diets induces hyperphagia and upregulates expression of liver fatty acid binding protein (L-FABP)-a protein intimately involved in fatty acid and glucose regulation of lipid metabolism. Wild-type (WT) and L-fabp gene ablated (LKO) mice were pair-fed either high-fat diet (HFD) or high-fat/high-glucose diet (HFGD) wherein total carbohydrate was maintained constant but the proportion of glucose was increased at the expense of fructose. In LKO mice, the pair-fed HFD increased body weight and lean tissue mass (LTM) but had no effect on fat tissue mass (FTM) or hepatic fatty vacuolation as compared to pair-fed WT counterparts. These LKO mice exhibited upregulation of hepatic proteins in fatty acid uptake and cytosolic transport (caveolin and sterol carrier protein-2), but lower hepatic fatty acid oxidation (decreased serum ß-hydroxybutyrate). LKO mice pair-fed HFGD also exhibited increased body weight; however, these mice had increased FTM, not LTM, and increased hepatic fatty vacuolation as compared to pair-fed WT counterparts. These LKO mice also exhibited upregulation of hepatic proteins in fatty acid uptake and cytosolic transport (caveolin and acyl-CoA binding protein, but not sterol carrier protein-2), but there was no change in hepatic fatty acid oxidation (serum ß-hydroxybutyrate) as compared to pair-fed WT counterparts.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Acid-Binding Proteins/genetics , Glucose/administration & dosage , 3-Hydroxybutyric Acid/blood , Animals , Body Weight/drug effects , Carrier Proteins/metabolism , Caveolin 2/metabolism , Fatty Acids/analysis , Female , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Male , Mice
9.
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
10.
Brain Res ; 1679: 155-170, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29217155

ABSTRACT

In this study, we analyze the neuropathological and biochemical alterations involved in the pathogenesis of a neurodegenerative/movement disorder during different developmental stages in juvenile rats with a mutant Myosin5a (Myo5a). In mutant rats, a spontaneous autosomal recessive mutation characterized by the absence of Myo5a protein expression in the brain is associated with a syndrome of locomotor dysfunction, altered coat color, and neuroendocrine abnormalities. Myo5a encodes a myosin motor protein required for transport and proper distribution of subcellular organelles in somatodendritic processes in neurons. Here we report marked hyperphosphorylation of alpha-synuclein and tau, as well as region-specific buildup of the autotoxic dopamine metabolite, 3,4-dihydroxyphenyl-acetaldehyde (DOPAL), related to decreased aldehyde dehydrogenases activity and neurodegeneration in mutant rats. Alpha-synuclein accumulation in mitochondria of dopaminergic neurons is associated with impaired enzymatic respiratory complex I and IV activity. The behavioral and biochemical lesions progress after 15 days postnatal, and by 30-40 days the animals must be euthanized because of neurological impairment. Based on the obtained results, we propose a pleiotropic pathogenesis that links the Myo5a gene mutation to deficient neuronal development and progressive neurodegeneration. This potential model of a neurodevelopmental disorder with neurodegeneration and motor deficits may provide further insight into molecular motors and their associated proteins responsible for altered neurogenesis and neuronal disease pathogenesis.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/pathology , Heredodegenerative Disorders, Nervous System , Mutation/genetics , Myosin Heavy Chains/genetics , Myosin Type V/genetics , tau Proteins/metabolism , 3,4-Dihydroxyphenylacetic Acid/analogs & derivatives , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Central Nervous System/ultrastructure , Disease Models, Animal , Electron Transport Chain Complex Proteins/metabolism , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Heredodegenerative Disorders, Nervous System/pathology , Microscopy, Electron, Transmission , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Phosphorylation/genetics , Rats , Rats, Mutant Strains , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , alpha-Synuclein/ultrastructure , tau Proteins/genetics , tau Proteins/ultrastructure
11.
Lipids ; 52(5): 385-397, 2017 05.
Article in English | MEDLINE | ID: mdl-28382456

ABSTRACT

Liver fatty acid binding protein (Fabp1) and sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) genes encode proteins that enhance hepatic uptake, cytosolic transport, and peroxisomal oxidation of toxic branched-chain fatty acids derived from dietary phytol. Since male wild-type (WT) mice express markedly higher levels of these proteins than females, the impact of ablating both genes (TKO) was examined in phytol-fed males. In WT males, high phytol diet alone had little impact on whole body weight and did not alter the proportion of lean tissue mass (LTM) versus fat tissue mass (FTM). TKO conferred on dietary phytol the ability to induce weight loss as well as reduce liver weight, FTM, and even more so LTM. Concomitantly TKO induced hepatic lipid accumulation, preferentially threefold increased phospholipid (PL) at the expense of decreased triacylglycerol (TG) and total cholesterol. Increased PL was associated with upregulation of membrane fatty acid transport/translocase proteins (FATP 2,4), cytosolic fatty acid/fatty acyl-CoA binding proteins (FABP2, ACBP), and the rate limiting enzyme in PL synthesis (Gpam). Decreased TG and cholesterol levels were not attributable to altered levels in respective synthetic enzymes or nuclear receptors. These data suggest that the higher level of Fabp1 and Scp2/Scpx gene products in WT males was protective against deleterious effects of dietary phytol, but TKO significantly exacerbated phytol effects in males.


Subject(s)
Body Weight/drug effects , Carrier Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Liver/drug effects , Phytol/administration & dosage , Animals , Fatty Acid-Binding Proteins/metabolism , Gene Knockout Techniques , Liver/chemistry , Liver/metabolism , Male , Mice , Organ Size/drug effects , Phenotype , Phospholipids/analysis , Phytol/pharmacology , Up-Regulation
12.
Exp Neurol ; 287(Pt 1): 21-33, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27771352

ABSTRACT

The presence of α-synuclein (α-syn) in Lewy bodies and Lewy neurites is an important characteristic of the neurodegenerative processes of substantia nigra pars compacta (SNpc) dopaminergic (DAergic) neurons in Parkinson's disease (PD) and other synucleinopathies. Here we report that Berlin-Druckrey rats carrying a spontaneous mutation in the 3' untranslated region of α-syn mRNA (m/m rats) display a marked accumulation of α-syn in the mesencephalic area, striatum and frontal cortex, accompanied to severe dysfunctions in the dorsolateral striatum. Despite a small reduction in the number of SNpc and ventral tegmental area DAergic cells, the surviving dopaminergic neurons of the m/m rats do not show clear-cut alterations of the spontaneous and evoked firing activity, DA responses and somatic amphetamine-induced firing inhibition. Interestingly, mutant DAergic neurons display diminished whole-cell Ih conductance and a reduced frequency of spontaneous excitatory synaptic currents. By contrast, m/m rats show a severe impairment of DA and glutamate release in the dorsolateral striatum, as revealed by amperometric measure of DA currents and by electrophysiological recordings of glutamatergic synaptic events in striatal medium spiny neurons. These functional impairments are paralleled by a decreased expression of the DA transporter and VGluT1 proteins in the same area. Thus, together with α-syn overload in the mesencephalic region, striatum and frontal cortex, the main functional alterations occur in the DAergic and glutamatergic terminals in the dorsal striatum of the m/m rats.


Subject(s)
Dopaminergic Neurons/physiology , Glutamic Acid/metabolism , Membrane Potentials/physiology , Mesencephalon/cytology , alpha-Synuclein/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Bicuculline/pharmacology , Cell Count , Dopamine/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , GABA-B Receptor Agonists/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Patch-Clamp Techniques , Picrotoxin/pharmacology , Rats , Synaptic Potentials/drug effects , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , alpha-Synuclein/genetics
13.
Biochemistry ; 55(37): 5243-55, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27552286

ABSTRACT

Endocannabinoids (ECs) and cannabinoids are very lipophilic molecules requiring the presence of cytosolic binding proteins that chaperone these molecules to intracellular targets. While three different fatty acid binding proteins (FABP3, -5, and -7) serve this function in brain, relatively little is known about how such hydrophobic ECs and cannabinoids are transported within the liver. The most prominent hepatic FABP, liver fatty acid binding protein (FABP1 or L-FABP), has high affinity for arachidonic acid (ARA) and ARA-CoA, suggesting that FABP1 may also bind ARA-derived ECs (AEA and 2-AG). Indeed, FABP1 bound ECs with high affinity as shown by displacement of FABP1-bound fluorescent ligands and by quenching of FABP1 intrinsic tyrosine fluorescence. FABP1 also had high affinity for most non-ARA-containing ECs, FABP1 inhibitors, EC uptake/hydrolysis inhibitors, and phytocannabinoids and less so for synthetic cannabinoid receptor (CBR) agonists and antagonists. The physiological impact was examined with liver from wild-type (WT) versus FABP1 gene-ablated (LKO) male mice. As shown by liquid chromatography and mass spectrometry, FABP1 gene ablation significantly increased hepatic levels of AEA, 2-AG, and 2-OG. These increases were not due to increased protein levels of EC synthetic enzymes (NAPEPLD and DAGL) or a decreased level of EC degradative enzyme (FAAH) but correlated with complete loss of FABP1, a decreased level of SCP2 (8-fold less prevalent than FABP1, but also binds ECs), and a decreased level of degradative enzymes (NAAA and MAGL). These data indicated that FABP1 not only is the most prominent endocannabinoid and cannabinoid binding protein but also impacts hepatic endocannabinoid levels.


Subject(s)
Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/metabolism , Receptors, Cannabinoid/metabolism , Animals , Female , Fluorescent Dyes , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL
14.
Lipids ; 51(9): 1007-20, 2016 09.
Article in English | MEDLINE | ID: mdl-27450559

ABSTRACT

Although liver fatty acid binding protein (FABP1, L-FABP) is not detectable in the brain, Fabp1 gene ablation (LKO) markedly increases endocannabinoids (EC) in brains of male mice. Since the brain EC system of females differs significantly from that of males, it was important to determine if LKO differently impacted the brain EC system. LKO did not alter brain levels of arachidonic acid (ARA)-containing EC, i.e. arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG), but decreased non-ARA-containing N-acylethanolamides (OEA, PEA) and 2-oleoylglycerol (2-OG) that potentiate the actions of AEA and 2-AG. These changes in brain potentiating EC levels were not associated with: (1) a net decrease in levels of brain membrane proteins associated with fatty acid uptake and EC synthesis; (2) a net increase in brain protein levels of cytosolic EC chaperones and enzymes in EC degradation; or (3) increased brain protein levels of EC receptors (CB1, TRVP1). Instead, the reduced or opposite responsiveness of female brain EC levels to loss of FABP1 (LKO) correlated with intrinsically lower FABP1 level in livers of WT females than males. These data show that female mouse brain endocannabinoid levels were unchanged (AEA, 2-AG) or decreased (OEA, PEA, 2-OG) by complete loss of FABP1 (LKO).


Subject(s)
Brain/metabolism , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/deficiency , Animals , Arachidonic Acid/metabolism , Ethanolamines/metabolism , Female , Glycerides/metabolism , Male , Mice
15.
J Neurochem ; 138(3): 407-22, 2016 08.
Article in English | MEDLINE | ID: mdl-27167970

ABSTRACT

Liver fatty acid-binding protein (FABP1, L-FABP) has high affinity for and enhances uptake of arachidonic acid (ARA, C20:4, n-6) which, when esterified to phospholipids, is the requisite precursor for synthesis of endocannabinoids (EC) such as arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG). The brain derives most of its ARA from plasma, taking up ARA and transporting it intracellularly via cytosolic fatty acid-binding proteins (FABPs 3,5, and 7) localized within the brain. In contrast, the much more prevalent cytosolic FABP1 is not detectable in the brain but is instead highly expressed in the liver. Therefore, the possibility that FABP1 outside the central nervous system may regulate brain AEA and 2-AG was examined in wild-type (WT) and FABP1 null (LKO) male mice. LKO increased brain levels of AA-containing EC (AEA, 2-AG), correlating with increased free and total ARA in brain and serum. LKO also increased brain levels of non-ARA that contain potentiating endocannabinoids (EC*) such as oleoyl ethanolamide (OEA), PEA, 2-OG, and 2-PG. Concomitantly, LKO decreased serum total ARA-containing EC, but not non-ARA endocannabinoids. LKO did not elicit these changes in the brain EC and EC* as a result of compensatory up-regulation of brain protein levels of enzymes in EC synthesis (NAPEPLD, DAGLα) or cytosolic EC chaperone proteins (FABPs 3, 5, 7, SCP-2, HSP70), or cannabinoid receptors (CB1, TRVP1). These data show for the first time that the non-CNS fatty acid-binding protein FABP1 markedly affected brain levels of both ARA-containing endocannabinoids (AEA, 2-AG) as well as their non-ARA potentiating endocannabinoids. Fatty acid-binding protein-1 (FABP-1) is not detectable in brain but instead is highly expressed in liver. The possibility that FABP1 outside the central nervous system may regulate brain endocannabinoids arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG) was examined in wild-type (WT) and FABP-1 null (LKO) male mice. LKO increased brain levels of arachidonic acid-containing endocannabinoids (AEA, 2-AG), correlating with increased free and total arachidonic acid in brain and serum. Read the Editorial Highlight for this article on page 371.


Subject(s)
Arachidonic Acids/metabolism , Brain/metabolism , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/genetics , Liver/metabolism , Oleic Acids/metabolism , Polyunsaturated Alkamides/metabolism , Animals , Arachidonic Acids/genetics , Brain/drug effects , Endocannabinoids/genetics , Glycerides/metabolism , Liver/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout
16.
Lipids ; 51(6): 655-76, 2016 06.
Article in English | MEDLINE | ID: mdl-27117865

ABSTRACT

The first discovered member of the mammalian FABP family, liver fatty acid binding protein (FABP1, L-FABP), occurs at high cytosolic concentration in liver, intestine, and in the case of humans also in kidney. While the rat FABP1 is well studied, the extent these findings translate to human FABP1 is not clear-especially in view of recent studies showing that endocannabinoids and cannabinoids represent novel rat FABP1 ligands and FABP1 gene ablation impacts the hepatic endocannabinoid system, known to be involved in non-alcoholic fatty liver (NAFLD) development. Although not detectable in brain, FABP1 ablation nevertheless also impacts brain endocannabinoids. Despite overall tertiary structure similarity, human FABP1 differs significantly from rat FABP1 in secondary structure, much larger ligand binding cavity, and affinities/specificities for some ligands. Moreover, while both mouse and human FABP1 mediate ligand induction of peroxisome proliferator activated receptor-α (PPARα), they differ markedly in pattern of genes induced. This is critically important because a highly prevalent human single nucleotide polymorphism (SNP) (26-38 % minor allele frequency and 8.3 ± 1.9 % homozygous) results in a FABP1 T94A substitution that further accentuates these species differences. The human FABP1 T94A variant is associated with altered body mass index (BMI), clinical dyslipidemias (elevated plasma triglycerides and LDL cholesterol), atherothrombotic cerebral infarction, and non-alcoholic fatty liver disease (NAFLD). Resolving human FABP1 and the T94A variant's impact on the endocannabinoid and cannabinoid system is an exciting challenge due to the importance of this system in hepatic lipid accumulation as well as behavior, pain, inflammation, and satiety.


Subject(s)
Dyslipidemias/genetics , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/genetics , Polymorphism, Single Nucleotide , Animals , Body Mass Index , Brain/metabolism , Cerebral Infarction/etiology , Cerebral Infarction/genetics , Dyslipidemias/metabolism , Fatty Acid-Binding Proteins/chemistry , Humans , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/genetics , PPAR alpha/metabolism , Protein Structure, Secondary , Rats , Species Specificity
17.
Arch Biochem Biophys ; 588: 25-32, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26541319

ABSTRACT

Both sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) and liver fatty acid binding protein (L-FABP) have been proposed to function in hepatobiliary bile acid metabolism/accumulation. To begin to address this issue, the impact of ablating L-FABP (LKO) or SCP-2/SCP-x (DKO) individually or both together (TKO) was examined in female mice. Biliary bile acid levels were decreased in LKO, DKO, and TKO mice; however, hepatic bile acid concentration was decreased in LKO mice only. In contrast, biliary phospholipid level was decreased only in TKO mice, while biliary cholesterol levels were unaltered regardless of phenotype. The loss of either or both genes increased hepatic expression of the major bile acid synthetic enzymes (CYP7A1 and/or CYP27A1). Loss of L-FABP and/or SCP-2/SCP-x genes significantly altered the molecular composition of biliary bile acids, but not the proportion of conjugated/unconjugated bile acids or overall bile acid hydrophobicity index. These data suggested that L-FABP was more important in hepatic retention of bile acids, while SCP-2/SCP-x more broadly affected biliary bile acid and phospholipid levels.


Subject(s)
Biliary Tract/metabolism , Carrier Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Liver/metabolism , Animals , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Carrier Proteins/genetics , Cholesterol/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phospholipids/metabolism
18.
Arch Biochem Biophys ; 580: 41-9, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26116377

ABSTRACT

Although roles for both sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) and liver fatty acid binding protein (L-FABP) have been proposed in hepatic lipid accumulation, individually ablating these genes has been complicated by concomitant alterations in the other gene product(s). For example, ablating SCP2/SCP-x induces upregulation of L-FABP in female mice. Therefore, the impact of ablating SCP-2/SCP-x (DKO) or L-FABP (LKO) individually or both together (TKO) was examined in female mice. Loss of SCP-2/SCP-x (DKO, TKO) more so than loss of L-FABP alone (LKO) increased hepatic total lipid and total cholesterol content, especially cholesteryl ester. Hepatic accumulation of nonesterified long chain fatty acids (LCFA) and phospholipids occurred only in DKO and TKO mice. Loss of SCP-2/SCP-x (DKO, TKO) increased serum total lipid primarily by increasing triglycerides. Altered hepatic level of proteins involved in cholesterol uptake, efflux, and/or secretion was observed, but did not compensate for the loss of L-FABP, SCP-2/SCP-x or both. However, synergistic responses were not seen with the combinatorial knock out animals-suggesting that inhibiting SCP-2/SCP-x is more correlative with hepatic dysfunction than L-FABP. The DKO- and TKO-induced hepatic accumulation of cholesterol and long chain fatty acids shared significant phenotypic similarities with non-alcoholic fatty liver disease (NAFLD).


Subject(s)
Carrier Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Lipid Metabolism/genetics , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Carrier Proteins/metabolism , Cholesterol/blood , Cholesterol Esters/blood , Disease Models, Animal , Fatty Acid-Binding Proteins/deficiency , Fatty Acids, Nonesterified/blood , Female , Gene Deletion , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Phospholipids/blood , Triglycerides/blood
19.
Am J Physiol Gastrointest Liver Physiol ; 309(5): G387-99, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26113298

ABSTRACT

While a high-cholesterol diet induces hepatic steatosis, the role of intracellular sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) proteins is unknown. We hypothesized that ablating SCP-2/SCP-x [double knockout (DKO)] would impact hepatic lipids (cholesterol and cholesteryl ester), especially in high-cholesterol-fed mice. DKO did not alter food consumption, and body weight (BW) gain decreased especially in females, concomitant with hepatic steatosis in females and less so in males. DKO-induced steatosis in control-fed wild-type (WT) mice was associated with 1) loss of SCP-2; 2) upregulation of liver fatty acid binding protein (L-FABP); 3) increased mRNA and/or protein levels of sterol regulatory element binding proteins (SREBP1 and SREBP2) as well as increased expression of target genes of cholesterol synthesis (Hmgcs1 and Hmgcr) and fatty acid synthesis (Acc1 and Fas); and 4) cholesteryl ester accumulation was also associated with increased acyl-CoA cholesterol acyltransferase-2 (ACAT2) in males. DKO exacerbated the high-cholesterol diet-induced hepatic cholesterol and glyceride accumulation, without further increasing SREBP1, SREBP2, or target genes. This exacerbation was associated both with loss of SCP-2 and concomitant downregulation of Ceh/Hsl, apolipoprotein B (ApoB), MTP, and/or L-FABP protein expression. DKO diminished the ability to secrete excess cholesterol into bile and oxidize cholesterol to bile acid for biliary excretion, especially in females. This suggested that SCP-2/SCP-x affects cholesterol transport to particular intracellular compartments, with ablation resulting in less to the endoplasmic reticulum for SREBP regulation, making more available for cholesteryl ester synthesis, for cholesteryl-ester storage in lipid droplets, and for bile salt synthesis and/or secretion. These alterations are significant findings, since they affect key processes in regulation of sterol metabolism.


Subject(s)
Carrier Proteins/metabolism , Cholesterol, Dietary/pharmacology , Lipid Metabolism , Liver/metabolism , Acetyltransferases/genetics , Acetyltransferases/metabolism , Animals , Apolipoprotein B-100 , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Carrier Proteins/genetics , Cholesterol, Dietary/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , fas Receptor/genetics , fas Receptor/metabolism , Sterol O-Acyltransferase 2
20.
Biochim Biophys Acta ; 1851(7): 946-55, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25732850

ABSTRACT

Although expression of the human liver fatty acid binding protein (FABP1) T94A variant alters serum lipoprotein cholesterol levels in human subjects, nothing is known whereby the variant elicits these effects. This issue was addressed by in vitro cholesterol binding assays using purified recombinant wild-type (WT) FABP1 T94T and T94A variant proteins and in cultured primary human hepatocytes expressing the FABP1 T94T (genotyped as TT) or T94A (genotyped as CC) proteins. The human FABP1 T94A variant protein had 3-fold higher cholesterol-binding affinity than the WT FABP1 T94T as shown by NBD-cholesterol fluorescence binding assays and by cholesterol isothermal titration microcalorimetry (ITC) binding assays. CC variant hepatocytes also exhibited 30% higher total FABP1 protein. HDL- and LDL-mediated NBD-cholesterol uptake was faster in CC variant than TT WT human hepatocytes. VLDL-mediated uptake of NBD-cholesterol did not differ between CC and TT human hepatocytes. The increased HDL- and LDL-mediated NBD-cholesterol uptake was not associated with any significant change in mRNA levels of SCARB1, LDLR, CETP, and LCAT encoding the key proteins in lipoprotein cholesterol uptake. Thus, the increased HDL- and LDL-mediated NBD-cholesterol uptake by CC hepatocytes may be associated with higher affinity of T94A protein for cholesterol and/or increased total T94A protein level.


Subject(s)
Cholesterol/metabolism , Fatty Acid-Binding Proteins/genetics , Hepatocytes/metabolism , Mutation, Missense , Alanine/genetics , Amino Acid Substitution , Animals , Biological Transport/genetics , Cells, Cultured , Fatty Acid-Binding Proteins/metabolism , Female , Humans , Lipid Metabolism/genetics , Middle Aged , Threonine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...