Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731803

ABSTRACT

This study explores the effects of normobaric hypoxia and intermittent hypoxic training (IHT) on the physiological condition of the cardiac muscle in swimmers. Hypoxia has been reported to elicit both beneficial and adverse changes in the cardiovascular system, but its impact on the myocardium during acute exercise and altitude/hypoxic training remains less understood. We aimed to determine how a single bout of intense interval exercise and a four-week period of high-intensity endurance training under normobaric hypoxia affect cardiac marker activity in swimmers. Sixteen young male swimmers were divided into two groups: one undergoing training in hypoxia and the other in normoxia. Cardiac markers, including troponin I and T (cTnI and cTnT), heart-type fatty acid-binding protein (H-FABP), creatine kinase-MB isoenzyme (CK-MB), and myoglobin (Mb), were analyzed to assess the myocardium's response. We found no significant differences in the physiological response of the cardiac muscle to intense physical exertion between hypoxia and normoxia. Four weeks of IHT did not alter the resting levels of cTnT, cTnI, and H-FABP, but it resulted in a noteworthy decrease in the resting concentration of CK-MB, suggesting enhanced cardiac muscle adaptation to exercise. In contrast, a reduction in resting Mb levels was observed in the control group training in normoxia. These findings suggest that IHT at moderate altitudes does not adversely affect cardiac muscle condition and may support cardiac muscle adaptation, affirming the safety and efficacy of IHT as a training method for athletes.


Subject(s)
Athletes , Biomarkers , Hypoxia , Humans , Male , Hypoxia/metabolism , Pilot Projects , Swimming/physiology , Young Adult , Myocardium/metabolism , Myoglobin/metabolism , Troponin I/metabolism , Fatty Acid Binding Protein 3/metabolism , Adolescent , Fatty Acid-Binding Proteins/metabolism , Physical Endurance/physiology , Creatine Kinase, MB Form/blood , Creatine Kinase, MB Form/metabolism , Adaptation, Physiological , Altitude
2.
J Pharmacol Sci ; 152(1): 30-38, 2023 May.
Article in English | MEDLINE | ID: mdl-37059489

ABSTRACT

Parkinson's disease (PD) is characterized by dopaminergic (DAergic) neuronal loss in the substantia nigra pars compacta (SNpc), resulting from α-synuclein (αSyn) toxicity. We previously reported that αSyn oligomerization and toxicity are regulated by the fatty-acid binding protein 3 (FABP3), and the therapeutic effects of the FABP3 ligand, MF1, was successfully demonstrated in PD models. Here, we developed a novel and potent ligand, HY-11-9, which has a higher affinity for FABP3 (Kd = 11.7 ± 8.8) than MF1 (Kd = 302.8 ± 130.3). We also investigated whether the FABP3 ligand can ameliorate neuropathological deterioration after the onset of disease in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism. Motor deficits were observed two weeks after MPTP treatment. Notably, oral administration of HY-11-9 (0.03 mg/kg) improved motor deficits in both beam-walking and rotarod tasks, whereas MF1 failed to improve the motor deficits in both tasks. Consistent with the behavioral tasks, HY-11-9 recovered dopamine neurons from MPTP toxicity in the substantia nigra and ventral tegmental areas. Furthermore, HY-11-9 reduced the accumulation of phosphorylated-serine129-α-synuclein (pS129-αSyn) and colocalization with FABP3 in tyrosine hydroxylase (TH)-positive DA neurons in the PD mouse model. Overall, HY-11-9 significantly improved MPTP-induced behavioral and neuropathological deterioration, suggesting that it may be a potential candidate for PD therapy.


Subject(s)
MPTP Poisoning , Parkinson Disease , Parkinsonian Disorders , Mice , Animals , alpha-Synuclein/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Ligands , Parkinsonian Disorders/drug therapy , Parkinson Disease/drug therapy , Substantia Nigra/metabolism , Substantia Nigra/pathology , Dopaminergic Neurons/metabolism , Mice, Inbred C57BL , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Disease Models, Animal , Fatty Acid Binding Protein 3/metabolism
3.
Int J Mol Sci ; 24(7)2023 Apr 02.
Article in English | MEDLINE | ID: mdl-37047614

ABSTRACT

We previously demonstrated that fatty acid-binding protein 3 null (FABP3-/-) mice exhibit resistance to nicotine-induced conditioned place preference (CPP). Here, we confirm that the FABP3 inhibitor, MF1 ((4-(2-(1-(2-chlorophenyl)-5-phenyl-1H-pyrazol-3-yl)phenoxy) butanoic acid), successfully reduces nicotine-induced CPP scores in mice. MF1 (0.3 or 1.0 mg/kg) was orally administered 30 min before nicotine, and CPP scores were assessed in the conditioning, withdrawal, and relapse phases. MF1 treatment decreased CPP scores in a dose-dependent manner. Failure of CPP induction by MF1 (1.0 mg/kg, p.o.) was associated with the inhibition of both CaMKII and ERK activation in the nucleus accumbens (NAc) and hippocampal CA1 regions. MF1 treatment reduced nicotine-induced increases in phosphorylated CaMKII and cAMP-response element-binding protein (CREB)-positive cells. Importantly, the increase in dopamine D2 receptor (D2R) levels following chronic nicotine exposure was inhibited by MF1 treatment. Moreover, the quinpirole (QNP)-induced increase in the level of CaMKII and ERK phosphorylation was significantly inhibited by MF1 treatment of cultured NAc slices from wild type (WT) mice; however, QNP treatment had no effect on CaMKII and ERK phosphorylation levels in the NAc of D2R null mice. Taken together, these results show that MF1 treatment suppressed D2R/FABP3 signaling, thereby preventing nicotine-induced CPP induction. Hence, MF1 can be used as a novel drug to block addiction to nicotine and other drugs by inhibiting the dopaminergic system.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Nicotine , Mice , Animals , Nicotine/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Nucleus Accumbens/metabolism , Signal Transduction , Mice, Knockout , Fatty Acid Binding Protein 3/metabolism
4.
Int J Mol Sci ; 24(6)2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36982599

ABSTRACT

Heart-type fatty-acid binding protein (FABP3) is an essential cytosolic lipid transport protein found in cardiomyocytes. FABP3 binds fatty acids (FAs) reversibly and with high affinity. Acylcarnitines (ACs) are an esterified form of FAs that play an important role in cellular energy metabolism. However, an increased concentration of ACs can exert detrimental effects on cardiac mitochondria and lead to severe cardiac damage. In the present study, we evaluated the ability of FABP3 to bind long-chain ACs (LCACs) and protect cells from their harmful effects. We characterized the novel binding mechanism between FABP3 and LCACs by a cytotoxicity assay, nuclear magnetic resonance, and isothermal titration calorimetry. Our data demonstrate that FABP3 is capable of binding both FAs and LCACs as well as decreasing the cytotoxicity of LCACs. Our findings reveal that LCACs and FAs compete for the binding site of FABP3. Thus, the protective mechanism of FABP3 is found to be concentration dependent.


Subject(s)
Fatty Acid-Binding Proteins , Fatty Acids , Fatty Acid Binding Protein 3/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Acids/pharmacology , Carnitine , Myocytes, Cardiac/metabolism
5.
Cytokine ; 162: 156090, 2023 02.
Article in English | MEDLINE | ID: mdl-36481477

ABSTRACT

BACKGROUND: Rotator cuff Tear (RCT) causes a lot of inconvenience for patients. In most cases, RCT injury does not heal back to bone after repair, and there is a high chance of retearing. Therefore, there is a need to explore more effective targeted therapies. Bone mesenchymal stem cell-derived exosome (BMSCs-Exo) has been proved to be beneficial to the proliferation of tendon cells, but its specific mechanism remains to be further explored. METHODS: BMSCs-Exo was isolated and identified by detecting the specific markers using flow cytometry and western blot assays. qRT-PCR and western blot were utilized to determine the gene or protein expressions, respectively. Cell proliferation, and migration in tenocytes were measured by CCK8, EdU and transwell assays. The interaction between miR-29a and FABP3 was analyzed using dual-luciferase reporter assay. RESULTS: Our findings demonstrated that miR-29a was expressed in BMSCs-Exo and could be significantly enriched after TGF-ß1 treatment. Moreover, TGF-ß1-modified BMSCs-Exo co-cultured could promote the proliferation, migration and fibrosis of tenocytes by carrying miR-29a. Upon miR-29a was reduced in BMSCs-Exo, the regulatory roles of BMSCs-Exo on tenocytes were reversed. Mechanistically, miR-29a negatively regulated FABP3 via interaction with its 3'-UTR. Enforced expression of FABP3 could reverse the modulation of exosomal miR-29a in tenocytes. CONCLUSION: Exosomal miR-29a derived from TGF-ß1-modified BMSCs facilitated the proliferation, migration and fibrosis of tenocytes through targeting FABP3.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Humans , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Tenocytes/metabolism , Mesenchymal Stem Cells/metabolism , Cell Proliferation/genetics , Fatty Acid Binding Protein 3/metabolism
6.
Drug Chem Toxicol ; 46(3): 597-608, 2023 May.
Article in English | MEDLINE | ID: mdl-35509154

ABSTRACT

Diclofenac is a widely prescribed anti-inflammatory drug having cardiovascular complications as one of the main liabilities that restrict its therapeutic use. We aimed to investigate for any role of rutin against diclofenac-induced cardiac injury with underlying mechanisms as there is no such precedent to date. The effect of rutin (10 and 20 mg/kg) was evaluated upon concomitant oral administration for fifteen days with diclofenac (10 mg/kg). Rutin significantly attenuated diclofenac-induced alterations in the serum cardiac markers (LDH, CK-MB, and SGOT), serum cytokine levels (TNF-α and IL-6), and oxidative stress markers (MDA and GSH) in the cardiac tissue. Histopathological examination and Scanning Electron Microscopy (SEM) findings displayed a marked effect of rutin to prevent diclofenac-mediated cardiac injury. Altered protein expression of myocardial injury markers (cTnT, FABP3, and ANP) and apoptotic markers (Bcl-2 and Caspase-3) in the cardiac tissue upon diclofenac treatment was considerably shielded by rutin treatment. MYL3 was unaffected due to diclofenac or rutin treatment. Rutin also significantly improved diclofenac-induced gastrointestinal and hepatic alterations based on the observed ameliorative effects in key mediators, oxidative stress markers, histopathology examination, and SEM findings. Overall results suggest that rutin can protect the diclofenac-induced cardiac injury by lowering oxidative stress, inhibiting inflammation, and reducing apoptosis. Further research work directs toward the development of phytotherapeutics for cardioprotection.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Antioxidants , Diclofenac , Inflammation , Rutin , Animals , Rats , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Diclofenac/pharmacology , Diclofenac/toxicity , Fatty Acid Binding Protein 3/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/prevention & control , Myosin Light Chains/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rutin/metabolism , Rutin/pharmacology , Rutin/therapeutic use
7.
Anim Biotechnol ; 34(6): 1960-1967, 2023 Nov.
Article in English | MEDLINE | ID: mdl-35416753

ABSTRACT

Fatty acid binding protein 3 (FABP3) is involved in signal transduction pathways, and in the uptake and utilization of long-chain fatty acids. However, the transcriptional regulation of FABP3 in goat is unclear. In this study, the FABP3 5' flanking region was amplified from goat (Capra hircus) genomic DNA. Luciferase reporter vectors containing promoter fragments of five different lengths were constructed and transfected into dairy goat mammary epithelial cells. The region of the promoter located between -1801 and -166 bp upstream of the transcription start site (TSS) exhibited the highest luciferase activity, and contained two cAMP response elements (CREs) located at -1632 bp and -189 bp. Interference with CREB1 significantly downregulated FABP3 promoter activity. In addition, FABP3 promoter activity was significantly reduced after mutation of the CRE1 (-1632 bp) and CRE2 (-189 bp) sites. Further analysis indicated that the CRE2 site was essential for the transcriptional activity induced by CREB1. These results demonstrated that CREB1 is involved in the transcriptional regulation of FABP3 expression in the goat mammary gland via a direct mechanism, thus revealing a novel signaling pathway involved in fatty acid metabolism in goat.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Goats , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Fatty Acid Binding Protein 3/genetics , Fatty Acid Binding Protein 3/metabolism , Goats/genetics , Goats/metabolism , Promoter Regions, Genetic/genetics , Epithelial Cells/metabolism
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 42(9): 1359-1366, 2022 Sep 20.
Article in Chinese | MEDLINE | ID: mdl-36210709

ABSTRACT

OBJECTIVE: To investigate the changes in myocardial calcium currents in rats subjected to forced running exercise during acute hypoxia and their association with myocardial injury. METHODS: Forty SD rats were randomized into quiescent group and running group either in normal oxygen (NQ and NR groups, respectively) or in acute hypoxia (HQ and HR groups, respectively). Hypoxia was induced by keeping the rats in a hypobaric oxygen chamber (PaO2=61.6kpa) for 4 h a day; the rats in the two running groups were forced to run on running wheels for 4 h each day. Rat ventricular myocytes was isolated by enzymatic digestion for recording action potentials and currents using patch clamp technique, and confocal Ca2+ imaging was used to monitor intracellular Ca2+ levels. The expressions of Cav1.2 channel and the cardiac ryanodine receptor (RyR2) were determined using Western blotting. RESULTS: Compared with those in NQ group, the rats in HR group showed significantly decreased SOD activity (P < 0.01), increased h-FABP, hs-CRP and IMA levels (P < 0.05 or 0.01), obvious myocardial pathology, and prolonged APD50 and APD90 (P < 0.05). Of the different stress conditions, forced running in acute hypoxia resulted in the most prominent increase of the densities of ICa, L currents, causing also a significant left shift of the steady state activation curve and a significant right shift of the steady state inactivation curve. Compared with those in NQ group, the rats in NR, HQ and HR groups all exhibited higher rates of spontaneous calcium wave events in the cardiac myocytes, increased frequency of calcium sparks with lowered amplitude, enhanced calcium release amplitude in the ventricular myocytes, and delayed calcium ion reabsorption; in particular, these changes were the most conspicuous in HR group (P < 0.05 or 0.01). There was also a significant increase in the protein levels of Cav1.2 channel and RyR2 receptor in HR group (P < 0.05 or 0.01). CONCLUSIONS: The mechanism of myocardial injury in rats subjected to forced running in acute hypoxia may involve the increase of oxidative stress and calcium current and intracellular calcium overload.


Subject(s)
Calcium , Heart Injuries , Animals , C-Reactive Protein/metabolism , Calcium/metabolism , Calcium Signaling , Fatty Acid Binding Protein 3/metabolism , Heart Injuries/metabolism , Hypoxia/metabolism , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/metabolism , Superoxide Dismutase/metabolism
9.
J Exp Biol ; 225(19)2022 10 01.
Article in English | MEDLINE | ID: mdl-36200468

ABSTRACT

Migratory birds undergo seasonal changes to muscle biochemistry. Nonetheless, it is unclear to what extent these changes are attributable to the exercise of flight itself versus endogenous changes. Using starlings (Sturnus vulgaris) flying in a wind tunnel, we tested the effects of exercise training, a single bout of flight and dietary lipid composition on pectoralis muscle oxidative enzymes and lipid transporters. Starlings were either unexercised or trained over 2 weeks to fly in a wind tunnel and sampled either immediately following a long flight at the end of this training or after 2 days recovery from this flight. Additionally, they were divided into dietary groups that differed in dietary fatty acid composition (high polyunsaturates versus high monounsaturates) and amount of dietary antioxidant. Trained starlings had elevated (19%) carnitine palmitoyl transferase and elevated (11%) hydroxyacyl-CoA dehydrogenase in pectoralis muscle compared with unexercised controls, but training alone had little effect on lipid transporters. Immediately following a long wind-tunnel flight, starling pectoralis had upregulated lipid transporter mRNA (heart-type fatty acid binding protein, H-FABP, 4.7-fold; fatty acid translocase, 1.9-fold; plasma membrane fatty acid binding protein, 1.6-fold), and upregulated H-FABP protein (68%). Dietary fatty acid composition and the amount of dietary antioxidants had no effect on muscle catabolic enzymes or lipid transporter expression. Our results demonstrate that birds undergo rapid upregulation of catabolic capacity that largely becomes available during flight itself, with minor effects due to training. These effects likely combine with endogenous seasonal changes to create the migratory phenotype observed in the wild.


Subject(s)
Starlings , Animal Migration/physiology , Animals , Antioxidants/metabolism , Carnitine/metabolism , Coenzyme A/metabolism , Fatty Acid Binding Protein 3/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Muscle, Skeletal/metabolism , Oxidative Stress , Oxidoreductases/metabolism , Pectoralis Muscles/metabolism , RNA, Messenger/genetics , Starlings/physiology , Transferases/metabolism
10.
Adv Med Sci ; 67(2): 283-290, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35843155

ABSTRACT

PURPOSE: Cryoablation is a recommended, modern and well-tolerated method of treating atrial fibrillation (AF). The study evaluates plasma biomarkers related to AF and the effectiveness of its treatment - cryoablation. Heart- and adipocyte-type fatty acid binding proteins (H-FABP and A-FABP, respectively) as well as fatty acids (FAs) were assessed in patients that underwent cryoballoon ablation (CBA) for AF. PATIENTS AND METHODS: Concentrations of plasma FABPs and FAs were measured in 33 AF patients on admission and 24 â€‹h after CBA (enzyme-linked immunoassay and gas liquid chromatography, respectively). The control group consisted of 20 volunteers. RESULTS: We showed that plasma H-FABP and A-FABP concentrations were significantly higher in the patients with AF than in the control group (1135 â€‹pg/mL vs 836 â€‹pg/mL, and 34.29 â€‹ng/mL vs 15.14 â€‹ng/mL, respectively; p â€‹< â€‹0.05). After CBA, H-FABP plasma concentration increased even further (1574 â€‹pg/mL vs 1135 â€‹pg/mL; p â€‹< â€‹0.05) and FAs levels decreased concomitantly. AF recurred in 8 patients (24.25%) after 3 months and in 13 patients (39.4%) after 6 months. Initially higher concentration of oleic acid (680.24 â€‹± â€‹189.768 vs 567.04 â€‹± â€‹70.002; p â€‹< â€‹0.05) correlated substantially with lower AF relapse rate in 6 months follow-up. CONCLUSIONS: The patients with AF showed increased concentration of H-FABP, whereas CBA triggered further elevation of H-FABP with a simultaneous decline in the total plasma FAs concentration. H-FABP and A-FABP could not be confirmed as new biomarkers of cryoablation efficiency, but this requires further investigation due to the limitations of the study.


Subject(s)
Atrial Fibrillation , Cryosurgery , Humans , Fatty Acid Binding Protein 3/metabolism , Fatty Acids/metabolism , Myocardium/metabolism , Fatty Acid-Binding Proteins/metabolism , Biomarkers , Oleic Acids/metabolism
11.
J Pharmacol Sci ; 148(2): 248-254, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35063140

ABSTRACT

An aging society leads to an increased number of patients with cognitive and movement disorders, such as Parkinson's disease and dementia with Lewy bodies. α-Synuclein accumulation in neuronal cells is a pathological hallmark of α-synucleinopathies. Aberrant soluble oligomeric units of α-synuclein are toxic and disrupt neuronal homeostasis. Fatty acids partially regulate α-synuclein accumulation as well as oligomerization, and fatty acid-binding protein (FABP) associates with the α-synuclein aggregates. Heart-type FABP (hFABP, FABP3) is rich in dopaminergic neurons and interacts with dopamine D2 receptors, specifically the long type (D2L), which is abundant in caveolae. We recently demonstrated that mesencephalic neurons require FABP3 and dopamine D2L receptors for the caveolae-mediated α-synuclein uptake. Accumulated α-synuclein gets fibrillized and tightly co-localizes with FABP3 and dopamine D2L receptors, which leads to mitochondrial dysfunction and loss of tyrosine hydroxylase, a rate-limiting enzyme in dopamine production. Furthermore, the inhibition of FABP3 using small-molecule ligands successfully prevents FABP3-induced neurotoxicity. In this review, we focus on the impact of FABP3, dopamine receptors, and other FABP family proteins in the process of α-synuclein propagation and the subsequent aggregate-induced cytotoxicity. We also propose the potential of FABP as a therapeutic target for α-synucleinopathies.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Receptors, Dopamine/metabolism , Synucleinopathies/etiology , Synucleinopathies/metabolism , alpha-Synuclein/metabolism , Dopamine/metabolism , Fatty Acid Binding Protein 3/metabolism , Humans , Mitochondria/metabolism , Molecular Targeted Therapy , Protein Aggregates , Synucleinopathies/therapy , alpha-Synuclein/toxicity
12.
J Oleo Sci ; 70(12): 1805-1814, 2021.
Article in English | MEDLINE | ID: mdl-34866110

ABSTRACT

Sterculia tragacantha (ST) Lindl leaf is commonly used locally in the management of diabetes mellitus (DM) and its complications. This study was aimed at assessing the valuable effects of ST leaf on streptozotocin-diabetic cardiomyopathy (DCM). Streptozotocin was administered intraperitoneally to the experimental animals to induce DM, and hence, placed on different doses of ST for 14 days. Thereafter, on the 15th day of the experiment, the animals were euthanized, and a number of cardiomyopathy indices were investigated. The diabetic rats exhibited a momentous increase in hyperlipidemia, lipid peroxidation as well as a significant (p < 0.05) decline in antioxidant enzyme activities. The serum creatine kinase MB (CK-MB), C-reactive protein (CRP), cardiac troponin I, tumour necrosis factor-alpha (TNF-α) and urotensin II expression revealed a significant (p < 0.05) upsurge in diabetic rats. Also, the expression of GLUT4 and fatty acid-binding protein 3 (FABP3) were significantly (p < 0.05) reduced in diabetic rats. However, at the conclusion of the experimental trial ST significantly (p < 0.05) attenuated hyperlipidemia, oxidative stress biomarkers by augmenting the antioxidant enzyme activities and decrease in lipid peroxidation, ameliorated CK-MB, CRP, cardiac troponin I, TNF-α, and urotensin-II levels, and improved GLUT4 and FABP3 expressions. Similarly, the administration of ST prevented histological alterations in the heart of diabetic animals. Therefore, the obtained results suggest that ST could mitigate DCM in streptozotocin-induced diabetic rats.


Subject(s)
Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , Diabetes Mellitus, Experimental/complications , Fatty Acid Binding Protein 3/genetics , Fatty Acid Binding Protein 3/metabolism , Gene Expression/drug effects , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Sterculia/chemistry , Urotensins/genetics , Urotensins/metabolism , Animals , Cardiomyopathies/etiology , Gene Expression/genetics , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Male , Oxidative Stress , Plant Extracts/isolation & purification , Rats, Inbred Strains , Streptozocin , Water
13.
Exp Cell Res ; 407(1): 112768, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34370993

ABSTRACT

Atherosclerosis is the underlying contributing factor of cardiovascular disease, which is a process of inflammation and lipid-rich lesion. Macrophage-derived foam cell is a key hallmark of atherosclerosis and connected with various factors of lipid metabolism. Here, we showed that fatty acid binding protein 3 (FABP3) was upregulated in the aorta of ApoE-/- mice with high-fat-diet (HFD) feeding. Knockdown of FABP3 in HFD-fed ApoE-/- mice notably facilitated cholesterol efflux, inhibited macrophage foam cell formation, and thus prevented atherogenesis. Furthermore, FABP3 silencing decreased the expression of peroxisome proliferator-activated receptor γ (PPARγ). Mechanistic studies had disclosed the involvement of PPARγ signaling in balancing cholesterol uptake and efflux and diminishing foam cell formation. These findings firstly revealed an anti-atherogenic role of FABP3 silencing in preventing foamy macrophage formation partly through PPARγ, which might be a beneficial approach for therapying atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol/metabolism , Fatty Acid Binding Protein 3/deficiency , Macrophages/metabolism , Animals , Fatty Acid Binding Protein 3/metabolism , Lipid Metabolism/physiology , Macrophages/pathology , Macrophages, Peritoneal/metabolism , PPAR gamma/metabolism
14.
Fa Yi Xue Za Zhi ; 37(2): 158-165, 2021 Apr.
Article in English, Chinese | MEDLINE | ID: mdl-34142475

ABSTRACT

ABSTRACT: Objective To observe the skin ultrastructure change of electric shock death rats and to test the expression changes of hypoxia-inducible factor-2α (HIF-2α) and heart type-fatty acid-binding protein (H-FABP) of myocardial cells, in order to provide basis for forensic identification of electric shock death. Methods The electric shock model of rats was established. The 72 rats were randomly divided into control group, electric shock death group and postmortem electric shock group. Each group was divided into three subgroups, immediate (0 min), 30 min and 60 min after death. The skin changes of rats were observed by HE staining, the changes of skin ultrastructure were observed by scanning electron microscopy, and the expression of HIF-2α and H-FABP in rats myocardium was tested by immunohistochemical staining. Results The skin in the electric shock death group and postmortem electric shock group had no significant difference through the naked eye or by HE staining. Under the scanning electron microscope, a large number of cellular debris, cells with unclear boundaries, withered cracks, circular or elliptical holes scattered on the cell surface and irregular edges were observed. A large number of spherical foreign body particles were observed. Compared with the control group, the expression of HIF-2α in all electric shock death subgroups increased, reaching the peak immediately after death. In the postmortem electric shock group, HIF-2α expression only increased immediately after death, but was lower than that of electric shock death group (P<0.05). Compared with the control group, the expression of H-FABP in all subgroups of electric shock death group and postmortem electric shock group significantly decreased. The expression of H-FABP in all subgroups of electric shock death group was lower than that of the postmortem electric shock group (P<0.05). Conclusion Electric shock can increase HIF-2α expression and decrease H-FABP expression in the myocardium, which may be of forensic significance for the determination of electric shock death and identification of antemortem and postmortem electric shock.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Fatty Acid Binding Protein 3/metabolism , Myocardium , Myocytes, Cardiac , Skin/ultrastructure , Animals , Autopsy , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Rats
15.
Article in English | MEDLINE | ID: mdl-34098488

ABSTRACT

While the processes governing docosahexaenoic acid (DHA) trafficking across the blood-brain barrier have been elucidated, factors governing DHA uptake into microglia, an essential step for this fatty acid to exert its anti-inflammatory effects, are unknown. This study assessed the mRNA and protein expression of fatty acid-binding proteins (FABPs) and fatty acid transport proteins (FATPs) in mouse BV-2 cells and their mRNA expression in primary mouse microglia. The microglial uptake of DHA-d5, a surrogate of DHA, was assessed by LC-MS/MS following interventions including temperature reduction, silencing of various FABP isoforms, competition with DHA, and metabolic inhibition. It was found that DHA-d5 uptake at 4°C was 39.6% lower than at 37°C, suggesting that microglial uptake of DHA-d5 likely involves passive and/or active uptake mechanisms. Of all FABP and FATP isoforms probed, only FABP3, FABP4, FABP5, FATP1, and FATP4 were expressed at both the mRNA and protein level. Silencing of FABP3, FABP4, and FABP5 resulted in no change in cellular DHA-d5 uptake, nor did concomitant DHA administration or the presence of 0.1% sodium azide/50 mM 2-deoxy-D-glucose. This study is the first to identify the presence of FABPs and FATPs in mouse microglia, albeit these proteins are not involved in the microglial uptake of DHA-d5.


Subject(s)
Blood-Brain Barrier/metabolism , Docosahexaenoic Acids/metabolism , Fatty Acid Transport Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Microglia/metabolism , Animals , Deuterium , Fatty Acid Binding Protein 3/genetics , Fatty Acid Binding Protein 3/metabolism , Fatty Acid Transport Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
16.
Heart Vessels ; 36(11): 1765-1774, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34028584

ABSTRACT

Ongoing myocardial damage at the acme of the sepsis status has not been sufficiently evaluated. The clinical data of 160 sepsis patients who require intensive care and 127 outpatients with chronic heart failure (HF) were compared as a retrospective cohort study. Thereafter, the sepsis patients were divided into 3 groups according to the serum heart-type fatty acid-binding protein (H-FABP) quartiles [low H-FABP = Q1 (n = 39), middle H-FABP = Q2/Q3 (n = 81), and high H-FABP = Q4 group (n = 40)]. The H-FABP level was measured within 15 min of admission. The serum H-FABP levels in the sepsis patients [26.6 (9.3-79.0) ng/ml] were significantly higher than in the choric HF patients [6.6 (4.6-9.7) ng/ml]. A Kaplan-Meier curve showed that the survival rate of the high-H-FABP group was significantly lower than that of the middle- and low-H-FABP groups. The multivariate Cox regression analysis for the 365-day mortality showed that the high-H-FABP group (hazard ratio: 6.544, 95% confidence interval [CI] 2.026-21.140; p = 0.002) was an independent predictor of the 365-day mortality. The same trend in the prognostic impact was significantly (p = 0.015) observed in the cohort that had not been suffering from the cardiac disease before admission. The serum H-FABP level was an independent predictor of the 365-day mortality in the patients who were emergently hospitalized in the intensive-care unit due to sepsis. Ongoing myocardial damage was detected in the majority of patients with sepsis, suggesting that ongoing myocardial damage might be a candidate predictor of adverse outcomes in sepsis patients.


Subject(s)
Fatty Acid Binding Protein 3/metabolism , Fatty Acid-Binding Proteins , Sepsis , Biomarkers , Fatty Acid Binding Protein 3/chemistry , Humans , Prognosis , Retrospective Studies , Sepsis/diagnosis
17.
J Biol Chem ; 296: 100663, 2021.
Article in English | MEDLINE | ID: mdl-33862084

ABSTRACT

α-synuclein (αSyn) is a protein known to form intracellular aggregates during the manifestation of Parkinson's disease. Previously, it was shown that αSyn aggregation was strongly suppressed in the midbrain region of mice that did not possess the gene encoding the lipid transport protein fatty acid binding protein 3 (FABP3). An interaction between these two proteins was detected in vitro, suggesting that FABP3 may play a role in the aggregation and deposition of αSyn in neurons. To characterize the molecular mechanisms that underlie the interactions between FABP3 and αSyn that modulate the cellular accumulation of the latter, in this report, we used in vitro fluorescence assays combined with fluorescence microscopy, transmission electron microscopy, and quartz crystal microbalance assays to characterize in detail the process and consequences of FABP3-αSyn interaction. We demonstrated that binding of FABP3 to αSyn results in changes in the aggregation mechanism of the latter; specifically, a suppression of fibrillar forms of αSyn and also the production of aggregates with an enhanced cytotoxicity toward mice neuro2A cells. Because this interaction involved the C-terminal sequence region of αSyn, we tested a peptide derived from this region of αSyn (αSynP130-140) as a decoy to prevent the FABP3-αSyn interaction. We observed that the peptide competitively inhibited binding of αSyn to FABP3 in vitro and in cultured cells. We propose that administration of αSynP130-140 might be used to prevent the accumulation of toxic FABP3-αSyn oligomers in cells, thereby preventing the progression of Parkinson's disease.


Subject(s)
Amyloid/antagonists & inhibitors , Fatty Acid Binding Protein 3/metabolism , Neuroblastoma/pathology , Peptide Fragments/pharmacology , Protein Aggregation, Pathological/prevention & control , alpha-Synuclein/metabolism , Amyloid/metabolism , Animals , Fatty Acid Binding Protein 3/genetics , Humans , Mice , Neuroblastoma/genetics , Neuroblastoma/metabolism , Tumor Cells, Cultured , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/genetics
18.
Int J Mol Sci ; 22(6)2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33804804

ABSTRACT

Nicotine is the predominant addictive compound of tobacco and causes the acquisition of dependence through its interactions with nicotinic acetylcholine receptors and various neurotransmitter releases in the central nervous system. The Ca2+/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase (ERK) play a pivotal role in synaptic plasticity in the hippocampus. CaMKII is involved in long-term potentiation induction, which underlies the consolidation of learning and memory; however, the roles of CaMKII in nicotine and other psychostimulant-induced addiction still require further investigation. This article reviews the molecular mechanisms and crucial roles of CaMKII and ERK in nicotine and other stimulant drug-induced addiction. We also discuss dopamine (DA) receptor signaling involved in nicotine-induced addiction in the brain reward circuitry. In the last section, we introduce the association of polyunsaturated fatty acids and cellular chaperones of fatty acid-binding protein 3 in the context of nicotine-induced addiction in the mouse nucleus accumbens and provide a novel target for the treatment of drug abuse affecting dopaminergic systems.


Subject(s)
Signal Transduction , Animals , Brain/metabolism , Brain/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Susceptibility , Dopamine/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fatty Acid Binding Protein 3/metabolism , Fatty Acids/metabolism , Humans , Nicotine/adverse effects , Nicotine/metabolism , Substance-Related Disorders/etiology , Substance-Related Disorders/metabolism
19.
Anim Sci J ; 92(1): e13537, 2021.
Article in English | MEDLINE | ID: mdl-33682250

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) is a critical transcription factor regulating lipid and glucose metabolism. However, the regulatory effect of PPARγ on milk fat synthesis in buffalo mammary gland is not clear. In order to explore the role of buffalo PPARG gene in milk fat synthesis, lentivirus-mediated interference was used to knock it down and then the recombinant fusion expression vector was transfected into buffalo mammary epithelial cell (BMEC) to overexpress it. PPARG gene knockdown significantly decreased the expression of CD36, FABP3, FABP4, ACSS2, ELOVL6, DGAT2, BTN1A1, AGPAT6, LPIN1, ABCG2, PPARGC1A, INSIG1, FASN, and SREBF2 genes and significantly upregulated the expression of INSIG2 gene but had no significant effect on the expression of ACSL1, GPAM, and SREBF1 genes. PPARG overexpression significantly increased the relative mRNA abundance of CD36, FABP3, FABP4, ACSS2, ELOVL6, DGAT2, BTN1A1, AGPAT6, LPIN1, PPARGC1A, INSIG1, and SREBF2 genes and significantly downregulated the expression of INSIG2 gene but had no significant effect on the expression of ACSL1, GPAM, ABCG2, FASN, and SREBF1 genes. In addition, knockdown/overexpression of PPARG gene significantly decreased/increased triacylglycerol (TAG) content in BMECs. This study revealed that buffalo PPARG gene is a key gene regulating buffalo milk fat synthesis.


Subject(s)
Buffaloes/genetics , Buffaloes/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation/genetics , Gene Expression/genetics , Glycolipids/metabolism , Glycoproteins/metabolism , Lipid Droplets/metabolism , Mammary Glands, Animal/cytology , Milk/metabolism , PPAR gamma/genetics , PPAR gamma/physiology , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Fatty Acid Binding Protein 3/genetics , Fatty Acid Binding Protein 3/metabolism , Female , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Triglycerides/metabolism
20.
Brain Res ; 1760: 147383, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33636166

ABSTRACT

Accumulation and aggregation of α-synuclein (αSyn) trigger neuronal loss in the substantia nigra pars compacta (SNpc), which in turn causes motor symptoms in Parkinson's disease. We previously demonstrated that fatty acid-binding protein 3 (FABP3), an intracellular fatty acid carrier protein, enhances αSyn neurotoxicity in SNpc and motor impairments after intranigral injection of αSyn fibrils. However, the temporal profile of αSyn fibril spread and their toxicity remains unclear. In the present study, we investigated the temporal profile of αSyn fibril spread and its toxicity, which induces intracellular fibril formation. Monomeric and fibrillar aSyn assemblies were labeled with ATTO550 to distinguish the exogenous form from the endogenous species and injected into bilateral striatum in Fabp3+/+ (wild type) and Fabp3-/- mice. Accumulation of both monomeric and fibrillar exogenous αSyn in the SNpc was drastically decreased in Fabp3-/- mice compared to that in the Fabp3+/+ counterparts. Deletion of Fabp3 also prevented exogenous αSyn fibril-induced seeding of the endogenous αSyn into aggregates containing phosphorylated and filamentous forms in the SNpc. Consistent with these results, loss of dopaminergic neurons and subsequent impaired motor behavior were attenuated in Fabp3-/- mice. These results highlight the crucial role of FABP3 in pathogenic αSyn accumulation and its seeding ability. Taken together, FABP3 could be a potential therapeutic target against αSyn propagation in synucleinopathies.


Subject(s)
Fatty Acid Binding Protein 3/metabolism , Pars Compacta/pathology , Synucleinopathies/pathology , alpha-Synuclein/metabolism , alpha-Synuclein/toxicity , Animals , Dopaminergic Neurons/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...