Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Diabetologia ; 65(2): 366-374, 2022 02.
Article in English | MEDLINE | ID: mdl-34806114

ABSTRACT

AIMS/HYPOTHESIS: Fatty acid-binding protein 4 (FABP4) is an adipokine with a key regulatory role in glucose and lipid metabolism. We prospectively evaluated the role of FABP4 in the pathophysiology of diabetic ketoacidosis (DKA) in new-onset type 1 diabetes. METHODS: Clinical and laboratory data were prospectively collected from consecutive children presenting with new-onset type 1 diabetes. In addition to blood chemistry and gases, insulin, C-peptide, serum FABP4 and NEFA were collected upon presentation and 48 h after initiation of insulin treatment. In a mouse model of type 1 diabetes, glucose, insulin, ß-hydroxybutyrate and weight were compared between FABP4 knockout (Fabp4-/-) and wild-type (WT) mice. RESULTS: Included were 33 children (mean age 9.3 ± 3.5 years, 52% male), of whom 14 (42%) presented with DKA. FABP4 levels were higher in the DKA group compared with the non-DKA group (median [IQR] 10.1 [7.9-14.2] ng/ml vs 6.3 [3.9-7] ng/ml, respectively; p = 0.005). The FABP4 level was positively correlated with HbA1c at presentation and inversely correlated with venous blood pH and bicarbonate levels (p < 0.05 for all). Following initiation of insulin therapy, a marked reduction in FABP4 was observed in all children. An FABP4 level of 7.22 ng/ml had a sensitivity of 86% and a specificity of 78% for the diagnosis of DKA, with an area under the receiver operating characteristic curve of 0.78 (95% CI 0.6, 0.95; p = 0.008). In a streptozotocin-induced diabetes mouse model, Fabp4-/- mice exhibited marked hypoinsulinaemia and hyperglycaemia similar to WT mice but displayed no significant increase in ß-hydroxybutyrate and were protected from ketoacidosis. CONCLUSIONS/INTERPRETATION: FABP4 is suggested to be a necessary regulator of ketogenesis in insulin-deficient states.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetic Ketoacidosis/metabolism , Fatty Acid-Binding Proteins/physiology , Animals , Blood Glucose/metabolism , Child , Diabetes Mellitus, Experimental , Female , Glycated Hemoglobin/metabolism , Humans , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prospective Studies
2.
Int J Cancer ; 150(1): 152-163, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34449874

ABSTRACT

Plasmacytoid dendritic cells (pDCs) promote viral elimination by producing large amounts of Type I interferon. Recent studies have shown that pDCs regulate the pathogenesis of diverse inflammatory diseases, such as cancer. Fatty acid-binding protein 5 (FABP5) is a cellular chaperone of long-chain fatty acids that induce biological responses. Although the effects of FABP-mediated lipid metabolism are well studied in various immune cells, its role in pDCs remains unclear. This study, which compares wild-type and Fabp5-/- mice, provides the first evidence that FABP5-mediated lipid metabolism regulates the commitment of pDCs to inflammatory vs tolerogenic gene expression patterns in the tumor microenvironment and in response to toll-like receptor stimulation. Additionally, we demonstrated that FABP5 deficiency in pDCs affects the surrounding cellular environment, and that FABP5 expression in pDCs supports the appropriate generation of regulatory T cells (Tregs). Collectively, our findings reveal that pDC FABP5 acts as an important regulator of tumor immunity by controlling lipid metabolism.


Subject(s)
Dendritic Cells/immunology , Fatty Acid-Binding Proteins/physiology , Forkhead Transcription Factors/metabolism , Interferon Type I/metabolism , Lipid Metabolism , Neoplasm Proteins/physiology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment , Animals , Forkhead Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptors/metabolism
3.
Biochem Biophys Res Commun ; 599: 87-92, 2022 04 09.
Article in English | MEDLINE | ID: mdl-35176630

ABSTRACT

Fatty acid-binding proteins (FABPs) are lipid chaperones that mediate the intracellular dynamics of the hydrophobic molecules that they physically bind to. FABPs are implicated in sleep and psychiatric disorders, as well as in various cellular processes, such as cell proliferation and survival. FABP is well conserved in insects, and Drosophila has one FABP ortholog, dFabp, in its genome. Although dFabp appears to be evolutionarily conserved in some brain functions, little is known about its development and physiological function. In the present study, we investigated the function of dFabp in Drosophila development and behavior. Knockdown or overexpression of dFabp in the developing brain, wing, and eye resulted in developmental defects, such as decreased survival, altered cell proliferation, and increased apoptosis. Glia-specific knockdown of dFabp affected neuronal development, and neuronal regulation of dFabp affected glial cell proliferation. Moreover, the behavioral phenotypes (circadian rhythm and locomotor activity) of flies with regulated dFabp expression in glia and flies with regulated dFabp expression in neurons were very similar. Collectively, our results suggest that dFabp is involved in the development of various tissues and brain functions to control behavior and is a mediator of neuron-glia interactions in the Drosophila nervous system.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Fatty Acid-Binding Proteins/physiology , Animals , Behavior, Animal/physiology , Brain/embryology , Brain/growth & development , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Embryo, Nonmammalian/physiology , Female , Gene Expression Regulation, Developmental , Male , Wings, Animal/growth & development
4.
J Neurosci Res ; 99(2): 561-572, 2021 02.
Article in English | MEDLINE | ID: mdl-32964457

ABSTRACT

Stroke severely impairs quality of life and has a high mortality rate. On the other hand, dietary docosahexaenoic acid (DHA) prevents neuronal damage. In this review, we describe the effects of dietary DHA on ischemic stroke-associated neuronal damage and its role in stroke prevention. Recent epidemiological studies have been conducted to analyze stroke prevention through DHA intake. The effects of dietary intake and supply of DHA to neuronal cells, DHA-mediated inhibition of neuronal damage, and its mechanism, including the effects of the DHA metabolite, neuroprotectin D1 (NPD1), were investigated. These studies revealed that DHA intake was associated with a reduced risk of stroke. Moreover, studies have shown that DHA intake may reduce stroke mortality rates. DHA, which is abundant in fish oil, passes through the blood-brain barrier to accumulate as a constituent of phospholipids in the cell membranes of neuronal cells and astrocytes. Astrocytes supply DHA to neuronal cells, and neuronal DHA, in turn, activates Akt and Raf-1 to prevent neuronal death or damage. Therefore, DHA indirectly prevents neuronal damage. Furthermore, NDP1 blocks neuronal apoptosis. DHA, together with NPD1, may block neuronal damage and prevent stroke. The inhibitory effect on neuronal damage is achieved through the antioxidant (via inducing the Nrf2/HO-1 system) and anti-inflammatory effects (via promoting JNK/AP-1 signaling) of DHA.


Subject(s)
Brain Damage, Chronic/prevention & control , Docosahexaenoic Acids/therapeutic use , Ischemic Stroke/diet therapy , Nerve Degeneration/prevention & control , Stroke/prevention & control , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Apoptosis/drug effects , Biological Availability , Biological Transport , Blood-Brain Barrier , Brain Damage, Chronic/etiology , Dietary Fats/administration & dosage , Dietary Fats/pharmacokinetics , Dietary Fats/therapeutic use , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacokinetics , Docosahexaenoic Acids/pharmacology , Fatty Acid-Binding Proteins/physiology , Fish Oils/administration & dosage , Fish Oils/pharmacokinetics , Humans , Incidence , Ischemic Stroke/complications , Ischemic Stroke/epidemiology , Membrane Lipids/metabolism , Mice , Neoplasm Proteins/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Plant Oils/administration & dosage , Plant Oils/pharmacokinetics , Signal Transduction/drug effects , Symporters/deficiency , Symporters/physiology , alpha-Linolenic Acid/pharmacokinetics
5.
Proc Natl Acad Sci U S A ; 115(13): 3482-3487, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29531087

ABSTRACT

Endocannabinoids (eCBs) are lipid-signaling molecules involved in the regulation of numerous behaviors and physiological functions. Released by postsynaptic neurons, eCBs mediate retrograde modulation of synaptic transmission and plasticity by activating presynaptic cannabinoid receptors. While the cellular mechanisms by which eCBs control synaptic function have been well characterized, the mechanisms controlling their retrograde synaptic transport remain unknown. Here, we demonstrate that fatty-acid-binding protein 5 (FABP5), a canonical intracellular carrier of eCBs, is indispensable for retrograde eCB transport in the dorsal raphe nucleus (DRn). Thus, pharmacological inhibition or genetic deletion of FABP5 abolishes both phasic and tonic eCB-mediated control of excitatory synaptic transmission in the DRn. The blockade of retrograde eCB signaling induced by FABP5 inhibition is not mediated by impaired cannabinoid receptor function or reduced eCB synthesis. These findings indicate that FABP5 is essential for retrograde eCB signaling and may serve as a synaptic carrier of eCBs at central synapses.


Subject(s)
Arachidonic Acids/metabolism , Endocannabinoids/pharmacology , Fatty Acid-Binding Proteins/physiology , Glutamic Acid/metabolism , Glycerides/metabolism , Neoplasm Proteins/physiology , Synapses/physiology , Synaptic Transmission/drug effects , Animals , Cells, Cultured , Endocannabinoids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , Synapses/drug effects
6.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34576221

ABSTRACT

RNA interference (RNAi) is a powerful tool for the analysis of gene function in nematodes. Fatty acid and retinol binding protein (FAR) is a protein that only exists in nematodes and plays an important role in their life activities. The rice white-tip nematode (RWTN), Aphelenchoides besseyi, is a migratory endoparasitic plant nematode that causes serious damage in agricultural production. In this study, the expression levels of eight RWTN genes were effectively decreased when RWTN was fed Ab-far-n (n: 1-8) hairpin RNA transgenic Botrytis cinerea (ARTBn). These functions of the far gene family were identified to be consistent and diverse through phenotypic changes after any gene was silenced. Such consistency indicates that the body lengths of the females were significantly shortened after silencing any of the eight Ab-far genes. The diversities were mainly manifested as follows: (1) Reproduction of nematodes was clearly inhibited after Ab-far-1 to Ab-far-4 were silenced. In addition, silencing Ab-far-2 could inhibit the pathogenicity of nematodes to Arabidopsis; (2) gonad length of female nematodes was significantly shortened after Ab-far-2 and Ab-far-4 were silenced; (3) proportion of male nematodes significantly increased in the adult population after Ab-far-1, Ab-far-3, and Ab-far-5 were silenced, whereas the proportion of adult nematodes significantly decreased in the nematode population after Ab-far-4 were silenced. (4) Fat storage of nematodes significantly decreased after Ab-far-3, Ab-far-4, and Ab-far-7 were silenced. To our knowledge, this is the first study to demonstrate that Ab-far genes affect sex formation and lipid metabolism in nematodes, which provides valuable data for further study and control of RWTNs.


Subject(s)
Botrytis/genetics , Fatty Acid-Binding Proteins/physiology , Gene Expression Profiling , Nematoda/metabolism , Nematoda/pathogenicity , RNA Interference , Retinol-Binding Proteins/physiology , Animals , Animals, Genetically Modified , Arabidopsis/parasitology , Fatty Acid-Binding Proteins/genetics , Fatty Acids/chemistry , Gene Silencing , Helminth Proteins/genetics , Phenotype , Retinol-Binding Proteins/genetics , Transcriptome
7.
J Biol Chem ; 294(44): 16416-16428, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31530637

ABSTRACT

Streptococcus pneumoniae is responsible for the majority of pneumonia, motivating ongoing searches for insights into its physiology that could enable new treatments. S. pneumoniae responds to exogenous fatty acids by suppressing its de novo biosynthetic pathway and exclusively utilizing extracellular fatty acids for membrane phospholipid synthesis. The first step in exogenous fatty acid assimilation is phosphorylation by fatty acid kinase (FakA), whereas bound by a fatty acid-binding protein (FakB). Staphylococcus aureus has two binding proteins, whereas S. pneumoniae expresses three. The functions of these binding proteins were not clear. We determined the SpFakB1- and SpFakB2-binding proteins were bioinformatically related to the two binding proteins of Staphylococcus aureus, and biochemical and X-ray crystallographic analysis showed that SpFakB1 selectively bound saturates, whereas SpFakB2 allows the activation of monounsaturates akin to their S. aureus counterparts. The distinct SpFakB3 enables the utilization of polyunsaturates. The SpFakB3 crystal structure in complex with linoleic acid reveals an expanded fatty acid-binding pocket within the hydrophobic interior of SpFakB3 that explains its ability to accommodate multiple cis double bonds. SpFakB3 also utilizes a different hydrogen bond network than other FakBs to anchor the fatty acid carbonyl and stabilize the protein. S. pneumoniae strain JMG1 (ΔfakB3) was deficient in incorporation of linoleate from human serum verifying the role of FakB3 in this process. Thus, the multiple FakBs of S. pneumoniae permit the utilization of the entire spectrum of mammalian fatty acid structures to construct its membrane.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Streptococcus pneumoniae/metabolism , Bacterial Proteins/metabolism , Biosynthetic Pathways , Fatty Acid-Binding Proteins/physiology , Fatty Acid-Binding Proteins/ultrastructure , Fatty Acids, Unsaturated/metabolism , Host-Pathogen Interactions/physiology , Humans , Phospholipids/metabolism , Phosphorylation , Serum/chemistry , Staphylococcus aureus/metabolism
8.
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
9.
Plant J ; 96(6): 1206-1217, 2018 12.
Article in English | MEDLINE | ID: mdl-30242928

ABSTRACT

The hydrophobic biopolymer suberin, which is deposited in the root endodermis and seed coats, functions as an extracellular barrier against uncontrolled water, gas, and ion loss. Suberin monomers synthesized in the endoplasmic reticulum (ER) are exported through the plasma membrane to the apoplast. However, limited information is available about the molecular mechanisms underlying suberin monomer export and assembly. In this study, we investigated the in planta role of LTPG15 encoding a glycosylphosphatidylinositol (GPI)-anchored lipid transfer protein. LTPG15 was predominantly expressed in the root endodermis and seed coat. Fluorescent signals from LTPG15:eYFP were detected in the plasma membrane in tobacco epidermis. Disruption of LTPG15 caused a significant decrease in the levels of fatty acids (C20-C24), primary alcohols (C20 and C22), ω-hydroxy fatty acids (C22 and C24), and α,ω-alkanediols (C20 and C22), but an increase in the amounts of primary alcohols and hydroxy fatty acids with C16 and C18 in seed coats. The mutant phenotype was restored to that of the wild type (WT) by the expression of LTPG15 driven by its own promoter. Seed coats of ltpg15 had an increase in permeability to tetrazolium salts compared with WT seed coats. ltpg15 seeds were more sensitive than WT seeds to inhibition of germination and seedling establishment by salt and osmotic stress treatments. Taken together, our results indicate that LTPG15 is involved in suberin monomer export in seed coats, and this highlights the role of Type G non-specific lipid transfer proteins (LTPGs) in very-long-chain fatty acids and their derivatives' export for suberin polyester formation.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/metabolism , Carrier Proteins/physiology , Fatty Acid-Binding Proteins/physiology , Glycosylphosphatidylinositols/metabolism , Seeds/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cell Membrane/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Permeability , Phylogeny , Plants, Genetically Modified , Transcriptome
10.
Plant Cell Environ ; 42(8): 2411-2421, 2019 08.
Article in English | MEDLINE | ID: mdl-31042812

ABSTRACT

Many plant pathogens gain entry to their host via stomata. On sensing attack, plants close these pores to restrict pathogen entry. Here, we show that plants exhibit a second longer term stomatal response to pathogens. Following infection, the subsequent development of leaves is altered via a systemic signal. This reduces the density of stomata formed, thus providing fewer entry points for pathogens on new leaves. Arabidopsis thaliana leaves produced after infection by a bacterial pathogen that infects through the stomata (Pseudomonas syringae) developed larger epidermal pavement cells and stomata and consequently had up to 20% reductions in stomatal density. The bacterial peptide flg22 or the phytohormone salicylic acid induced similar systemic reductions in stomatal density suggesting that they might mediate this effect. In addition, flagellin receptors, salicylic acid accumulation, and the lipid transfer protein AZI1 were all required for this developmental response. Furthermore, manipulation of stomatal density affected the level of bacterial colonization, and plants with reduced stomatal density showed slower disease progression. We propose that following infection, development of new leaves is altered by a signalling pathway with some commonalities to systemic acquired resistance. This acts to reduce the potential for future infection by providing fewer stomatal openings.


Subject(s)
Arabidopsis/microbiology , Plant Stomata/microbiology , Pseudomonas syringae/physiology , Abscisic Acid/metabolism , Arabidopsis/cytology , Arabidopsis/immunology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/physiology , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/physiology , Host-Pathogen Interactions , Peronospora/physiology , Pipecolic Acids/metabolism , Plant Diseases/immunology , Plant Diseases/microbiology , Plant Leaves/cytology , Plant Leaves/immunology , Plant Leaves/microbiology
11.
Curr Opin Clin Nutr Metab Care ; 22(6): 407-412, 2019 11.
Article in English | MEDLINE | ID: mdl-31503024

ABSTRACT

PURPOSE OF REVIEW: Fatty acid-binding proteins (FABPs) are a family of small, abundant proteins with highly tissue-specific expression patterns whose different functions remain incompletely understood. The purpose of this review is to summarize recent findings regarding FABP functions and mechanisms of action, including their potential utilization as serum markers of tissue-specific metabolic diseases. RECENT FINDINGS: FABPs are important not only in their tissues of origin but also appear to influence the metabolism and function of tissues distal to their sites of expression. This may be secondary to metabolic changes in their primary tissues, and/or a result of FABP secretion from these tissues leading to effects on distal sites. Their levels in the circulation are increasingly explored as potential biomarkers for tissue-specific disease prognosis and progression. SUMMARY: The nine fatty acid-binding members of the FABP family have unique tissue-specific functions and important secondary effects on tissues in which they are not expressed. For many of the FABPs, circulating levels may be indicative of disease processes related to their primary tissues, and may influence physiological function in distal tissues.


Subject(s)
Fatty Acid-Binding Proteins , Animals , Biomarkers/analysis , Biomarkers/metabolism , Fatty Acid-Binding Proteins/analysis , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/physiology , Fatty Acids/metabolism , Humans , Mice , Neoplasms/diagnosis , Neoplasms/metabolism , Obesity/diagnosis , Obesity/metabolism , Organ Specificity
12.
J Neurochem ; 144(1): 81-92, 2018 01.
Article in English | MEDLINE | ID: mdl-29105065

ABSTRACT

Lower levels of the cognitively beneficial docosahexaenoic acid (DHA) are often observed in Alzheimer's disease (AD) brains. Brain DHA levels are regulated by the blood-brain barrier (BBB) transport of plasma-derived DHA, a process facilitated by fatty acid-binding protein 5 (FABP5). This study reports a 42.1 ± 12.6% decrease in the BBB transport of 14 C-DHA in 8-month-old AD transgenic mice (APPswe,PSEN1∆E9) relative to wild-type mice, associated with a 34.5 ± 6.7% reduction in FABP5 expression in isolated brain capillaries of AD mice. Furthermore, short-term spatial and recognition memory deficits were observed in AD mice on a 6-month n-3 fatty acid-depleted diet, but not in AD mice on control diet. This intervention led to a dramatic reduction (41.5 ± 11.9%) of brain DHA levels in AD mice. This study demonstrates FABP5 deficiency and impaired DHA transport at the BBB are associated with increased vulnerability to cognitive deficits in mice fed an n-3 fatty acid-depleted diet, in line with our previous studies demonstrating a crucial role of FABP5 in BBB transport of DHA and cognitive function.


Subject(s)
Blood-Brain Barrier , Cognition Disorders/etiology , Docosahexaenoic Acids/pharmacokinetics , Fatty Acid-Binding Proteins/physiology , Neoplasm Proteins/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain Chemistry , Cognition Disorders/genetics , Cognition Disorders/metabolism , Dietary Fats/administration & dosage , Docosahexaenoic Acids/deficiency , Escherichia coli Proteins , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acids, Omega-3/deficiency , Female , Humans , Male , Maze Learning , Memory Disorders/etiology , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Neoplasm Proteins/biosynthesis , Polysaccharide-Lyases , Presenilin-1/genetics , Presenilin-1/metabolism , Recognition, Psychology , Recombinant Fusion Proteins/metabolism
13.
Biochim Biophys Acta ; 1861(8 Pt B): 913-923, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26825693

ABSTRACT

The tubular lipid-binding (TULIP) superfamily has emerged in recent years as a major mediator of lipid sensing and transport in eukaryotes. It currently encompasses three protein families, SMP-like, BPI-like, and Takeout-like, which share a common fold. This fold consists of a long helix wrapped in a highly curved anti-parallel ß-sheet, enclosing a central, lipophilic cavity. The SMP-like proteins, which include subunits of the ERMES complex and the extended synaptotagmins (E-Syts), appear to be mainly located at membrane contacts sites (MCSs) between organelles, mediating inter-organelle lipid exchange. The BPI-like proteins, which include the bactericidal/permeability-increasing protein (BPI), the LPS (lipopolysaccharide)-binding protein (LBP), the cholesteryl ester transfer protein (CETP), and the phospholipid transfer protein (PLTP), are either involved in innate immunity against bacteria through their ability to sense lipopolysaccharides, as is the case for BPI and LBP, or in lipid exchange between lipoprotein particles, as is the case for CETP and PLTP. The Takeout-like proteins, which are comprised of insect juvenile hormone-binding proteins and arthropod allergens, transport, where known, lipid hormones to target tissues during insect development. In all cases, the activity of these proteins is underpinned by their ability to bind large, hydrophobic ligands in their central cavity and segregate them away from the aqueous environment. Furthermore, where they are involved in lipid exchange, recent structural studies have highlighted their ability to establish lipophilic, tubular channels, either between organelles in the case of SMP domains or between lipoprotein particles in the case of CETP. Here, we review the current knowledge on the structure, versatile functions, and evolution of the TULIP superfamily. We propose a deep evolutionary split in this superfamily, predating the Last Eukaryotic Common Ancestor, between the SMP-like proteins, which act on lipids endogenous to the cell, and the BPI-like proteins (including the Takeout-like proteins of arthropods), which act on exogenous lipids. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.


Subject(s)
Eukaryotic Cells/metabolism , Fatty Acid-Binding Proteins/physiology , Lipid Metabolism , Acute-Phase Proteins/chemistry , Acute-Phase Proteins/physiology , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/physiology , Biological Transport/genetics , Blood Proteins/chemistry , Blood Proteins/physiology , Carrier Proteins/chemistry , Carrier Proteins/physiology , Cholesterol Ester Transfer Proteins/chemistry , Cholesterol Ester Transfer Proteins/physiology , Fatty Acid-Binding Proteins/chemistry , Humans , Lipid Metabolism/genetics , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/physiology , Models, Molecular , Multigene Family/physiology , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/physiology , Phylogeny
14.
Subcell Biochem ; 81: 179-199, 2016.
Article in English | MEDLINE | ID: mdl-27830505

ABSTRACT

It has long been established that the transcriptional activity of retinoic acid (RA) is mediated by members of the nuclear receptor family of ligand-activated transcription factors termed RA receptors (RARs). More recent observations have established that RA also activates an additional nuclear receptor, PPARß/δ. Partitioning RA between RARs and PPARß/δ is governed by different intracellular lipid-binding proteins: cellular RA binding protein 2 (CRABP2) delivers RA to nuclear RARs and a fatty acid binding protein (FABP5) delivers the hormone from the cytosol to nuclear PPARß/δ. Consequently, RA signals through RARs in CRABP2-expressing cells, but activates PPARß/δ in cells that express a high level of FABP5. RA elicits different and sometimes opposing responses in cells that express different FABP5/CRABP2 ratios because PPARß/δ and RARs regulate the expression of distinct sets of genes. An overview of the observations that led to the discovery of this non-classical activity of RA are presented here, along with a discussion of evidence demonstrating the involvement of the dual transcriptional activities of RA in regulating energy homeostasis, insulin responses, and adipocyte and neuron differentiation.


Subject(s)
Gene Expression Regulation/drug effects , PPAR delta/physiology , PPAR-beta/physiology , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Adipogenesis/drug effects , Adipogenesis/genetics , Adipose Tissue/metabolism , Animals , Biological Transport , Fatty Acid-Binding Proteins/physiology , Forecasting , Gene Expression Regulation/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Models, Molecular , Neoplasm Proteins/physiology , Neurogenesis/drug effects , Neurogenesis/genetics , Obesity/metabolism , PPAR delta/drug effects , PPAR-beta/drug effects , Protein Conformation , Receptors, Retinoic Acid/physiology
15.
J Biol Chem ; 290(14): 8711-21, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25666611

ABSTRACT

Δ(9)-Tetrahydrocannabinol (THC) and cannabidiol (CBD) occur naturally in marijuana (Cannabis) and may be formulated, individually or in combination in pharmaceuticals such as Marinol or Sativex. Although it is known that these hydrophobic compounds can be transported in blood by albumin or lipoproteins, the intracellular carrier has not been identified. Recent reports suggest that CBD and THC elevate the levels of the endocannabinoid anandamide (AEA) when administered to humans, suggesting that phytocannabinoids target cellular proteins involved in endocannabinoid clearance. Fatty acid-binding proteins (FABPs) are intracellular proteins that mediate AEA transport to its catabolic enzyme fatty acid amide hydrolase (FAAH). By computational analysis and ligand displacement assays, we show that at least three human FABPs bind THC and CBD and demonstrate that THC and CBD inhibit the cellular uptake and catabolism of AEA by targeting FABPs. Furthermore, we show that in contrast to rodent FAAH, CBD does not inhibit the enzymatic actions of human FAAH, and thus FAAH inhibition cannot account for the observed increase in circulating AEA in humans following CBD consumption. Using computational molecular docking and site-directed mutagenesis we identify key residues within the active site of FAAH that confer the species-specific sensitivity to inhibition by CBD. Competition for FABPs may in part or wholly explain the increased circulating levels of endocannabinoids reported after consumption of cannabinoids. These data shed light on the mechanism of action of CBD in modulating the endocannabinoid tone in vivo and may explain, in part, its reported efficacy toward epilepsy and other neurological disorders.


Subject(s)
Cannabidiol/metabolism , Carrier Proteins/physiology , Dronabinol/metabolism , Fatty Acid-Binding Proteins/physiology , Amino Acid Sequence , Animals , Cannabidiol/chemistry , Carrier Proteins/chemistry , Dronabinol/chemistry , Fatty Acid-Binding Proteins/chemistry , HeLa Cells , Humans , Mice , Molecular Docking Simulation , Molecular Sequence Data , Rats , Sequence Homology, Amino Acid , Signal Transduction
16.
Am J Physiol Endocrinol Metab ; 308(7): E583-91, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25628425

ABSTRACT

Gastric inhibitory polypeptide (GIP) is an incretin released from enteroendocrine K cells in response to nutrient intake, especially fat. GIP is one of the contributing factors inducing fat accumulation that results in obesity. A recent study shows that fatty acid-binding protein 5 (FABP5) is expressed in murine K cells and is involved in fat-induced GIP secretion. We investigated the mechanism of fat-induced GIP secretion and the impact of FABP5-related GIP response on diet-induced obesity (DIO). Single oral administration of glucose and fat resulted in a 40% reduction of GIP response to fat but not to glucose in whole body FABP5-knockout (FABP5(-/-)) mice, with no change in K cell count or GIP content in K cells. In an ex vivo experiment using isolated upper small intestine, oleic acid induced only a slight increase in GIP release, which was markedly enhanced by coadministration of bile and oleic acid together with attenuated GIP response in the FABP5(-/-) sample. FABP5(-/-) mice exhibited a 24% reduction in body weight gain and body fat mass under a high-fat diet compared with wild-type (FABP5(+/+)) mice; the difference was not observed between GIP-GFP homozygous knock-in (GIP(gfp/gfp))-FABP5(+/+) mice and GIP(gfp/gfp)-FABP5(-/-) mice, in which GIP is genetically deleted. These results demonstrate that bile efficiently amplifies fat-induced GIP secretion and that FABP5 contributes to the development of DIO in a GIP-dependent manner.


Subject(s)
Diet, High-Fat , Dietary Fats/pharmacology , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism , Fatty Acid-Binding Proteins/physiology , Gastric Inhibitory Polypeptide/metabolism , Neoplasm Proteins/physiology , Obesity/genetics , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Eating , Glucose/pharmacology , Mice , Mice, Transgenic , Obesity/metabolism
17.
J Neurochem ; 132(1): 85-98, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25147052

ABSTRACT

Epidermal fatty acid-binding protein (E-FABP/FABP5/DA11) binds and transport long-chain fatty acids in the cytoplasm and may play a protecting role during neuronal injury. We examined whether E-FABP protects nerve growth factor-differentiated PC12 cells (NGFDPC12 cells) from lipotoxic injury observed after palmitic acid (C16:0; PAM) overload. NGFDPC12 cells cultures treated with PAM/bovine serum albumin at 0.3 mM/0.15 mM show PAM-induced lipotoxicity (PAM-LTx) and apoptosis. The apoptosis was preceded by a cellular accumulation of reactive oxygen species (ROS) and higher levels of E-FABP. Antioxidants MCI-186 and N-acetyl cysteine prevented E-FABP's induction in expression by PAM-LTx, while tert-butyl hydroperoxide increased ROS and E-FABP expression. Non-metabolized methyl ester of PAM, methyl palmitic acid (mPAM), failed to increase cellular ROS, E-FABP gene expression, or trigger apoptosis. Treatment of NGFDPC12 cultures with siE-FABP showed reduced E-FABP levels correlating with higher accumulation of ROS and cell death after exposure to PAM. In contrast, increasing E-FABP cellular levels by pre-loading the cells with recombinant E-FABP diminished the PAM-induced ROS and cell death. Finally, agonists for PPARß (GW0742) or PPARγ (GW1929) increased E-FABP expression and enhanced the resistance of NGFDPC12 cells to PAM-LTx. We conclude that E-FABP protects NGFDPC12 cells from lipotoxic injury through mechanisms that involve reduction of ROS. Epidermal fatty acid-binding protein (E-FABP) may protect nerve cells from the damaging exposure to high levels of free fatty acids (FA). We show that E-FABP can neutralize the effects of reactive oxygen species (ROS) generated by the high levels of FA in the cell and protect PC12 cells from lipotoxic injuries common in Type 2 diabetes neuropathy. Potentially, E-FABP gene up-regulation may be mediated through the NFkB pathway and future studies are needed to further evaluate this proposition.


Subject(s)
Eye Proteins/physiology , Fatty Acid-Binding Proteins/physiology , Lipids/antagonists & inhibitors , Lipids/toxicity , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/physiology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Eye Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Nerve Tissue Proteins/genetics , PC12 Cells , Palmitic Acid/antagonists & inhibitors , Palmitic Acid/toxicity , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology , Transfection
18.
Clin Sci (Lond) ; 129(7): 547-59, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26186740

ABSTRACT

Clinical evidence shows that circulating levels of adipocyte fatty-acid-binding protein (A-FABP) are elevated in patients with diabetes and closely associated with ischaemic heart disease. Patients with diabetes are more susceptible to myocardial ischaemia/reperfusion (MI/R) injury. The experiments in the present study investigated the role of A-FABP in MI/R injury with or without diabetes. Non-diabetic and diabetic (streptozotocin-induced) A-FABP knockout and wild-type mice were subjected to MI/R or sham intervention. After MI/R, A-FABP knockout mice exhibited reductions in myocardial infarct size, apoptotic index, oxidative and nitrative stress, and inflammation. These reductions were accompanied by an improved left ventricular function compared with the relative controls under non-diabetic or diabetic conditions. After diabetes induction, A-FABP knockout mice exhibited a preserved cardiac function compared with that in wild-type mice. Endothelial cells, but not cardiomyocytes, were identified as the most likely source of cardiac A-FABP. Cardiac and circulating A-FABP levels were significantly increased in mice with diabetes or MI/R. Diabetes-induced superoxide anion production was significantly elevated in wild-type mice, but diminished in A-FABP knockout mice, and this elevation contributed to the exaggeration of MI/R-induced cardiac injury. Phosphorylation of endothelial nitric oxide synthase (eNOS) and production of nitric oxide (NO) were enhanced in both diabetic and non-diabetic A-FABP knockout mice after MI/R injury, but diminished in wild-type mice. The beneficial effects of A-FABP deficiency on MI/R injury were abolished by the NOS inhibitor N(G)-nitro-L-arginine methyl ester. Thus, A-FABP deficiency protects mice against MI/R-induced and/or diabetes-induced cardiac injury at least partially through activation of the eNOS/NO pathway and reduction in superoxide anion production.


Subject(s)
Diabetes Mellitus/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/physiology , Myocardial Ischemia/therapy , Myocardium/pathology , Animals , Anions , Apoptosis , Blood Pressure , Endothelium, Vascular/metabolism , Immunohistochemistry , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Myocardial Reperfusion Injury/physiopathology , Oxidative Stress , Reperfusion Injury , Superoxides/metabolism
19.
Cell Biol Int ; 39(5): 540-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25572488

ABSTRACT

The aims of this study were to delineate the expression of fatty-acid binding protein (FABP) 4 in human uterine endometrium and its function in the regulation of proliferation, migration and invasion of epithelial cells. Immunohistochenistry, immunofluorence and Western blotting were used to determine the expression and cellular localization of FABP4 in endometrium and endometrial epithelial cell lines. Interference of small ribonuclear acid (siRNA) and specific FABP4 inhibitor were used to inhibit FABP4. The proliferation, migration and invasion of epithelial cells were evaluated with CCK-8 assay, wound-healing test and transwell analysis respectively. We found that FABP4 was expressed by epithelial cells of proliferative endometrium and epithelial and stromal cells of secrectory endometrium. Epithelial cell lines Ishikawa and RL-952 expressed FABP4 and this expression was decreased by FABP4 siRNA. FABP4 siRNA and specific FABP4 inhibition significantly decreased the proliferation, migration and invasion of epithelial cell lines. We concluded that FABP4 is functionally expressed in endometrial epithelium and is necessary for maintaining the cell function of epithelial cells of endometrium.


Subject(s)
Endometrium/metabolism , Epithelial Cells/metabolism , Fatty Acid-Binding Proteins/physiology , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Endometrium/drug effects , Epithelial Cells/drug effects , Fatty Acid-Binding Proteins/antagonists & inhibitors , Fatty Acid-Binding Proteins/genetics , Female , Humans , RNA, Small Interfering/pharmacology
20.
Yi Chuan ; 37(2): 174-182, 2015 Feb.
Article in Zh | MEDLINE | ID: mdl-25665644

ABSTRACT

FABP4 (Fatty acid binding protein 4) is a hot candidate gene in fat deposition and lipid metabolism and participates in the transport and metabolism of intracellular free fatty acids. We aim to study the role of FABP4 in fat deposition and metabolism of the rump fat in Altay sheep. In this study, bioinformatics method was used to analyze the protein sequence homology among 10 species, and RT-PCR was employed to detect FABP4 tissue profiling of Altay sheep. An animal model simulating the rump fat deposition and metabolism of Altay sheep was established by continuous starvation, and qPCR and iTRAQ (isobaric tags for relative and absolute quantitation) were used to detecte FABP4 mRNA and protein expression changes in the control and continuous starvation groups, respectively. Sequence analysis showed that FABP4 protein sequence is highly conserved among species, suggesting an important biological function during evolution for FABP4. The RT-PCR result confirmed that FABP4 mRNA was highly expressed in intestinal and rump fat, suggesting that FABP4 plays an important physiological role in fat tissues. We did not find significant differences in FABP4 mRNA and protein between control and continuous starvation groups (P>0.05), which indicates that FABP4 may not be the key gene in fat deposition and metabolism in Altay sheep.The results above lay a foundation for further studies of FABP4 in rump or tail fat.


Subject(s)
Fatty Acid-Binding Proteins/genetics , Lipid Metabolism , Animals , Fatty Acid-Binding Proteins/physiology , Female , Phylogeny , RNA, Messenger/analysis , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL