Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Clin Transl Immunology ; 11(2): e1375, 2022.
Article in English | MEDLINE | ID: mdl-35228870

ABSTRACT

OBJECTIVE: Pre-existing neutralising antibodies (NAbs) to adeno-associated viruses (AAVs) remain an impediment for systemically administered AAV-mediated gene therapy treatment in many patients, and various strategies are under investigation to overcome this limitation. Here, IgG-degrading enzymes (Ides) derived from bacteria of the genus Streptococcus were tested for their ability to cleave human IgG and allow AAV-mediated transduction in individuals with pre-existing NAbs. METHODS: Cleavage activity of three different Ides was evaluated in vitro in serum from different species. Passively immunised mice or non-human primates (NHP) with naturally occurring anti-AAV NAbs were used to define the optimal IdeS dose and administration window for AAVAnc80 and AAV8 vectors in mice and AAV3B in NHPs. RESULTS: The selected candidate, IdeS, was found to be highly efficient at cleaving human IgG, less efficient against NHP IgG and inefficient against mouse IgG. In vivo, we observed differences in how IdeS affected liver transduction in the presence of NAbs depending on the AAV serotype. For AAVAnc80 and AAV3B, the best transduction levels were achieved when the vector was administered after IgG digestion products were cleared from circulation. However, for AAV8 we only observed a modest and transient inhibition of transduction by IdeS cleavage products. CONCLUSION: Preconditioning with IdeS represents a unique treatment opportunity for patients primarily excluded from participation in gene therapy clinical trials because of elevated circulating anti-AAV NAb levels. However, careful determination of the optimal IdeS dose and timing for the administration of each AAV serotype is essential for optimal transduction.

2.
J Lipid Res ; 63(1): 100147, 2022 01.
Article in English | MEDLINE | ID: mdl-34752805

ABSTRACT

The myelin sheath, which is wrapped around axons, is a lipid-enriched structure produced by mature oligodendrocytes. Disruption of the myelin sheath is observed in several neurological diseases, such as multiple sclerosis. A crucial component of myelin is sphingomyelin, levels of which can be increased by ABCA8, a member of the ATP-binding cassette transporter family. ABCA8 is highly expressed in the cerebellum, specifically in oligodendroglia. However, whether ABCA8 plays a role in myelination and mechanisms that would underlie this role remain unknown. Here, we found that the absence of Abca8b, a mouse ortholog of ABCA8, led to decreased numbers of cerebellar oligodendrocyte precursor cells (OPCs) and mature oligodendrocytes in mice. We show that in oligodendrocytes, ABCA8 interacts with chondroitin sulfate proteoglycan 4 (CSPG4), a molecule essential for OPC proliferation, migration, and myelination. In the absence of Abca8b, localization of CSPG4 to the plasma membrane was decreased, contributing to reduced cerebellar CSPG4 expression. Cerebellar CSPG4+ OPCs were also diminished, leading to decreased mature myelinating oligodendrocyte numbers and cerebellar myelination levels in Abca8b-/- mice. In addition, electron microscopy analyses showed that the number of nonmyelinated cerebellar axons was increased, whereas cerebellar myelin thickness (g-ratio), myelin sheath periodicity, and axonal diameter were all decreased, indicative of disordered myelin ultrastructure. In line with disrupted cerebellar myelination, Abca8b-/- mice showed lower cerebellar conduction velocity and disturbed locomotion. In summary, ABCA8 modulates cerebellar myelination, in part through functional regulation of the ABCA8-interacting protein CSPG4. Our findings suggest that ABCA8 disruption may contribute to the pathophysiology of myelin disorders.


Subject(s)
Oligodendrocyte Precursor Cells
3.
J Inherit Metab Dis ; 44(6): 1369-1381, 2021 11.
Article in English | MEDLINE | ID: mdl-33896013

ABSTRACT

Phenylketonuria (PKU) is the most common inborn error of metabolism of the liver, and results from mutations of both alleles of the phenylalanine hydroxylase gene (PAH). As such, it is a suitable target for gene therapy via gene delivery with a recombinant adeno-associated virus (AAV) vector. Here we use the synthetic AAV vector Anc80 via systemic administration to deliver a functional copy of a codon-optimized human PAH gene, with or without an intron spacer, to the Pahenu2 mouse model of PKU. Dose-dependent transduction of the liver and expression of PAH mRNA were present with both vectors, resulting in significant and durable reduction of circulating phenylalanine, reaching near control levels in males. Coat color of treated Pahenu2 mice reflected an increase in pigmentation from brown to the black color of control animals, further indicating functional restoration of phenylalanine metabolism and its byproduct melanin. There were no adverse effects associated with administration of AAV up to 5 × 1012 VG/kg, the highest dose tested. Only minor and/or transient variations in some liver enzymes were observed in some of the AAV-dosed animals which were not associated with pathology findings in the liver. Finally, there was no impact on cell turnover or apoptosis as evaluated by Ki-67 and TUNEL staining, further supporting the safety of this approach. This study demonstrates the therapeutic potential of AAV Anc80 to safely and durably cure PKU in a mouse model, supporting development for clinical consideration.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Phenylalanine Hydroxylase/genetics , Phenylketonurias/therapy , Animals , Cell Line , DNA, Recombinant/administration & dosage , Disease Models, Animal , Female , Genetic Vectors/genetics , Hair Color , Humans , Injections, Intravenous , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Phenylalanine/blood , Phenylalanine Hydroxylase/immunology , Phenylalanine Hydroxylase/metabolism , Transduction, Genetic/methods
4.
Arterioscler Thromb Vasc Biol ; 39(3): 387-401, 2019 03.
Article in English | MEDLINE | ID: mdl-30651003

ABSTRACT

Objective- Atherosclerotic coronary artery disease is the leading cause of death worldwide, and current treatment options are insufficient. Using systems-level network cluster analyses on a large coronary artery disease case-control cohort, we previously identified PCSK3 (proprotein convertase subtilisin/kexin family member 3; FURIN) as a member of several coronary artery disease-associated pathways. Thus, our objective is to determine the role of FURIN in atherosclerosis. Approach and Results- In vitro, FURIN inhibitor treatment resulted in reduced monocyte migration and reduced macrophage and vascular endothelial cell inflammatory and cytokine gene expression. In vivo, administration of an irreversible inhibitor of FURIN, α-1-PDX (α1-antitrypsin Portland), to hyperlipidemic Ldlr-/- mice resulted in lower atherosclerotic lesion area and a specific reduction in severe lesions. Significantly lower lesional macrophage and collagen area, as well as systemic inflammatory markers, were observed. MMP2 (matrix metallopeptidase 2), an effector of endothelial function and atherosclerotic lesion progression, and a FURIN substrate was significantly reduced in the aorta of inhibitor-treated mice. To determine FURIN's role in vascular endothelial function, we administered α-1-PDX to Apoe-/- mice harboring a wire injury in the common carotid artery. We observed significantly decreased carotid intimal thickness and lower plaque cellularity, smooth muscle cell, macrophage, and inflammatory marker content, suggesting protection against vascular remodeling. Overexpression of FURIN in this model resulted in a significant 67% increase in intimal plaque thickness, confirming that FURIN levels directly correlate with atherosclerosis. Conclusions- We show that systemic inhibition of FURIN in mice decreases vascular remodeling and atherosclerosis. FURIN-mediated modulation of MMP2 activity may contribute to the atheroprotection observed in these mice.


Subject(s)
Atherosclerosis/prevention & control , Furin/antagonists & inhibitors , Plaque, Atherosclerotic/drug therapy , alpha 1-Antitrypsin/therapeutic use , Animals , Aorta/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Artery, Common , Disease Progression , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Furin/genetics , Furin/physiology , Gene Expression Regulation/drug effects , Macrophages/physiology , Male , Matrix Metalloproteinase 2/analysis , Mice , Mice, Inbred C57BL , Monocytes/physiology , Plaque, Atherosclerotic/pathology , Receptors, LDL/deficiency , Tunica Intima/drug effects , Tunica Intima/pathology , Vascular Remodeling , alpha 1-Antitrypsin/pharmacology
5.
FASEB J ; 32(7): 3792-3802, 2018 07.
Article in English | MEDLINE | ID: mdl-29481310

ABSTRACT

Bile acids (BAs) are surfactant molecules that regulate the intestinal absorption of lipids. Thus, the modulation of BAs represents a potential therapy for nonalcoholic fatty liver disease (NAFLD), which is characterized by hepatic accumulation of fat and is a major cause of liver disease worldwide. Cyp8b1 is a critical modulator of the hydrophobicity index of the BA pool. As a therapeutic proof of concept, we aimed to determine the impact of Cyp8b1 inhibition in vivo on BA pool composition and as protection against NAFLD. Inhibition of Cyp8b1 expression in mice led to a remodeling of the BA pool, which altered its signaling properties and decreased intestinal fat absorption. In a model of cholesterol-induced NAFLD, Cyp8b1 knockdown significantly decreased steatosis and hepatic lipid content, which has been associated with an increase in fecal lipid and BA excretion. Moreover, inhibition of Cyp8b1 not only decreased hepatic lipid accumulation, but also resulted in the clearance of previously accumulated hepatic cholesterol, which led to a regression in hepatic steatosis. Taken together, our data demonstrate that Cyp8b1 inhibition is a viable therapeutic target of crucial interest for metabolic diseases, such as NAFLD.-Chevre, R., Trigueros-Motos, L., Castaño, D., Chua, T., Corlianò, M., Patankar, J. V., Sng, L., Sim, L., Juin, T. L., Carissimo, G., Ng, L. F. P., Yi, C. N. J., Eliathamby, C. C., Groen, A. K., Hayden, M. R., Singaraja, R. R. Therapeutic modulation of the bile acid pool by Cyp8b1 knockdown protects against nonalcoholic fatty liver disease in mice.


Subject(s)
Bile Acids and Salts/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Steroid 12-alpha-Hydroxylase/genetics , Animals , Female , HEK293 Cells , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/therapy , RNAi Therapeutics , Steroid 12-alpha-Hydroxylase/metabolism
6.
Arterioscler Thromb Vasc Biol ; 37(11): 2147-2155, 2017 11.
Article in English | MEDLINE | ID: mdl-28882873

ABSTRACT

OBJECTIVE: High-density lipoproteins (HDL) are considered to protect against atherosclerosis in part by facilitating the removal of cholesterol from peripheral tissues. However, factors regulating lipid efflux are incompletely understood. We previously identified a variant in adenosine triphosphate-binding cassette transporter A8 (ABCA8) in an individual with low HDL cholesterol (HDLc). Here, we investigate the role of ABCA8 in cholesterol efflux and in regulating HDLc levels. APPROACH AND RESULTS: We sequenced ABCA8 in individuals with low and high HDLc and identified, exclusively in low HDLc probands, 3 predicted deleterious heterozygous ABCA8 mutations (p.Pro609Arg [P609R], IVS17-2 A>G and p.Thr741Stop [T741X]). HDLc levels were lower in heterozygous mutation carriers compared with first-degree family controls (0.86±0.34 versus 1.17±0.26 mmol/L; P=0.005). HDLc levels were significantly decreased by 29% (P=0.01) in Abca8b-/- mice on a high-cholesterol diet compared with wild-type mice, whereas hepatic overexpression of human ABCA8 in mice resulted in significant increases in plasma HDLc and the first steps of macrophage-to-feces reverse cholesterol transport. Overexpression of wild-type but not mutant ABCA8 resulted in a significant increase (1.8-fold; P=0.01) of cholesterol efflux to apolipoprotein AI in vitro. ABCA8 colocalizes and interacts with adenosine triphosphate-binding cassette transporter A1 and further potentiates adenosine triphosphate-binding cassette transporter A1-mediated cholesterol efflux. CONCLUSIONS: ABCA8 facilitates cholesterol efflux and modulates HDLc levels in humans and mice.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol, Dietary/blood , Cholesterol, HDL/blood , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Adult , Aged , Animals , Apolipoprotein A-I/blood , Apolipoprotein B-100/blood , Biological Transport , Biomarkers/blood , COS Cells , Case-Control Studies , Chlorocebus aethiops , DNA Mutational Analysis , Diet, High-Fat , Feces/chemistry , Female , HEK293 Cells , Heredity , Heterozygote , Humans , Liver/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mutation , Pedigree , Phenotype , Transfection
7.
J Lipid Res ; 56(10): 1993-2001, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26255038

ABSTRACT

A low level of HDL cholesterol (HDL-C) is a common clinical scenario and an important marker for increased cardiovascular risk. Many patients with very low or very high HDL-C have a rare mutation in one of several genes, but identification of the molecular abnormality in patients with extreme HDL-C is rarely performed in clinical practice. We investigated the accuracy and diagnostic yield of a targeted next-generation sequencing (NGS) assay for extreme levels of HDL-C. We developed a targeted NGS panel to capture the exons, intron/exon boundaries, and untranslated regions of 26 genes with highly penetrant effects on plasma lipid levels. We sequenced 141 patients with extreme HDL-C levels and prioritized variants in accordance with medical genetics guidelines. We identified 35 pathogenic and probably pathogenic variants in HDL genes, including 21 novel variants, and performed functional validation on a subset of these. Overall, a molecular diagnosis was established in 35.9% of patients with low HDL-C and 5.2% with high HDL-C, and all prioritized variants identified by NGS were confirmed by Sanger sequencing. Our results suggest that a molecular diagnosis can be identified in a substantial proportion of patients with low HDL-C using targeted NGS.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Cardiovascular Diseases/genetics , Cholesterol, HDL/blood , Cholesterol, HDL/genetics , High-Throughput Nucleotide Sequencing/methods , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , ATP Binding Cassette Transporter 1/blood , Alleles , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Exons , Female , Genetic Association Studies , Humans , Introns , Male , Middle Aged , Risk Factors
8.
Sci Signal ; 7(322): ra37, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24757177

ABSTRACT

In many cell types, nuclear A-type lamins regulate multiple cellular functions, including higher-order genome organization, DNA replication and repair, gene transcription, and signal transduction; however, their role in specialized immune cells remains largely unexplored. We showed that the abundance of A-type lamins was almost negligible in resting naïve T lymphocytes, but was increased upon activation of the T cell receptor (TCR). The increase in lamin-A was an early event that accelerated formation of the immunological synapse between T cells and antigen-presenting cells. Polymerization of F-actin in T cells is a critical step for immunological synapse formation, and lamin-A interacted with the linker of nucleoskeleton and cytoskeleton (LINC) complex to promote F-actin polymerization. We also showed that lamin-A expression accelerated TCR clustering and led to enhanced downstream signaling, including extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, as well as increased target gene expression. Pharmacological inhibition of the ERK pathway reduced lamin-A-dependent T cell activation. Moreover, mice lacking lamin-A in immune cells exhibited impaired T cell responses in vivo. These findings underscore the importance of A-type lamins for TCR activation and identify lamin-A as a previously unappreciated regulator of the immune response.


Subject(s)
Actin Cytoskeleton/immunology , Actins/immunology , Immunological Synapses/immunology , Lamin Type A/immunology , Lymphocyte Activation/physiology , T-Lymphocytes/immunology , Actin Cytoskeleton/genetics , Actins/genetics , Animals , Humans , Immunological Synapses/genetics , Jurkat Cells , Lamin Type A/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/cytology
9.
Arterioscler Thromb Vasc Biol ; 33(6): 1248-56, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23448971

ABSTRACT

OBJECTIVE: Different vascular beds show differing susceptibility to the development of atherosclerosis, but the molecular mechanisms underlying these differences are incompletely understood. This study aims to identify factors that contribute to the phenotypic heterogeneity of distinct regions of the adult vasculature. APPROACH AND RESULTS: High-throughput mRNA profiling in adult mice reveals higher expression of the homeobox paralogous genes 6 to 10 (Hox6-10) in the athero-resistant thoracic aorta (TA) than in the athero-susceptible aortic arch (AA). Higher homeobox gene expression also occurs in rat and porcine TA, and is maintained in primary smooth muscle cells isolated from TA (TA-SMCs) compared with cells from AA (AA-SMCs). This region-specific homeobox gene expression pattern is also observed in human embryonic stem cells differentiated into neuroectoderm-SMCs and paraxial mesoderm-SMCs, which give rise to AA-SMCs and TA-SMCs, respectively. We also find that, compared with AA and AA-SMCs, TA and TA-SMCs have lower activity of the proinflammatory and proatherogenic nuclear factor-κB (NF-κB) and lower expression of NF-κB target genes, at least in part attributable to HOXA9-dependent inhibition. Conversely, NF-κB inhibits HOXA9 promoter activity and mRNA expression in SMCs. CONCLUSION: Our findings support a model of Hox6-10-specified positional identity in the adult vasculature that is established by embryonic cues independently of environmental factors and is conserved in different mammalian species. Differential homeobox gene expression contributes to maintaining phenotypic differences between SMCs from athero-resistant and athero-susceptible regions, at least in part through feedback regulatory mechanisms involving inflammatory mediators, for example, reciprocal inhibition between HOXA9 and NF-κB.


Subject(s)
Aorta, Thoracic/embryology , Gene Expression Regulation, Developmental , Genes, Homeobox/genetics , NF-kappa B/genetics , Phenotype , Adult , Animals , Aorta/embryology , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Cell Differentiation/genetics , Cells, Cultured , Humans , Mice , Models, Animal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Species Specificity , Swine
10.
BMC Cancer ; 12: 434, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-23017148

ABSTRACT

BACKGROUND: Nucleoside analogs used in the chemotherapy of solid tumors, such as the capecitabine catabolite 5'-deoxy-5-fluorouridine (5'-DFUR) trigger a transcriptomic response that involves the aquaglyceroporin aquaporin 3 along with other p53-dependent genes. Here, we examined whether up-regulation of aquaporin 3 (AQP3) mRNA in cancer cells treated with 5'-DFUR represents a collateral transcriptomic effect of the drug, or conversely, AQP3 participates in the activity of genotoxic agents. METHODS: The role of AQP3 in cell volume increase, cytotoxicity and cell cycle arrest was analyzed using loss-of-function approaches. RESULTS: 5'-DFUR and gemcitabine, but not cisplatin, stimulated AQP3 expression and cell volume, which was partially and significantly blocked by knockdown of AQP3. Moreover, AQP3 siRNA significantly blocked other effects of nucleoside analogs, including G1/S cell cycle arrest, p21 and FAS up-regulation, and cell growth inhibition. Short incubations with 5-fluorouracil (5-FU) also induced AQP3 expression and increased cell volume, and the inhibition of AQP3 expression significantly blocked growth inhibition triggered by this drug. To further establish whether AQP3 induction is related to cell cycle arrest and apoptosis, cells were exposed to long incubations with escalating doses of 5-FU. AQP3 was highly up-regulated at doses associated with cell cycle arrest, whereas at doses promoting apoptosis induction of AQP3 mRNA expression was reduced. CONCLUSIONS: Based on the results, we propose that the aquaglyceroporin AQP3 is required for cytotoxic activity of 5'-DFUR and gemcitabine in the breast cancer cell line MCF7 and the colon adenocarcinoma cell line HT29, and is implicated in cell volume increase and cell cycle arrest.


Subject(s)
Antineoplastic Agents/pharmacology , Aquaporin 3/genetics , Gene Expression Regulation, Neoplastic/drug effects , Nucleosides/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Aquaporin 3/metabolism , Blotting, Western , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Size/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Floxuridine/pharmacology , Fluorouracil/pharmacology , HT29 Cells , Humans , MCF-7 Cells , Nucleosides/chemistry , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , fas Receptor/genetics , fas Receptor/metabolism , Gemcitabine
11.
Front Biosci (Schol Ed) ; 3(4): 1285-97, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622271

ABSTRACT

Hutchinson-Gilford Progeria Syndrome (HGPS), a rare human disease characterized by premature aging, is mainly caused by the abnormal accumulation of progerin, a mutant form of the mammalian nuclear envelope component lamin A. HGPS patients exhibit vascular alterations and die at an average age of 13 years, predominantly from myocardial infarction or stroke. Animal models of HGPS have been a valuable tool in the study of the pathological processes implicated in the origin of this disease and its associated cardiovascular alterations. Some of the molecular mechanisms of HGPS might be relevant to the process of normal aging, since progerin is detected in cells from normal elderly humans. Conversely, processes linked to normal aging, such as the increase in oxidative stress, might be relevant to the pathogenic mechanisms of HGPS. In this review, we discuss recent advances in the understanding of the molecular mechanisms underlying the cardiovascular alterations associated with HGPS, the potential role of oxidative stress, and therapeutic approaches for the treatment of this devastating disease.


Subject(s)
Aging/physiology , Cardiovascular Diseases/physiopathology , Genetic Therapy/methods , Nuclear Proteins/metabolism , Oxidative Stress/physiology , Progeria/physiopathology , Protein Precursors/metabolism , Alternative Splicing , Animals , Cholesterol/biosynthesis , Diphosphonates/therapeutic use , Humans , Lamin Type A/genetics , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Oligonucleotides/therapeutic use , Progeria/drug therapy , Protein Precursors/genetics , Terpenes/metabolism
12.
Haematologica ; 93(12): 1843-51, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18945750

ABSTRACT

BACKGROUND: The nucleoside analogue fludarabine is used in the treatment of chronic lymphocytic leukemia. It triggers p53-mediated apoptosis, although the mutational status of p53 does not fully account for heterogeneity in responsiveness to treatment. The aim of this study was to identify new genes implicated in fludarabine action as well as to determine the role of equilibrative nucleoside transporters (ENT) in the transcriptomic response triggered by this drug in chronic lymphocytic leukemia cells bearing wild type p53. DESIGN AND METHODS: We performed gene expression profiling in cells from two fludarabine-sensitive and two fludarabine-resistant cases of chronic lymphocytic leukemia treated with fludarabine either in the presence or the absence of nitrobenzylthioinosine, a hENT1-specific blocker. Twenty selected fludarabine-inducible genes were validated using Taqman low-density arrays in cells from 20 chronic lymphocytic leukemia patients with the same experimental design. RESULTS: Sixteen of the twenty genes (DDB2, GADD45A, TYMS, BAX, TIGAR, FAS, TNFSF7, TNFSF9, CCNG1, CDKN1A, MDM2, SESN1, MAP4K4, PPM1D, OSBPL3 and WIG1) correlated with the ex vivo sensitivity of chronic lymphocytic leukemia cells to fludarabine, TIGAR (TP53-induced glycolysis and apoptosis regulator) being the gene that showed the strongest correlation (p<0.0001; r2= 0.6022).We observed that the transcriptomic response was weakly sensitive to the hENT1 blocker nitrobenzylthioinosine. Interestingly, we also found a correlation between hENT2 expression and induction of TIGAR after fludarabine treatment. CONCLUSIONS: We demonstrate a correlation between the recently described p53-inducible apoptosis gene TIGAR and both sensitivity to fludarabine and hENT2 expression in chronic lymphocytic leukemia cells. These results, as well as the variability in fludarabine response among chronic lymphocytic leukemia patients with wild type p53, support the major role of hENT2 in the uptake of fludarabine into chronic lymphocytic leukemia cells.


Subject(s)
Equilibrative-Nucleoside Transporter 2/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Vidarabine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins , Equilibrative-Nucleoside Transporter 2/metabolism , Female , Gene Expression Profiling , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Phosphoric Monoester Hydrolases , Tumor Cells, Cultured , Vidarabine/pharmacokinetics , Vidarabine/pharmacology
13.
Nucleosides Nucleotides Nucleic Acids ; 27(6): 769-78, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18600539

ABSTRACT

Nucleoside transporter proteins, CNT and ENT, encoded by gene families SLC28 and SLC29, respectively, mediate the uptake of natural nucleosides (among them adenosine) and are major routes of entry for a variety of nucleoside analogs used in anticancer and antiviral therapies. Expression of NT proteins is apparently redundant in most cell types, and the elucidation of their particular physiological roles still remains elusive. Moreover, transporter-mediated uptake of nucleoside-derived anticancer drugs is crucial for the pharmacogenomic response triggered by these molecules in tumor cells. This review focuses on recent data demonstrating that nucleoside transporters, particularly CNTs, can play physiological roles other than salvage, whereas particular NT isoforms can significantly contribute to the transcriptomic response triggered by nucleoside analogs in cancer cells.


Subject(s)
Nucleoside Transport Proteins/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Equilibrative Nucleoside Transport Proteins/metabolism , Humans , Nucleosides/chemistry , Nucleosides/metabolism , Nucleosides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL