Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(2): 101554, 2022 02.
Article in English | MEDLINE | ID: mdl-34973337

ABSTRACT

The mitochondrial pyruvate carrier (MPC) is an inner mitochondrial membrane complex that plays a critical role in intermediary metabolism. Inhibition of the MPC, especially in liver, may have efficacy for treating type 2 diabetes mellitus. Herein, we examined the antidiabetic effects of zaprinast and 7ACC2, small molecules which have been reported to act as MPC inhibitors. Both compounds activated a bioluminescence resonance energy transfer-based MPC reporter assay (reporter sensitive to pyruvate) and potently inhibited pyruvate-mediated respiration in isolated mitochondria. Furthermore, zaprinast and 7ACC2 acutely improved glucose tolerance in diet-induced obese mice in vivo. Although some findings were suggestive of improved insulin sensitivity, hyperinsulinemic-euglycemic clamp studies did not detect enhanced insulin action in response to 7ACC2 treatment. Rather, our data suggest acute glucose-lowering effects of MPC inhibition may be due to suppressed hepatic gluconeogenesis. Finally, we used reporter sensitive to pyruvate to screen a chemical library of drugs and identified 35 potentially novel MPC modulators. Using available evidence, we generated a pharmacophore model to prioritize which hits to pursue. Our analysis revealed carsalam and six quinolone antibiotics, as well as 7ACC1, share a common pharmacophore with 7ACC2. We validated that these compounds are novel inhibitors of the MPC and suppress hepatocyte glucose production and demonstrated that one quinolone (nalidixic acid) improved glucose tolerance in obese mice. In conclusion, these data demonstrate the feasibility of therapeutic targeting of the MPC for treating diabetes and provide scaffolds that can be used to develop potent and novel classes of MPC inhibitors.


Subject(s)
Anion Transport Proteins , Mitochondrial Membrane Transport Proteins , Monocarboxylic Acid Transporters , Obesity , Quinolones , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet , Glucose/metabolism , Mice , Mice, Obese , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Obesity/drug therapy , Obesity/metabolism , Pyruvic Acid/metabolism , Quinolones/pharmacology
2.
Cell Metab ; 33(3): 629-648.e10, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33333007

ABSTRACT

The metabolic rewiring of cardiomyocytes is a widely accepted hallmark of heart failure (HF). These metabolic changes include a decrease in mitochondrial pyruvate oxidation and an increased export of lactate. We identify the mitochondrial pyruvate carrier (MPC) and the cellular lactate exporter monocarboxylate transporter 4 (MCT4) as pivotal nodes in this metabolic axis. We observed that cardiac assist device-induced myocardial recovery in chronic HF patients was coincident with increased myocardial expression of the MPC. Moreover, the genetic ablation of the MPC in cultured cardiomyocytes and in adult murine hearts was sufficient to induce hypertrophy and HF. Conversely, MPC overexpression attenuated drug-induced hypertrophy in a cell-autonomous manner. We also introduced a novel, highly potent MCT4 inhibitor that mitigated hypertrophy in cultured cardiomyocytes and in mice. Together, we find that alteration of the pyruvate-lactate axis is a fundamental and early feature of cardiac hypertrophy and failure.


Subject(s)
Anion Transport Proteins/metabolism , Cardiomegaly/pathology , Heart Failure/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , Cardiomegaly/chemically induced , Cardiomegaly/complications , Heart Failure/etiology , Heart-Assist Devices , Humans , Lactic Acid/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/genetics , Muscle Proteins/antagonists & inhibitors , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pyruvic Acid/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Ventricular Function, Left/physiology
3.
Biol Pharm Bull ; 43(11): 1792-1798, 2020.
Article in English | MEDLINE | ID: mdl-33132325

ABSTRACT

Xanthine and hypoxanthine are intermediate metabolites of uric acid and a source of reactive oxidative species (ROS) by xanthine oxidoreductase (XOR), suggesting that facilitating their elimination is beneficial. Since they are reabsorbed in renal proximal tubules, we investigated their reabsorption mechanism by focusing on the renal uric acid transporters URAT1 and GLUT9, and examined the effect of clinically used URAT1 inhibitor on their renal clearance when their plasma concentration is increased by XOR inhibitor. Uptake study for [3H]xanthine and [3H]hypoxanthine was performed using URAT1- and GLUT9-expressing Xenopus oocytes. Transcellular transport study for [3H]xanthine was carried out using Madin-Darby canine kidney (MDCK)II cells co-expressing URAT1 and GLUT9. In in vivo pharmacokinetic study, renal clearance of xanthine was estimated based on plasma concentration and urinary recovery. Uptake by URAT1- and GLUT9-expressing oocytes demonstrated that xanthine is a substrate of URAT1 and GLUT9, while hypoxanthine is not. Transcellular transport of xanthine in MDCKII cells co-expressing URAT1 and GLUT9 was significantly higher than those in mock cells and cells expressing URAT1 or GLUT9 alone. Furthermore, dotinurad, a URAT1 inhibitor, increased renal clearance of xanthine in rats treated with topiroxostat to inhibit XOR. It was suggested that xanthine is reabsorbed in the same manner as uric acid through URAT1 and GLUT9, while hypoxanthine is not. Accordingly, it is expected that treatment with XOR and URAT1 inhibitors will effectively decrease purine pools in the body and prevent cell injury due to ROS generated during XOR-mediated reactions.


Subject(s)
Anion Transport Proteins/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Monosaccharide Transport Proteins/metabolism , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Xanthine/pharmacokinetics , Animals , Anion Transport Proteins/antagonists & inhibitors , Benzothiazoles/administration & dosage , Dogs , Glucose Transport Proteins, Facilitative/genetics , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Madin Darby Canine Kidney Cells , Models, Animal , Nitriles/administration & dosage , Oocytes , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Pyridines/administration & dosage , Rats , Rats, Wistar , Reactive Oxygen Species , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Renal Elimination/drug effects , Uric Acid/metabolism , Xanthine/blood , Xanthine/metabolism , Xanthine/urine , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/metabolism , Xenopus laevis
4.
Int J Mol Sci ; 21(11)2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32545297

ABSTRACT

Proteins are relevant antimicrobial drug targets, and among them, enzymes represent a significant group, since most of them catalyze reactions essential for supporting the central metabolism, or are necessary for the pathogen vitality. Genomic exploration of pathogenic and non-pathogenic microorganisms has revealed genes encoding for a superfamily of metalloenzymes, known as carbonic anhydrases (CAs, EC 4.2.1.1). CAs catalyze the physiologically crucial reversible reaction of the carbon dioxide hydration to bicarbonate and protons. Herein, we investigated the sulfonamide inhibition profile of the recombinant ß-CA (CynT2) identified in the genome of the Gram-negative bacterium Escherichia coli. This biocatalyst is indispensable for the growth of the microbe at atmospheric pCO2. Surprisingly, this enzyme has not been investigated for its inhibition with any class of CA inhibitors. Here, we show that CynT2 was strongly inhibited by some substituted benzene-sulfonamides and the clinically used inhibitor sulpiride (KIs in the range of 82-97 nM). This study may be relevant for identifying novel CA inhibitors, as well as for another essential part of the drug discovery pipeline, such as the structure-activity relationship for this class of enzyme inhibitors.


Subject(s)
Anion Transport Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Escherichia coli Proteins/metabolism , Sulfonamides/chemistry , Sulfonamides/pharmacology , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , Anti-Bacterial Agents/chemistry , Benzene/chemistry , Carbon Dioxide/chemistry , Carbon Dioxide/metabolism , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrases/genetics , Drug Evaluation, Preclinical/methods , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Humans , Structure-Activity Relationship
5.
Int J Biol Macromol ; 159: 570-576, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32442571

ABSTRACT

Volume-regulated anion channel (VRAC) is ubiquitously expressed in vertebrate cells and in various types of cancer cells. Leucine-rich repeat containing 8A (LRRC8A) and its four homologous family members (LRRC8B-E) assemble into heterogeneous VRAC complexes of ~800 kDa. The main components of VRAC, LRRC8A and LRRC8D have been implicated in the proliferation, migration, death, and multidrug resistance of cancer cells through their involvement in various signal pathways. This review summarizes recent findings concerning the involvement of VRAC in cancer development and progression, including the molecular structure, function, and regulation of VRAC and its roles in various cancers, and highlights the remaining challenges in the field. Our aim is to evaluate the potential of VRAC as a therapeutic target for cancer therapies and to discuss the major problems to be solved.


Subject(s)
Anion Transport Proteins/metabolism , Biomarkers, Tumor , Neoplasms/etiology , Neoplasms/metabolism , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/chemistry , Anion Transport Proteins/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Disease Progression , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Protein Binding , Protein Multimerization , Signal Transduction/drug effects
6.
Am J Physiol Renal Physiol ; 318(4): F870-F877, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31984792

ABSTRACT

Adenosine plays an important role in various aspects of kidney physiology, but the specific targets and mechanisms of actions are not completely understood. The collecting duct has the highest expression of adenosine receptors, particularly adenosine A1 receptors (A1Rs). Interstitial adenosine levels are greatly increased up to a micromolar range in response to dietary salt loading. We have previously shown that the basolateral membrane of principal cells has primarily K+ conductance mediated by Kir4.1/5.1 channels to mediate K+ recycling and to set up a favorable driving force for Na+/K+ exchange (47). Intercalated cells express the Cl- ClC-K2/b channel mediating transcellular Cl- reabsorption. Using patch-clamp electrophysiology in freshly isolated mouse collecting ducts, we found that acute application of adenosine reversely inhibits ClC-K2/b open probability from 0.31 ± 0.04 to 0.17 ± 0.06 and to 0.10 ± 0.05 for 1 and 10 µM, respectively. In contrast, adenosine (10 µM) had no measureable effect on Kir4.1/5.1 channel activity in principal cells. The inhibitory effect of adenosine on ClC-K2/b was abolished in the presence of the A1R blocker 8-cyclopentyl-1,3-dipropylxanthine (10 µM). Consistently, application of the A1R agonist N6-cyclohexyladenosine (1 µM) recapitulated the inhibitory action of adenosine on ClC-K2/b open probability. The effects of adenosine signaling in the collecting duct were independent from its purinergic counterpartner, ATP, having no measurable actions on ClC-K2/b and Kir4.1/5.1. Overall, we demonstrated that adenosine selectively inhibits ClC-K2/b activity in intercalated cells by targeting A1Rs. We propose that inhibition of transcellular Cl- reabsorption in the collecting duct by adenosine would aid in augmenting NaCl excretion during high salt intake.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Adenosine/pharmacology , Anion Transport Proteins/antagonists & inhibitors , Chloride Channels/antagonists & inhibitors , Chlorides/metabolism , Kidney Tubules, Collecting/drug effects , Receptor, Adenosine A1/drug effects , Renal Reabsorption/drug effects , Animals , Anion Transport Proteins/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Chloride Channels/metabolism , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Male , Membrane Potentials/drug effects , Mice, Inbred C57BL , Receptor, Adenosine A1/metabolism
7.
Physiol Rep ; 7(23): e14232, 2019 12.
Article in English | MEDLINE | ID: mdl-31833218

ABSTRACT

Slc4a11, a member of the Slc4 HCO3- transporter family, has a wide tissue distribution. In mouse salivary glands, the expression of Slc4a11 mRNA was more than eightfold greater than the other nine members of the Slc4 gene family. The Slc4a11 protein displayed a diffuse subcellular distribution in both the acinar and duct cells of mouse submandibular glands (SMG). Slc4a11 disruption induced a significant increase in the Na+ and Cl- concentrations of stimulated SMG saliva, whereas it did not affect the fluid secretion rate in response to either ß-adrenergic or cholinergic receptor stimulation. Heterologous expressed mouse Slc4a11 acted as a H+ /OH- transporter that was uncoupled of Na+ or Cl- movement, and this activity was blocked by ethyl-isopropyl amiloride (EIPA) but not 4,4'-Diisothiocyanato-2,2'-stilbenedisulfonic acid (DIDS). Slc4a11 disruption revealed that Slc4a11 does not play a major role in intracellular pH regulation in mouse salivary gland cells. In contrast, NaCl reabsorption was impaired in the SMG saliva of female compared to male Slc4a11 null mice, which correlated with the loss of duct cells and a decrease in expression of the duct-cell-specific transcription factor Ascl3. Together, our results suggest that Slc4a11 expression regulates the number of ducts cells in the mouse SMG and consequently NaCl reabsorption.


Subject(s)
Absorption, Physiological , Anion Transport Proteins/metabolism , Protons , Sodium Chloride/metabolism , Submandibular Gland/metabolism , Symporters/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Submandibular Gland/cytology , Symporters/antagonists & inhibitors , Symporters/genetics
8.
Mol Nutr Food Res ; 63(12): e1801402, 2019 06.
Article in English | MEDLINE | ID: mdl-30913372

ABSTRACT

SCOPE: Conjugated linoleic acid (CLA), a bioactive substance predominantly found in ruminant products, improves insulin resistance and exhibits anti-inflammatory activity. The chief objective of the study is to investigate the effects and potential mechanisms of CLA on high fructose-induced hyperuricemia and renal inflammation. METHODS AND RESULTS: Hyperuricemia and renal inflammation are induced in rats by 10% fructose. Hyperuricemia, insulin resistance, and renal inflammation are evaluated. CLA potently ameliorates fructose-induced hyperuricemia with insulin resistance and significantly reduces the levels of inflammation factors in serum and kidney. It reverses fructose-induced upregulation of glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1) in the kidney. Moreover, CLA dramatically inhibits the activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. Additionally, CLA suppresses toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88) signaling activation to inhibit nuclear factor-kB (NF-kB) signaling in the kidney of fructose-fed rats. CONCLUSION: CLA ameliorates hyperuricemia along with insulin resistance and renal inflammatory, which may be associated with the suppression of renal GLUT9 and URAT1 in fructose-fed rats. Its molecular mechanism may be related to the inhibition of NLRP3 inflammasome and TLR4/MyD88 signaling pathway. Therefore, CLA may be a promising candidate for preventing fructose-induced hyperuricemia and renal inflammation.


Subject(s)
Fructose/administration & dosage , Hyperuricemia/drug therapy , Inflammasomes/physiology , Inflammation/drug therapy , Kidney/drug effects , Linoleic Acids, Conjugated/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Toll-Like Receptor 4/physiology , Animals , Anion Transport Proteins/antagonists & inhibitors , Linoleic Acids, Conjugated/therapeutic use , Male , Monosaccharide Transport Proteins/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
9.
Cancer Res ; 78(10): 2513-2523, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29510993

ABSTRACT

Glycolysis and fatty acid synthesis are highly active in cancer cells through cytosolic citrate metabolism, with intracellular citrate primarily derived from either glucose or glutamine via the tricarboxylic acid cycle. We show here that extracellular citrate is supplied to cancer cells through a plasma membrane-specific variant of the mitochondrial citrate transporter (pmCiC). Metabolomic analysis revealed that citrate uptake broadly affected cancer cell metabolism through citrate-dependent metabolic pathways. Treatment with gluconate specifically blocked pmCiC and decreased tumor growth in murine xenografts of human pancreatic cancer. This treatment altered metabolism within tumors, including fatty acid metabolism. High expression of pmCiC was associated with invasion and advanced tumor stage across many human cancers. These findings support the exploration of extracellular citrate transport as a novel potential target for cancer therapy.Significance: Uptake of extracellular citrate through pmCiC can be blocked with gluconate to reduce tumor growth and to alter metabolic characteristics of tumor tissue. Cancer Res; 78(10); 2513-23. ©2018 AACR.


Subject(s)
Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/metabolism , Cell Proliferation/drug effects , Citric Acid/metabolism , Gluconates/pharmacology , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Epithelial Cells/metabolism , Fatty Acids/biosynthesis , Glycolysis/physiology , Humans , Male , Mice , Organic Anion Transporters , Prostate/cytology , Prostate/metabolism , RNA Interference , RNA, Small Interfering/genetics
10.
J Cell Physiol ; 233(2): 1414-1423, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28543431

ABSTRACT

Existing studies on the mechanism of cell volume regulation are mainly relevant to ion channels and osmosis in extracellular fluid. Recently, accumulating evidence has shown that cellular mechanical microenvironment also influences the cell volume. Herein, we investigated the regulation of substrate stiffness on the cell volume homeostasis of MCF-7 cells and their following migration behaviors. We found that cell volume increases with increasing substrate stiffness, which could be affected by blocking the cell membrane anion permeability and dopamine receptor. In addition, the cell migration is significantly inhibited by decreasing the cell volume using tamoxifen and such inhibition effect on migration is enhanced by increasing substrate stiffness. The cell membrane anion permeability might be the linker between cellular mechanical microenvironment and cellular volume homeostasis regulation. This work revealed the regulation of substrate stiffness on cell volume homeostasis for the first time, which would provide a new perspective into the understanding of cancer metastasis and a promising anti-cancer therapy through regulation of cell volume homeostasis.


Subject(s)
Acrylic Resins/chemistry , Breast Neoplasms/metabolism , Cell Membrane/metabolism , Cell Size , Osmoregulation , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Benzazepines/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Adhesion , Cell Membrane/drug effects , Cell Membrane/pathology , Cell Membrane Permeability , Cell Movement , Cell Size/drug effects , Collagen/metabolism , Female , Humans , Hydrogels , Hypotonic Solutions/pharmacology , MCF-7 Cells , Osmoregulation/drug effects , Porosity , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism , Tamoxifen/pharmacology , Tumor Microenvironment
11.
Nat Cell Biol ; 19(9): 1017-1026, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28812580

ABSTRACT

Although normally dormant, hair follicle stem cells (HFSCs) quickly become activated to divide during a new hair cycle. The quiescence of HFSCs is known to be regulated by a number of intrinsic and extrinsic mechanisms. Here we provide several lines of evidence to demonstrate that HFSCs utilize glycolytic metabolism and produce significantly more lactate than other cells in the epidermis. Furthermore, lactate generation appears to be critical for the activation of HFSCs as deletion of lactate dehydrogenase (Ldha) prevented their activation. Conversely, genetically promoting lactate production in HFSCs through mitochondrial pyruvate carrier 1 (Mpc1) deletion accelerated their activation and the hair cycle. Finally, we identify small molecules that increase lactate production by stimulating Myc levels or inhibiting Mpc1 carrier activity and can topically induce the hair cycle. These data suggest that HFSCs maintain a metabolic state that allows them to remain dormant and yet quickly respond to appropriate proliferative stimuli.


Subject(s)
Cell Proliferation , Cellular Senescence , Glycolysis , Hair Follicle/enzymology , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Stem Cells/enzymology , Acrylates/pharmacology , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Female , Genotype , Glycolysis/drug effects , Hair Follicle/cytology , Hair Follicle/drug effects , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/metabolism , L-Lactate Dehydrogenase/deficiency , L-Lactate Dehydrogenase/genetics , Lactate Dehydrogenase 5 , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Stem Cells/drug effects , Time Factors
12.
Nat Cell Biol ; 19(9): 1027-1036, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28812582

ABSTRACT

Most differentiated cells convert glucose to pyruvate in the cytosol through glycolysis, followed by pyruvate oxidation in the mitochondria. These processes are linked by the mitochondrial pyruvate carrier (MPC), which is required for efficient mitochondrial pyruvate uptake. In contrast, proliferative cells, including many cancer and stem cells, perform glycolysis robustly but limit fractional mitochondrial pyruvate oxidation. We sought to understand the role this transition from glycolysis to pyruvate oxidation plays in stem cell maintenance and differentiation. Loss of the MPC in Lgr5-EGFP-positive stem cells, or treatment of intestinal organoids with an MPC inhibitor, increases proliferation and expands the stem cell compartment. Similarly, genetic deletion of the MPC in Drosophila intestinal stem cells also increases proliferation, whereas MPC overexpression suppresses stem cell proliferation. These data demonstrate that limiting mitochondrial pyruvate metabolism is necessary and sufficient to maintain the proliferation of intestinal stem cells.


Subject(s)
Cell Proliferation , Drosophila melanogaster/metabolism , Glycolysis , Intestinal Mucosa/metabolism , Mitochondria/metabolism , Pyruvic Acid/metabolism , Stem Cells/metabolism , Acrylates/pharmacology , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Cell Differentiation , Cell Proliferation/drug effects , Cells, Cultured , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Genotype , Humans , Intestines/cytology , Intestines/drug effects , Lactic Acid/metabolism , Mice, Knockout , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Monocarboxylic Acid Transporters , Phenotype , RNA Interference , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stem Cells/drug effects , Time Factors , Tissue Culture Techniques , Transfection
13.
FEBS J ; 284(3): 451-465, 2017 02.
Article in English | MEDLINE | ID: mdl-27987376

ABSTRACT

Pioglitazone is used globally for the treatment of type 2 diabetes mellitus (T2DM) and is one of the most effective therapies for improving glucose homeostasis and insulin resistance in T2DM patients. However, its mechanism of action in the tissues and pathways that regulate glucose metabolism are incompletely defined. Here we investigated the direct effects of pioglitazone on hepatocellular pyruvate metabolism and the dependency of these observations on the purported regulators of mitochondrial pyruvate transport, MPC1 and MPC2. In cultured H4IIE hepatocytes, pioglitazone inhibited [2-14 C]-pyruvate oxidation and pyruvate-driven oxygen consumption and, in mitochondria isolated from both hepatocytes and human skeletal muscle, pioglitazone selectively and dose-dependently inhibited pyruvate-driven ATP synthesis. Pioglitazone also suppressed hepatocellular glucose production (HGP), without influencing the mRNA expression of key HGP regulatory genes. Targeted siRNA silencing of MPC1 and 2 caused a modest inhibition of pyruvate oxidation and pyruvate-driven ATP synthesis, but did not alter pyruvate-driven HGP and, importantly, it did not influence the actions of pioglitazone on either pathway. In summary, these findings outline a novel mode of action of pioglitazone relevant to the pathogenesis of T2DM and suggest that targeting pyruvate metabolism may lead to the development of effective new T2DM therapies.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Glucose/antagonists & inhibitors , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Mitochondria/drug effects , Thiazolidinediones/pharmacology , Adenosine Triphosphate/biosynthesis , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Biological Transport/drug effects , Carbon Radioisotopes , Cell Line , Gluconeogenesis/drug effects , Glucose/biosynthesis , Glycolysis/drug effects , Hepatocytes/cytology , Hepatocytes/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Oxidation-Reduction , Oxidative Phosphorylation/drug effects , Pioglitazone , Pyruvic Acid/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats
14.
Physiol Plant ; 159(3): 340-353, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27787914

ABSTRACT

Miscanthus is a vigorous perennial Gramineae genus grown throughout the world as a promising bioenergy crop and generally regarded as heavy metal tolerant due to its ability to absorb heavy metals. However, little is known about the mechanism for heavy metal tolerance in Miscanthus. In this study, two Miscanthus species (Miscanthus sacchariflorus and Miscanthus floridulus) exhibiting different cadmium (Cd) sensitivity were used to address the mechanisms of Cd tolerance. Under the same Cd stress, M. sacchariflorus showed higher Cd tolerance with better growth and lower Cd accumulation in both shoots and roots than M. floridulus. The malate (MA) content significantly increased in root exudates of M. sacchariflorus following Cd treatment while it was almost unchanged in M. floridulus. Cellular Cd analysis and flux data showed that exogenous MA application markedly restricted Cd influx and accumulation while an anion-channel inhibitor (phenylglyoxal) effectively blocked Cd-induced MA secretion and increased Cd influx in M. sacchariflorus, indicating that MA secretion could alleviate Cd toxicity by reducing Cd uptake. The genes of malate dehydrogenases (MsMDHs) and Al-activated malate transporter 1 (MsALMT1) in M. sacchariflorus were highly upregulated under Cd stress, compared with that in M. floridulus. The results indicate that Cd-induced MA synthesis and secretion efficiently alleviate Cd toxicity by reducing Cd influx in M. sacchariflorus.


Subject(s)
Cadmium/toxicity , Malates/metabolism , Poaceae/physiology , Soil Pollutants/toxicity , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Cadmium/metabolism , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , Malates/pharmacology , Phenylglyoxal/pharmacology , Plant Proteins/antagonists & inhibitors , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Roots/cytology , Plant Roots/drug effects , Plant Roots/genetics , Plant Roots/physiology , Poaceae/cytology , Poaceae/drug effects , Poaceae/genetics , Soil Pollutants/metabolism , Stress, Physiological
15.
Hepatology ; 65(5): 1543-1556, 2017 05.
Article in English | MEDLINE | ID: mdl-28027586

ABSTRACT

Diseases of the liver related to metabolic syndrome have emerged as the most common and undertreated hepatic ailments. The cause of nonalcoholic fatty liver disease is the aberrant accumulation of lipid in hepatocytes, though the mechanisms whereby this leads to hepatocyte dysfunction, death, and hepatic fibrosis are still unclear. Insulin-sensitizing thiazolidinediones have shown efficacy in treating nonalcoholic steatohepatitis (NASH), but their widespread use is constrained by dose-limiting side effects thought to be due to activation of the peroxisome proliferator-activated receptor γ. We sought to determine whether a next-generation thiazolidinedione with markedly diminished ability to activate peroxisome proliferator-activated receptor γ (MSDC-0602) would retain its efficacy for treating NASH in a rodent model. We also determined whether some or all of these beneficial effects would be mediated through an inhibitory interaction with the mitochondrial pyruvate carrier 2 (MPC2), which was recently identified as a mitochondrial binding site for thiazolidinediones, including MSDC-0602. We found that MSDC-0602 prevented and reversed liver fibrosis and suppressed expression of markers of stellate cell activation in livers of mice fed a diet rich in trans-fatty acids, fructose, and cholesterol. Moreover, mice with liver-specific deletion of MPC2 were protected from development of NASH on this diet. Finally, MSDC-0602 directly reduced hepatic stellate cell activation in vitro, and MSDC-0602 treatment or hepatocyte MPC2 deletion also limited stellate cell activation indirectly by affecting secretion of exosomes from hepatocytes. CONCLUSION: Collectively, these data demonstrate the effectiveness of MSDC-0602 for attenuating NASH in a rodent model and suggest that targeting hepatic MPC2 may be an effective strategy for pharmacologic development. (Hepatology 2017;65:1543-1556).


Subject(s)
Acetophenones/therapeutic use , Anion Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/drug therapy , Thiazolidinediones/therapeutic use , Acetophenones/pharmacology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Exosomes/drug effects , Hepatic Stellate Cells/drug effects , Male , Mice, Inbred C57BL , Molecular Targeted Therapy , Random Allocation , Thiazolidinediones/pharmacology
16.
Sci Rep ; 6: 34995, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713539

ABSTRACT

Gout is caused by elevated serum urate levels, which can be treated using inhibitors of the uric acid transporter, URAT1. We exploited affinity differences between the human and rat transporters to map inhibitor binding sites in URAT1. Human-rat transporter chimeras revealed that human URAT1 serine-35, phenylalanine-365 and isoleucine-481 are necessary and sufficient to provide up to a 100-fold increase in affinity for inhibitors. Moreover, serine-35 and phenylalanine-365 are important for high-affinity interaction with the substrate urate. A novel URAT1 binding assay provides support for direct interaction with these amino acids; thus, current clinically important URAT1 inhibitors likely bind the same site in URAT1. A structural model suggests that these three URAT1 residues are in close proximity potentially projecting within the channel. Our results indicate that amino acids from several transmembrane segments functionally cooperate to form a high-affinity URAT1 inhibitor binding site that, when occupied, prevents substrate interactions.


Subject(s)
Organic Anion Transporters/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Amino Acid Substitution , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/chemistry , Anion Transport Proteins/genetics , Binding Sites/genetics , HEK293 Cells , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Organic Anion Transport Protein 1/chemistry , Organic Anion Transporters/chemistry , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/chemistry , Organic Cation Transport Proteins/genetics , Protein Interaction Domains and Motifs , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Uric Acid/metabolism
17.
J Am Soc Nephrol ; 27(12): 3706-3714, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27153921

ABSTRACT

Pendrin is a Cl-/HCO3- exchanger expressed in type B and non-A, non-B intercalated cells in the distal nephron, where it facilitates Cl- absorption and is involved in Na+ absorption and acid-base balance. Pendrin-knockout mice show no fluid-electrolyte abnormalities under baseline conditions, although mice with double knockout of pendrin and the Na+/Cl- cotransporter (NCC) manifest profound salt wasting. Thus, pendrin may attenuate diuretic-induced salt loss, but this function remains unconfirmed. To clarify the physiologic role of pendrin under conditions not confounded by gene knockout, and to test the potential utility of pendrin inhibitors for diuretic therapy, we tested in mice a small-molecule pendrin inhibitor identified from a high-throughput screen. In vitro, a pyrazole-thiophenesulfonamide, PDSinh-C01, inhibited Cl-/anion exchange mediated by mouse pendrin with a 50% inhibitory concentration of 1-3 µM, without affecting other major kidney tubule transporters. Administration of PDSinh-C01 to mice at predicted therapeutic doses, determined from serum and urine pharmacokinetics, did not affect urine output, osmolality, salt excretion, or acid-base balance. However, in mice treated acutely with furosemide, administration of PDSinh-C01 produced a 30% increase in urine output, with increased Na+ and Cl- excretion. In mice treated long term with furosemide, in which renal pendrin is upregulated, PDSinh-C01 produced a 60% increase in urine output. Our findings clarify the role of pendrin in kidney function and suggest pendrin inhibition as a novel approach to potentiate the action of loop diuretics. Such combination therapy might enhance diuresis and salt excretion for treatment of hypertension and edema, perhaps including diuretic-resistant edema.


Subject(s)
Anion Transport Proteins/antagonists & inhibitors , Diuretics/pharmacology , Furosemide/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Animals , Drug Synergism , Female , Mice , Sulfate Transporters
18.
Am J Physiol Cell Physiol ; 310(11): C857-73, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26984736

ABSTRACT

The leucine-rich repeat containing 8A (LRRC8A) protein is an essential component of the volume-sensitive organic anion channel (VSOAC), and using pharmacological anion channel inhibitors (NS3728, DIDS) and LRRC8A siRNA we have investigated its role in development of Cisplatin resistance in human ovarian (A2780) and alveolar (A549) carcinoma cells. In Cisplatin-sensitive cells Cisplatin treatment increases p53-protein level as well as downstream signaling, e.g., expression of p21(Waf1/Cip1), Bax, Noxa, MDM2, and activation of Caspase-9/-3. In contrast, Cisplatin-resistant cells do not enter apoptosis, i.e., their p53 and downstream signaling are reduced and caspase activity unaltered following Cisplatin exposure. Reduced LRRC8A expression and VSOAC activity are previously shown to correlate with Cisplatin resistance, and here we demonstrate that pharmacological inhibition and transient knockdown of LRRC8A reduce the protein level of p53, MDM2, and p21(Waf1/Cip1) as well as Caspase-9/-3 activation in Cisplatin-sensitive cells. Cisplatin resistance is accompanied by reduction in total LRRC8A expression (A2780) or LRRC8A expression in the plasma membrane (A549). Activation of Caspase-3 dependent apoptosis by TNFα-exposure or hyperosmotic cell shrinkage is almost unaffected by pharmacological anion channel inhibition. Our data indicate 1) that expression/activity of LRRC8A is essential for Cisplatin-induced increase in p53 protein level and its downstream signaling, i.e., Caspase-9/-3 activation, expression of p21(Waf1/Cip1) and MDM2; and 2) that downregulation of LRRC8A-dependent osmolyte transporters contributes to acquirement of Cisplatin resistance in ovarian and lung carcinoma cells. Activation of LRRC8A-containing channels is upstream to apoptotic volume decrease as hypertonic cell shrinkage induces apoptosis independent of the presence of LRRC8A.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Agents/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Lung Neoplasms/drug therapy , Membrane Proteins/metabolism , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/enzymology , Adenocarcinoma, Bronchiolo-Alveolar/genetics , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/metabolism , Apoptosis/drug effects , Caspase 3/genetics , Caspase 9/genetics , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/metabolism , Cell Size , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Proteins/genetics , Membrane Transport Modulators/pharmacology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics
19.
PLoS One ; 11(3): e0150918, 2016.
Article in English | MEDLINE | ID: mdl-26963391

ABSTRACT

BACKGROUND: The absence of NCC does not cause significant salt wasting in NCC deficient mice under basal conditions. We hypothesized that ENaC and pendrin play important roles in compensatory salt absorption in the setting of NCC inactivation, and their inhibition and/or downregulation can cause significant salt wasting in NCC KO mice. METHODS: WT and NCC KO mice were treated with a daily injection of either amiloride, an inhibitor of ENaC, or acetazolamide (ACTZ), a blocker of salt and bicarbonate reabsorption in the proximal tubule and an inhibitor of carbonic anhydrases in proximal tubule and intercalated cells, or a combination of acetazolamide plus amiloride for defined durations. Animals were subjected to daily balance studies. At the end of treatment, kidneys were harvested and examined. Blood samples were collected for electrolytes and acid base analysis. RESULTS: Amiloride injection significantly increased the urine output (UO) in NCC KO mice (from 1.3 ml/day before to 2.5 ml/day after amiloride, p<0.03, n = 4) but caused only a slight change in UO in WT mice (p>0.05). The increase in UO in NCC KO mice was associated with a significant increase in sodium excretion (from 0.25 mmol/24 hrs at baseline to 0.35 mmol/24 hrs after amiloride injection, p<0.05, n = 4). Daily treatment with ACTZ for 6 days resulted in >80% reduction of kidney pendrin expression in both WT and NCC KO mice. However, ACTZ treatment noticeably increased urine output and salt excretion only in NCC KO mice (with urine output increasing from a baseline of 1.1 ml/day to 2.3 ml/day and sodium excretion increasing from 0.22 mmole/day before to 0.31 mmole/day after ACTZ) in NCC KO mice; both parameters were significantly higher than in WT mice. Western blot analysis demonstrated significant enhancement in ENaC expression in medulla and cortex of NCC KO and WT mice in response to ACTZ injection for 6 days, and treatment with amiloride in ACTZ-pretreated mice caused a robust increase in salt excretion in both NCC KO and WT mice. Pendrin KO mice did not display a significant increase in urine output or salt excretion after treatment with amiloride or ACTZ. CONCLUSION: 1. ENaC plays an important role in salt reabsorption in NCC KO mice. 2. NCC contributes to compensatory salt reabsorption in the setting of carbonic anhydrase inhibition, which is associated with increased delivery of salt from the proximal tubule and the down regulation of pendrin. 3. ENaC is upregulated by ACTZ treatment and its inhibition by amiloride causes significant diuresis in NCC KO and WT mice. Despite being considered mild agents individually, we propose that the combination of acetazolamide and amiloride in the setting of NCC inhibition (i.e., hydrochlorothiazide) will be a powerful diuretic regimen.


Subject(s)
Anion Transport Proteins/metabolism , Epithelial Sodium Channels/metabolism , Kidney Tubules, Proximal/metabolism , Sodium Chloride/urine , Acetazolamide/pharmacology , Amiloride/pharmacology , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , Epithelial Sodium Channel Blockers/pharmacology , Epithelial Sodium Channels/genetics , Mice , Mice, Knockout , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Sulfate Transporters
20.
Biol Trace Elem Res ; 172(1): 193-200, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26660892

ABSTRACT

It is well known that excess iodide can lead to thyroid colloid retention, a classic characteristic of iodide-induced goiter. However, the mechanism has not been fully unrevealed. Iodide plays an important role in thyroid function at multiple steps of thyroid colloid synthesis and transport among which sodium/iodide symporter (NIS) and pendrin are essential. In our study, we fed female BALB/c mice with different concentrations of high-iodine water including group A (control group, 0 µg/L), group B (1500 µg/L), group C (3000 µg/L), group D (6000 µg/L), and group E (12,000 µg/L). After 7 months of feeding, we found that excess iodide could lead to different degrees of thyroid colloid retention. Besides, NIS and pendrin expression were downregulated in the highest dose group. The thyroid iodide intake function detected by urine iodine assay and thyroidal (125)I experiments showed that the urine level of iodine increased, while the iodine intake rate decreased when the concentration of iodide used in feeding water increased (all p < 0.05 vs. control group). In addition, transmission electron microscopy (TEM) indicated a reduction in the number of intracellular mitochondria of thyroid cells. Based on these findings, we concluded that the occurrence of thyroid colloid retention exacerbated by excess iodide was associated with the suppression of NIS and pendrin expression, providing an additional insight of the potential mechanism of action of excess iodide on thyroid gland.


Subject(s)
Anion Transport Proteins/antagonists & inhibitors , Iodides/pharmacology , Symporters/antagonists & inhibitors , Thyroid Gland/drug effects , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Colloids/chemistry , Colloids/metabolism , Female , Iodides/administration & dosage , Mice , Mice, Inbred BALB C , Sulfate Transporters , Symporters/genetics , Symporters/metabolism , Thyroid Gland/metabolism , Thyroid Gland/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...