Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1357716, 2024.
Article in English | MEDLINE | ID: mdl-38384461

ABSTRACT

Objectives: Despite their efficacy, some immunotherapies have been shown to induce immune-related adverse events, including the potentially life-threatening cytokine release syndrome (CRS), calling for reliable and translational preclinical models to predict potential safety issues and investigate their rescue. Here, we tested the reliability of humanized BRGSF mice for the assessment of therapeutics-induced CRS features in preclinical settings. Methods: BRGSF mice reconstituted with human umbilical cord blood CD34+ cells (BRGSF-CBC) were injected with anti-CD3 antibody (OKT3), anti-CD3/CD19 bispecific T-cell engager Blinatumomab, or VISTA-targeting antibody. Human myeloid and dendritic cells' contribution was investigated in hFlt3L-boosted BRGSF-CBC mice. OKT3 treatment was also tested in human PBMC-reconstituted BRGSF mice (BRGSF-PBMC). Cytokine release, immune cell distribution, and clinical signs were followed. Results: OKT3 injection in BRGSF-CBC mice induced hallmark features of CRS, specifically inflammatory cytokines release, modifications of immune cell distribution and activation, body weight loss, and temperature drop. hFlt3L-boosted BRGSF-CBC mice displayed enhanced CRS features, revealing a significant role of myeloid and dendritic cells in this process. Clinical CRS-managing treatment Infliximab efficiently attenuated OKT3-induced toxicity. Comparison of OKT3 treatment's effect on BRGSF-CBC and BRGSF-PBMC mice showed broadened CRS features in BRGSF-CBC mice. CRS-associated features were also observed in hFlt3L-boosted BRGSF-CBC mice upon treatment with other T-cell or myeloid-targeting compounds. Conclusions: These data show that BRGSF-CBC mice represent a relevant model for the preclinical assessment of CRS and CRS-managing therapies. They also confirm a significant role of myeloid and dendritic cells in CRS development and exhibit the versatility of this model for therapeutics-induced safety assessment.


Subject(s)
Cytokine Release Syndrome , Muromonab-CD3 , Humans , Mice , Animals , Muromonab-CD3/pharmacology , Leukocytes, Mononuclear , Reproducibility of Results , Cytokines , Dendritic Cells
2.
Mol Imaging Biol ; 18(5): 768-75, 2016 10.
Article in English | MEDLINE | ID: mdl-27122234

ABSTRACT

PURPOSE: Nectin-4 is selectively overexpressed in a variety of cancers and is currently under clinical investigation as a therapeutic target. A monoclonal antibody against nectin-4 (AGS-22M6) was evaluated as an Immuno-positron emission tomography (ImmunoPET) reagent. Its ability to assay nectin-4 expression as well as detect nectin-4 positive tumors in the liver and bone was evaluated using mouse models. PROCEDURES: The biodistribution of [(89)Zr]AGS-22M6 was evaluated in mice bearing tumors with varying levels of nectin-4 expression. An isogenic breast cancer tumor line was used to model metastatic liver and bone disease in mice. The biodistribution of [(18)F]AGS-22M6 in cynomolgus monkeys was evaluated. RESULTS: A positive correlation was demonstrated between tumor nectin-4 expression and [(89)Zr]AGS-22M6 uptake. Tumors in the liver and bone were detected and differentiated based on nectin-4 expression. [(18)F]AGS-22M6 showed limited uptake in cynomolgus monkey tissues. CONCLUSIONS: [(89)Zr]AGS-22M6 is a promising ImmunoPET reagent that can assay nectin-4 expression in both primary and metastatic lesions.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Adhesion Molecules/immunology , Positron-Emission Tomography/methods , Xenograft Model Antitumor Assays , Animals , Bone Neoplasms/secondary , Cell Line, Tumor , Humans , Immunohistochemistry , Indicators and Reagents , Liver Neoplasms/secondary , Macaca fascicularis , Mice , Nectins , Tissue Distribution , Zirconium/chemistry
3.
J Exp Med ; 211(3): 473-86, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24567448

ABSTRACT

Pharmacological targeting of metabolic processes in cancer must overcome redundancy in biosynthetic pathways. Deoxycytidine (dC) triphosphate (dCTP) can be produced both by the de novo pathway (DNP) and by the nucleoside salvage pathway (NSP). However, the role of the NSP in dCTP production and DNA synthesis in cancer cells is currently not well understood. We show that acute lymphoblastic leukemia (ALL) cells avoid lethal replication stress after thymidine (dT)-induced inhibition of DNP dCTP synthesis by switching to NSP-mediated dCTP production. The metabolic switch in dCTP production triggered by DNP inhibition is accompanied by NSP up-regulation and can be prevented using DI-39, a new high-affinity small-molecule inhibitor of the NSP rate-limiting enzyme dC kinase (dCK). Positron emission tomography (PET) imaging was useful for following both the duration and degree of dCK inhibition by DI-39 treatment in vivo, thus providing a companion pharmacodynamic biomarker. Pharmacological co-targeting of the DNP with dT and the NSP with DI-39 was efficacious against ALL models in mice, without detectable host toxicity. These findings advance our understanding of nucleotide metabolism in leukemic cells, and identify dCTP biosynthesis as a potential new therapeutic target for metabolic interventions in ALL and possibly other hematological malignancies.


Subject(s)
Biosynthetic Pathways/physiology , Deoxycytidine Kinase/antagonists & inhibitors , Deoxycytosine Nucleotides/biosynthesis , Disease Eradication/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Biosynthetic Pathways/drug effects , Deoxycytosine Nucleotides/metabolism , Mice , Positron-Emission Tomography , Thymidine/pharmacology
4.
J Med Chem ; 56(17): 6696-708, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23947754

ABSTRACT

Combined inhibition of ribonucleotide reductase and deoxycytidine kinase (dCK) in multiple cancer cell lines depletes deoxycytidine triphosphate pools leading to DNA replication stress, cell cycle arrest, and apoptosis. Evidence implicating dCK in cancer cell proliferation and survival stimulated our interest in developing small molecule dCK inhibitors. Following a high throughput screen of a diverse chemical library, a structure-activity relationship study was carried out. Positron Emission Tomography (PET) using (18)F-L-1-(2'-deoxy-2'-FluoroArabinofuranosyl) Cytosine ((18)F-L-FAC), a dCK-specific substrate, was used to rapidly rank lead compounds based on their ability to inhibit dCK activity in vivo. Evaluation of a subset of the most potent compounds in cell culture (IC50 = ∼1-12 nM) using the (18)F-L-FAC PET pharmacodynamic assay identified compounds demonstrating superior in vivo efficacy.


Subject(s)
Deoxycytidine Kinase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Positron-Emission Tomography/methods , Cell Line, Tumor , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Monte Carlo Method , Spectrometry, Mass, Electrospray Ionization
5.
J Nucl Med ; 54(10): 1820-4, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23978446

ABSTRACT

UNLABELLED: We report on a radiopharmaceutical imaging platform designed to capture the kinetics of cellular responses to drugs. METHODS: A portable in vitro molecular imaging system comprising a microchip and a ß-particle imaging camera permitted routine cell-based radioassays of small numbers of either suspended or adherent cells. We investigated the kinetics of responses of model lymphoma and glioblastoma cancer cell lines to (18)F-FDG uptake after drug exposure. Those responses were correlated with kinetic changes in the cell cycle or with changes in receptor tyrosine kinase signaling. RESULTS: The platform enabled direct radioassays of multiple cell types and yielded results comparable to those from conventional approaches; however, the platform used smaller sample sizes, permitted a higher level of quantitation, and did not require cell lysis. CONCLUSION: The kinetic analysis enabled by the platform provided a rapid (≈ 1 h) drug screening assay.


Subject(s)
Drug Evaluation, Preclinical/instrumentation , Miniaturization/instrumentation , Molecular Imaging/instrumentation , Systems Integration , Biological Transport/drug effects , Cell Line, Tumor , Fluorodeoxyglucose F18/metabolism , Glycolysis/drug effects , Humans , Kinetics
6.
Mol Imaging Biol ; 15(3): 273-81, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23054556

ABSTRACT

PURPOSE: This study aims to use a simple, quantitative method to compare the HSV1sr39TK/(18) F-FHBG PET reporter gene/PET reporter probe (PRG/PRP) system with PRGs derived from human nucleoside kinases. PROCEDURES: The same adenovirus vector is used to express alternative PRGs. Equal numbers of vectors are injected intravenously into mice. After PRP imaging, quantitative hepatic PET signals are normalized for transduction by measuring hepatic viral genomes. RESULTS: The same adenovirus vector was used to express equivalent amounts of HSV1sr39TK, mutant human thymidine kinase 2 (TK2-DM), and mutant human deoxycytidine kinase (dCK-A100VTM) in mouse liver. HSV1sr39TK expression was measured with (18) F-FHBG, TK2-DM and dCK-A100VTM with (18) F-L-FMAU. TK2-DM/(18) F-L-FMAU and HSV1sr39TK/(18) F-FHBG had equivalent sensitivities; dCK-A100VTM/(18) F-L-FMAU was twice as sensitive as HSV1sr39TK/(18) F-FHBG. CONCLUSIONS: The human PRG/PRP sensitivities are comparable and/or better than HSV1sr39TK/(18) F-FHBG. However, for clinical use, identification of the best PRP substrate for each enzyme, characterization of probe distribution, and consequences of overexpressing nucleoside kinases must be evaluated.


Subject(s)
Adenoviridae/genetics , Genes, Reporter/genetics , Molecular Probes/metabolism , Positron-Emission Tomography/methods , Animals , Cloning, Molecular , Deoxycytidine Kinase/genetics , Female , Gene Expression , Genetic Vectors , Genome, Viral/genetics , HEK293 Cells , HeLa Cells , Humans , Liver/metabolism , Mice , Plasmids/genetics , Time Factors , Viral Load
7.
J Exp Med ; 209(12): 2215-28, 2012 Nov 19.
Article in English | MEDLINE | ID: mdl-23148236

ABSTRACT

Nucleotide deficiency causes replication stress (RS) and DNA damage in dividing cells. How nucleotide metabolism is regulated in vivo to prevent these deleterious effects remains unknown. In this study, we investigate a functional link between nucleotide deficiency, RS, and the nucleoside salvage pathway (NSP) enzymes deoxycytidine kinase (dCK) and thymidine kinase (TK1). We show that inactivation of dCK in mice depletes deoxycytidine triphosphate (dCTP) pools and induces RS, early S-phase arrest, and DNA damage in erythroid, B lymphoid, and T lymphoid lineages. TK1(-/-) erythroid and B lymphoid lineages also experience nucleotide deficiency but, unlike their dCK(-/-) counterparts, they still sustain DNA replication. Intriguingly, dCTP pool depletion, RS, and hematopoietic defects induced by dCK inactivation are almost completely reversed in a newly generated dCK/TK1 double-knockout (DKO) mouse model. Using NSP-deficient DKO hematopoietic cells, we identify a previously unrecognized biological activity of endogenous thymidine as a strong inducer of RS in vivo through TK1-mediated dCTP pool depletion. We propose a model that explains how TK1 and dCK "tune" dCTP pools to both trigger and resolve RS in vivo. This new model may be exploited therapeutically to induce synthetic sickness/lethality in hematological malignancies, and possibly in other cancers.


Subject(s)
DNA Replication/physiology , Hematopoiesis/physiology , Metabolic Networks and Pathways/physiology , Models, Biological , Nucleosides/metabolism , Nucleotides/deficiency , Stress, Physiological/physiology , Animals , Blotting, Western , Bromodeoxyuridine , Deoxycytidine Kinase/genetics , Deoxycytidine Kinase/metabolism , Deoxycytosine Nucleotides/metabolism , Flow Cytometry , Immunophenotyping , Mice , Mice, Knockout , Nucleotides/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/metabolism
8.
Theranostics ; 2(4): 374-91, 2012.
Article in English | MEDLINE | ID: mdl-22509201

ABSTRACT

Positron emission tomography (PET) imaging reporter genes (IRGs) and PET reporter probes (PRPs) are amongst the most valuable tools for gene and cell therapy. PET IRGs/PRPs can be used to non-invasively monitor all aspects of the kinetics of therapeutic transgenes and cells in all types of living mammals. This technology is generalizable and can allow long-term kinetics monitoring. In gene therapy, PET IRGs/PRPs can be used for whole-body imaging of therapeutic transgene expression, monitoring variations in the magnitude of transgene expression over time. In cell or cellular gene therapy, PET IRGs/PRPs can be used for whole-body monitoring of therapeutic cell locations, quantity at all locations, survival and proliferation over time and also possibly changes in characteristics or function over time. In this review, we have classified PET IRGs/PRPs into two groups based on the source from which they were derived: human or non-human. This classification addresses the important concern of potential immunogenicity in humans, which is important for expansion of PET IRG imaging in clinical trials. We have then discussed the application of this technology in gene/cell therapy and described its use in these fields, including a summary of using PET IRGs/PRPs in gene and cell therapy clinical trials. This review concludes with a discussion of the future direction of PET IRGs/PRPs and recommends cell and gene therapists collaborate with molecular imaging experts early in their investigations to choose a PET IRG/PRP system suitable for progression into clinical trials.

9.
J Nucl Med ; 53(2): 275-80, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22302964

ABSTRACT

UNLABELLED: The ability to measure tumor determinants of response to nucleoside analog (NA) chemotherapy agents such as gemcitabine and related compounds could significantly affect the management of several types of cancer. Previously we showed that the accumulation in tumors of the new PET tracer 1-(2'-deoxy-2'-(18)F-fluoro-ß-d-arabinofuranosyl)cytosine ((18)F-FAC) is predictive of responses to gemcitabine. (18)F-FAC retention in cells requires deoxycytidine kinase (dCK), a rate-limiting enzyme in the deoxyribonucleoside salvage metabolism and in gemcitabine conversion from an inactive prodrug to a cytotoxic compound. The objectives of the current study were to determine whether (18)F-FAC tumor uptake is also influenced by cytidine deaminase (CDA), a determinant of resistance to gemcitabine; to develop a new PET assay using (18)F-FAC and the related probe 1-(2'-deoxy-2'-(18)F-fluoro-ß-l-arabinofuranosyl)-5-methylcytosine (l-(18)F-FMAC) to profile tumor lesions for both dCK and CDA enzymatic activities; and to determine whether this PET assay can identify the most effective NA chemotherapy against tumors with differential expression of dCK and CDA. METHODS: Isogenic murine leukemic cell lines with defined dCK and CDA activities were generated by retroviral transduction. A cell viability assay was used to determine the sensitivity of the isogenic cell lines to the dCK-dependent NA prodrugs gemcitabine and clofarabine. In vitro enzymatic and cell-based tracer uptake assays and in vivo PET with (18)F-FAC and l-(18)F-FMAC were used to predict tumor responses to gemcitabine and clofarabine. RESULTS: dCK and CDA activities measured by kinase and tracer uptake assays correlated with the sensitivity of isogenic cell lines to gemcitabine and clofarabine. Coexpression of CDA decreased the sensitivity of dCK-positive cells to gemcitabine treatment in vitro by 15-fold but did not affect responses to clofarabine. Coexpression of CDA decreased (18)F-FAC but not l-(18)F-FMAC, phosphorylation, and uptake by dCK-positive cells. (18)F-FAC and l-(18)F-FMAC PET estimates of the enzymatic activities of dCK and CDA in tumor implants in mice were predictive of responses to gemcitabine and clofarabine treatment in vivo. CONCLUSION: These findings support the utility of PET-based phenotyping of tumor nucleoside metabolism for guiding the selection of NA prodrugs.


Subject(s)
Adenine Nucleotides/therapeutic use , Arabinonucleosides/therapeutic use , Cytarabine/analogs & derivatives , Cytosine , Deoxycytidine/analogs & derivatives , Fluorine Radioisotopes , Positron-Emission Tomography/methods , Adenine Nucleotides/pharmacology , Animals , Arabinonucleosides/pharmacology , Cell Line, Tumor , Clofarabine , Cytidine Deaminase/metabolism , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Deoxycytidine Kinase/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Lymphoid/diagnostic imaging , Leukemia, Lymphoid/drug therapy , Leukemia, Lymphoid/genetics , Leukemia, Lymphoid/metabolism , Mice , Treatment Outcome , Gemcitabine
10.
J Biol Chem ; 287(1): 446-454, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22074768

ABSTRACT

Positron emission tomography (PET) reporter gene imaging can be used to non-invasively monitor cell-based therapies. Therapeutic cells engineered to express a PET reporter gene (PRG) specifically accumulate a PET reporter probe (PRP) and can be detected by PET imaging. Expanding the utility of this technology requires the development of new non-immunogenic PRGs. Here we describe a new PRG-PRP system that employs, as the PRG, a mutated form of human thymidine kinase 2 (TK2) and 2'-deoxy-2'-18F-5-methyl-1-ß-L-arabinofuranosyluracil (L-18F-FMAU) as the PRP. We identified L-18F-FMAU as a candidate PRP and determined its biodistribution in mice and humans. Using structure-guided enzyme engineering, we generated a TK2 double mutant (TK2-N93D/L109F) that efficiently phosphorylates L-18F-FMAU. The N93D/L109F TK2 mutant has lower activity for the endogenous nucleosides thymidine and deoxycytidine than wild type TK2, and its ectopic expression in therapeutic cells is not expected to alter nucleotide metabolism. Imaging studies in mice indicate that the sensitivity of the new human TK2-N93D/L109F PRG is comparable with that of a widely used PRG based on the herpes simplex virus 1 thymidine kinase. These findings suggest that the TK2-N93D/L109F/L-18F-FMAU PRG-PRP system warrants further evaluation in preclinical and clinical applications of cell-based therapies.


Subject(s)
Genes, Reporter/genetics , Positron-Emission Tomography/methods , Protein Engineering/methods , Thymidine Kinase/chemistry , Thymidine Kinase/genetics , Thymidine/analogs & derivatives , Thymidine/metabolism , Adult , Animals , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/chemistry , Arabinofuranosyluracil/metabolism , Arabinofuranosyluracil/pharmacokinetics , Female , Fluorine Radioisotopes , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/metabolism , Guanine/pharmacokinetics , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Humans , Male , Mice , Middle Aged , Models, Molecular , Phosphorylation , Protein Conformation , Thymidine/pharmacokinetics , Thymidine Kinase/metabolism
11.
J Nucl Med ; 51(7): 1092-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20554721

ABSTRACT

UNLABELLED: Deoxycytidine kinase (dCK) is a rate-limiting enzyme in the deoxyribonucleoside salvage pathway and a critical determinant of therapeutic activity for several nucleoside analog prodrugs. We have previously reported the development of 1-(2'-deoxy-2'-(18)F-fluoro-beta-D-arabinofuranosyl)cytosine ((18)F-FAC), a new probe for PET of dCK activity in immune disorders and certain cancers. The objective of the current study was to develop PET probes with improved metabolic stability and specificity for dCK. Toward this goal, several candidate PET probes were synthesized and evaluated in vitro and in vivo. METHODS: High-pressure liquid chromatography was used to analyze the metabolic stability of (18)F-FAC and several newly synthesized analogs with the natural D-enantiomeric sugar configuration or the corresponding unnatural L-configuration. In vitro kinase and uptake assays were used to determine the affinity of the (18)F-FAC L-nucleoside analogs for dCK. The biodistribution of selected L-analogs in mice was determined by small-animal PET/CT. RESULTS: Candidate PET probes were selected using the following criteria: low susceptibility to deamination, high affinity for purified recombinant dCK, high uptake in dCK-expressing cell lines, and biodistribution in mice reflective of the tissue-expression pattern of dCK. Among the 10 newly developed candidate probes, 1-(2'-deoxy-2'-(18)F-fluoro-beta-L-arabinofuranosyl)cytosine (L-(18)F-FAC) and 1-(2'-deoxy-2'-(18)F-fluoro-beta-L-arabinofuranosyl)-5-methylcytosine (L-(18)F-FMAC) most closely matched the selection criteria. The selection of L-(18)F-FAC and L-(18)F-FMAC was validated by showing that these two PET probes could be used to image animal models of leukemia and autoimmunity. CONCLUSION: Promising in vitro and in vivo data warrant biodistribution and dosimetry studies of L-(18)F-FAC and L-(18)F-FMAC in humans.


Subject(s)
Deoxycytidine Kinase/metabolism , Radiopharmaceuticals , Adenine Nucleotides , Animals , Arabinonucleosides , Binding, Competitive , Biotransformation , Cell Line , Chromatography, High Pressure Liquid , Clofarabine , Humans , L-Lactate Dehydrogenase/metabolism , Lymphoproliferative Disorders/diagnostic imaging , Mice , Phosphorylation , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Recombinant Proteins , Substrate Specificity , Tomography, Emission-Computed , Whole Body Imaging
12.
Gastroenterology ; 138(4): 1266-75, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20080095

ABSTRACT

BACKGROUND & AIMS: Uptake of [18F]1-(2'-deoxy-2'-arabinofuranosyl)cytosine (D-FAC) is a trait of activated lymphocytes; its biodistribution predominates in the spleen, thymus, and bone marrow. In addition, D-FAC is taken up at high levels by the intestine. We analyzed the regional specificity of uptake and cell types that mediate it. METHODS: In mice, 3-dimensional isocontour regions of interest were drawn based on computed tomographic images to quantify intestinal signals from micro-positron emission tomography scans. To ascertain the cell type responsible, intestinal epithelium and immune cells were isolated and D-FAC uptake was analyzed in vitro. Mice deficient in mucosal homing (beta7 integrin-/-), enteric microbiota (germ-free), or active for immune colitis (G alpha i2-/- CD3+ transferred into Rag-/- recipients) were studied. RESULTS: Strong uptake of D-FAC was detected throughout the intestine, with greatest signal per region of interest in the duodenum. Fractionation of intestinal cell types after in vivo uptake revealed that the signal was almost entirely from epithelial cells. Among resident immune cell types, CD4+ T cells showed the greatest per-cell and total uptake. D-FAC uptake increased in both intestinal and systemic lymphoid sites during colitis. Compared with fluorodeoxyglucose, increased uptake of D-FAC in the small and large intestine occurred at an earlier stage of disease development. CONCLUSIONS: Uptake of D-FAC is a prominent trait of normal mouse intestinal epithelial cells, which is useful for their noninvasive visualization by positron emission tomography. Increased uptake of D-FAC reflects the activity of the epithelium and lymphocytes, providing a unique early marker of intestinal inflammation.


Subject(s)
Colitis/diagnostic imaging , Cytarabine/analogs & derivatives , Intestinal Mucosa/metabolism , Radiopharmaceuticals/pharmacokinetics , Animals , Apoptosis , Cytarabine/pharmacokinetics , Duodenum/metabolism , Female , Fluorodeoxyglucose F18 , Germ-Free Life , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Positron-Emission Tomography , Tissue Distribution
13.
Proc Natl Acad Sci U S A ; 107(12): 5551-6, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20080663

ABSTRACT

Deoxycytidine kinase (dCK) is a rate-limiting enzyme in deoxyribonucleoside salvage, a metabolic pathway that recycles products of DNA degradation. dCK phosphorylates and therefore activates nucleoside analog prodrugs frequently used in cancer, autoimmunity, and viral infections. In contrast to its well established therapeutic relevance, the biological function of dCK remains enigmatic. Highest levels of dCK expression are found in thymus and bone marrow, indicating a possible role in lymphopoiesis. To test this hypothesis we generated and analyzed dCK knockout (KO) mice. dCK inactivation selectively and profoundly affected T and B cell development. A 90-fold decrease in thymic cellularity was observed in the dCK KO mice relative to wild-type littermates. Lymphocyte numbers in the dCK KO mice were 5- to 13-fold below normal values. The severe impact of dCK inactivation on lymphopoiesis was unexpected given that nucleoside salvage has been thought to play a limited, "fine-tuning" role in regulating deoxyribonucleotide triphosphate pools produced by the de novo pathway. The dCK KO phenotype challenges this view and indicates that, in contrast to the great majority of other somatic cells, normal lymphocyte development critically requires the deoxyribonucleoside salvage pathway.


Subject(s)
B-Lymphocytes/enzymology , Deoxycytidine Kinase/physiology , Lymphopoiesis/physiology , T-Lymphocytes/enzymology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Deoxycytidine Kinase/deficiency , Deoxycytidine Kinase/genetics , Exons , Gene Targeting , Lymphoid Tissue/abnormalities , Lymphopoiesis/immunology , Mice , Mice, Knockout , Models, Biological , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
Proc Natl Acad Sci U S A ; 106(8): 2847-52, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19196993

ABSTRACT

Gemcitabine (2',2'-difluorodeoxycytidine, dFdC) and cytosine arabinoside (cytarabine, ara-C) represent a class of nucleoside analogs used in cancer chemotherapy. Administered as prodrugs, dFdC and ara-C are transported across cell membranes and are converted to cytotoxic derivatives through consecutive phosphorylation steps catalyzed by endogenous nucleoside kinases. Deoxycytidine kinase (DCK) controls the rate-limiting step in the activation cascade of dFdC and ara-C. DCK activity varies significantly among individuals and across different tumor types and is a critical determinant of tumor responses to these prodrugs. Current assays to measure DCK expression and activity require biopsy samples and are prone to sampling errors. Noninvasive methods that can detect DCK activity in tumor lesions throughout the body could circumvent these limitations. Here, we demonstrate an approach to detecting DCK activity in vivo by using positron emission tomography (PET) and (18)F-labeled 1-(2'-deoxy-2'-fluoroarabinofuranosyl) cytosine] ([(18)F]FAC), a PET probe recently developed by our group. We show that [(18)F]FAC is a DCK substrate with an affinity similar to that of dFdC. In vitro, accumulation of [(18)F]FAC in murine and human leukemia cell lines is critically dependent on DCK activity and correlates with dFdC sensitivity. In mice, [(18)F]FAC accumulates selectively in DCK-positive vs. DCK-negative tumors, and [(18)F]FAC microPET scans can predict responses to dFdC. We suggest that [(18)F]FAC PET might be useful for guiding treatment decisions in certain cancers by enabling individualized chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Deoxycytidine/analogs & derivatives , Leukemia/diagnostic imaging , Animals , Antineoplastic Agents/pharmacokinetics , Cytosine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , Humans , Leukemia/drug therapy , Mice , Positron-Emission Tomography , Gemcitabine
15.
Biochemistry ; 45(35): 10591-605, 2006 Sep 05.
Article in English | MEDLINE | ID: mdl-16939211

ABSTRACT

In the Neurospora VS ribozyme, magnesium ions facilitate formation of a loop-loop interaction between stem-loops I and V, which is important for recognition and activation of the stem-loop I substrate. Here, we present the high-resolution NMR structure of stem-loop V (SL5) in the presence of Mg(2+) (SL5(Mg)) and demonstrate that Mg(2+) induces a conformational change in which the SL5 loop adopts a compact structure with most characteristics of canonical U-turn structures. Divalent cation-binding sites were probed with Mn(2+)-induced paramagnetic line broadening and intermolecular NOEs to Co(NH(3))(6)(3+). Structural modeling of Mn(H(2)O)(6)(2+) in SL5(Mg) revealed four divalent cation-binding sites in the loop. Sites 1, 3, and 4 are located in the major groove near multiple phosphate groups, whereas site 2 is adjacent to N7 of G697 and N7 of A698 in the minor groove. Cation-binding sites equivalent to sites 1-3 in SL5 are present in other U-turn motifs, and these metal-binding sites may represent a common feature of the U-turn fold. Although magnesium ions affect the loop conformation, they do not significantly change the conformation of residues 697-699 involved in the proposed Watson-Crick base pairs with stem-loop I. In both the presence and the absence of Mg(2+), G697, A698, and C699 adopt an A-form structure that exposes their Watson-Crick faces, and this is compatible with their proposed interaction with stem-loop I. In SL5(Mg), however, U700 becomes exposed on the minor groove face of the loop in the proximity of the bases of G697, A698, and C699, suggesting that the Mg(2+)-bound conformation of stem-loop V allows additional contacts with stem-loop I. These studies improve our understanding of the role of Mg(2+) in U-turn structures and in substrate recognition by the VS ribozyme.


Subject(s)
Endoribonucleases/chemistry , Magnesium/chemistry , Magnetic Resonance Spectroscopy , RNA, Catalytic/chemistry , Base Pairing , Base Sequence , Binding Sites , Cations, Divalent/chemistry , Models, Molecular , Molecular Sequence Data , Neurospora/enzymology , Neurospora/genetics , Nucleic Acid Conformation , Protein Structure, Tertiary , RNA Stability , Structure-Activity Relationship
16.
J Biol Chem ; 281(3): 1817-26, 2006 Jan 20.
Article in English | MEDLINE | ID: mdl-16269411

ABSTRACT

Many virulence factors in gram-positive bacteria are covalently anchored to the cell-wall peptidoglycan by sortase enzymes, a group of widely distributed cysteine transpeptidases. The Staphylococcus aureus Sortase A protein (SrtA) is the archetypal member of the Sortase family and is activated by Ca2+, an adaptation that may facilitate host colonization as elevated concentrations of this ion are encountered in human tissue. Here we show that a single Ca2+ ion bound to an ordered pocket on SrtA allosterically activates catalysis by modulating both the structure and dynamics of a large active site loop. Detailed nitrogen-15 relaxation measurements indicate that Ca2+ may facilitate the adaptive recognition of the substrate by inducing slow micro- to millisecond time-scale dynamics in the active site. Interestingly, relaxation compensated Carr-Purcell-Meiboom-Gill experiments suggest that the time scale of these motions is directly correlated with ion binding. The results of site-directed mutagenesis indicate that this motional coupling is mediated by the side chain of Glu-171, which is positioned within the beta6/beta7 loop and shown to contribute to Ca2+ binding. The available structural and dynamics data are compatible with a loop closure model of Ca2+ activation, in which the beta6/beta7 loop fluctuates between a binding competent closed form that is stabilized by Ca2+, and an open, highly flexible state that removes key substrate contacting residues from the active site.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Calcium/physiology , Peptidyl Transferases/metabolism , Staphylococcus aureus/enzymology , Allosteric Regulation , Amino Acid Substitution , Aminoacyltransferases/chemistry , Aminoacyltransferases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Base Sequence , Binding Sites , Cysteine Endopeptidases , DNA Primers , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Peptidyl Transferases/chemistry , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/physiology , Virulence
17.
Bioorg Med Chem Lett ; 15(22): 5076-9, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16169722

ABSTRACT

L-Threonine 2 was converted in seven steps into the protected aminomercaptoalcohol 8, a threonine mimic. This compound 8 was coupled to various oligopeptides to produce two different tetrapeptide analogues, for example, 11 and 17, which were shown to inhibit the Sortase enzymes (SrtA and SrtB) via covalent attachment of the thiol groups of 11 and 17 to the catalytically active cysteine residue of the Sortase enzymes.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Aminoacyltransferases/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Butanols/chemical synthesis , Butanols/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Threonine/analogs & derivatives , Butanols/chemistry , Chromatography, High Pressure Liquid , Cysteine Endopeptidases , Enzyme Inhibitors/chemistry , Molecular Structure , Threonine/chemistry
18.
Biochemistry ; 44(11): 4157-70, 2005 Mar 22.
Article in English | MEDLINE | ID: mdl-15766243

ABSTRACT

An important step in the substrate recognition of the Neurospora Varkud Satellite (VS) ribozyme is the formation of a magnesium-dependent loop/loop interaction between the terminal loops of stem-loops I and V. We have studied the structure of stem-loop V by nuclear magnetic resonance spectroscopy and shown that it adopts a U-turn conformation, a common motif found in RNA. Structural comparisons indicate that the U-turn of stem-loop V fulfills some but not all of the structural characteristics found in canonical U-turn structures. This U-turn conformation exposes the Watson-Crick faces of the bases within stem-loop V (G697, A698, and C699) and makes them accessible for interaction with stem-loop I. Using chemical-shift mapping, we show that magnesium ions interact with the loop of the isolated stem-loop V and induce a conformational change that may be important for interaction with stem-loop I. This study expands our understanding of the role of U-turn motifs in RNA structure and function and provides insights into the mechanism of substrate recognition in the VS ribozyme.


Subject(s)
Endoribonucleases/chemistry , Endoribonucleases/metabolism , Magnesium/metabolism , Nuclear Magnetic Resonance, Biomolecular/methods , Nucleic Acid Conformation , RNA, Catalytic/chemistry , RNA, Catalytic/metabolism , RNA, Fungal/chemistry , RNA/chemistry , Base Pairing , Base Sequence , Binding Sites , Carbon Isotopes/chemistry , Cations, Divalent/chemistry , Cations, Divalent/metabolism , Crystallography, X-Ray , Magnesium/chemistry , Molecular Sequence Data , Neurospora/enzymology , RNA/metabolism , RNA, Mitochondrial , Substrate Specificity , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...