Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Healthc Manage Forum ; 34(6): 311-315, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34535064

ABSTRACT

An awakening to systemic anti-Black racism, anti-Indigenous racism, and harmful colonial structures in the context of a pandemic has made health inequities and injustices impossible to ignore, and is driving healthcare organizations to establish and strengthen approaches to inclusion, diversity, equity, and accessibility (IDEA). Health research and care organizations, which are shaping the future of healthcare, have a responsibility to make IDEA central to their missions. Many organizations are taking concrete action critically important to embedding IDEA principles, but durable change will not be achieved until IDEA becomes a core leadership competency. Drawing from the literature and consultation with individuals recognized for excellence in IDEA-informed leadership, this study will help Canadian healthcare and health research leaders-particularly those without lived experience-understand what it means to embed IDEA within traditional leadership competencies and propose opportunities to achieve durable change by rethinking governance, mentorship, and performance management through an IDEA lens.


Subject(s)
Leadership , Racism , Canada , Delivery of Health Care , Humans , Population Groups
2.
Lab Chip ; 21(9): 1788-1797, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33734246

ABSTRACT

Biological research and many cell-based therapies rely on the successful delivery of cargo materials into cells. Intracellular delivery in an in vitro setting refers to a variety of physical and biochemical techniques developed for conducting rapid and efficient transport of materials across the plasma membrane. Generally, the techniques that are time-efficient (e.g., electroporation) suffer from heterogeneity and low cellular viability, and those that are precise (e.g., microinjection) suffer from low-throughput and are labor-intensive. Here, we present a novel in vitro microfluidic strategy for intracellular delivery, which is based on the acoustic excitation of adherent cells. Strong mechanical oscillations, mediated by Lamb waves, inside a microfluidic channel facilitate the cellular uptake of different size (e.g., 3-500 kDa, plasmid encoding EGFP) cargo materials through endocytic pathways. We demonstrate successful delivery of 500 kDa dextran to various adherent cell lines with unprecedented efficiency in the range of 65-85% above control. We also show that actuation voltage and treatment duration can be tuned to control the dosage of delivered substances. High viability (≥91%), versatility across different cargo materials and various adherent cell lines, scalability to hundreds of thousands of cells per treatment, portability, and ease-of-operation are among the unique features of this acoustofluidic strategy. Potential applications include targeting through endocytosis-dependant pathways in cellular disorders, such as lysosomal storage diseases, which other physical methods are unable to address. This novel acoustofluidic method achieves rapid, uniform, and scalable delivery of material into cells, and may find utility in lab-on-a-chip applications.


Subject(s)
Electroporation , Lab-On-A-Chip Devices , Acoustics , Cell Membrane , Cell Survival
3.
RSC Adv ; 11(52): 32824-32829, 2021 Oct 04.
Article in English | MEDLINE | ID: mdl-35493567

ABSTRACT

Adherent cultured cells are widely used biological tools for a variety of biochemical and biotechnology applications, including drug screening and gene expression analysis. One critical step in culturing adherent cells is the dissociation of cell monolayers into single-cell suspensions. Different enzymatic and non-enzymatic methods have been proposed for this purpose. Trypsinization, the most common enzymatic method for dislodging adhered cells, can be detrimental to cells, as it can damage cell membranes and ultimately cause cell death. Additionally, all available techniques require a prolonged treatment duration, typically on the order of minutes (5-10 min). Dissociation of cells becomes even more challenging in microfluidic devices, where, due to the nature of low Reynolds number flow and reduced mixing efficiency, multiple washing steps and prolonged trypsinization may be necessary to treat all cells. Here, we report a novel acoustofluidic method for the detachment of cells adhered onto a microchannel surface without exposing the cells to any enzymatic or non-enzymatic chemicals. This method enables a rapid (i.e., on the order of seconds), cost-effective, and easy-to-operate cell detachment strategy, yielding a detachment efficiency of ∼99% and cellular viability similar to that of the conventional trypsinization method. Also, as opposed to biochemical-based techniques (e.g., enzymatic), in our approach, cells are exposed to the dissociating agent (i.e., substrate-mediated acoustic excitation and microstreaming flow) only for as long as they remain attached to the substrate. After dissociation, the effect of acoustic excitation is reduced to microstreaming flow, therefore, minimizing unwanted effects of the dissociating agent on the cell phenotype. Additionally, our results suggest that cell excitation at acoustic powers lower than that required for complete cell detachment can potentially be employed for probing the adhesion strength of cell-substrate attachment. This novel approach can, therefore, be used for a wide range of lab-on-a-chip applications.

4.
Lancet ; 393(10171): 587-593, 2019 02 09.
Article in English | MEDLINE | ID: mdl-30739694

ABSTRACT

In August 2018, the president of the World Bank noted that "'Human capital'-the potential of individuals-is going to be the most important long-term investment any country can make for its people's future prosperity and quality of life". Nevertheless, leaders and practitioners in academic science and medicine continue to be unaware of and poorly educated about the nature, extent, and impact of barriers to full participation of women and minorities in science and medicine around the world. This lack of awareness and education results in failures to fully mobilise the human capital of half the population and limits global technological and medical advancements. The chronic lack of recruitment, promotion, and retention of women in science and medicine is due to systemic, structural, organisational, institutional, cultural, and societal barriers to equity and inclusion. These barriers must be identified and removed through increased awareness of the challenges combined with evidence-based, data-driven approaches leading to measurable targets and outcomes. In this Review, we discuss these issues and highlight actions that could achieve gender equality in science and medicine. We survey approaches and insights that have helped to identify and remove systemic bias and barriers in science and medicine, and propose tools that will help organisational change toward gender equality. We describe tools that include formal legislation and mandated quotas at national or large-scale levels (eg, gender parity), techniques that increase fairness (eg, gender equity) through facilitated organisational cultural change at institutional levels, and professional development of core competencies at individual levels. This Review is not intended to be an extensive analysis of all the literature currently available on achieving gender equality in academic medicine and science, but rather, a reflection on finding multifactorial solutions.


Subject(s)
Medicine , Science , Sexism/prevention & control , Women's Rights , Career Choice , Career Mobility , Female , Humans , Leadership , Organizational Innovation , Organizational Objectives
5.
FASEB J ; 33(3): 3841-3850, 2019 03.
Article in English | MEDLINE | ID: mdl-30521377

ABSTRACT

Equilibrative nucleoside transporters (ENTs) translocate nucleosides and nucleobases across plasma membranes, as well as a variety of anti-cancer, -viral, and -parasite nucleoside analogs. They are also key members of the purinome complex and regulate the protective and anti-inflammatory effects of adenosine. Despite their important role, little is known about the mechanisms involved in their regulation. We conducted membrane yeast 2-hybrid and coimmunoprecipitation studies and identified, for the first time to our knowledge, the existence of protein-protein interactions between human ENT1 and ENT2 (hENT1 and hENT2) proteins in human cells and the formation of hetero- and homo-oligomers at the plasma membrane and the submembrane region. The use of NanoLuc Binary Technology allowed us to analyze changes in the oligomeric status of hENT1 and hENT2 and how they rapidly modify the uptake profile for nucleosides and nucleobases and allow cells to respond promptly to external signals or changes in the extracellular environment. These changes in hENTs oligomerization are triggered by PKC activation and subsequent action of protein phosphatase 1.-Grañe-Boladeras, N., Williams, D., Tarmakova, Z., Stevanovic, K., Villani, L. A., Mehrabi, P., Siu, K. W. M., Pastor-Anglada, M., Coe, I. R. Oligomerization of equilibrative nucleoside transporters: a novel regulatory and functional mechanism involving PKC and PP1.


Subject(s)
Equilibrative Nucleoside Transporter 1/metabolism , Equilibrative-Nucleoside Transporter 2/metabolism , Protein Multimerization , HEK293 Cells , Humans , Protein Binding , Protein Kinase C/metabolism , Protein Phosphatase 1/metabolism
6.
Ultrasonics ; 89: 110-117, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29775835

ABSTRACT

Ultrasonically-stimulated microbubbles enhance the therapeutic effects of various chemotherapy drugs. However, the application of ultrasound and microbubbles (USMB) for enhancing the therapeutic effect of nucleoside analogs, which are used as front-line treatments in a range of cancers, and its underlying mechanism is not well understood. This study investigated the effect of gemcitabine, a nucleoside analog drug, in combination with USMB in increasing cell cytotoxicity relative to either treatment alone in BxPC3 pancreatic cancer cells. Cells were sonicated using low frequency (0.5 MHz) ultrasound in combination with Definity® microbubbles (1.7% v/v) in the presence of 1 µM of gemcitabine for a total of 2 h. USMB in combination with gemcitabine decreased cell viability (48 h) to 44.7 ±â€¯5.2%, 27.7 ±â€¯4.3%, and 12.5 ±â€¯3.4% with increasing ultrasound peak negative pressures (220, 360, 530 kPa) from 84.7 ±â€¯3.6%, 54.2 ±â€¯3.8%, and 26.8 ±â€¯3.0%, respectively, when USMB was applied in the absence of drug. We further confirmed that USMB did not enhance the internalization of 1 µM of a radiolabeled nucleoside analog (2-chloroadenosine) at each of the three chosen ultrasound PNPs, determined by radiolabeled scintillation counting. These data suggest that USMB in combination with nucleoside analog drugs leads to an additive effect on cell toxicity and that USMB does not impair transporter-mediated uptake of nucleoside analogs.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Fluorocarbons/pharmacology , Microbubbles , Pancreatic Neoplasms/drug therapy , Ultrasonic Therapy/methods , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/pharmacology , Flow Cytometry , Humans , In Vitro Techniques , Gemcitabine
7.
Science ; 360(6385): 162-163, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29650665
9.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 631-642, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27906634

ABSTRACT

Human equilibrative nucleoside transporter 1 (hENT1) is a major route of entry of nucleosides and nucleoside analog drugs. The regulation of hENT1 is poorly understood in spite of its clinical importance as a drug transporter. Immunofluorescence microscopy and fluorescence-activated cell sorting suggested that cytidine pre-treatment (40 µM, 6 h) promotes hENT1 internalization in a way that does not affect either hENT1-mediated nucleoside uptake or gemcitabine-induced cytotoxicity. The Scatchard plot analyses of our NBTI binding data support previous speculations that hENT1 proteins exist as two sub-populations, and suggest that cytidine pre-treatment leads to the internalization of one population.


Subject(s)
Equilibrative Nucleoside Transporter 1/physiology , Antimetabolites, Antineoplastic/pharmacology , Binding Sites , Cytidine/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , HEK293 Cells , Humans , Protein Transport , Thioinosine/analogs & derivatives , Thioinosine/metabolism , Gemcitabine
10.
Biosci Rep ; 36(4)2016 08.
Article in English | MEDLINE | ID: mdl-27480168

ABSTRACT

Human equilibrative nucleoside transporter 1 (hENT1) transports nucleosides and nucleoside analogue drugs across cellular membranes and is necessary for the uptake of many anti-cancer, anti-parasitic and anti-viral drugs. Previous work, and in silico prediction, suggest that hENT1 is glycosylated at Asn(48) in the first extracellular loop of the protein and that glycosylation plays a role in correct localization and function of hENT1. Site-directed mutagenesis of wild-type (wt) hENT1 removed potential glycosylation sites. Constructs (wt 3xFLAG-hENT1, N48Q-3xFLAG-hENT1 or N288Q-3xFLAG-hENT2) were transiently transfected into HEK293 cells and cell lysates were treated with or without peptide-N-glycosidase F (PNGase-F), followed by immunoblotting analysis. Substitution of N48 prevents hENT1 glycosylation, confirming a single N-linked glycosylation site. N48Q-hENT1 protein is found at the plasma membrane in HEK293 cells but at lower levels compared with wt hENT1 based on S-(4-nitrobenzyl)-6-thioinosine (NBTI) binding analysis (wt 3xFLAG-ENT1 Bmax, 41.5±2.9 pmol/mg protein; N48Q-3xFLAG-ENT1 Bmax, 13.5±0.45 pmol/mg protein) and immunofluorescence microscopy. Although present at the membrane, chloroadenosine transport assays suggest that N48Q-hENT1 is non-functional (wt 3xFLAG-ENT1, 170.80±44.01 pmol/mg protein; N48Q-3xFLAG-ENT1, 57.91±17.06 pmol/mg protein; mock-transfected 74.31±19.65 pmol/mg protein). Co-immunoprecipitation analyses suggest that N48Q ENT1 is unable to interact with self or with wt hENT1. Based on these data we propose that glycosylation at N48 is critical for the localization, function and oligomerization of hENT1.


Subject(s)
Equilibrative Nucleoside Transporter 1/metabolism , Biological Transport/physiology , Cell Line , Glycosylation , HEK293 Cells , Humans , Mutagenesis, Site-Directed/methods , Thioinosine/analogs & derivatives , Thioinosine/metabolism
11.
Cell Mol Life Sci ; 73(23): 4559-4575, 2016 12.
Article in English | MEDLINE | ID: mdl-27271752

ABSTRACT

Nucleosides participate in many cellular processes and are the fundamental building blocks of nucleic acids. Nucleoside transporters translocate nucleosides across plasma membranes although the mechanism by which nucleos(t)ides are translocated into the nucleus during DNA replication is unknown. Here, we identify two novel functional splice variants of equilibrative nucleoside transporter 2 (ENT2), which are present at the nuclear envelope. Under proliferative conditions, these splice variants are up-regulated and recruit wild-type ENT2 to the nuclear envelope to translocate nucleosides into the nucleus for incorporation into DNA during replication. Reduced presence of hENT2 splice variants resulted in a dramatic decrease in cell proliferation and dysregulation of cell cycle due to a lower incorporation of nucleotides into DNA. Our findings support a novel model of nucleoside compartmentalisation at the nuclear envelope and translocation into the nucleus through hENT2 and its variants, which are essential for effective DNA synthesis and cell proliferation.


Subject(s)
Cell Cycle , Cell Nucleus/metabolism , Equilibrative-Nucleoside Transporter 2/metabolism , Nucleosides/metabolism , Alternative Splicing/genetics , Biological Transport , Cell Cycle/genetics , Cell Proliferation , Equilibrative-Nucleoside Transporter 2/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Neoplasms/genetics , Neoplasms/pathology , Nuclear Envelope/metabolism , Protein Interaction Mapping , Protein Isoforms/genetics , Protein Isoforms/metabolism , Thymidine/metabolism
12.
Am J Physiol Cell Physiol ; 310(10): C808-20, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27009875

ABSTRACT

Equilibrative nucleoside transporters (ENTs) facilitate the flux of nucleosides, such as adenosine, and nucleoside analog (NA) drugs across cell membranes. A correlation between adenosine flux and calcium-dependent signaling has been previously reported; however, the mechanistic basis of these observations is not known. Here we report the identification of the calcium signaling transducer calmodulin (CaM) as an ENT1-interacting protein, via a conserved classic 1-5-10 motif in ENT1. Calcium-dependent human ENT1-CaM protein interactions were confirmed in human cell lines (HEK293, RT4, U-87 MG) using biochemical assays (HEK293) and the functional assays (HEK293, RT4), which confirmed modified nucleoside uptake that occurred in the presence of pharmacological manipulations of calcium levels and CaM function. Nucleoside and NA drug uptake was significantly decreased (∼12% and ∼39%, respectively) by chelating calcium (EGTA, 50 µM; BAPTA-AM, 25 µM), whereas increasing intracellular calcium (thapsigargin, 1.5 µM) led to increased nucleoside uptake (∼26%). Activation of N-methyl-d-aspartate (NMDA) receptors (in U-87 MG) by glutamate (1 mM) and glycine (100 µM) significantly increased nucleoside uptake (∼38%) except in the presence of the NMDA receptor antagonist, MK-801 (50 µM), or CaM antagonist, W7 (50 µM). These data support the existence of a previously unidentified novel receptor-dependent regulatory mechanism, whereby intracellular calcium modulates nucleoside and NA drug uptake via CaM-dependent interaction of ENT1. These findings suggest that ENT1 is regulated via receptor-dependent calcium-linked pathways resulting in an alteration of purine flux, which may modulate purinergic signaling and influence NA drug efficacy.


Subject(s)
Calcium/metabolism , Calmodulin/chemistry , Calmodulin/metabolism , Equilibrative Nucleoside Transporter 1/chemistry , Equilibrative Nucleoside Transporter 1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Binding Sites , Calcium/chemistry , HEK293 Cells , Humans , Protein Binding , Receptors, N-Methyl-D-Aspartate/chemistry
13.
Biochim Biophys Acta ; 1853(7): 1636-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25862406

ABSTRACT

The human amino acid transporter SLC1A5 (ASCT2) contains two N-glycosylation sites (N163 and N212) located in the large extracellular loop. In the homology structural model of ASCT2 these Asn residues are extracellularly exposed. Mutants of the two Asn exhibited altered electrophoretic mobility. N163Q and N212Q displayed multiple bands with apparent molecular masses from 80kDa to 50kDa. N163/212Q displayed a single band of 50kDa corresponding to the unglycosylated protein. The presence in membrane of WT and mutants was evaluated by protein biotinylation assay followed by immunoblotting. The double mutation significantly impaired the presence of the protein in membrane, without impairment in protein synthesis. [(3)H]glutamine transport was measured in cells transiently transfected with the WT or mutants. N163/212Q exhibited a strongly reduced transport activity correlating with reduced surface expression. The same proteins extracted from cells and reconstituted in liposomes showed comparable transport activities demonstrating that the intrinsic transport function of the mutants was not affected. The rate of endocytosis of ASCT2 was assayed by a reversible biotinylation strategy. N212Q and N163/212Q showed strongly increased rates of endocytosis respect to WT. ASCT2 stability was determined using cycloheximide. N163Q or N163/212Q showed a slightly or significantly lower stability with respect to WT. To assess trafficking to the membrane, a brefeldin-based assay, which caused retention of proteins in ER, was performed. One hour after brefeldin removal WT protein was localized to the plasma membrane while the double mutant was localized in the cytosol. The results demonstrate that N-glycosylation is critical for trafficking.


Subject(s)
Amino Acid Transport System ASC/metabolism , Cell Membrane/metabolism , Amino Acid Transport System ASC/chemistry , Animals , Biological Assay , Biotinylation , Computational Biology , Endocytosis , Endoplasmic Reticulum/metabolism , Glycosylation , HEK293 Cells , Humans , Minor Histocompatibility Antigens , Models, Molecular , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Protein Stability , Protein Transport , Rats , Structural Homology, Protein , Time Factors
14.
J Virol ; 89(1): 626-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25339775

ABSTRACT

UNLABELLED: Ribavirin (RBV) continues to be an important component of interferon-free hepatitis C treatment regimens, as RBV alone does not inhibit hepatitis C virus (HCV) replication effectively; the reason for this ineffectiveness has not been established. In this study, we investigated the RBV resistance mechanism using a persistently HCV-infected cell culture system. The antiviral activity of RBV against HCV was progressively impaired in the persistently infected culture, whereas interferon lambda 1 (IFN-λ1), a type III IFN, showed a strong antiviral response and induced viral clearance. We found that HCV replication in persistently infected cultures induces an autophagy response that impairs RBV uptake by preventing the expression of equilibrative nucleoside transporter 1 (ENT1). The Huh-7.5 cell line treated with an autophagy inducer, Torin 1, downregulated membrane expression of ENT1 and terminated RBV uptake. In contrast, the autophagy inhibitors hydroxychloroquine (HCQ), 3-methyladenine (3-MA), and bafilomycin A1 (BafA1) prevented ENT1 degradation and enhanced RBV antiviral activity. The HCV-induced autophagy response, as well as treatment with Torin 1, degrades clathrin heavy chain expression in a hepatoma cell line. Reduced expression of the clathrin heavy chain by HCV prevents ENT1 recycling to the plasma membrane and forces ENT1 to the lysosome for degradation. This study provides a potential mechanism for the impairment of RBV antiviral activity in persistently HCV-infected cell cultures and suggests that inhibition of the HCV-induced autophagy response could be used as a strategy for improving RBV antiviral activity against HCV infection. IMPORTANCE: The results from this work will allow a review of the competing theories of antiviral therapy development in the field of HCV virology. Ribavirin (RBV) remains an important component of interferon-free hepatitis C treatment regimens. The reason why RBV alone does not inhibit HCV replication effectively has not been established. This study provides a potential mechanism for why RBV antiviral activity is impaired in persistently HCV-infected cell cultures and suggests that inhibition of the HCV-induced autophagy response could be used as a strategy to increase RBV antiviral activity against HCV infection. Therefore, it is anticipated that this work would generate a great deal of interest, not only among virologists but also among the general public.


Subject(s)
Antiviral Agents/metabolism , Clathrin/metabolism , Drug Resistance , Equilibrative Nucleoside Transporter 1/metabolism , Hepacivirus/drug effects , Ribavirin/metabolism , Cell Line , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Protein Transport
15.
Neurochem Int ; 73: 229-37, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24704797

ABSTRACT

The purinome is a rich complex of proteins and cofactors that are involved in fundamental aspects of cellular homeostasis and cellular responses. The purinome is evolutionarily ancient and is made up of thousands of members. Our understanding of the mechanisms linking some parts of this complex network and the physiological relevance of the various connections is well advanced. However, our understanding of other parts of the purinome is less well developed. Our research focuses on the adenosine or nucleoside transporters (NTs), which are members of the membrane purinome. Nucleoside transporters are integral membrane proteins that are responsible for the flux of nucleosides, such as adenosine, and nucleoside analog drugs, used in a variety of anti-cancer, anti-viral and anti-parasite therapies, across cell membranes. Nucleoside transporters form the SLC28 and SLC29 families of solute carriers and the protein members of these families are widely distributed in human tissues including the central nervous system (CNS). NTs modulate purinergic signaling in the CNS primarily through their effects on modulating prevailing adenosine levels inside and outside the cell. By clearing the extracellular milieu of adenosine, NTs can terminate adenosine receptor-dependent signaling and this raises the possibility of regulatory feedback loops that tie together receptor signaling with transporter function. Despite the important role of NTs as modulators of purinergic signaling in the human body, very little is known about the nature or underlying mechanisms of regulation of either the SLC28 or SLC29 families, particularly within the context of the CNS purinome. Here we provide a brief overview of our current understanding of the regulation of members of the SLC29 family and highlight some interesting avenues for future research.


Subject(s)
Nucleoside Transport Proteins/physiology , Purines/metabolism , Humans , Nervous System Diseases/genetics , Nervous System Diseases/physiopathology , Nucleoside Transport Proteins/genetics , Receptors, Purinergic/genetics , Receptors, Purinergic/physiology
16.
Biochem Biophys Res Commun ; 443(2): 505-10, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24333872

ABSTRACT

Nucleoside transport is important for nucleic acid synthesis in cells that cannot synthesize nucleosides de novo, and for entry of many cytotoxic nucleoside analog drugs used in chemotherapy. This study demonstrates that various steroid hormones induce inhibition of nucleoside transport in mammalian cells. We analyzed the inhibitory effects of estradiol (E2) on nucleoside transport using SH-SY5Y human neuroblastoma cells. We observed inhibitory effects after acute treatment with E2, which lasted in the presence of E2. However, when E2 was removed, the effect immediately disappeared, suggesting that E2 effects are not mediated through the canonical regulatory pathway of steroid hormones, such as transcriptional regulation. We also discovered that E2 could competitively inhibit thymidine uptake and binding of the labeled nucleoside transporter inhibitor, S-[4-nitrobenzyl]-6-thioinosine (NBTI), indicating that E2 binds to endogenous nucleoside transporters, leading to inhibition of nucleoside transport. We then tested the effects of various steroids on nucleoside uptake in NBTI-sensitive cells, SH-SY5Y and NBTI-insensitive cells H9c2 rat cardiomyoblasts. We found E2 and progesterone clearly inhibited both NBTI-sensitive and insensitive uptake at micromolar concentrations. Taken together, we concluded that steroid hormones function as novel nucleoside transport inhibitors by competition with nucleosides for their transporters.


Subject(s)
Estradiol/administration & dosage , Myocytes, Cardiac/metabolism , Neuroblastoma/metabolism , Nucleoside Transport Proteins/antagonists & inhibitors , Nucleoside Transport Proteins/metabolism , Nucleosides/metabolism , Progesterone/administration & dosage , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Gonadal Steroid Hormones/administration & dosage , Humans , Myocytes, Cardiac/drug effects , Rats
17.
Purinergic Signal ; 10(2): 305-12, 2014.
Article in English | MEDLINE | ID: mdl-24163005

ABSTRACT

The adenosine transporter 1 (ENT1) transports nucleosides, such as adenosine, and cytotoxic nucleoside analog drugs. ENT1 is well established to play a role in adenosinergic signaling in the cardiovascular system by modulating adenosine levels. Moderate ethanol consumption is cardioprotective and underlying mechanisms of action are not clear although adenosinergic signaling has been implicated. Here, we show that ethanol (5-200 mM) significantly reduces ENT1-dependent [(3)H] 2-chloroadenosine uptake (by up to 27 %) in the cardiomyocyte cell line, HL-1. Inhibition or absence of ENT1 is known to be cardioprotective, suggesting that the interaction of ethanol with ENT1 may promote adenosinergic cardioprotective pathways in the cardiovasculature.Ethanol sensitivity of adenosine uptake is altered by pharmacological activation of PKA and PKC. Primary cardiomyocytes from PKCε-null mice have significantly greater sensitivity to inhibition (by approximately 37 %) of adenosine uptake by ethanol than controls. These data suggest that the presence of ethanol may compromise ENT1-dependent nucleoside analog drug cytotoxicity, and indeed, ethanol (5 mM) reduces the cytotoxic effects of gemcitabine (2 nM), an anti-cancer drug, in the human cancer cell line, HTB2. Thus, the pharmacological inhibition of ENT1 by ethanol may contribute to ethanol-dependent cardioprotection but compromise gemcitabine cytotoxicity.


Subject(s)
Central Nervous System Depressants/pharmacology , Equilibrative Nucleoside Transporter 1/metabolism , Ethanol/pharmacology , Myocytes, Cardiac/metabolism , Protein Kinase C-epsilon/metabolism , Adenosine/metabolism , Animals , Antimetabolites, Antineoplastic/toxicity , Cell Line , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/toxicity , Humans , Mice , Mice, Knockout , Myocytes, Cardiac/drug effects , Gemcitabine
18.
J Clin Invest ; 122(2): 693-710, 2012 02.
Article in English | MEDLINE | ID: mdl-22269324

ABSTRACT

A complex biologic network regulates kidney perfusion under physiologic conditions. This system is profoundly perturbed following renal ischemia, a leading cause of acute kidney injury (AKI) - a life-threatening condition that frequently complicates the care of hospitalized patients. Therapeutic approaches to prevent and treat AKI are extremely limited. Better understanding of the molecular pathways promoting postischemic reflow could provide new candidate targets for AKI therapeutics. Due to its role in adapting tissues to hypoxia, we hypothesized that extracellular adenosine has a regulatory function in the postischemic control of renal perfusion. Consistent with the notion that equilibrative nucleoside transporters (ENTs) terminate adenosine signaling, we observed that pharmacologic ENT inhibition in mice elevated renal adenosine levels and dampened AKI. Deletion of the ENTs resulted in selective protection in Ent1-/- mice. Comprehensive examination of adenosine receptor-knockout mice exposed to AKI demonstrated that renal protection by ENT inhibitors involves the A2B adenosine receptor. Indeed, crosstalk between renal Ent1 and Adora2b expressed on vascular endothelia effectively prevented a postischemic no-reflow phenomenon. These studies identify ENT1 and adenosine receptors as key to the process of reestablishing renal perfusion following ischemic AKI. If translatable from mice to humans, these data have important therapeutic implications.


Subject(s)
Acute Kidney Injury/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Ischemia/metabolism , Regional Blood Flow/physiology , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Adenosine/metabolism , Animals , Cell Line , Chimerism , Dipyridamole/therapeutic use , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Equilibrative Nucleoside Transporter 1/genetics , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , No-Reflow Phenomenon , Nucleoside Transport Proteins/antagonists & inhibitors , Nucleoside Transport Proteins/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism
19.
Life Sci ; 89(17-18): 621-30, 2011 Oct 24.
Article in English | MEDLINE | ID: mdl-21872611

ABSTRACT

AIMS: Equilibrative nucleoside transporters (ENT) modulate the flux of adenosine. The ENT1-null (KO) mouse heart is endogenously cardioprotected but the cellular basis of this phenotype is unknown. Therefore, we investigated the cellular mechanisms underlying ENT1-mediated cardioprotection. MAIN METHODS: Circulating adenosine levels were measured in WT and KO mice. Cellular levels of nucleosides and nucleotides were investigated in isolated adult cardiomyocytes from WT and KO mice using HPLC following hypoxic challenge (30 min, 2% O(2)). Changes in hypoxic gene expression were analyzed by PCR arrays and cAMP levels were measured to investigate contributions from adenosine receptors. KEY FINDINGS: Circulating adenosine levels were significantly higher in KO (416±42nmol/l, n=12) compared to WT animals (208±21, n=13, p<0.001). Absence of ENT1 led to an elevated expression of genes involved in cardioprotective pathways compared to WT cardiomyocytes. Following hypoxic challenge, extracellular adenosine levels were significantly elevated in KO (4360±1840 pmol/mg protein) versus WT cardiomyocytes (3035±730 pmol/mg protein, n≥12, p<0.05). This effect was enhanced in the presence of dipyridamole (30 µM), which inhibits ENT1 and ENT2. Enhanced extracellular adenosine levels in ENT1-null cardiomyocytes appeared to come from a pool of extracellular nucleotides including IMP, AMP and ADP. Adenosine receptor (AR) activation mimicked increases in cAMP levels due to hypoxic challenge suggesting that ENT1 modulates AR-dependent signaling. SIGNIFICANCE: ENT1 contributes to modulation of extracellular adenosine levels and subsequent purinergic signaling via ARs. ENT1-null mice possess elevated circulating adenosine levels and reduced cellular uptake resulting in a perpetually cardioprotected phenotype.


Subject(s)
Equilibrative Nucleoside Transporter 1/genetics , Myocytes, Cardiac/metabolism , Nucleosides/metabolism , Adenosine/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Cyclic AMP/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Female , Gene Deletion , Gene Expression Regulation , Inosine/metabolism , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Protein Kinases/metabolism , Receptors, Purinergic/metabolism
20.
Mol Membr Biol ; 28(6): 412-26, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21809900

ABSTRACT

Equilibrative Nucleoside Transporters (SLC29) are a family of proteins that transport nucleosides, nucleobases and nucleoside analogue drugs across cellular membranes. ENT1 is expressed ubiquitously in mammalian tissues and responsible for a significant portion of nucleoside analog drug uptake in humans. Despite the important clinical role of ENT1, many aspects of the regulation of this protein remain unknown. A major outstanding question in this field is the whether ENT1 is phosphorylated directly. To answer this question, we overexpressed tagged human (h) and mouse (m) ENT1, affinity purified protein using the tag, conducted phosphoamino acid analysis and found that m/hENT1 is predominantly phosphorylated at serine residues. The large intracellular loop of ENT1, between transmembrane domains 6 and 7, has been suggested to be a site of regulation by phosphorylation, therefore we generated His/Ubiquitin tagged peptides of this region and used them for in vitro kinase assays to identify target serines. Our data support a role for PKA and PKC in the phosphorylation of ENT1 within the intracellular loop and show that PKA can phosphorylate multiple sites within this loop while PKC specifically targets serines 279 and 286 and threonine 274. These data demonstrate, for the first time, that ENT1 is a phosphoprotein that can be directly phosphorylated at several sites by more than one kinase. The presence of multiple kinase targets within the loop suggests that ENT1 phosphorylation is considerably more complex than previously thought and thus ENT1 may be subject to phosphorylation by multiple pathways.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Protein Kinase C/metabolism , Animals , COS Cells , Chlorocebus aethiops , Equilibrative Nucleoside Transporter 1/chemistry , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...