Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
J Nutr Biochem ; 108: 109087, 2022 10.
Article in English | MEDLINE | ID: mdl-35691593

ABSTRACT

Although the role of mechanistic target of rapamycin complex 1 (mTORC1) in lipid metabolism has been the subject of previous research, its function in chylomicron production is not known. In this study, we created three stable human colorectal adenocarcinoma Caco-2 cell lines exhibiting normal, low, or high mTORC1 kinase activity, and used these cells to investigate the consequences of manipulating mTORC1 activity on enterocyte differentiation and chylomicron-like particle production. Constitutively active mTORC1 induced Caco-2 cell proliferation and differentiation (as judged by alkaline phosphatase activity) but weakened transepithelial electrical resistance (TEER). Repressed mTORC1 activity due to the knockdown of RPTOR significantly decreased the expression of lipogenic genes FASN, DGAT1, and DGAT2, lipoprotein assembly genes APOB and MTTP, reduced protein expression of APOB, MTTP, and FASN, downregulated the gene expression of very long-chain fatty acyl-CoA ligase (FATP2), acyl-CoA binding protein (DBI), and prechylomicron transport vesicle-associated proteins VAMP7 (vesicle-associated membrane protein 7) and SAR1B (secretion associated Ras related GTPase 1B) resulting in the repression of apoB-containing triacylglycerol-rich lipoprotein secretion. Exposure of Caco-2 cells harboring a constitutively active mTORC1 to short-chain fatty acid derivatives, R-α-lipoic acid and 4-phenylbutyric acid, downregulated chylomicron-like particle secretion by interfering with the lipidation and assembly of the particles, and concomitantly repressed mTORC1 activity with no change to Raptor abundance or PRAS40 (Thr246) phosphorylation. R-α-lipoic acid and 4-phenylbutyric acid may be useful to mitigate intestinal lipoprotein overproduction and associated postprandial inflammation.


Subject(s)
Chylomicrons , Enterocytes , Monomeric GTP-Binding Proteins , Phenylbutyrates , Regulatory-Associated Protein of mTOR , Thioctic Acid , Apolipoproteins B/metabolism , Caco-2 Cells , Chylomicrons/metabolism , Coenzyme A Ligases/metabolism , Enterocytes/drug effects , Enterocytes/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Phenylbutyrates/metabolism , Phenylbutyrates/pharmacology , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Thioctic Acid/metabolism , Thioctic Acid/pharmacology
2.
Life Sci Alliance ; 5(4)2022 04.
Article in English | MEDLINE | ID: mdl-35064074

ABSTRACT

The human Sec61 complex is a widely distributed and abundant molecular machine. It resides in the membrane of the endoplasmic reticulum to channel two types of cargo: protein substrates and calcium ions. The SEC61A1 gene encodes for the pore-forming Sec61α subunit of the Sec61 complex. Despite their ubiquitous expression, the idiopathic SEC61A1 missense mutations p.V67G and p.T185A trigger a localized disease pattern diagnosed as autosomal dominant tubulointerstitial kidney disease (ADTKD-SEC61A1). Using cellular disease models for ADTKD-SEC61A1, we identified an impaired protein transport of the renal secretory protein renin and a reduced abundance of regulatory calcium transporters, including SERCA2. Treatment with the molecular chaperone phenylbutyrate reversed the defective protein transport of renin and the imbalanced calcium homeostasis. Signal peptide substitution experiments pointed at targeting sequences as the cause for the substrate-specific impairment of protein transport in the presence of the V67G or T185A mutations. Similarly, dominant mutations in the signal peptide of renin also cause ADTKD and point to impaired transport of this renal hormone as important pathogenic feature for ADTKD-SEC61A1 patients as well.


Subject(s)
Phenylbutyrates/pharmacology , Renin/metabolism , SEC Translocation Channels/genetics , Calcium/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Kidney Diseases/physiopathology , Molecular Chaperones/metabolism , Mutation, Missense , Phenylbutyrates/metabolism , Polycystic Kidney Diseases , Protein Transport/genetics , Renin/genetics , SEC Translocation Channels/chemistry , SEC Translocation Channels/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
3.
Int J Mol Sci ; 22(13)2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34281222

ABSTRACT

There are two types of cytochrome P450 enzymes in nature, namely, the monooxygenases and the peroxygenases. Both enzyme classes participate in substrate biodegradation or biosynthesis reactions in nature, but the P450 monooxygenases use dioxygen, while the peroxygenases take H2O2 in their catalytic cycle instead. By contrast to the P450 monooxygenases, the P450 peroxygenases do not require an external redox partner to deliver electrons during the catalytic cycle, and also no external proton source is needed. Therefore, they are fully self-sufficient, which affords them opportunities in biotechnological applications. One specific P450 peroxygenase, namely, P450 OleTJE, reacts with long-chain linear fatty acids through oxidative decarboxylation to form hydrocarbons and, as such, has been implicated as a suitable source for the biosynthesis of biofuels. Unfortunately, the reactions were shown to produce a considerable amount of side products originating from Cα and Cß hydroxylation and desaturation. These product distributions were found to be strongly dependent on whether the substrate had substituents on the Cα and/or Cß atoms. To understand the bifurcation pathways of substrate activation by P450 OleTJE leading to decarboxylation, Cα hydroxylation, Cß hydroxylation and Cα-Cß desaturation, we performed a computational study using 3-phenylpropionate and 2-phenylbutyrate as substrates. We set up large cluster models containing the heme, the substrate and the key features of the substrate binding pocket and calculated (using density functional theory) the pathways leading to the four possible products. This work predicts that the two substrates will react with different reaction rates due to accessibility differences of the substrates to the active oxidant, and, as a consequence, these two substrates will also generate different products. This work explains how the substrate binding pocket of P450 OleTJE guides a reaction to a chemoselectivity.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Fatty Acids/metabolism , Models, Chemical , Phenylbutyrates/metabolism , Phenylpropionates/metabolism
4.
Exp Physiol ; 106(3): 585-592, 2021 03.
Article in English | MEDLINE | ID: mdl-33369803

ABSTRACT

NEW FINDINGS: What is the central question of this study? The compound sodium phenylbutyrate (PB) has been shown to promote branched-chain amino acid (BCAA) catabolism, and as such has been proposed as a treatment for disorders with enhanced BCAA levels: does PB induce muscle protein catabolism by forcing BCAA degradation away from muscle protein synthesis and mechanistic target of rapamycin (mTOR) inhibition? What is the main finding and its importance? Accelerated BCAA catabolism using PB resulted in adverse effects related to mTOR signalling and muscle protein metabolism in skeletal muscle cells, which may limit its application in conditions where muscle wasting is a risk. ABSTRACT: The compound sodium phenylbutyrate (PB) has been used for reducing ammonia in patients with urea cycle disorders and proposed as a treatment for disorders with enhanced branched-chain amino acid (BCAA) levels, due to its effects on promoting BCAA catabolism. In skeletal muscle cells, we hypothesised that PB would induce muscle protein catabolism due to forcing BCAA degradation away from muscle protein synthesis and downregulating mechanistic target of rapamycin (mTOR). PB reduced medium BCAA and branched-chain keto acid (BCKA) concentrations, while total cell protein (-21%; P < 0.001 vs. control) and muscle protein synthesis (-25%; P < 0.001 vs. control; assessed by measurement of puromycin incorporation into polypeptides) were decreased with PB. The regulator of anabolic pathways mTOR and its downstream components were impaired with PB treatment. The present results indicate that accelerated BCAA catabolism using PB resulted in adverse effects related to mTOR signalling and muscle protein metabolism, which may limit its application in settings where muscle wasting is a risk.


Subject(s)
Muscle, Skeletal , Phenylbutyrates , Amino Acids, Branched-Chain/metabolism , Animals , Cell Line , Mice , Muscle, Skeletal/metabolism , Oxidoreductases/metabolism , Phenylbutyrates/metabolism , Phenylbutyrates/pharmacology
5.
Acta Neurobiol Exp (Wars) ; 80(3): 305-321, 2020.
Article in English | MEDLINE | ID: mdl-32990288

ABSTRACT

In humans, pyruvate dehydrogenase complex (PDC) deficiency impairs brain energy metabolism by reducing the availability of the functional acetyl­CoA pool. This "hypometabolic defect" results in congenital lactic acidosis and abnormalities of brain morphology and function, ranging from mild ataxia to profound psychomotor retardation. Our previous study showed reduction in total cell number and dendritic arbors in the cerebellar Purkinje cells in systemic PDC­deficient mice. Phenylbutyrate has been shown to increase PDC activity in cultured fibroblasts from PDC­deficient patients. Hence, we investigated the effects of postnatal (days 2­35) phenylbutyrate administration on the cerebellar Purkinje cell population in PDC­deficient female mice. Histological analyses of different regions of cerebellar cortex from the brain­specific PDC­deficient saline­injected mice revealed statistically significant reduction in the Purkinje cell density and increased cell size of the individual Purkinje cell soma compared to control PDC­normal, saline­injected group. Administration of phenylbutyrate to control mice did not cause significant changes in the Purkinje cell density and cell size in the studied regions. In contrast, administration of phenylbutyrate variably lessened the ill effects of PDC deficiency on Purkinje cell populations in different areas of the cerebellum. Our results lend further support for the possible use of phenylbutyrate as a potential treatment for PDC deficiency.


Subject(s)
Brain/drug effects , Neurons/drug effects , Phenylbutyrates/pharmacology , Purkinje Cells/drug effects , Animals , Cerebellar Cortex/drug effects , Cerebellum/drug effects , Disease Models, Animal , Mice, Transgenic , Phenylbutyrates/metabolism , Purkinje Cells/cytology
6.
Biochim Biophys Acta Mol Basis Dis ; 1866(11): 165899, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32698045

ABSTRACT

Aging impairs the mitochondrial electron transport chain (ETC), especially in interfibrillar mitochondria (IFM). Mitochondria are in close contact with the endoplasmic reticulum (ER). Induction of ER stress leads to ETC injury in adult heart mitochondria. We asked if ER stress contributes to the mitochondrial dysfunction during aging. Subsarcolemmal mitochondria (SSM) and IFM were isolated from 3, 18, and 24 mo. C57Bl/6 mouse hearts. ER stress progressively increased with age, especially in 24 mo. mice that manifest mitochondrial dysfunction. OXPHOS was decreased in 24 mo. IFM oxidizing complex I and complex IV substrates. Proteomic analysis showed that the content of multiple complex I subunits was decreased in IFM from 24 mo. hearts, but remained unchanged in in 18 mo. IFM without a decrease in OXPHOS. Feeding 24 mo. old mice with 4-phenylbutyrate (4-PBA) for two weeks attenuated the ER stress and improved mitochondrial function. These results indicate that ER stress contributes to the mitochondrial dysfunction in aged hearts. Attenuation of ER stress is a potential approach to improve mitochondrial function in aged hearts.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardium/metabolism , Aging/physiology , Animals , Electron Transport Complex I/metabolism , Endoplasmic Reticulum Stress/genetics , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Oxidative Phosphorylation , Phenylbutyrates/metabolism , Real-Time Polymerase Chain Reaction
7.
Molecules ; 25(9)2020 Apr 28.
Article in English | MEDLINE | ID: mdl-32354057

ABSTRACT

The use of baker's yeast to reduce ethyl 2-oxo-4-phenylbutyrate (EOPB) in conventional biphasic systems is hindered by low productivities due to mass transfer resistance between the biocatalyst and the substrate partitioned into two different phases. To overcome the limitation, a new reaction-separation coupling process (RSCP) was configured in this study, based on the novel thermosensitive ionic liquids (ILs) with polyoxyethylene-tail. The solubility of ILs in common solvents was investigated to configure the unique thermosensitive ionic liquids-solvent biphasic system (TIBS) in which the reduction was performed. [(CH3)2N(C2H5)(CH2CH2O)2H][PF6] (c2) in 1,2-dimethoxyethane possesses the thermosensitive function of homogeneous at lower temperatures and phase separating at higher temperatures. The phase transformation temperature (PTT) of the mixed system of c2/1,2-dimethoxyethane (v/v, 5:18) was about 33 °C. The bioreaction takes place in a "homogeneous" liquid phase at 30 °C. At the end of each reduction run, the system temperature is increased upon to the PTT, while c2 is separated from 1,2-dimethoxyethane with turning the system into two phases. The enantiomeric excesses (e.e.) of ethyl (R)-2-hydroxy-4-phenylbutyrate ((R)-EHPB) increased about 25~30% and the yield of ethyl-2-hydroxy-4-phenylbutyrate (EHPB) increased 35% in TIBS, compared with the reduction in 1,2-dimethoxyethane. It is expected that the TIBS established in this study could provide many future opportunities in the biocatalysis.


Subject(s)
Ionic Liquids , Phenylbutyrates/metabolism , Polyethylene Glycols/chemistry , Saccharomyces cerevisiae/metabolism , Biocatalysis , Catalysis , Chlorides/chemistry , Green Chemistry Technology , Solubility , Solvents , Stereoisomerism , Temperature , Water
8.
Chembiochem ; 21(18): 2680-2688, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32324965

ABSTRACT

Glucose dehydrogenase (GDH) is a general tool for driving nicotinamide (NAD(P)H) regeneration in synthetic biochemistry. An increasing number of synthetic bioreactions are carried out in media containing high amounts of organic cosolvents or hydrophobic substrates/products, which often denature native enzymes, including those for cofactor regeneration. In this work, we attempted to improve the chemical stability of Bacillus megaterium GDH (BmGDHM0 ) in the presence of large amounts of 1-phenylethanol by directed evolution. Among the resulting mutants, BmGDHM6 (Q252L/E170K/S100P/K166R/V72I/K137R) exhibited a 9.2-fold increase in tolerance against 10 % (v/v) 1-phenylethanol. Moreover, BmGDHM6 was also more stable than BmGDHM0 when exposed to hydrophobic and enzyme-inactivating compounds such as acetophenone, ethyl 2-oxo-4-phenylbutyrate, and ethyl (R)-2-hydroxy-4-phenylbutyrate. Coupled with a Candida glabrata carbonyl reductase, BmGDHM6 was successfully used for the asymmetric reduction of deactivating ethyl 2-oxo-4-phenylbutyrate with total turnover number of 1800 for the nicotinamide cofactor, thus making it attractive for commercial application. Overall, the evolution of chemically robust GDH facilitates its wider use as a general tool for NAD(P)H regeneration in biocatalysis.


Subject(s)
Glucose 1-Dehydrogenase/metabolism , Niacinamide/metabolism , Bacillus megaterium/enzymology , Benzyl Alcohols/chemistry , Benzyl Alcohols/metabolism , Glucose 1-Dehydrogenase/chemistry , Glucose 1-Dehydrogenase/genetics , Molecular Structure , Mutation , Niacinamide/chemistry , Oxidation-Reduction , Phenylbutyrates/chemistry , Phenylbutyrates/metabolism
9.
Biochemistry ; 59(13): 1367-1377, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32207963

ABSTRACT

More than 80 loss-of-function (LOF) mutations in the SLC6A8 creatine transporter (hCRT1) are responsible for cerebral creatine deficiency syndrome (CCDS), which gives rise to a spectrum of neurological defects, including intellectual disability, epilepsy, and autism spectrum disorder. To gain insight into the nature of the molecular defects caused by these mutations, we quantitatively profiled the cellular processing, trafficking, expression, and function of eight pathogenic CCDS variants in relation to the wild type (WT) and one neutral isoform. All eight CCDS variants exhibit measurable proteostatic deficiencies that likely contribute to the observed LOF. However, the magnitudes of their specific effects on the expression and trafficking of hCRT1 vary considerably, and we find that the LOF associated with two of these variants primarily arises from the disruption of the substrate-binding pocket. In conjunction with an analysis of structural models of the transporter, we use these data to suggest mechanistic classifications for these variants. To evaluate potential avenues for therapeutic intervention, we assessed the sensitivity of these variants to temperature and measured their response to the proteostasis regulator 4-phenylbutyrate (4-PBA). Only one of the tested variants (G132V) is sensitive to temperature, though its response to 4-PBA is negligible. Nevertheless, 4-PBA significantly enhances the activity of WT hCRT1 in HEK293T cells, which suggests it may be worth evaluating as a therapeutic for female intellectual disability patients carrying a single CCDS mutation. Together, these findings reveal that pathogenic SLC6A8 mutations cause a spectrum of molecular defects that should be taken into consideration in future efforts to develop CCDS therapeutics.


Subject(s)
Brain Diseases, Metabolic, Inborn/metabolism , Creatine/deficiency , Mental Retardation, X-Linked/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Brain Diseases, Metabolic, Inborn/genetics , Creatine/genetics , Creatine/metabolism , HEK293 Cells , Humans , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mental Retardation, X-Linked/genetics , Mutation, Missense , Nerve Tissue Proteins/chemistry , Phenylbutyrates/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/metabolism
10.
Brain Dev ; 42(2): 231-235, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31604595

ABSTRACT

An adult female patient was diagnosed with arginase 1 deficiency (ARG1-D) at 4 years of age, and had been managed with protein restriction combined with sodium benzoate therapy. Though the treatment was successful in ameliorating hyperammonemia, hyperargininemia persisted. After being under control with a strict restriction of dietary protein, severe fall of serum albumin levels appeared and her condition became strikingly worsened. However, after sodium phenylbutyrate (NaPB) therapy was initiated, the clinical condition and metabolic stability was greatly improved. Current management of ARG1-D is aimed at lowering plasma arginine levels. The nitrogen scavengers, such as NaPB can excrete the waste nitrogen not through the urea cycle but via the alternative pathway. The removal of nitrogen via alternative pathway lowers the flux of arginine in the urea cycle. Thereby, the clinical complications due to insufficient amount of protein intake can be prevented. Thus, NaPB therapy can be expected as a useful therapeutic option, particularly in patients with ARG1-D.


Subject(s)
Arginase/genetics , Hyperargininemia/drug therapy , Phenylbutyrates/therapeutic use , Adult , Arginase/metabolism , Arginine/metabolism , Female , Humans , Hyperammonemia/blood , Hyperargininemia/blood , Hyperargininemia/genetics , Phenylbutyrates/metabolism
11.
Biomater Sci ; 7(11): 4624-4635, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31451819

ABSTRACT

4-Phenylbutyric acid (PBA)-installed hyaluronic acid (HA)-based nanoparticles (NPs) were developed for amplifying the anticancer potential of curcumin (CUR) for lung cancer therapy. PBA was introduced to the HA backbone as a hydrophobic segment of a nanoassembled structure and as a histone deacetylase (HDAC) inhibitor for cancer therapy. PBA was released from the HA-PBA conjugate (HAPBA) via an esterase-responsive cleavage of ester bonds in cancer cells and may affect the dissociation of NP structure. CUR-entrapped HAPBA-based NPs, with 265 nm hydrodynamic size, unimodal size distribution, negative zeta potential, and sustained drug release, were fabricated. Co-treatment of A549 cells by PBA and CUR elevated the antiproliferation efficiency compared with CUR-treatment. CUR-loaded HAPBA NPs also exhibited a significantly lower IC50 value compared with the CUR and HAPBA10 + CUR groups (p < 0.05). Cy5.5-labeled HAPBA NPs containing CUR group displayed higher accumulation in tumor tissue and less distribution in liver and spleen after intravenous injection compared with the Cy5.5-injected group in A549 tumor-bearing mouse model. Multiple dosing of CUR-loaded HAPBA NPs in A549 tumor-bearing mouse model exhibited efficient tumor growth suppression and apoptosis-inducing effects. CD44 receptor targeting and HDAC inhibiting HAPBA NPs can be used to boost the anticancer potentials of drug cargo for the therapy of CD44 receptor-expressed cancers.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Esterases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hyaluronic Acid/pharmacology , Lung Neoplasms/drug therapy , Nanoparticles/chemistry , Phenylbutyrates/pharmacology , A549 Cells , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Apoptosis/drug effects , Female , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylases/metabolism , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Phenylbutyrates/chemistry , Phenylbutyrates/metabolism
12.
Pharmacol Res ; 144: 116-131, 2019 06.
Article in English | MEDLINE | ID: mdl-30954630

ABSTRACT

In the past two decades, significant advances have been made in the etiology of lipid disorders. Concomitantly, the discovery of liporegulatory functions of certain short-chain fatty acids has generated interest in their clinical applications. In particular, butyric acid (BA) and its derivative, 4-phenylbutyric acid (PBA), which afford health benefits against lipid disorders while being generally well tolerated by animals and humans have been assessed clinically. This review examines the evidence from cell, animal and human studies pertaining to the lipid-regulating effects of BA and PBA, their molecular mechanisms and therapeutic potential. Collectively, the evidence supports the view that intakes of BA and PBA benefit lipid homeostasis across biological systems. We reviewed the evidence that BA and PBA downregulate de novo lipogenesis, ameliorate lipotoxicity, slow down atherosclerosis progression, and stimulate fatty acid ß-oxidation. Central to their mode of action, BA appears to function as a histone deacetylase (HDAC) inhibitor while PBA acts as a chemical chaperone and/or a HDAC inhibitor. Areas of further inquiry include the effects of BA and PBA on adipogenesis, lipolysis and apolipoprotein metabolism.


Subject(s)
Butyric Acid/pharmacology , Lipid Metabolism/drug effects , Phenylbutyrates/pharmacology , Adipogenesis/drug effects , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Butyric Acid/metabolism , Butyric Acid/therapeutic use , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/therapeutic use , Humans , Phenylbutyrates/metabolism , Phenylbutyrates/therapeutic use
13.
Comput Biol Chem ; 80: 128-137, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30952039

ABSTRACT

Phenylbutyrate (PB), a small aromatic fatty acid, has been known as an interesting compound with the ability of anti-proliferation and cell growth inhibition in cancer cells. In the present study, a series of PB derivatives were synthesized by Passerini multicomponent reaction and their cytotoxic activities against various human cancer cell lines including A549 (non-small cell lung cancer), MDA-MB-231 (breast cancer), and SW1116 (colon cancer) were evaluated. The results revealed that B9, displayed significantly higher in vitro cytotoxicity with IC50 of 6.65, 8.44 and 24.71 µM, against A549, MDA-MB-231 and, SW1116, respectively, in comparison to PB. The effects of these compounds on the proliferation of MCF-10A as non-tumoral breast cell line, showed good selectivity of the compounds between tumorigenic and non-tumorigenic cell lines. Moreover, B9 has indicated apoptosis-inducing activities to MDA-MB-231 cancer cell line in a dose-dependent manner. The molecular docking studies of the synthesized compounds on pyruvate dehydrogenase kinase 2 (PDK2; PDB ID: 2BU8) and histone deacetylase complex (HDAC; PDB ID: 1C3R), as the main targets of PB were applied to predict the binding sites and binding orientation of the compounds to these targets.


Subject(s)
Antineoplastic Agents/pharmacology , Phenylbutyrates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Bacteria/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Phenylbutyrates/chemical synthesis , Phenylbutyrates/chemistry , Phenylbutyrates/metabolism , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Structure-Activity Relationship
14.
Psychoneuroendocrinology ; 104: 286-299, 2019 06.
Article in English | MEDLINE | ID: mdl-30927713

ABSTRACT

Antipsychotics are the most important treatment for schizophrenia. However, antipsychotics, particularly olanzapine and clozapine, are associated with severe weight gain/obesity side-effects. Although numerous studies have been carried out to identify the exact mechanisms of antipsychotic-induced weight gain, it is still important to consider other pathways. Endoplasmic reticulum (ER) stress signaling and its associated inflammation pathway is one of the most important pathways involved in regulation of energy balance. In the present study, we examined the role of hypothalamic protein kinase R like endoplasmic reticulum kinase- eukaryotic initiation factor 2α (PERK-eIF2α) signaling and the inflammatory IkappaB kinase ß- nuclear factor kappa B (IKKß-NFκB) signaling pathway in olanzapine-induced weight gain in female rats. In this study, we found that olanzapine significantly activated PERK-eIF2α and IKKß-NFκB signaling in SH-SY5Y cells in a dose-dependent manner. Olanzapine treatment for 8 days in rats was associated with activated PERK-eIF2α signaling and IKKß-NFκB signaling in the hypothalamus, accompanied by increased food intake and weight gain. Co-treatment with an ER stress inhibitor, 4-phenylbutyrate (4-PBA), decreased olanzapine-induced food intake and weight gain in a dose- and time-dependent manner. Moreover, 4-PBA dose-dependently inhibited olanzapine-induced activated PERK-eIF2α and IKKß-NFκB signaling in the hypothalamus. These results suggested that hypothalamic ER stress may play an important role in antipsychotic-induced weight gain.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Phenylbutyrates/pharmacology , Animals , Antipsychotic Agents/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Eukaryotic Initiation Factor-2/metabolism , Female , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , I-kappa B Kinase/metabolism , Inflammation/metabolism , NF-kappa B/metabolism , Olanzapine/pharmacology , Phenylbutyrates/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , eIF-2 Kinase/metabolism
15.
Nat Chem Biol ; 15(1): 18-26, 2019 01.
Article in English | MEDLINE | ID: mdl-30510193

ABSTRACT

Prostaglandin E receptor EP4, a G-protein-coupled receptor, is involved in disorders such as cancer and autoimmune disease. Here, we report the crystal structure of human EP4 in complex with its antagonist ONO-AE3-208 and an inhibitory antibody at 3.2 Å resolution. The structure reveals that the extracellular surface is occluded by the extracellular loops and that the antagonist lies at the interface with the lipid bilayer, proximal to the highly conserved Arg316 residue in the seventh transmembrane domain. Functional and docking studies demonstrate that the natural agonist PGE2 binds in a similar manner. This structural information also provides insight into the ligand entry pathway from the membrane bilayer to the EP4 binding pocket. Furthermore, the structure reveals that the antibody allosterically affects the ligand binding of EP4. These results should facilitate the design of new therapeutic drugs targeting both orthosteric and allosteric sites in this receptor family.


Subject(s)
Receptors, Prostaglandin E, EP4 Subtype/chemistry , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Allosteric Regulation , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Binding Sites , Caprylates/chemistry , Caprylates/metabolism , Crystallography, X-Ray , Epoprostenol/analogs & derivatives , Epoprostenol/chemistry , Epoprostenol/metabolism , Humans , Ligands , Lipid Bilayers , Molecular Docking Simulation , Naphthalenes/chemistry , Naphthalenes/metabolism , Phenyl Ethers/chemistry , Phenyl Ethers/metabolism , Phenylbutyrates/chemistry , Phenylbutyrates/metabolism , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/genetics , Spodoptera/genetics
16.
Bioprocess Biosyst Eng ; 41(9): 1383-1390, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29948210

ABSTRACT

(R)-2-hydroxy-4-phenylbutyric acid (R-HPBA) is a valuable intermediate for the synthesis of angiotensin-converting enzyme inhibitors. The asymmetric reduction of 2-oxo-4-phenylbutyric acid (OPBA) by oxidoreductases is an efficient approach for its synthesis. Here, we report a novel biocatalytic approach for asymmetric synthesis of R-HPBA using recombinant Pichia pastoris expressing the Tyr52Leu variant of D-lactate dehydrogenase (D-LDH) from Lactobacillus plantarum. The recombinant yeast cells showed impressive catalytic activity at a high concentration of NaOPBA (380 mM, 76 g/L) and achieved full conversion starting with 40 g/L NaOPBA or even at higher concentration. Under optimized reaction conditions (pH 7.5, 37 °C, and 2% glucose), a full conversion with > 95% reaction yield and ~ 100% product enantiomeric excess (ee) was achieved for the preparation of R-HPBA on a 2-g scale. The findings of this study promote both the biotransformation of R-HPBA and an extension of the application of recombinant yeast as biocatalysts.


Subject(s)
Bacterial Proteins , L-Lactate Dehydrogenase , Lactobacillus plantarum/genetics , Microorganisms, Genetically-Modified , Phenylbutyrates/metabolism , Pichia , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , L-Lactate Dehydrogenase/biosynthesis , L-Lactate Dehydrogenase/genetics , Lactobacillus plantarum/enzymology , Microorganisms, Genetically-Modified/genetics , Microorganisms, Genetically-Modified/metabolism , Pichia/genetics , Pichia/metabolism
17.
Biol Pharm Bull ; 41(6): 961-966, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29526885

ABSTRACT

Sodium 4-phenylbutyrate (PBA), which exerts a wide range of anti-inflammatory effects, is rapidly cleared from the body (approximately 98%) by urinary excretion by 24 h after oral treatment in humans. PBA was almost entirely excreted to urine as phenylacetyl glutamine (PAGln). However, no data describe the potential anti-inflammatory effects of PAGln. The purpose of this study was to evaluate the anti-inflammatory effects of PAGln on mouse spleen cells and peritoneal cavity cells, and explore the potential mechanism underlying this effect. PAGln was added to mouse spleen cell cultures stimulated by concanavalin A, or mouse peritoneal cavity cell cultures stimulated by lipopolysaccharide. After 72 h of culture, levels of inflammatory cytokines in culture supernatants were measured using a sandwich enzyme-linked immunosorbent assay system, and levels of inflammatory proteins were assessed by Western blotting. PAGln significantly inhibited inflammatory cytokine (interferon-γ, interleukin-6, and tumor necrosis factor-α) production, decrease of cell number in the spleen cell, and suppressed the expression of inflammatory proteins (nuclear factor κB, and inducible nitric oxide synthase). These results suggest that PAGln possesses anti-inflammatory activity via inhibition of T cell activation and Toll-like receptor 4 signaling. This study of the anti-inflammatory mechanism of PAGln provides useful information about its potential for therapeutic applications.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glutamine/analogs & derivatives , Animals , Antineoplastic Agents/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Concanavalin A/pharmacology , Glutamine/pharmacology , Interferon-gamma/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Male , Mice, Inbred ICR , Peritoneal Cavity/cytology , Phenylbutyrates/metabolism , Signal Transduction/drug effects , Spleen/cytology , T-Lymphocytes/drug effects , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Fish Shellfish Immunol ; 72: 247-258, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29108970

ABSTRACT

Maintaining fish health is one of the most important aims in aquaculture. Prevention of fish diseases therefore is crucial and can be achieved by various different strategies, including most often a combination of different methods such as optimal feed and fish density, as well as strengthening the immune system. Understanding the fish innate immune system and developing methods to activate it, in an effort to prevent infections in the first place, has been a goal in recent years. In this study we choose different inducers of the innate immune system and examined their effects in vitro on the salmon cell line CHSE-214. We found that the butyrate derivatives 4-phenyl butyrate (PBA) and ß-hydroxy-ß-methyl butyrate (HMB) induce the expression of various innate immune genes differentially over 24-72 h. Similarly, lipids generated from fish oils were found to have an effect on the expression of the antimicrobial peptides cathelicidin and hepcidin, as well as iNOS and the viral receptor RIG-1. Interestingly we found that vitamin D3, similar as in mammals, was able to increase cathelicidin expression in fish cells. The observed induction of these different innate immune factors correlated with antibacterial activity against Aeromonas salmonicida and antiviral activity against IPNV and ISAV in vitro. To relate this data to the in vivo situation we examined cathelicidin expression in juvenile salmon and found that salmon families vary greatly in their basal cathelicidin levels. Examining cathelicidin levels in families known to be resistant to IPNV showed that these QTL-families had lower basal levels of cathelicidin in gills, than non QTL-families. Feeding fish with HMB caused a robust increase in cathelicidin expression in gills, but not skin and this was independent of the fish being resistant to IPNV. These findings support the use of fish cell lines as a tool to develop new inducers of the fish innate immune system, but also highlight the importance of the tissue studied in vivo. Understanding the response of the innate immune system in different tissues and what effect this might have on infections and downstream cellular pathways is an interesting research topic for the future.


Subject(s)
Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Immunity, Innate , Salmo salar/genetics , Salmo salar/immunology , Aeromonas salmonicida/physiology , Animals , Birnaviridae Infections/immunology , Birnaviridae Infections/veterinary , Cell Line , Cholecalciferol/administration & dosage , Cholecalciferol/metabolism , Furunculosis/immunology , Gene Expression , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Infectious pancreatic necrosis virus/physiology , Lipids/administration & dosage , Phenylbutyrates/administration & dosage , Phenylbutyrates/metabolism , Valerates/administration & dosage , Valerates/metabolism
19.
J Inorg Biochem ; 177: 1-7, 2017 12.
Article in English | MEDLINE | ID: mdl-28918353

ABSTRACT

Five new platinum(IV) derivatives of carboplatin each incorporating the histone deacetylase inhibitor 4-phenylbutyrate in axial position were synthesized and characterized by 1H and 195Pt NMR spectroscopy, electrospray ionization mass spectrometry and elemental analysis, namely cis,trans-[Pt(CBDCA)(NH3)2(PBA)(OH)] (1), cis,trans-[Pt(CBDCA)(NH3)2(PBA)2] (2), cis,trans-[Pt(CBDCA)(NH3)2(PBA)(bz)] (3), cis,trans-[Pt(CBDCA)(NH3)2(PBA)(suc)] (4) and cis,trans-[Pt(CBDCA)(NH3)2)(PBA)(ac)] (5) (PBA=4-phenylbutyrate, CBDCA=1,1-cyclobutane dicarboxylate, bz=benzoate, suc=succinate and ac=acetate). The reduction behavior in the presence of ascorbic acid was studied by high performance liquid chromatography. The cytotoxicity against a panel of human tumor cell lines, histone deacetylase (HDAC) inhibitory activity, cellular accumulation and the ability to induce apoptosis were evaluated. The most effective complex, compound 3, was found to be up to ten times more effective than carboplatin and to decrease cellular basal HDAC activity by approximately 18% in A431 human cervical cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carboplatin/analogs & derivatives , Carboplatin/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Phenylbutyrates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Carboplatin/chemical synthesis , Carboplatin/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Humans , Inhibitory Concentration 50 , Phenylbutyrates/chemical synthesis , Phenylbutyrates/metabolism
20.
Virus Res ; 242: 24-29, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28916365

ABSTRACT

African swine fever virus (ASFV) causes a highly lethal disease in swine for which neither a vaccine nor treatment are available. Recently, a new class of drugs that inhibit histone deacetylases enzymes (HDACs) has received an increasing interest as antiviral agents. Considering studies by others showing that valproic acid, an HDAC inhibitor (HDACi), blocks the replication of enveloped viruses and that ASFV regulates the epigenetic status of the host cell by promoting heterochromatinization and recruitment of class I HDACs to viral cytoplasmic factories, the antiviral activity of four HDACi against ASFV was evaluated in this study. Results showed that the sodium phenylbutyrate fully abrogates the ASFV replication, whereas the valproic acid leads to a significant reduction of viral progeny at 48h post-infection (-73.9%, p=0.046), as the two pan-HDAC inhibitors tested (Trichostatin A: -82.2%, p=0.043; Vorinostat: 73.9%, p=0.043). Further evaluation showed that protective effects of NaPB are dose-dependent, interfering with the expression of late viral genes and reversing the ASFV-induced histone H3 lysine 9 and 14 (H3K9K14) hypoacetylation status, compatible to an open chromatin state and possibly enabling the expression of host genes non-beneficial to infection progression. Additionally, a synergic antiviral effect was detected when NaPB is combined with an ASFV-topoisomerase II poison (Enrofloxacin). Altogether, our results strongly suggest that cellular HDACs are involved in the establishment of ASFV infection and emphasize that further in vivo studies are needed to better understand the antiviral activity of HDAC inhibitors.


Subject(s)
African Swine Fever Virus/drug effects , African Swine Fever Virus/physiology , Antiviral Agents/metabolism , Histones/metabolism , Phenylbutyrates/metabolism , Protein Processing, Post-Translational/drug effects , Virus Replication/drug effects , Acetylation/drug effects , Animals , Chlorocebus aethiops , Histone Deacetylase Inhibitors/metabolism , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL