Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 268
Filter
1.
Nutrients ; 14(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35276934

ABSTRACT

Hyperuricemia, a lifestyle-related disease characterized by elevated serum urate levels, is the main risk factor for gout; therefore, the serum urate-lowering effects of human diets or dietary ingredients have attracted widespread interest. As Urate transporter 1 (URAT1) governs most urate reabsorption from primary urine into blood, URAT1 inhibition helps decrease serum urate levels by increasing the net renal urate excretion. In this study, we used a cell-based urate transport assay to investigate the URAT1-inhibitory effects of 162 extracts of plant materials consumed by humans. Among these, we focused on Aspalathus linearis, the source of rooibos tea, to explore its active ingredients. Using liquid-liquid extraction with subsequent column chromatography, as well as spectrometric analyses for chemical characterization, we identified quercetin as a URAT1 inhibitor. We also investigated the URAT1-inhibitory activities of 23 dietary ingredients including nine flavanols, two flavanonols, two flavones, two isoflavonoids, eight chalcones, and a coumarin. Among the tested authentic chemicals, fisetin and quercetin showed the strongest and second-strongest URAT1-inhibitory activities, with IC50 values of 7.5 and 12.6 µM, respectively. Although these effects of phytochemicals should be investigated further in human studies, our findings may provide new clues for using nutraceuticals to promote health.


Subject(s)
Aspalathus , Organic Anion Transporters , Health Promotion , Humans , Organic Anion Transporters/physiology , Plant Leaves , Polyphenols , Uric Acid
2.
Dev Cell ; 56(19): 2741-2751.e7, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34610327

ABSTRACT

Cancer cachexia is associated with many types of tumors and is characterized by a combination of anorexia, loss of body weight, catabolic alterations, and systemic inflammation. We developed a tumor model in Drosophila larvae that causies cachexia-like syndrome, and we found that cachectic larvae show reduced levels of the circulating steroid ecdysone (Ec). Artificially importing Ec in the tumor through the use of the EcI/Oatp74D importer aggravated cachexia, whereas feeding animals with Ec rescued cachectic defects. This suggests that a steroid sink induced by the tumor promotes catabolic alterations in healthy tissues. We found that Oatp33Eb, a member of the Oatp transporter family, is specifically induced in tumors promoting cachexia. The overexpression of Oatp33Eb in noncachectic tumors induced cachexia, whereas its inhibition in cachectic tumors restored circulating Ec and reversed cachectic alterations. Oatp transporters are induced in several types of hormone-dependent tumors, and this result suggests that a similar sink effect could modify hormonal balance in cachectic cancer patients.


Subject(s)
Cachexia/metabolism , Ecdysone/metabolism , Organic Anion Transporters/metabolism , Animals , Body Weight , Cachexia/physiopathology , Drosophila Proteins , Drosophila melanogaster , Larva/metabolism , Neoplasms , Organic Anion Transporters/physiology , Steroids/metabolism
3.
Biomolecules ; 11(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33499062

ABSTRACT

The mitochondrial citrate/isocitrate carrier, CIC, has been shown to play an important role in a growing list of human diseases. CIC belongs to a large family of nuclear-encoded mitochondrial transporters that serve the fundamental function of allowing the transit of ions and metabolites through the impermeable mitochondrial membrane. Citrate is central to mitochondrial metabolism and respiration and plays fundamental activities in the cytosol, serving as a metabolic substrate, an allosteric enzymatic regulator and, as the source of Acetyl-Coenzyme A, also as an epigenetic modifier. In this review, we highlight the complexity of the mechanisms of action of this transporter, describing its involvement in human diseases and the therapeutic opportunities for targeting its activity in several pathological conditions.


Subject(s)
Citrates/metabolism , Inflammation/metabolism , Mitochondrial Proteins/physiology , Neoplasms/metabolism , Organic Anion Transporters/physiology , Allosteric Site , Animals , Chromosomes, Human, Pair 22/metabolism , Citric Acid , Cytosol/metabolism , Diabetes Mellitus/metabolism , Epigenesis, Genetic , Humans , Liver Diseases/metabolism , Metabolic Diseases/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Phosphorylation
4.
Neuromolecular Med ; 23(2): 267-284, 2021 06.
Article in English | MEDLINE | ID: mdl-32865657

ABSTRACT

Intracerebral hemorrhage (ICH) is a devastating disease with high rates of mortality and morbidity. Galactose lectin-9 (Gal-9) belongs to the family of ß-galactoside-binding lectins, which has been shown to play a vital role in immune tolerance and inflammation. However, the function of Gal-9 in ICH has not been fully studied in details. Several experiments were carried out to explore the role of Gal-9 in the late period of ICH. Primarily, ICH models were established in male adult Sprague Dawley (SD) rats. Next, the relative protein levels of Gal-9 at different time points after ICH were examined and the result showed that the level of Gal-9 increased and peaked at the 7th day after ICH. Then we found that when the content of Gal-9 increased, both the number of M2-type microglia and the corresponding anti-inflammatory factors also increased. Through co-immunoprecipitation (CO-IP) analysis, it was found that Gal-9 combines with Toll-like receptor-4 (TLR-4) during the period of the recovery after ICH. TUNEL staining and Fluoro-Jade B staining (FJB) proved that the amount of cell death decreased with the increase of Gal-9 content. Additionally, several behavioral experiments also demonstrated that when the level of Gal-9 increased, the motor, sensory, learning, and memory abilities of the rats recovered better compared to the ICH group. In short, this study illustrated that Gal-9 takes a crucial role after ICH. Enhancing Gal-9 could alleviate brain injury and promote the recovery of ICH-induced injury, so that Gal-9 may exploit a new pathway for clinical treatment of ICH.


Subject(s)
Cerebral Hemorrhage/physiopathology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Organic Anion Transporters/physiology , Toll-Like Receptor 4/physiology , Animals , Apoptosis , Astrocytes/metabolism , Basal Ganglia/physiopathology , Disease Models, Animal , Male , Microglia/metabolism , Morris Water Maze Test , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/pathology , Organic Anion Transporters/biosynthesis , Organic Anion Transporters/genetics , Protein Binding , Protein Interaction Mapping , Psychomotor Performance , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Recovery of Function , Rotarod Performance Test , Signal Transduction , Single-Blind Method , Time Factors
5.
Plant Cell Environ ; 43(10): 2443-2459, 2020 10.
Article in English | MEDLINE | ID: mdl-32666573

ABSTRACT

Malate exudation through wheat (Triticum aestivum L) aluminium-activated malate transporter 1 (TaALMT1) confers Al3+ tolerance at low pH, but is also activated by alkaline pH, and is regulated by and facilitates significant transport of gamma-aminobutyric acid (GABA, a zwitterionic buffer). Therefore, TaALMT1 may facilitate acidification of an alkaline rhizosphere by promoting exudation of both malate and GABA. Here, the performance of wheat near isogenic lines ET8 (Al+3 -tolerant, high TaALMT1 expression) and ES8 (Al+3 -sensitive, low TaALMT1 expression) are compared. Root growth (at 5 weeks) was higher for ET8 than ES8 at pH 9. ET8 roots exuded more malate and GABA at high pH and acidified the rhizosphere more rapidly. GABA and malate exudation was enhanced at high pH by the addition of aluminate in both ET8 and transgenic barley expressing TaALMT1. Xenopus laevis oocytes expressing TaALMT1 acidified an alkaline media more rapidly than controls corresponding to higher GABA efflux. TaALMT1 expression did not change under alkaline conditions but key genes involved in GABA turnover changed in accordance with a high rate of GABA synthesis. We propose that TaALMT1 plays a role in alkaline tolerance by exuding malate and GABA, possibly coupled to proton efflux.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Malates/metabolism , Organic Anion Transporters/metabolism , Plant Proteins/metabolism , Triticum/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Genetically Modified , Chlorophyll/metabolism , GABA Plasma Membrane Transport Proteins/physiology , Hordeum , Hydrogen-Ion Concentration , Oocytes , Organic Anion Transporters/physiology , Plant Leaves/metabolism , Plant Proteins/physiology , Plant Roots/metabolism , Plant Roots/physiology , Plants, Genetically Modified , Rhizosphere , Seedlings/metabolism , Seedlings/physiology , Stress, Physiological , Triticum/physiology , Xenopus
6.
Nutrients ; 12(6)2020 May 29.
Article in English | MEDLINE | ID: mdl-32486008

ABSTRACT

The beneficial effects of fatty acids (FAs) on human health have attracted widespread interest. However, little is known about the impact of FAs on the handling of urate, the end-product of human purine metabolism, in the body. Increased serum urate levels occur in hyperuricemia, a disease that can lead to gout. In humans, urate filtered by the glomerulus of the kidney is majorly re-absorbed from primary urine into the blood via the urate transporter 1 (URAT1)-mediated pathway. URAT1 inhibition, thus, contributes to decreasing serum urate concentration by increasing net renal urate excretion. Here, we investigated the URAT1-inhibitory effects of 25 FAs that are commonly contained in foods or produced in the body. For this purpose, we conducted an in vitro transport assay using cells transiently expressing URAT1. Our results showed that unsaturated FAs, especially long-chain unsaturated FAs, inhibited URAT1 more strongly than saturated FAs. Among the tested unsaturated FAs, eicosapentaenoic acid, α-linolenic acid, and docosahexaenoic acid exhibited substantial URAT1-inhibitory activities, with half maximal inhibitory concentration values of 6.0, 14.2, and 15.2 µM, respectively. Although further studies are required to investigate whether the ω-3 polyunsaturated FAs can be employed as uricosuric agents, our findings further confirm FAs as nutritionally important substances influencing human health.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Kidney Glomerulus/metabolism , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/physiology , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/physiology , Renal Reabsorption/drug effects , Uric Acid/metabolism , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Eicosapentaenoic Acid/pharmacology , Humans , Hyperuricemia/blood , Renal Elimination/drug effects , Uric Acid/blood , alpha-Linolenic Acid/pharmacology
7.
Cell Physiol Biochem ; 54(4): 538-555, 2020 May 23.
Article in English | MEDLINE | ID: mdl-32442363

ABSTRACT

BACKGROUND/AIMS: Maxi-anion channel (Maxi-Cl) is ubiquitously expressed and involved in a number of important cell functions especially by serving as an ATP release pathway. We recently identified SLCO2A1 as its essential core component. However, the regulatory component required for the channel activation/inactivation remains unidentified. METHODS: In the present study, to identify the regulatory component, we made genome-wide analysis combined with siRNA screening and performed patch-clamp studies and ATP release assay after gene silencing and overexpression. RESULTS: Comparative microarray analysis between Maxi-Cl-rich C127 and -deficient C1300 cells revealed highly differential expression not only of SLCO2A1 but also of four annexin family members. Gene silencing study showed that Anxa2 is involved in Maxi-Cl activity. The Maxi-Cl events appeared in C1300 cells by overexpression of Slco2a1 and more efficiently by that of Slco2a1 plus Anxa2. Immunoprecipitation assay supported the interaction between ANXA2 and SLCO2A1. Suppressive effects of overexpression of a phospho-mimicking mutant of Anxa2, Anxa2-Y23E, indicated that protein tyrosine dephosphorylation dependence of Maxi-Cl is conferred by ANXA2. Maxi-Cl activity was suppressed by gene silencing of S100A10, a binding partner of ANXA2, and by applying a synthetic ANXA2 peptide, Ac-(1-14), which interferes with the ANXA2-S100A10 complex formation. Intracellular Ca2+ dependence of Maxi-Cl activity was abolished by S100a10 knockdown. CONCLUSION: The ANXA2-S100A10 complex represents the regulatory component of Maxi-Cl conferring protein tyrosine dephosphorylation dependence and intracellular Ca2+ sensitivity on this channel.


Subject(s)
Annexin A2/metabolism , Calcium/metabolism , Organic Anion Transporters/metabolism , S100 Proteins/metabolism , Tyrosine/metabolism , Animals , Anions , Annexin A2/genetics , Cell Line, Tumor , Gene Silencing , HEK293 Cells , Humans , Mice , Oligonucleotide Array Sequence Analysis , Organic Anion Transporters/genetics , Organic Anion Transporters/physiology , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , S100 Proteins/genetics , Up-Regulation
8.
Thyroid ; 30(1): 161-174, 2020 01.
Article in English | MEDLINE | ID: mdl-31797746

ABSTRACT

Background: The thyroid hormones (THs) triiodothyronine (T3) and thyroxine (T4) are crucial regulators of brain development and function. Cell-specific transporter proteins facilitate TH uptake and efflux across the cell membrane, and insufficient TH transport causes hypothyroidism and mental retardation. Mutations in the TH transporters monocarboxylate transporter 8 (MCT8, SLC16A2) and the organic anion-transporting polypeptide 1C1 (OATP1C1, SLCO1C1) are associated with the psychomotor retardation Allan-Herndon-Dudley syndrome and juvenile neurodegeneration, respectively. Methods: To understand the mechanisms and test potential treatments for the recently discovered OATP1C1 deficiency, we established an oatp1c1 mutant (oatp1c1-/-) zebrafish. Results:oatp1c1 is expressed in endothelial cells, neurons, and astrocytes in zebrafish. The activity of the hypothalamic-pituitary-thyroid axis and behavioral locomotor activity increased in oatp1c1-/- larvae. Neuropathological analysis revealed structural alteration in radial glial cells and shorter neuronal axons in oatp1c1-/- larvae and adults. Notably, oatp1c1-/- and oatp1c1-/-Xmct8-/- adults exhibit an enlarged thyroid gland (goiter). Pharmacological assays showed that TH analogs, but not THs, can reduce the size and improve the color of the thyroid gland in adult mutant zebrafish. Conclusion: These results establish a vertebrate model for OATP1C1 deficiency that demonstrates endocrinological, neurological, and behavioral alterations mimicking findings observed in an OATP1C1-deficient patient. Further, the curative effect of TH analogs in the oatp1c1-/- zebrafish model may provide a lead toward a treatment modality in human patients.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Mutation , Neurons/physiology , Organic Anion Transporters/genetics , Thyroid Gland/physiology , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Astrocytes/metabolism , Behavior, Animal , Brain/metabolism , Cell Membrane/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Gene Knockout Techniques , Microscopy, Fluorescence , Organic Anion Transporters/deficiency , Organic Anion Transporters/physiology , Zebrafish , Zebrafish Proteins/physiology
9.
Endocr Rev ; 41(2)2020 04 01.
Article in English | MEDLINE | ID: mdl-31754699

ABSTRACT

Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).


Subject(s)
Biological Transport/physiology , Membrane Transport Proteins/physiology , Mental Retardation, X-Linked , Monocarboxylic Acid Transporters/physiology , Muscle Hypotonia , Muscular Atrophy , Organic Anion Transporters/physiology , Symporters/physiology , Thyroid Hormones/metabolism , Animals , Humans , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/therapy , Monocarboxylic Acid Transporters/deficiency , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/genetics , Muscle Hypotonia/metabolism , Muscle Hypotonia/physiopathology , Muscle Hypotonia/therapy , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Muscular Atrophy/therapy , Organic Anion Transporters/deficiency , Organic Anion Transporters/genetics , Symporters/deficiency , Symporters/genetics , Thyroid Hormones/therapeutic use
10.
Microb Cell Fact ; 18(1): 154, 2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31506101

ABSTRACT

BACKGROUND: Mitochondrial and cytoplasmic malate transporter proteins are responsible for transmembrane transport of malate, thereby linking malate metabolism in various subcellular regions of the cell. These transporters play an important role in fatty acid biosynthesis of oleaginous microorganisms. Our previous studies have found that lipid content of the recombinant Mucor circinelloides (M. circinelloides) strain with mitochondrial malate transporter (mt) gene overexpression was increased by 70%, while that of strain with mt gene knockout was decreased by 27%. However, the mechanism of malate transporter promoting the transport of mitochondrial malate and citrate related to lipid accumulation is not clear. Therefore, 13C-labeled glucose metabolic flux analysis was carried out to identify the metabolic network topology and estimate intracellular fluxes of genetically engineered M. circinelloides strains for the purpose of better understanding the roles of malate transporters in citrate transport systems and lipid accumulation. RESULTS: The metabolic flux distribution analysis suggested that tricarboxylic acid (TCA) cycle flux ratio of mt-overexpression strains was decreased compared to that of the control strain, but in contrast, glyoxylic acid (GOX) cycle flux ratio was increased. Accordingly, the mt-knockout strain showed an opposite phenomenon with a higher TCA cycle flux ratio and a lower GOX cycle flux ratio than the control strain. GOX cycle might be more effective than TCA cycle in producing malate and oxaloacetate replenishment. Moreover, a relatively higher flux ratio of the pentose phosphate (PP) pathway was obtained in mt-overexpression strains, but no significant difference in the malic enzyme flux between recombinant strains and the control strain. Our results confirmed that PP pathway might play an important role for supplying NADPH and malic enzyme is not a limiting factor for fatty acid synthesis in oleaginous fungus M. circinelloides strains. CONCLUSION: Intracellular metabolic flux information suggested that mt-overexpression strains had higher flux in PP pathway and GOX cycle, lower flux in TCA cycle, and no difference in malic enzyme cycle. Together, the role of malate transporter was assumed to further participate in transporting cycle of acetyl-CoA and drive PP pathway to supply NADPH required for lipid accumulation in recombinant M. circinelloides strains.


Subject(s)
Lipid Metabolism , Malates/metabolism , Mucor/metabolism , Organic Anion Transporters/physiology , Biological Transport , Glyoxylates/metabolism , Metabolic Flux Analysis/methods , Pentose Phosphate Pathway
11.
Drug Metab Dispos ; 47(11): 1343-1351, 2019 11.
Article in English | MEDLINE | ID: mdl-31519697

ABSTRACT

Both nonsteroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids have been widely used for the treatment of gout, a disease promoted by an excess body burden of uric acid (UA); however, their effects on the homeostasis of UA remain poorly understood. The present study showed that 1-week treatments with three NSAIDs (ibuprofen, diclofenac, and indomethacin) had little effect on UA homeostasis in mice, whereas 1-week low doses (1 and 5 mg/kg) of dexamethasone (DEX) significantly decreased serum UA by about 15%. Additionally, low doses of DEX also resulted in increases in hepatic UA concentration and urinary UA excretion, which were associated with an induction of xanthine oxidoreductase (XOR) in the liver and a downregulation of urate transporter 1 (URAT1) in the kidney, respectively. Neither 75 mg/kg DEX nor 100 mg/kg pregnenolone-16α-carbonitrile altered UA concentrations in serum and livers of mice, suggesting that the effect of DEX on UA homeostasis was not due to the pregnane X receptor pathway. Further in vitro studies demonstrated that glucocorticoid receptor (GR) was involved in DEX-mediated downregulation of URAT1. Knockdown of both p65 and c-Jun completely blocked the effect of DEX on URAT1, suggesting that GR regulates URAT1 via its interaction with both nuclear factor κB and activator protein 1 signaling pathways. To conclude, the present study identifies, for the first time, a critical role of glucocorticoids in regulating UA homeostasis and elucidates the mechanism for GR-mediated regulation of URAT1 in mice. SIGNIFICANCE STATEMENT: This study demonstrates, for the first time, a critical role of glucocorticoid receptor in regulating urate transporter 1 in mouse kidney.


Subject(s)
Dexamethasone/pharmacology , Kidney/metabolism , Organic Anion Transporters/genetics , Uric Acid/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Down-Regulation , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Organic Anion Transporters/physiology , Pregnane X Receptor/physiology , Receptors, Glucocorticoid/physiology , Signal Transduction/physiology , Xanthine Dehydrogenase/physiology
12.
Hepatology ; 70(6): 2156-2170, 2019 12.
Article in English | MEDLINE | ID: mdl-31102415

ABSTRACT

Organic anion transport proteins (OATPs) on the basolateral surface of hepatocytes mediate uptake of a number of drugs and endogenous compounds. Previous studies showed that rat OATP1A1 (rOATP1A1) has a postsynaptic density protein, drosophila disc large tumor suppressor, zonula occludens-1 protein (PDZ) consensus binding motif at its C-terminus and binds to PDZ domain containing 1 (PDZK1), which is required for its cell-surface localization. PDZK1 associates with rOATP1A1-containing endocytic vesicles within cells, mediating recruitment of motor proteins required for microtubule-based trafficking to the plasma membrane. rOATP1A4 also traffics to the plasma membrane, although it lacks a PDZ binding consensus sequence. The current study was designed to test the hypothesis that trafficking of rOATP1A4 to the plasma membrane requires its direct interaction with rOATP1A1 resulting in a complex that traffics through the cell in common subcellular vesicles in which the cytosolic tail of rOATP1A1 is bound to PDZK1. We found that 74% of rOATP1A4-containing rat liver endocytic vesicles (n = 12,044) also contained rOATP1A1. Studies in transfected HEK293 cells showed surface localization of rOATP1A1 only when coexpressed with PDZK1 whereas rOATP1A4 required coexpression with rOATP1A1 and PDZK1. Studies in stably transfected HeLa cells that constitutively expressed PDZK1 showed that coexpression of rOATP1A4 with rOATP1A1 resulted in more rapid appearance of rOATP1A4 on the plasma membrane and faster maturation to its fully glycosylated form. Similar results were observed on immunofluorescence analysis of single cells. Immunoprecipitation of rat liver or transfected HeLa cell lysates with rOATP1A1 antibody specifically co-immunoprecipitated rOATP1A4 as determined by western blotting. Conclusion: These studies indicate that optimal rOATP1A4 trafficking to the cell surface is dependent upon coexpression and interaction with rOATP1A1. As rOATP1A1 binds to the chaperone protein, PDZK1, rOATP1A4 functionally hitchhikes through the cell with this complex.


Subject(s)
Hepatocytes/metabolism , Organic Anion Transporters, Sodium-Independent/physiology , Organic Anion Transporters/physiology , Protein Transport , Animals , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Fluorescent Antibody Technique , HEK293 Cells , HeLa Cells , Humans , Rats
13.
Clin J Am Soc Nephrol ; 14(4): 540-548, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30885911

ABSTRACT

BACKGROUND AND OBJECTIVES: The neonatal and juvenile human kidney can be exposed to a variety of potentially toxic drugs (e.g., nonsteroidal anti-inflammatory drugs, antibiotics, antivirals, diuretics), many of which are substrates of the kidney organic anion transporters, OAT1 (SLC22A6, originally NKT) and OAT3 (SLC22A8). Despite the immense concern about the consequences of drug toxicity in this vulnerable population, the developmental regulation of OATs in the immature postnatal kidney is poorly understood. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Recognizing that today it is difficult to obtain rich data on neonatal kidney handling of OAT probes due to technical, logistic, and ethical considerations, multiple older physiologic studies that used the prototypical organic anion substrate para-aminohippurate (PAH) were reanalyzed in order to provide a quantitative description of OAT-mediated tubular secretion across the pediatric age continuum. Parametric and semiparametric models were evaluated for kidney function outcome variables of interest (maximum tubular secretory capacity of PAH [TmPAH], effective renal plasma flow [ERPF], and GFR). RESULTS: Data from 119 neonates, infants, and children ranging in age from 1 day to 11.8 years were used to fit TmPAH, ERPF, and GFR as functions of postnatal age. TmPAH is low in the immediate postnatal period and increases markedly after birth, reaching 50% of the adult value (80 mg/min) at 8.3 years of age. During the first 2 years of life, TmPAH is lower than that of GFR when viewed as the fraction of the adult value. CONCLUSIONS: During postnatal human kidney development, proximal tubule secretory function-as measured using PAH, a surrogate for OAT-mediated secretion of organic anion drugs, metabolites, and toxins-is low initially but increases rapidly. Despite developmental differences between species, this overall pattern is roughly consistent with animal studies. The human data raise the possibility that the acquisition of tubular secretory function may not closely parallel glomerular filtration.


Subject(s)
Kidney Tubules, Proximal/metabolism , Kidney/growth & development , Organic Anion Transporters/physiology , Child , Child, Preschool , Humans , Infant , Infant, Newborn
14.
Gene ; 693: 69-75, 2019 Apr 20.
Article in English | MEDLINE | ID: mdl-30708027

ABSTRACT

The human SLC25A13 gene encodes the liver type aspartate/glutamate carrier isoform 2 (AGC2, commonly named as citrin), which plays a key role in the main NADH-shuttle of human hepatocyte. Biallelic SLC25A13 mutations result in Citrin deficiency (CD). In order to identify the important regulatory region of SLC25A13 gene and elucidate the way how potential promoter mutations affect the citrin expression, we performed promoter deletion analysis and established the reporter constructs of luciferase gene-carrying SLC25A13 promoter containing several mutations located in putative transcription factor-binding sites. The luciferase activities of all promoter constructs were measured using a Dual-Luciferase Reporter Assay System. Bioinformatic analysis showed that the promoter of SLC25A13 gene lacks TATA box and obviously typical initiator element, but contains a CCAAT box and two GC box. Promoter deletion analysis confirmed the region from -221 to -1 upstream ATG was essential for SLC25A13 to maintain the promoter activity. We utilized dual-luciferase reporter system as function analytical model to tentatively assess the effect of artificially constructed promoter mutations on citrin expression, and our analysis revealed that mutated putative CCAAT box and GC box could significantly affect the citrin expression. Our study confirmed the important SLC25A13 promoter regions that influenced citrin expression in HL7702 cells, and constructed a function analytical model. This work may be useful to further identify the pathogenic mutations leading to CD in the promoter region.


Subject(s)
Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Base Sequence , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Computational Biology , Humans , Mutation , Organic Anion Transporters/genetics , Organic Anion Transporters/physiology , Promoter Regions, Genetic/genetics
15.
Nutrients ; 10(10)2018 Sep 28.
Article in English | MEDLINE | ID: mdl-30274153

ABSTRACT

Chrysanthemum indicum Linne flower (CF) and Cinnamomum cassia (L.) J. Presl bark (CB) extracts have been used as the main ingredients in several prescriptions to treat the hyperuricemia and gout in traditional medicine. In the present study, we investigated the antihyperuricemic effects of DKB114, a CF, and CB mixture, and the underlying mechanisms in vitro and in vivo. DKB114 markedly reduced serum uric acid levels in normal rats and rats with PO-induced hyperuricemia, while increasing renal uric acid excretion. Furthermore, it inhibited the activity of xanthine oxidase (XOD) in vitro and in the liver in addition to reducing hepatic uric acid production. DKB114 decreased cellular uric acid uptake in oocytes and HEK293 cells expressing human urate transporter (hURAT)1 and decreased the protein expression levels of urate transporters, URAT1, and glucose transporter, GLUT9, associated with the reabsorption of uric acid in the kidney. DKB114 exerts antihyperuricemic effects and uricosuric effects, which are accompanied, partially, by a reduction in the production of uric acid and promotion of uric acid excretion via the inhibition of XOD activity and reabsorption of uric acid. Therefore, it may have potential as a treatment for hyperuricemia and gout.


Subject(s)
Chrysanthemum/chemistry , Cinnamomum/chemistry , Hyperuricemia/drug therapy , Plant Extracts/administration & dosage , Uric Acid/urine , Xanthine Oxidase/antagonists & inhibitors , Animals , Enzyme Inhibitors/pharmacology , Flowers/chemistry , Gene Expression , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/physiology , HEK293 Cells , Hep G2 Cells , Humans , Liver/chemistry , Male , Membrane Potential, Mitochondrial/drug effects , Oocytes/drug effects , Oocytes/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/physiology , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/physiology , Plant Bark/chemistry , Plant Extracts/toxicity , Rats , Rats, Sprague-Dawley , Transfection , Urate Oxidase/antagonists & inhibitors , Uric Acid/analysis , Uric Acid/metabolism
16.
Gastroenterology ; 155(5): 1578-1592.e16, 2018 11.
Article in English | MEDLINE | ID: mdl-30063921

ABSTRACT

BACKGROUND & AIMS: Bile acid transporters maintain bile acid homeostasis. Little is known about the functions of some transporters in cholestasis or their regulatory mechanism. We investigated the hepatic expression of solute carrier organic anion transporter family member 3A1 (SLCO3A1, also called OATP3A1) and assessed its functions during development of cholestasis. METHODS: We measured levels of OATP3A1 protein and messenger RNA and localized the protein in liver tissues from 22 patients with cholestasis and 21 patients without cholestasis, using real-time quantitative polymerase chain reaction, immunoblot, and immunofluorescence analyses. We performed experiments with Slco3a1-knockout and C57BL/6J (control) mice. Mice and Sprague-Dawley rats underwent bile duct ligation (BDL) or a sham operation. Some mice were placed on a 1% cholic acid (CA) diet to induce cholestasis or on a control diet. Serum and liver tissues were collected and analyzed; hepatic levels of bile acids and 7-α-C4 were measured using liquid chromatography/mass spectrometry. Human primary hepatocytes and hepatoma (PLC/PRF/5) cell lines were used to study mechanisms that regulate OATP3A1 expression and transport. RESULTS: Hepatic levels of OATP3A1 messenger RNA and protein were significantly increased in liver tissues from patients with cholestasis and from rodents with BDL or 1% CA diet-induced cholestasis. Levels of fibroblast growth factor 19 (FGF19, FGF15 in rodents) were also increased in liver tissues from patients and rodents with cholestasis. FGF19 signaling activated the Sp1 transcription factor and nuclear factor κB to increase expression of OATP3A1 in hepatocytes; we found binding sites for these factors in the SLCO3A1 promoter. Slco3a1-knockout mice had shorter survival times and increased hepatic levels of bile acid, and they developed more liver injury after the 1% CA diet or BDL than control mice. In hepatoma cell lines, we found OATP3A1 to take prostaglandin E2 and thyroxine into cells and efflux bile acids. CONCLUSIONS: We found levels of OATP3A1 to be increased in cholestatic liver tissues from patients and rodents compared with healthy liver tissues. We show that OATP3A1 functions as a bile acid efflux transporter that is up-regulated as an adaptive response to cholestasis.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/metabolism , Organic Anion Transporters/physiology , Animals , Extracellular Signal-Regulated MAP Kinases/physiology , Fibroblast Growth Factors/analysis , Fibroblast Growth Factors/physiology , Humans , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Organic Anion Transporters/analysis , Rats , Rats, Sprague-Dawley , Signal Transduction , Sp1 Transcription Factor/physiology , Transcription Factor RelA/physiology
17.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 43(3): 327-332, 2018 Mar 28.
Article in Chinese | MEDLINE | ID: mdl-29701197

ABSTRACT

Pharmacokinetics plays a key role in rational use of medicines. Many factors can affect the drug's pharmacokinetics. Previous studies mainly focused on the impact of hypoxia on hepatic drug metabolizing enzyme, but uncommon on drug transporters. Actually, drug transporter is a key factor for activation of the drugs transport across the cell membrane into the inside of cells, such as multidrug resistance protein (MDR), breast cancer resistance protein (BCRP), multidrug resistance associated protein (MRP), organic cation transporter (OCT), organic anion-transporting polypeptide (OATP), organic anion transporter (OAT), qligopeptide transporter (PEPT), etc. They are widely present in the small intestine villus epithelial cells, renal tubular epithelial cells, hepatocytes and biliary epithelial cells. They play a very important role in drug absorption, distribution, metabolism and excretion. The changes in drug transporters under hypoxia in intestinal could affect the bioavailability of drugs; the changes in drug transporters in organs could affect drug's distribution, subsequent drug's indications and adverse reactions; the changes in drug transporters in liver and kidney could affect the metabolism and excretion rate of drugs, thereby the drug's residence time and half-life.


Subject(s)
Altitude , Membrane Transport Proteins/physiology , Pharmacokinetics , Cell Hypoxia/physiology , Hepatocytes , Humans , Multidrug Resistance-Associated Proteins/physiology , Organic Anion Transporters/physiology , Organic Cation Transport Proteins/physiology
18.
AAPS J ; 20(1): 13, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29204966

ABSTRACT

Solute carrier organic anion transporter family member 2A1 (OATP2A1, encoded by the SLCO2A1 gene), which was initially identified as prostaglandin transporter (PGT), is expressed ubiquitously in tissues and mediates the distribution of prostanoids, such as PGE2, PGF2α, PGD2 and TxB2. It is well known to play a key role in the metabolic clearance of prostaglandins, which are taken up into the cell by OATP2A1 and then oxidatively inactivated by 15-ketoprostaglandin dehydrogenase (encoded by HPGD); indeed, OATP2A1-mediated uptake is the rate-limiting step of PGE2 catabolism. Consequently, since OATP2A1 activity is required for termination of prostaglandin signaling via prostanoid receptors, its inhibition can enhance such signaling. On the other hand, OATP2A1 can also function as an organic anion exchanger, mediating efflux of prostaglandins in exchange for import of anions such as lactate, and in this context, it plays a role in the release of newly synthesized prostaglandins from cells. These different functions likely operate in different compartments within the cell. OATP2A1 is reported to function at cytoplasmic vesicle/organelle membranes. As a regulator of the levels of physiologically active prostaglandins, OATP2A1 is implicated in diverse physiological and pathophysiological processes in many organs. Recently, whole exome analysis has revealed that recessive mutations in SLCO2A1 cause refractory diseases in humans, including primary hypertrophic osteoarthropathy (PHO) and chronic non-specific ulcers in small intestine (CNSU). Here, we review and summarize recent information on the molecular functions of OATP2A1 and on its physiological and pathological significance.


Subject(s)
Organic Anion Transporters/physiology , Prostaglandins/physiology , Drug Interactions , Endothelial Cells/physiology , Humans , Kidney/metabolism , Lung/metabolism , Organic Anion Transporters/antagonists & inhibitors
19.
Curr Atheroscler Rep ; 19(12): 65, 2017 Nov 25.
Article in English | MEDLINE | ID: mdl-29177971

ABSTRACT

PURPOSE OF REVIEW: Statin drug-drug interactions (DDIs) are both troublesome to patients as well as costly to medical resources. The ability to predict and avoid these events could lead to improved outcomes as well as patient satisfaction. This review will explore efforts to better understand and predict these interactions specifically related to one drug transport system, the organic anion-transporting polypeptides (OATPs) specifically OATP1B1 and OATP1B3. RECENT FINDINGS: Since the publication of the discovery of OATPs, there have been various pharmacokinetic models that have been proposed to explain the variation in pharmacokinetic and clinical effects related to the OATPs. The effects in transport activity appear to be partially related to the individual polymorphisms studied. Drug-drug interactions can occur when other drugs compete for the metabolic site on the OATPs. Various medications are identified as substrates and/or inhibitors of the OATPs, thereby complicating the ability to fully predict the impact on levels and effects. All of the models reviewed claim successes but show limited clinical utility. There are specific populations that have been identified, predominately various Asian descendants that require lower doses of statins to avoid adverse events. The concept of attributing these actions to the OATPs has been explored, but current models cannot accurately predict statin blood levels or elimination constants. The current research only points to the differences in the human genome and the single-nucleotide polymorphisms that exist between us. Based upon the currently available studies, there is beginning to be a glimmer in the understanding how different populations respond to statin transport and elimination. Additionally and unfortunately, there are other enzymes to be studied to better predict patient differences. Clearly, there has been much work completed, yet many more questions require answering to better understand these transport proteins.


Subject(s)
Drug Interactions/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Liver/metabolism , Organic Anion Transporters/physiology , Animals , Asian People , Biological Transport , Drug Interactions/ethnology , Humans , White People
20.
AAPS J ; 19(6): 1878-1889, 2017 11.
Article in English | MEDLINE | ID: mdl-29019117

ABSTRACT

In the present investigations, we evaluate in vitro hepatocyte uptake and partitioning for the prediction of in vivo clearance and liver partitioning. Monkeys were intravenously co-dosed with rosuvastatin and bosentan, substrates of the organic anion transporting polypeptides (OATPs), and metformin, a substrate of organic cation transporter 1 (OCT1). Serial plasma and liver samples were collected over time. Liver and plasma unbound fraction was determined using equilibrium dialysis. In vivo unbound partitioning (Kpu,u) for rosuvastatin, bosentan, and metformin, calculated from total concentrations in the liver and plasma, were 243, 553, and 15, respectively. A physiologically based pharmacokinetic monkey model that incorporates active and passive hepatic uptake was developed to fit plasma and liver concentrations. In addition, a two-compartment model was used to fit in vitro hepatic uptake curves in suspended monkey hepatocyte to determine active uptake, passive diffusion, and intracellular unbound fraction parameters. At steady-state in the model, in vitro Kpu,u was determined. The results demonstrated that in vitro values under-predicted in vivo active uptake for rosuvastatin, bosentan, and metformin by 6.7-, 28-, and 1.5-fold, respectively, while passive diffusion was over-predicted. In vivo Kpu,u values were under-predicted from in vitro data by 30-, 79-, and 3-fold. In conclusion, active uptake and liver partitioning in monkeys for OATP substrates were greatly under-predicted from in vitro hepatocyte uptake, while OCT-mediated uptake and partitioning scaled reasonably well from in vitro, demonstrating substrate- and transporter-dependent scaling factors. The combination of in vitro experimental and modeling approaches proved useful for assessing prediction of in vivo intracellular partitioning.


Subject(s)
Liver/metabolism , Organic Anion Transporters/physiology , Organic Cation Transporter 1/physiology , Animals , Bosentan , Macaca fascicularis , Metformin/pharmacokinetics , Models, Biological , Rosuvastatin Calcium/pharmacokinetics , Sulfonamides/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...