Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(30): e2122158119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858418

ABSTRACT

Eicosapentaenoic acid (EPA), an omega-3 (ω-3) polyunsaturated fatty acid, is an essential nutrient that exhibits antiinflammatory, neuroprotective, and cardiovascular-protective activities. Although EPA is used as a nutrient-based pharmaceutical agent or dietary supplement, its molecular target(s) is debatable. Here, we showed that EPA and its metabolites strongly and reversibly inhibit vesicular nucleotide transporter (VNUT), a key molecule for vesicular storage and release of adenosine triphosphate (ATP) in purinergic chemical transmission. In vitro analysis showed that EPA inhibits human VNUT-mediated ATP uptake at a half-maximal inhibitory concentration (IC50) of 67 nM, acting as an allosteric modulator through competition with Cl-. EPA impaired vesicular ATP release from neurons without affecting the vesicular release of other neurotransmitters. In vivo, VNUT-/- mice showed a delay in the onset of neuropathic pain and resistance to both neuropathic and inflammatory pain. EPA potently attenuated neuropathic and inflammatory pain in wild-type mice but not in VNUT-/- mice without affecting the basal nociception. The analgesic effect of EPA was canceled by the intrathecal injection of purinoceptor agonists and was stronger than that of existing drugs used for neuropathic pain treatment, with few side effects. Neuropathic pain impaired insulin sensitivity in previous studies, which was improved by EPA in the wild-type mice but not in the VNUT-/- mice. Our results showed that VNUT is a molecular target of EPA that attenuates neuropathic and inflammatory pain and insulin resistance. EPA may represent a unique nutrient-based treatment and prevention strategy for neurological, immunological, and metabolic diseases by targeting purinergic chemical transmission.


Subject(s)
Eicosapentaenoic Acid , Neuralgia , Nucleotide Transport Proteins , Adenosine Triphosphate/metabolism , Animals , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Humans , Insulin Resistance , Mice , Neuralgia/drug therapy , Neuralgia/genetics , Nociception , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism
2.
Yakugaku Zasshi ; 141(4): 517-526, 2021.
Article in Japanese | MEDLINE | ID: mdl-33790119

ABSTRACT

ATP, used in cells as an energy currency, also acts as an extracellular signaling molecule. Studies of purinergic receptor subtypes have revealed that purinergic chemical transmission plays important roles in various cell types. The vesicular nucleotide transporter (VNUT), the ninth transporter in the SLC17 organic anion transporter family, is essential for vesicular ATP storage and its subsequent release. The VNUT is localized on the membrane of secretory vesicles and actively transports ATP into vesicles using an electrochemical gradient of protons supplied by vacuolar proton ATPase (V-ATPase) as a driving force. ATP acts as a damage-associated molecular pattern (DAMPs), contributing to the persistence of chronic inflammation. Chronic inflammation induces systemic insulin resistance, which is the underlying pathology of type 2 diabetes and non-alcoholic fatty liver disease (NAFLD), ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). We previously demonstrated that ATP transported in insulin granules via the VNUT negatively regulates insulin secretion. We also found that hepatocytes release ATP in a VNUT-dependent manner, which elicits hepatic insulin resistance and inflammation. VNUT-knockout mice exhibited improved glucose tolerance and were resistant to the development of high fat diet-induced NAFLD. In this article, we summarize recent advances in our understanding of the mechanism of the VNUT, the development of inhibitors, and its pathological involvement in type 2 diabetes and NAFLD. The pharmacological inhibition of the VNUT may represent a potential therapeutic approach for the treatment of metabolic diseases.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Nucleotide Transport Proteins/physiology , Adenosine Triphosphate/metabolism , Animals , Clodronic Acid/pharmacology , Clodronic Acid/therapeutic use , Drug Discovery , Humans , Insulin Resistance/genetics , Insulin Secretion/genetics , Mice, Knockout , Nucleotide Transport Proteins/antagonists & inhibitors , Secretory Vesicles/metabolism
3.
Sci Rep ; 11(1): 5192, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664289

ABSTRACT

The vesicular nucleotide transporter (VNUT) is responsible for the vesicular storage and release of ATP from various ATP-secreting cells, and it plays an essential role in purinergic signaling. Although extracellular ATP and its degradation products are known to mediate various inflammatory responses via purinoceptors, whether vesicular ATP release affects steatohepatitis and acute liver injury is far less understood. In the present study, we investigated the effects of clodronate, a potent and selective VNUT inhibitor, on acute and chronic liver inflammation in mice. In a model of methionine/choline-deficient diet-induced non-alcoholic steatohepatitis (NASH), the administration of clodronate reduced hepatic inflammation, fibrosis, and triglyceride accumulation. Clodronate also protected mice against high-fat/high-cholesterol diet-induced steatohepatitis. Moreover, prophylactic administration of clodronate prevented D-galactosamine and lipopolysaccharide-induced acute liver injury by reducing inflammatory cytokines and hepatocellular apoptosis. In vitro, clodronate inhibited glucose-induced vesicular ATP release mediated by VNUT and reduced the intracellular level and secretion of triglycerides in isolated hepatocytes. These results suggest that VNUT-dependent vesicular ATP release plays a crucial role in the recruitment of immune cells, cytokine production, and the aggravation of steatosis in the liver. Pharmacological inhibition of VNUT may provide therapeutic benefits in liver inflammatory disorders, including NASH and acute toxin-induced injury.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Clodronic Acid/pharmacology , Fatty Liver/drug therapy , Nucleotide Transport Proteins/genetics , Adenosine Triphosphate/metabolism , Animals , Chemical and Drug Induced Liver Injury/pathology , Diet/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Humans , Inflammation/drug therapy , Inflammation/etiology , Inflammation/genetics , Inflammation/pathology , Lipopolysaccharides/toxicity , Mice , Nucleotide Transport Proteins/antagonists & inhibitors , Receptors, Purinergic/genetics
4.
Biochim Biophys Acta Biomembr ; 1862(12): 183408, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32652056

ABSTRACT

Vesicular nucleotide transporter (VNUT) is the last identified member of the SLC17 organic anion transporter family, which plays a central role in vesicular storage in ATP-secreting cells. The discovery of VNUT demonstrated that, despite having been neglected for a long time, vesicular ATP release represents a major pathway for purinergic chemical transmission, which had been mainly attributed to ATP permeation channels. This article summarizes recent advances in our understanding of the mechanism of VNUT and its physiopathological roles as well as the development of inhibitors. Regulating the activity and/or the expression of VNUT represents a new and promising therapeutic strategy for the treatment of multiple diseases.


Subject(s)
Adenosine Triphosphate/metabolism , Nucleotide Transport Proteins/metabolism , Animals , Circadian Rhythm , Clodronic Acid/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Neurons/metabolism , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Pain Perception/physiology , Porokeratosis/genetics , Porokeratosis/pathology
5.
Digestion ; 101(1): 6-11, 2020.
Article in English | MEDLINE | ID: mdl-31770754

ABSTRACT

BACKGROUND: Transient receptor potential vanilloid 4 (TRPV4) is activated by stretch (mechanical), warm temperature, some epoxyeicosatrienoic acids, and lipopolysaccharide. TRPV4 is expressed throughout the gastrointestinal epithelia and its activation induces adenosine triphosphate (ATP) exocytosis that is involved in visceral hypersensitivity. As an ATP transporter, vesicular nucleotide transporter (VNUT) mediates ATP storage in secretory vesicles and ATP release via exocytosis upon stimulation. SUMMARY: TRPV4 is sensitized under inflammatory conditions by a variety of factors, including proteases and serotonin, whereas methylation-dependent silencing of TRPV4 expression is associated with various pathophysiological conditions. Gastrointestinal epithelia also release ATP in response to hypo-osmolality or acid through molecular mechanisms that remain unclear. These synergistically released ATP could be involved in visceral hypersensitivity. Low concentrations of the first generation bisphosphate, clodronate, were recently reported to inhibit VNUT activity and thus clodronate may be a safe and potent therapeutic option to treat visceral pain. Key Messages: This review focuses on: (1) ATP and TRPV4 activities in gastrointestinal epithelia; (2) factors that could modulate TRPV4 activity in gastrointestinal epithelia; and (3) the inhibition of VNUT as a potential novel therapeutic strategy for functional gastrointestinal disorders.


Subject(s)
Adenosine Triphosphate/metabolism , Gastrointestinal Tract/metabolism , Nucleotide Transport Proteins/metabolism , TRPV Cation Channels/metabolism , Abdominal Pain/drug therapy , Abdominal Pain/etiology , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Chronic Disease , Clodronic Acid/pharmacology , Clodronic Acid/therapeutic use , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/physiopathology , Humans , Inflammation/metabolism , Inflammation/physiopathology , Mice , Mucous Membrane/drug effects , Mucous Membrane/metabolism , Mucous Membrane/physiopathology , Nucleotide Transport Proteins/antagonists & inhibitors , Pressoreceptors/drug effects , Pressoreceptors/metabolism , Pressoreceptors/physiopathology , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/metabolism
6.
Biochem J ; 476(11): 1585-1604, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31036718

ABSTRACT

Mitochondria play a central role in the maintenance of the naive state of embryonic stem cells. Many details of the mechanism remain to be fully elucidated. Solute carrier family 25 member 36 (Slc25a36) might regulate mitochondrial function through transporting pyrimidine nucleotides for mtDNA/RNA synthesis. Its physical role in this process remains unknown; however, Slc25a36 was recently found to be highly expressed in naive mouse embryonic stem cells (mESCs). Here, the function of Slc25a36 was characterized as a maintenance factor of mESCs pluripotency. Slc25a36 deficiency (via knockdown) has been demonstrated to result in mitochondrial dysfunction, which induces the differentiation of mESCs. The expression of key pluripotency markers (Pou5f1, Sox2, Nanog, and Utf1) decreased, while that of key TE genes (Cdx2, Gata3, and Hand1) increased. Cdx2-positive cells emerged in Slc25a36-deficient colonies under trophoblast stem cell culture conditions. As a result of Slc25a36 deficiency, mtDNA of knockdown cells declined, leading to impaired mitochondria with swollen morphology, decreased mitochondrial membrane potential, and low numbers. The key transcription regulators of mitochondrial biogenesis also decreased. These results indicate that mitochondrial dysfunction leads to an inability to support the pluripotency maintenance. Moreover, down-regulated glutathione metabolism and up-regulated focal adhesion reinforced and stabilized the process of differentiation by separately enhancing OCT4 degradation and promoting cell spread. This study improves the understanding of the function of Slc25a36, as well as the relationship of mitochondrial function with naive pluripotency maintenance and stem cell fate decision.


Subject(s)
Glutathione/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nucleotide Transport Proteins/metabolism , Animals , CDX2 Transcription Factor/metabolism , Cell Differentiation/genetics , Cells, Cultured , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Focal Adhesions , Gene Expression Regulation , Gene Knockdown Techniques , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Octamer Transcription Factor-3/metabolism
7.
PLoS One ; 13(11): e0207521, 2018.
Article in English | MEDLINE | ID: mdl-30458018

ABSTRACT

SLC35B4 belongs to the solute carrier 35 (SLC35) family whose best-characterized members display a nucleotide sugar transporting activity. Using an experimental model of HepG2 cells and indirect immunofluorescent staining, we verified that SLC35B4 was localized to the endoplasmic reticulum (ER). We demonstrated that dilysine motif, especially lysine at position 329, is crucial for the ER localization of this protein in human cells and therefore one should use protein C-tagging with caution. To verify the importance of the protein in glycoconjugates synthesis, we generated SLC35B4-deficient HepG2 cell line using CRISPR-Cas9 approach. Our data showed that knock-out of the SLC35B4 gene does not affect major UDP-Xyl- and UDP-GlcNAc-dependent glycosylation pathways.


Subject(s)
Amino Acid Motifs/genetics , Endoplasmic Reticulum/chemistry , Golgi Apparatus/chemistry , Nucleotide Transport Proteins/chemistry , Amino Acid Sequence/genetics , CRISPR-Cas Systems/genetics , Dipeptides/chemistry , Dipeptides/genetics , Endoplasmic Reticulum/genetics , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Glycosylation , Golgi Apparatus/genetics , Hep G2 Cells , Humans , Lysine/chemistry , Lysine/genetics , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Signal Transduction , Uridine Diphosphate Sugars/chemistry
8.
J Biol Chem ; 293(10): 3770-3779, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29363573

ABSTRACT

Neutrophils migrate to sites infected by pathogenic microorganisms. This migration is regulated by neutrophil-secreted ATP, which stimulates neutrophils in an autocrine manner through purinergic receptors on the plasma membrane. Although previous studies have shown that ATP is released through channels at the plasma membrane of the neutrophil, it remains unknown whether it is also released through alternate secretory systems involving vesicular mechanisms. In this study, we investigated the possible involvement of vesicular nucleotide transporter (VNUT), a key molecule for vesicular storage and nucleotide release, in ATP secretion from neutrophils. RT-PCR and Western blotting analysis indicated that VNUT is expressed in mouse neutrophils. Immunohistochemical analysis indicated that VNUT mainly colocalized with matrix metalloproteinase-9 (MMP-9), a marker of tertiary granules, which are secretory organelles. In mouse neutrophils, ATP release was inhibited by clodronate, which is a potent VNUT inhibitor. Furthermore, neutrophils from VNUT-/- mice did not release ATP and exhibited significantly reduced migration in vitro and in vivo These findings suggest that tertiary granule-localized VNUT is responsible for vesicular ATP release and subsequent neutrophil migration. Thus, these findings suggest an additional mechanism through which ATP is released by neutrophils.


Subject(s)
Adenosine Triphosphate/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Nucleotide Transport Proteins/metabolism , Secretory Vesicles/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Biological Transport/drug effects , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Movement/drug effects , Freund's Adjuvant/pharmacology , Gene Expression Regulation , Humans , Male , Matrix Metalloproteinase 9/metabolism , Membrane Transport Modulators/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Activation/drug effects , Neutrophil Infiltration/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/immunology , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Protein Transport/drug effects , Secretory Vesicles/drug effects , Secretory Vesicles/immunology
9.
Trends Pharmacol Sci ; 39(1): 13-23, 2018 01.
Article in English | MEDLINE | ID: mdl-29146440

ABSTRACT

Clodronate is a first-generation bisphosphonate used worldwide for antiresorptive therapy for osteoporosis. Although clodronate is analgesic in nature, its mechanism and efficacy were unknown for some time. Recently, clodronate was identified as a selective and potent inhibitor for vesicular nucleotide transporter (VNUT), a transporter responsible for vesicular storage of ATP. Clodronate inhibits vesicular ATP release from neurons and reduces chronic neuropathic and inflammatory pain following blockade of purinergic chemical transmission. Its effectiveness is stronger, faster acting, and longer lasting than that of existing drugs such as pregabalin. Thus, clodronate might be a promising drug for attenuating chronic neuropathic pain and opens a new field of drug discovery as a presynaptic blocker for purinergic chemical transmission.


Subject(s)
Adenosine Triphosphate/metabolism , Analgesics/pharmacology , Chronic Pain/drug therapy , Clodronic Acid/pharmacology , Nucleotide Transport Proteins/antagonists & inhibitors , Analgesics/therapeutic use , Animals , Clodronic Acid/therapeutic use , Humans , Nucleotide Transport Proteins/metabolism
10.
Cell Rep ; 21(5): 1169-1179, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29091757

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a lethal brain disease caused by uncontrolled replication of JC polyomavirus (JCV). JCV strains recovered from the brains of PML patients carry mutations that prevent the engagement of sialylated glycans, which are thought to serve as receptors for the infectious entry of wild-type JCV. In this report, we show that non-sialylated glycosaminoglycans (GAGs) can serve as alternative attachment receptors for the infectious entry of both wild-type and PML mutant JCV strains. After GAG-mediated attachment, PML mutant strains engage non-sialylated non-GAG co-receptor glycans, such as asialo-GM1. JCV-neutralizing monoclonal antibodies isolated from patients who recovered from PML appear to block infection by preventing the docking of post-attachment co-receptor glycans in an apical pocket of the JCV major capsid protein. Identification of the GAG-dependent/sialylated glycan-independent alternative entry pathway should facilitate the development of infection inhibitors, including recombinant neutralizing antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , JC Virus/physiology , Virus Internalization , Antibodies, Neutralizing/pharmacology , Capsid Proteins/genetics , Capsid Proteins/immunology , Capsid Proteins/metabolism , Cell Line, Tumor , Gangliosides/pharmacology , Genotype , Glycosaminoglycans/metabolism , Hemagglutination/drug effects , Humans , JC Virus/genetics , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/metabolism , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Mutation , Neuraminidase/metabolism , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Sialic Acids/pharmacology , Virus Internalization/drug effects
11.
Glycoconj J ; 34(3): 411-420, 2017 06.
Article in English | MEDLINE | ID: mdl-27744520

ABSTRACT

Proteoglycans and glycosaminoglycans modulate numerous cellular processes relevant to tumour progression, including cell proliferation, cell-matrix interactions, cell motility and invasive growth. Among the glycosaminoglycans with a well-documented role in tumour progression are heparan sulphate, chondroitin/dermatan sulphate and hyaluronic acid/hyaluronan. While the mode of biosynthesis differs for sulphated glycosaminoglycans, which are synthesised in the ER and Golgi compartments, and hyaluronan, which is synthesized at the plasma membrane, these polysaccharides partially compete for common substrates. In this study, we employed a siRNA knockdown approach for heparan sulphate (EXT1) and heparan/chondroitin/dermatan sulphate-biosynthetic enzymes (ß4GalT7) in the aggressive human breast cancer cell line MDA-MB-231 to study the impact on cell behaviour and hyaluronan biosynthesis. Knockdown of ß4GalT7 expression resulted in a decrease in cell viability, motility and adhesion to fibronectin, while these parameters were unchanged in EXT1-silenced cells. Importantly, these changes were associated with a decreased expression of syndecan-1, decreased signalling response to HGF and an increase in the synthesis of hyaluronan, due to an upregulation of the hyaluronan synthases HAS2 and HAS3. Interestingly, EXT1-depleted cells showed a downregulation of the UDP-sugar transporter SLC35D1, whereas SLC35D2 was downregulated in ß4GalT7-depleted cells, indicating an intricate regulatory network that connects all glycosaminoglycans synthesis. The results of our in vitro study suggest that a modulation of breast cancer cell behaviour via interference with heparan sulphate biosynthesis may result in a compensatory upregulation of hyaluronan biosynthesis. These findings have important implications for the development of glycosaminoglycan-targeted therapeutic approaches for malignant diseases.


Subject(s)
Chondroitin Sulfates/biosynthesis , Dermatan Sulfate/analogs & derivatives , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Heparitin Sulfate/biosynthesis , Hyaluronic Acid/biosynthesis , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Chondroitin Sulfates/antagonists & inhibitors , Chondroitin Sulfates/genetics , Dermatan Sulfate/antagonists & inhibitors , Dermatan Sulfate/biosynthesis , Dermatan Sulfate/genetics , Epithelial Cells/pathology , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Heparitin Sulfate/antagonists & inhibitors , Heparitin Sulfate/genetics , Humans , Hyaluronan Synthases/antagonists & inhibitors , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Monosaccharide Transport Proteins/antagonists & inhibitors , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , N-Acetyllactosamine Synthase/antagonists & inhibitors , N-Acetyllactosamine Synthase/genetics , N-Acetyllactosamine Synthase/metabolism , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
12.
Biol Pharm Bull ; 36(11): 1688-91, 2013.
Article in English | MEDLINE | ID: mdl-24189413

ABSTRACT

Vesicular nucleotide transporter (VNUT) is responsible for vesicular ATP storage in ATP-secreting cells. In the present study, we examined the effects on VNUT-mediated transport of ATP release inhibitors such as ATP-binding cassette (ABC) proteins, hemichannels, maxi anion channels and P2X7 receptor. The ATP transport activity of proteoliposomes containing purified human VNUT was blocked by glibenclamide, carbenoxolone, 18 α-glycyrrhetinic acid, flufenamic acid, arachidonic acid and A438079 without the formation of Δψ (positive inside) as a driving force being affected. Thus, inhibitors of ATP release may inhibit VNUT and subsequent ATP release, since the previous works proved that inhibitors of ATP release blocked VNUT-mediated ATP release at the cell level.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Nucleotide Transport Proteins/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Arachidonic Acid/pharmacology , Carbenoxolone/pharmacology , Cloning, Molecular , DNA, Complementary/genetics , Escherichia coli/genetics , Flufenamic Acid/pharmacology , Glyburide/pharmacology , Glycyrrhetinic Acid/pharmacology , Humans , Liposomes , Molecular Sequence Data , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Polymerase Chain Reaction , Pyridines/pharmacology , Tetrazoles/pharmacology
13.
Endocrinology ; 154(2): 675-84, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23254199

ABSTRACT

Extracellular ATP plays a critical role in regulating insulin secretion in pancreatic ß cells. The ATP released from insulin secretory vesicles has been proposed to be a major source of extracellular ATP. Currently, the mechanism by which ATP accumulates into insulin secretory granules remains elusive. In this study, the authors identified the expression of a vesicular nucleotide transporter (VNUT) in mouse pancreas, isolated mouse islets, and MIN6 cells, a mouse ß cell line. Immunohistochemistry and immunofluorescence revealed that VNUT colocalized extensively with insulin secretory granules. Functional studies showed that suppressing endogenous VNUT expression in ß cells by small hairpin RNA knockdown greatly reduced basal- and glucose-induced ATP release. Importantly, knocking down VNUT expression by VNUT small hairpin RNA in MIN6 cells and isolated mouse islets dramatically suppressed basal insulin release and glucose-stimulated insulin secretion (GSIS). Moreover, acute pharmacologic blockade of VNUT with Evans blue, a VNUT antagonist, greatly attenuated GSIS in a dose-dependent manner. Exogenous ATP treatment effectively reversed the insulin secretion defect induced by both VNUT knockdown and functional inhibition, indicating that VNUT-mediated ATP release is essential for maintaining normal insulin secretion. In contrast to VNUT knockdown, overexpression of VNUT in ß cells resulted in excessive ATP release and enhanced basal insulin secretion and GSIS. Elevated insulin secretion induced by VNUT overexpression was reversed by pharmacologic inhibition of P2X but not P2Y purinergic receptors. This study reveals VNUT is expressed in pancreatic ß cells and plays an essential and novel role in regulating insulin secretion through vesicular ATP release and extracellular purinergic signaling.


Subject(s)
Adenosine Triphosphate/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Nucleotide Transport Proteins/metabolism , Secretory Vesicles/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Evans Blue/pharmacology , Gene Knockdown Techniques , Glucose/pharmacology , Insulin Secretion , Male , Mice , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics
14.
Biol Reprod ; 87(6): 145, 2012 Jun.
Article in English | MEDLINE | ID: mdl-23100621

ABSTRACT

The yolk and white of eggs from chickens contain proteins and other molecules either secreted or transported by cells of the reproductive tract, or secreted by the liver and transported to the ovarian follicles of laying hens. Nutrients transported by solute carriers (SLCs) include glucose, electrolytes, and amino acids. Although SLC genes have been investigated in mammals, there are few studies of expression of SLC genes in the chicken oviduct. Therefore, we investigated temporal and cell-specific expression of selected SLC genes at 3 h and 20 h postovulation and regulation of their expression by microRNAs (miRs). Expression of SLC1A4 (glutamate and neutral amino acid transporter), SLC13A2 (dicarboxylate transporter), and SLC35B4 (UDP-xylose: UDP-N-acetylglucosamine transporter) mRNAs was limited to glandular epithelium (GE), while SLC4A5 (sodium bicarbonate cotransporter) and SLC7A3 (cationic amino acid transporter) mRNAs were expressed predominantly in the luminal epithelium of the magnum. Interestingly, SLC1A4, SLC4A5, SLC13A2 and SLC35B4 mRNAs were abundant only in GE of the shell gland, whereas SLC7A3 was not detected in the shell gland. In the magnum, SLC7A3 and SLC4A5 were expressed, but SLC1A4, SLC35B4, and SLC13A2 were not expressed at 20 h postovulation. In the shell gland, all SLC mRNAs were expressed at both time points, except for SLC7A3. The miRNA target validation assay revealed that miR-1764 and miR-1700 bind directly to SLC13A2 and SLC35B4 transcripts, respectively, to regulate expression. Results of this study demonstrate cell-specific and temporal changes in expression of selected SLC genes and regulation of SLC13A2 and SLC35B4 expression by miRs in the oviduct of laying hens.


Subject(s)
Dicarboxylic Acid Transporters/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Nucleotide Transport Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Oviducts/metabolism , Symporters/metabolism , Animals , Animals, Inbred Strains , Avian Proteins/genetics , Avian Proteins/metabolism , Cell Line , Chickens , Dicarboxylic Acid Transporters/antagonists & inhibitors , Dicarboxylic Acid Transporters/genetics , Egg Shell/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , MicroRNAs/biosynthesis , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Organ Specificity , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Dependent/genetics , Oviducts/cytology , Ovulation/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Symporters/genetics
15.
Exp Cell Res ; 318(17): 2226-36, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22677043

ABSTRACT

In cycling cells cytosolic de novo synthesis of deoxynucleotides is the main source of precursors for mitochondrial (mt) DNA synthesis. The transfer of deoxynucleotides across the inner mt membrane requires protein carriers. PNC1, a SLC25 family member, exchanges pyrimidine nucleoside triphosphates in liposomes and its downregulation decreases mtUTP concentration in cultured cells. By an isotope-flow protocol we confirmed transport of uridine nucleotides by PNC1 in intact cultured cells and investigated PNC1 involvement in the mt trafficking of thymidine phosphates. Key features of our approach were the manipulation of PNC1 expression by RNA interference or inducible overexpression, the employment of cells proficient or deficient for cytosolic thymidine kinase (TK1) to distinguish the direction of flow of thymidine nucleotides across the mt membrane during short pulses with [(3)H]-thymidine, the determination of mtdTTP specific radioactivity to quantitate the rate of mtdTTP export to the cytoplasm. Downregulation of PNC1 in TK1(-) cells increased labeled dTTP in mitochondria due to a reduced rate of export. Overexpression of PNC1 in TK1(+) cells increased mtdTTP pool size and radioactivity, suggesting an involvement in the import of thymidine phosphates. Thus PNC1 is a component of the network regulating the mtdTTP pool in human cells.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nucleotide Transport Proteins/metabolism , Thymidine Kinase/physiology , Thymine Nucleotides/metabolism , Biological Transport , Blotting, Western , Cells, Cultured , Cytosol/enzymology , Humans , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Hum Mol Genet ; 21(9): 2005-16, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22279085

ABSTRACT

FANCM is the most highly conserved protein within the Fanconi anaemia (FA) tumour suppressor pathway. However, although FANCM contains a helicase domain with translocase activity, this is not required for its role in activating the FA pathway. Instead, we show here that FANCM translocaseactivity is essential for promoting replication fork stability. We demonstrate that cells expressing translocase-defective FANCM show altered global replication dynamics due to increased accumulation of stalled forks that subsequently degenerate into DNA double-strand breaks, leading to ATM activation, CTBP-interacting protein (CTIP)-dependent end resection and homologous recombination repair. Accordingly, abrogation of ATM or CTIP function in FANCM-deficient cells results in decreased cell survival. We also found that FANCM translocase activity protects cells from accumulating 53BP1-OPT domains, which mark lesions resulting from problems arising during replication. Taken together, these data show that FANCM plays an essential role in maintaining chromosomal integrity by promoting the recovery of stalled replication forks and hence preventing tumourigenesis.


Subject(s)
DNA Helicases/metabolism , DNA Replication/physiology , Nucleotide Transport Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line , DNA Breaks, Double-Stranded , DNA Helicases/antagonists & inhibitors , DNA Helicases/genetics , DNA Repair , DNA Replication/genetics , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Homologous Recombination , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...