Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Mol Syst Biol ; 18(9): e10490, 2022 09.
Article in English | MEDLINE | ID: mdl-36124745

ABSTRACT

Dose-response relationships are a general concept for quantitatively describing biological systems across multiple scales, from the molecular to the whole-cell level. A clinically relevant example is the bacterial growth response to antibiotics, which is routinely characterized by dose-response curves. The shape of the dose-response curve varies drastically between antibiotics and plays a key role in treatment, drug interactions, and resistance evolution. However, the mechanisms shaping the dose-response curve remain largely unclear. Here, we show in Escherichia coli that the distinctively shallow dose-response curve of the antibiotic trimethoprim is caused by a negative growth-mediated feedback loop: Trimethoprim slows growth, which in turn weakens the effect of this antibiotic. At the molecular level, this feedback is caused by the upregulation of the drug target dihydrofolate reductase (FolA/DHFR). We show that this upregulation is not a specific response to trimethoprim but follows a universal trend line that depends primarily on the growth rate, irrespective of its cause. Rewiring the feedback loop alters the dose-response curve in a predictable manner, which we corroborate using a mathematical model of cellular resource allocation and growth. Our results indicate that growth-mediated feedback loops may shape drug responses more generally and could be exploited to design evolutionary traps that enable selection against drug resistance.


Subject(s)
Anti-Bacterial Agents , Tetrahydrofolate Dehydrogenase , Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics , Feedback , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/pharmacology , Trimethoprim/pharmacology
2.
Neurotoxicology ; 92: 15-24, 2022 09.
Article in English | MEDLINE | ID: mdl-35779630

ABSTRACT

Methotrexate (MTX) is a drug widely used for chemotherapy and can reduce cancer cell production by inhibiting dihydrofolate reductase and decreasing cancer cell growth. MTX has a neurotoxic effect on neural stem and glial cells, leading to memory deficits. Chrysin is a natural flavonoid that contains essential biological activities, such as neuroprotective and cognitive-improving properties. Therefore, the aim of the present study was to investigate the protective effect of chrysin against MTX-induced memory impairments related to hippocampal neurogenesis. Seventy-two male Sprague Dawley rats were divided into six groups: control, MTX, chrysin (10 and 30 mg/kg), and MTX+ chrysin (10 and 30 mg/kg) groups. Chrysin (10 and 30 mg/kg) was administered by oral gavage for 15 days. MTX (75 mg/kg) was administered by intravenous injection on days 8 and 15. Spatial and recognition memories were evaluated using the novel object location (NOL) and novel object recognition (NOR) tests, respectively. Moreover, cell proliferation, neuronal cell survival, and immature neurons in the subgranular zone of the hippocampal dentate gyrus were quantified by Ki-67, bromodeoxyuridine/neuronal nuclear protein (BrdU/NeuN), and doublecortin (DCX) immunohistochemistry staining. The results of the MTX group demonstrated that spatial and recognition memories were both impaired. Furthermore, cell division reduction, neuronal cell survival reduction, and immature neuron decreases were detected in the MTX group and not observed in the co-administration groups. Therefore, these results revealed that chrysin could alleviate memory and neurogenesis impairments in MTX-treated rats.


Subject(s)
Methotrexate , Tetrahydrofolate Dehydrogenase , Animals , Bromodeoxyuridine , Cell Proliferation , Cell Survival , Cognition , Dentate Gyrus , Doublecortin Domain Proteins , Flavonoids/pharmacology , Hippocampus , Ki-67 Antigen , Male , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Memory Disorders/prevention & control , Methotrexate/toxicity , Neurogenesis , Neurons , Rats , Rats, Sprague-Dawley , Tetrahydrofolate Dehydrogenase/pharmacology
3.
Hum Mol Genet ; 31(7): 1151-1158, 2022 03 31.
Article in English | MEDLINE | ID: mdl-34788822

ABSTRACT

BACKGROUND: Higher serum homocysteine is associated with cognitive decline in older people. But homocysteine-lowering trials including folic acid (FA) show inconsistent results on cognitive decline. The reduction of FA to dihydrofolate by dihydrofolate reductase (DHFR) is slow in humans. OBJECTIVE: We examined the effects of the DHFR 19-bp deletion/insertion (del/ins) polymorphism on FA-containing treatment on cognitive decline and brain atrophy in older people with mild cognitive impairment (MCI). METHODS: This study used pooled data from two randomized B-vitamin trials on 545 MCI subjects who received either FA-containing B vitamins or placebo for 24 months. Subjects were typed for the DHFR genotype. Primary outcome was the Clinical Dementia Rating scale-global score (CDR-global). Secondary outcomes were CDR-sum of boxes score (CDR-SOB), memory and executive Z-scores and whole brain atrophy rate by serial MRI. RESULTS: The proportions of subjects with del/del, del/ins and ins/ins genotype were 29.5, 44.3 and 26.1%, respectively. DHFR genotypes modified the effects of B vitamins on CDR-global, CDR-SOB and executive function Z-score (Pinteraction = 0.017, 0.014 and 0.052, respectively), with significant benefits being observed only in those with ins/ins genotype (Beta = -1.367, -0.614 and 0.315, P = 0.004, 0.014 and 0.012, respectively). The interaction was not significant for memory Z-score and whole brain atrophy rate. Notably, the supplements only slowed brain atrophy in members of the 'ins/ins' group who were not using aspirin. CONCLUSIONS: Our data indicate that the beneficial effects of B vitamins including FA on cognitive function are only apparent in those with ins/ins genotype, i.e. relatively better preserved DHFR activity.


Subject(s)
Cognition Disorders , Cognitive Dysfunction , Vitamin B Complex , Aged , Cognition , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/genetics , Humans , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/pharmacology , Vitamin B Complex/therapeutic use
4.
ACS Chem Biol ; 15(4): 1004-1015, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32162909

ABSTRACT

Most cell behaviors are the outcome of processing information from multiple signals generated upon cell stimulation. Thus, a systematic understanding of cellular systems requires methods that allow the activation of more than one specific signaling molecule or pathway within a cell. However, the construction of tools suitable for such multiplexed signal control remains challenging. In this work, we aimed to develop a platform for chemically manipulating multiple signaling molecules/pathways in living mammalian cells based on self-localizing ligand-induced protein translocation (SLIPT). SLIPT is an emerging chemogenetic tool that controls protein localization and cell signaling using synthetic self-localizing ligands (SLs). Focusing on the inner leaflet of the plasma membrane (PM), where there is a hub of intracellular signaling networks, here we present the design and engineering of two new PM-specific SLIPT systems based on an orthogonal eDHFR and SNAP-tag pair. These systems rapidly induce translocation of eDHFR- and SNAP-tag-fusion proteins from the cytoplasm to the PM specifically in a time scale of minutes upon addition of the corresponding SL. We then show that the combined use of the two systems enables chemically inducible, individual translocation of two distinct proteins in the same cell. Finally, by integrating the orthogonal SLIPT systems with fluorescent reporters, we demonstrate simultaneous multiplexed activation and fluorescence imaging of endogenous ERK and Akt activities in a single cell. Collectively, orthogonal PM-specific SLIPT systems provide a powerful new platform for multiplexed chemical signal control in living single cells, offering new opportunities for dissecting cell signaling networks and synthetic cell manipulation.


Subject(s)
MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , O(6)-Methylguanine-DNA Methyltransferase/pharmacology , Protein Transport/drug effects , Pyrimidines/pharmacology , Tetrahydrofolate Dehydrogenase/pharmacology , Cell Membrane/metabolism , Escherichia coli/enzymology , HeLa Cells , Humans , Membrane Proteins/genetics , O(6)-Methylguanine-DNA Methyltransferase/chemistry , O(6)-Methylguanine-DNA Methyltransferase/genetics , Protein Engineering , Pyrimidines/chemistry , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/genetics
5.
ACS Chem Biol ; 13(11): 3153-3160, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30278129

ABSTRACT

Immunotoxins are proteins containing a cell-targeting element linked to a toxin that are under investigation for next-generation cancer treatment. However, these agents are difficult to synthesize, chemically heterogeneous, expensive, and show toxicity toward healthy cells. In this work, we describe the synthesis and characterization of a new type of immunotoxin that showed exquisite selectivity toward targeted cells. In our construct, targeting molecules were covalently attached or genetically fused to oligomeric pore-forming toxins. The activity of the immunotoxin was then caged by fusing a soluble protein to the transmembrane domain and activated via cleavage with furin, which is a protease that is overexpressed in many cancer cells. During the several coupling steps, directed evolution allowed the efficient synthesis of the molecules in E. coli cells, as well as selection for further specificity toward targeted cells. The final construct showed no off-target activity, while acquiring an additional degree of specificity toward the targeted cells upon activation. The pore-forming toxins described here do not require internalization to operate, while the many protomeric subunits can be individually modified to refine target specificity.


Subject(s)
Cnidarian Venoms/pharmacology , Immunotoxins/pharmacology , Pore Forming Cytotoxic Proteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Tetrahydrofolate Dehydrogenase/pharmacology , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cnidarian Venoms/genetics , Directed Molecular Evolution/methods , Drug Design , Escherichia coli/genetics , Escherichia coli/metabolism , Folic Acid/chemistry , Furin/metabolism , Humans , Immunotoxins/chemistry , Immunotoxins/genetics , Immunotoxins/metabolism , Mutagenesis , Pore Forming Cytotoxic Proteins/genetics , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Salmonella typhi/chemistry , Sea Anemones/chemistry , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism
6.
Invest Ophthalmol Vis Sci ; 58(10): 4126-4137, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28829844

ABSTRACT

Purpose: Nuclear factor κB (NFκB) is a ubiquitously expressed, proinflammatory transcription factor that controls the expression of genes involved in cell survival, angiogenesis, complement activation, and inflammation. Studies have implicated NFκB-dependent cytokines or complement-related factors as being detrimentally involved in retinal diseases, thus making inhibition of NFκB signaling a potential therapeutic target. We sought to develop a conditional and reversible method that could regulate pathogenic NFκB signaling by the addition of a small molecule. Methods: We developed a genetically based, trimethoprim (TMP)-regulated approach that conditionally inhibits NFκB signaling by fusing a destabilized dihydrofolate reductase (DHFR) domain to an inhibitor of NFκB, IκBα, in ARPE-19 cells. We then challenged ARPE-19 cells with a number of stimuli that have been demonstrated to trigger NFκB signaling, including LPS, TNFα, IL-1α, and A2E. Western blotting, electrophoretic mobility shift assay, quantitative PCR, ELISA, and NFκB reporter assays were used to evaluate the effectiveness of this DHFR-IκBα approach. Results: This destabilized domain approach, coupled with doxycycline-inducibility, allowed for accurate control over the abundance of DHFR-IκBα. Stabilization of DHFR-IκBα with TMP prevented IL-1α-, A2E-, LPS-, and TNFα-induced NFκB-mediated upregulation and release of the proinflammatory cytokines IL-1ß and IL-6 from ARPE-19 cells (by as much as 93%). This strategy is dosable, completely reversible, and can be cycled "on" or "off" within the same cell population repeatedly to confer protection at desired time points. Conclusions: These studies lay the groundwork for the use of destabilized domains in retinal pigment epithelium (RPE) cells in vivo and in this context, demonstrate their utility for preventing inflammatory signaling.


Subject(s)
Cytochrome P-450 CYP2C8 Inhibitors/pharmacology , NF-kappa B/antagonists & inhibitors , Retinal Pigment Epithelium/metabolism , Tetrahydrofolate Dehydrogenase/pharmacology , Trimethoprim/pharmacology , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cytochrome P-450 CYP2C8 Inhibitors/chemistry , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Humans , NF-kappa B/metabolism , Protein Domains/drug effects , Signal Transduction/drug effects , Tetrahydrofolate Dehydrogenase/chemistry , Trimethoprim/chemistry
7.
Exp Parasitol ; 163: 38-45, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26821296

ABSTRACT

Existing antifolate antimalarial drugs have shown resistance due to the mutations at some amino acid positions of Plasmodium falciparum DHFR-TS. In the present study, to overcome this resistance, a new series of hybrid 4-aminoquinoline-triazine derivatives were designed and docked into the active site of Pf-DHFR-TS (PDB i.d. 1J3K) using validated CDOCKER protocol. Binding energy was calculated by applying CHARMm forcefield. Binding energy and the pattern of interaction of the docked compounds were analysed. Fifteen compounds were selected for synthesis based on their binding energy values and docking poses. Synthesized compounds were characterised by FTIR, (1)H NMR, (13)C NMR, mass spectroscopy and were screened for antimalarial activity against 3D7 strain of Plasmodium falciparum.


Subject(s)
Aminoquinolines/chemistry , Antimalarials/chemistry , Multienzyme Complexes/chemistry , Plasmodium falciparum/drug effects , Tetrahydrofolate Dehydrogenase/chemistry , Thymidylate Synthase/chemistry , Triazines/chemistry , Aminoquinolines/pharmacology , Antimalarials/pharmacology , Crystallography, X-Ray , Inhibitory Concentration 50 , Magnetic Resonance Imaging/methods , Molecular Docking Simulation , Molecular Structure , Multienzyme Complexes/pharmacology , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Tetrahydrofolate Dehydrogenase/pharmacology , Thymidylate Synthase/pharmacology , Transition Temperature , Triazines/pharmacology
8.
Yao Xue Xue Bao ; 47(5): 580-7, 2012 May.
Article in Chinese | MEDLINE | ID: mdl-22811999

ABSTRACT

Pyrimidine derivatives have been the subject of much attention in pesticide and medicine fields owing to their unique biological properties. Particularly, a large number of these compounds have recently been reported to show substantial antitumor activities, and some of them have been investigated in clinical trials. Although these structurally novel compounds have a common chemical moiety of a pyrimidine ring, there are a variety of mechanisms of their antitumor action, such as, inhibition of cyclin-dependent-kinases, inhibition of protein tyrosine kinase, inhibition of carbonic anhydrases, inhibition of dihydrofolate reductase and disruption of microtubule assembly. In this paper, we described the latest advances in the research of such pyrimidine derivatives as antitumor drug according to their action on targets.


Subject(s)
Antineoplastic Agents , Cyclin-Dependent Kinases/antagonists & inhibitors , Neoplasms , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carbonic Anhydrase Inhibitors/pharmacology , Cell Proliferation/drug effects , Folic Acid Antagonists/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Tetrahydrofolate Dehydrogenase/pharmacology , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use
9.
Lett Appl Microbiol ; 54(5): 483-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22394198

ABSTRACT

AIMS: Present report describes the in vitro antimalarial activity and docking analysis of seven 4-aminoquinoline-clubbed 1,3,5-triazine derivatives on pf-DHFR-TS. METHODS AND RESULTS: The antimalarial activity was evaluated in vitro against chloroquine-sensitive 3D7 strain of Plasmodium falciparum. Compounds were docked onto the active site of pf-DHFR-TS using docking server to explicate necessary structural requirements for antimalarial activity. CONCLUSION: Title molecules demonstrated considerable bioactivity against the malaria parasite. Docking analysis revealed deep engulfment of the molecules into the inner groove of pf-DHFR-TS active site by making stable ligand-receptor posses. Hydrophobic interaction was identified as the only major interacting force playing a role between ligand-receptor interaction and minor with hydrogen bonds. SIGNIfiCANCE AND IMPACT OF THE STUDY: The study provided the novel insight into the necessary structural requirement for rationale-based antimalarial drug discovery.


Subject(s)
Antimalarials/pharmacology , Multienzyme Complexes/pharmacology , Tetrahydrofolate Dehydrogenase/pharmacology , Thymidylate Synthase/pharmacology , Triazines/pharmacology , Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Antimalarials/chemistry , Hydrogen Bonding , Multienzyme Complexes/chemistry , Plasmodium falciparum/drug effects , Tetrahydrofolate Dehydrogenase/chemistry , Thymidylate Synthase/chemistry , Triazines/chemistry
10.
Klin Onkol ; 25 Suppl 2: 2S87-92, 2012.
Article in English | MEDLINE | ID: mdl-23581023

ABSTRACT

Methotrexate, a structural analogue of folic acid, is one of the most frequently used chemotherapeutics, especially in haematological malignancies, various solid tumours and also inflammatory disorders. Methotrexate interferes with folate metabolism, mainly by inhibition of dihydrofolate reductase, resulting in the suppression of purine and pyrimidine precursor synthesis. The depletion of nucleic acid precursors seems to be responsible for the cytostatic, cytotoxic and differentiation effects of methotrexate. Methylation of biomolecules represents another folate-dependent pathway that is also affected by methotrexate. Furthermore, methotrexate is able to modify metabolic pathways and cellular processes independently of folate metabolism. Based on the similar structure of methotrexate and of functional groups of certain histone deacetylase inhibitors, the ability of methotrexate to inhibit histone deacetylases was predicted and consequently verified. Recently published findings also suggest that methotrexate affects glyoxalase and antioxidant systems. Although methotrexate has been used as a folate metabolism antagonist in anticancer therapy for more than 60 years, the identification of its'other molecular targets in cellular metabolism still continues.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Methotrexate/pharmacology , Cell Death/drug effects , Cell Differentiation/drug effects , Folic Acid/metabolism , Folic Acid Antagonists/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Humans , Methylation/drug effects , Nucleotides/biosynthesis , Tetrahydrofolate Dehydrogenase/pharmacology
11.
J Pharmacol Exp Ther ; 322(3): 989-97, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17586722

ABSTRACT

Methotrexate (MTX) dose-escalation studies were conducted in C57BL/6 mice to determine the chemoprotective effect of transplantation using bone marrow transduced with lentivirus vectors expressing a drug-resistant variant of murine dihydrofolate reductase (DHFR). Methotrexate-resistant dihydrofolate reductase [tyrosine-22 (Tyr22)DHFR] and enhanced green fluorescent protein (GFP) coding sequences were inserted into self-inactivating lentiviral vectors as part of a genetic fusion or within the context of a bicistronic expression cassette. MTX-treated animals that received Tyr22DHFR-transduced marrow recovered to normal hematocrit levels by 3 weeks post-transplant and exhibited significant GFP marking in myeloid and lymphoid lineage-derived peripheral blood mononuclear cells (PBMCs). In contrast, MTX-treated animals transplanted with control GFP-transduced marrow exhibited extremely reduced hematocrits with severe marrow hypoplasia and did not survive MTX dose escalation. To minimize cell manipulation, we treated unfractionated marrow in an overnight exposure. Transduction at a multiplicity of infection of 10 resulted in up to 11% vector-modified PBMCs in primary recipients and successful repopulation of secondary recipients with vector-marked cells. Experimental cohorts exhibited sustained proviral expression with stable GFP fluorescence intensity. These results demonstrate the effectiveness of lentivirus vectors for chemoprotection in a well developed animal model, with the potential for further preclinical development toward human application.


Subject(s)
Antidotes/administration & dosage , Drug Resistance/genetics , Methotrexate/toxicity , Tetrahydrofolate Dehydrogenase/administration & dosage , Tetrahydrofolate Dehydrogenase/pharmacology , Animals , Bone Marrow Diseases/chemically induced , Bone Marrow Transplantation , Genetic Vectors , Hematocrit , Lentivirus , Mice , Mice, Inbred C57BL , Mutation , Tetrahydrofolate Dehydrogenase/genetics , Transduction, Genetic
12.
Bioorg Med Chem Lett ; 14(18): 4691-5, 2004 Sep 20.
Article in English | MEDLINE | ID: mdl-15324889

ABSTRACT

The spherical truncation of electrostatic interactions between aminoacids makes it possible to break down long-range spatial electrostatic interactions, resulting in short-range interactions. As a result, a Markov Chain model may be used to calculate the probabilities with which the effect of a given interaction reaches aminoacids at different distances within the backbone. The entropies of a Markov Chain model of this type may then be used to codify information about the spatial distribution of charges in the protein used in this study exploring the structure-activity relationship. In this paper, a linear discriminant analysis is reported, which correctly classified 92.3% of 26 under investigation in training and leave-one-out cross validation, purely for illustrative purposes. Classification was carried out for three possible activities: lysozymes, dihydrofolate reductases, and alcohol dehydrogenases. The discriminant analysis equations were contracted into two canonical roots. These simple canonical roots have high regression coefficients (R(c1)=0.903 and R(c2)=0.70). Root1 explains the biological activity of alcohol dehydrogenases while Root2 discriminates between lysozymes and dihydrofolate reductases. It was possible to profile the effect of core, middle, and surface aminoacids on biological activity. In contrast, a model considering classic physicochemical parameters such as: polarizability, refractivity, and partition coefficient classify correctly only the 80.8% of the proteins.


Subject(s)
Markov Chains , Proteins/chemistry , Proteins/pharmacology , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/pharmacology , Linear Models , Muramidase/chemistry , Muramidase/pharmacology , Probability , Quantitative Structure-Activity Relationship , Static Electricity , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/pharmacology
13.
J Chemother ; 16(1): 3-12, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15077993

ABSTRACT

Despite all progress made in the fight against infections caused by bacteria, fungi, protozoa or viruses, there is a need for more and new active agents. Intensive efforts are currently directed against many new and attractive targets, and are hoped to result in new useful agents. The opportunities offered by some known and validated targets are, however, by far not exhausted. Dihydrofolate reductase (DHFR, EC 1.5.1.3) attracted much attention over several decades, which yielded several useful agents. There are excellent chances for new drugs in this field, and they are thought to increase by limiting the spectrum of activity. Whereas trimethoprim seems to present the optimum which can be achieved for a broad spectrum antibacterial agent, specific agents could probably be designed for well defined groups or specific organisms, such as staphylococci among the bacteria, or for a number of parasites, such as Plasmodium falciparum, the fungus Pneumocystis carinii, and several protozoa, such as Trypanosoma, Toxoplasma, and others. This would even extend to herbicides or specific plant pathogens. Achievements and current efforts directed against new DHFR-inhibitors are reviewed, considering only the most recent literature.


Subject(s)
Anti-Infective Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Folic Acid Antagonists/pharmacology , Tetrahydrofolate Dehydrogenase/pharmacology , Humans
14.
Exp Hematol ; 31(12): 1215-22, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14662327

ABSTRACT

OBJECTIVE: While retrovirally mediated gene transfer of dihydrofolate reductase mutants (mutDHFR) has convincingly been demonstrated to confer methotrexate (MTX) resistance to murine hematopoietic cells, clinical application of this technology will require high efficacy in human cells. Therefore, we investigated retroviral constructs expressing various point mutants of human DHFR for their ability to confer MTX resistance to human clonogenic progenitor cells (CFU-C) and to allow for in vitro selection of transduced CFU-C. METHODS: Primary human hematopoietic cells were retrovirally transduced using MMLV- and SFFV/MESV-based vectors expressing DHFR(Ser31), DHFR(Phe22/Ser31), or DHFR(Tyr22/Gly31). MTX resistance of unselected and in vitro-selected CFU-C was determined using MTX-supplemented methylcellulose cultures and gene transfer efficiency was assesed by single-colony PCR analysis. RESULTS: While less than 1% mock-transduced CFU-C survived the presence of > or =5 x 10(-8) M MTX, MMLV- and SFFV/MESV-based vectors expressing DHFR(Ser31) significantly protected CFU-C from MTX at doses ranging from 2.5 to 30 x 10(-8) M. Vectors expressing DHFR(Phe22/Ser31) or DHFR(Tyr22/Gly31) were even more protective and MTX-resistant CFU-C were observed up to 1 x 10(-5) M MTX. Three-day suspension cultures in the presence of 10-20 x 10(-8) M MTX resulted in significant selection of mutDHFR-transduced CFU-C. The percentage of CFU-C resistant to 10 x 10(-8) M MTX increased fourfold to 20-fold and provirus-containing CFU-C increased from 27% to 79-100%. CONCLUSION: Gene transfer of DHFR using suitable retroviral backbones and DHFR mutants significantly increases MTX resistance of human CFU-C and allows efficient in vitro selection of transduced cells using a short-term selection procedure.


Subject(s)
Drug Resistance, Neoplasm/genetics , Hematopoietic Stem Cells/metabolism , Methotrexate/pharmacology , Tetrahydrofolate Dehydrogenase/genetics , Transduction, Genetic/methods , Cell Separation/methods , Cell Survival/drug effects , Cell Survival/genetics , Drug Resistance, Neoplasm/drug effects , Genetic Vectors , Humans , Point Mutation , Tetrahydrofolate Dehydrogenase/pharmacology , Transduction, Genetic/standards
15.
Zhonghua Yi Xue Za Zhi ; 82(19): 1342-5, 2002 Oct 10.
Article in Chinese | MEDLINE | ID: mdl-12509940

ABSTRACT

OBJECTIVE: Investigating the bio-activities of peptides selected from phage display peptide library with vascular endothelial growth factor receptor Flt-1. METHODS: Activities of DHFR-F56/F90 binding to human ubilial vein endothelial cells were detected by immunocytochemistry, and the activity of antiangiogenesis was determined with chick embryo chorioallantoric membrane (CAM) assay. Balb/c nude mice were used as model to detect the activity of DHFR-F56/F90 on inhibiting tumor growth, and immunohistochemistry was employed to determine the localization of the DHFR-F56/F90 in tumor. RESULTS: DHFR-F56/F90 can bind to HUVEC, and DHFR-F56 inhibite angiogenesis in CAM. Meanwhile DHFR-F56 can bind with tumor cells, induce tumor necrosis and inhibit tumor growth in vivo. CONCLUSION: The peptide F56 is an effective antagonist of VEGF binding to Flt-1 and has a potent utility in antiangiogenesis and inhibiting tumor growth.


Subject(s)
Antineoplastic Agents/pharmacology , Peptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Animals , Chick Embryo , Endothelial Growth Factors/antagonists & inhibitors , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Lymphokines/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic/drug effects , Peptide Library , Peptides/metabolism , Tetrahydrofolate Dehydrogenase/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factors
16.
Biochemistry ; 37(45): 15737-46, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9843379

ABSTRACT

Solving the crystallographic structure of the ring-shaped heptamer formed by protective antigen (PA), the B moiety of anthrax toxin, has focused attention on understanding how this oligomer mediates membrane translocation of the toxin's A moieties. We have developed an assay for translocation in which radiolabeled ligands are bound to proteolytically activated PA (PA63) at the surface of CHO or L6 cells, and translocation across the plasma membrane is induced by lowering the pH. The cells are then treated with Pronase E to degrade residual surface-bound material, and protected ligands are quantified after fractionation by SDS-PAGE. Translocation was most efficient (35%-50%) with LFN, the N-terminal PA binding domain of the anthrax lethal factor (LF). Intact LF, edema factor (EF), or fusion proteins containing LFN fused to certain heterologous proteins [the diphtheria toxin A chain (DTA) or dihydrofolate reductase (DHFR)] were less efficiently translocated (15%-20%); and LFN fusions to several other proteins were not translocated at all. LFN with different N-terminal residues was found to be degraded according to the N-end rule by the proteasome, and translocation of LFN fused to a mutant form of DHFR with a low affinity for methotrexate (MTX) protected cells from the effects of MTX. Both results are consistent with a cytosolic location of protected proteins. Evidence that a protein must unfold to be translocated was obtained in experiments showing that (i) translocation of LFNDTA was blocked by introduction of an artificial disulfide into the DTA moiety, and (ii) translocation of LFNDHFR and LFNDTA was blocked by their ligands (MTX and adenine, respectively). These results demonstrate that the acid-induced translocation by anthrax toxin closely resembles that of diphtheria toxin, despite the fact that these two toxins are unrelated and form pores by different mechanisms.


Subject(s)
Antigens, Bacterial/metabolism , Bacillus anthracis/immunology , Bacterial Toxins/metabolism , Amino Acid Sequence , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Biological Transport/genetics , Biological Transport/immunology , Biomarkers , CHO Cells , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/microbiology , Cricetinae , Methotrexate/antagonists & inhibitors , Methotrexate/toxicity , Molecular Sequence Data , Pronase/metabolism , Protein Folding , Rats , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/pharmacology
17.
J Med Chem ; 41(9): 1367-70, 1998 Apr 23.
Article in English | MEDLINE | ID: mdl-9554869

ABSTRACT

Pyrimethamine acts by selectively inhibiting malarial dihydrofolate reductase-thymidylate synthase (DHFR-TS). Resistance in the most important human parasite, Plasmodium falciparum, initially results from an S108N mutation in the DHFR domain, with additional mutation (most commonly C59R or N51I or both) imparting much greater resistance. From a homology model of the 3-D structure of DHFR-TS, rational drug design techniques have been used to design and subsequently synthesize inhibitors able to overcome malarial pyrimethamine resistance. Compared to pyrimethamine (Ki 1.5 nM) with purified recombinant DHFR fromP. falciparum, the Ki value of the m-methoxy analogue of pyrimethamine was 1.07 nM, but against the DHFR bearing the double mutation (C59R + S108N), the Ki values for pyrimethamine and the m-methoxy analogue were 71.7 and 14.0 nM, respectively. The m-chloro analogue of pyrimethamine was a stronger inhibitor of both wild-type DHFR (with Ki 0.30 nM) and the doubly mutant (C59R +S108N) purified enzyme (with Ki 2.40 nM). Growth of parasite cultures of P. falciparum in vitro was also strongly inhibited by these compounds with 50% inhibition of growth occurring at 3.7 microM for the m-methoxy and 0.6 microM for the m-chloro compounds with the K1 parasite line bearing the double mutation (S108N + C59R), compared to 10.2 microM for pyrimethamine. These inhibitors were also found in preliminary studies to retain antimalarial activity in vivo in P. berghei-infected mice.


Subject(s)
Antimalarials/pharmacology , Drug Design , Folic Acid Antagonists/pharmacology , Plasmodium falciparum/drug effects , Pyrimethamine/analogs & derivatives , Pyrimethamine/pharmacology , Animals , Drug Resistance , Malaria/drug therapy , Male , Mice , Multienzyme Complexes/genetics , Multienzyme Complexes/pharmacology , Mutation , Plasmodium berghei , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Pyrimethamine/chemical synthesis , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/pharmacology , Thymidylate Synthase/genetics , Thymidylate Synthase/pharmacology
18.
Hum Gene Ther ; 8(17): 2069-77, 1997 Nov 20.
Article in English | MEDLINE | ID: mdl-9414255

ABSTRACT

Retroviral transduction of antifolate-resistant variants of human dihydrofolate reductase (hDHFR) into cells can increase their resistance to the cytotoxic effects of these drugs. We evaluated the ability of wild-type hDHFR and 20 mutant enzymes (13 with single-amino acid substitutions, 7 with two substitutions) to prevent growth inhibition in antifolate-treated CCRF-CEM cells. The wild-type enzyme and all of the variants significantly protected transduced cells from trimetrexate (TMTX)-induced growth inhibition. However, only half of the variants conferred more protection than does the wild-type enzyme. For the variants tested, the observed protective effect was higher for TMTX than for methotrexate (< or =7.5-fold increased resistance), piritrexim (< or =16-fold), and edatrexate (negligible). Transduction of the variants L22Y-F31S and L22Y-F31R led to the greatest protection against TMTX (approximately 200-fold). Protection from loss of cell viability was similar to protection from growth inhibition. The protection associated with a particular mutant hDHFR did not result from the level of expression: Efficient protection resulted from low affinity of the variant for antifolates, reasonable catalytic activity, and good thermal stability. Clones isolated from a polyclonal population of transduced cells varied by as much as 30-fold in their resistance to TMTX, the resistance differences depending on hDHFR expression levels.


Subject(s)
Folic Acid Antagonists/pharmacology , Tetrahydrofolate Dehydrogenase/genetics , Aminopterin/analogs & derivatives , Aminopterin/antagonists & inhibitors , Animals , Cell Survival , Drug Resistance, Neoplasm/genetics , Gene Expression , Genetic Variation , Growth Inhibitors/antagonists & inhibitors , Humans , Kinetics , Methotrexate/antagonists & inhibitors , Pyrimidines/antagonists & inhibitors , Rabbits , Tetrahydrofolate Dehydrogenase/pharmacology , Thymidine/metabolism , Transfection , Trimetrexate/antagonists & inhibitors
19.
Biochemistry ; 36(40): 12317-22, 1997 Oct 07.
Article in English | MEDLINE | ID: mdl-9315871

ABSTRACT

Previous studies suggest that dihydrofolate reductase (DHFR) regulates its own translation. Moreover, intracellular levels of DHFR protein increase following exposure to the antifolate methotrexate (MTX), suggesting that MTX may release the translational inhibition mediated by DHFR [Chu et al. (1993) Biochemistry 32,4756-4760; Ercikan et al. (1993) Adv. Exp. Med. Biol. 338, 537-540]. To further investigate the role of DHFR in translational autoregulation, we have considered the possibility that DHFR directly contacts its cognate mRNA. Binding studies using a series of truncated DHFR mRNAs as probes localized the DHFR/RNA interaction to a 100-base-pair region containing two putative stem-loop structures; initial studies indicated that both of these loop structures are involved in protein binding. Moreover, the binding of MTX to DHFR prevents interaction of the protein with its cognate mRNA, thereby relieving translational autoregulation.


Subject(s)
Codon/metabolism , Folic Acid Antagonists/metabolism , Folic Acid Antagonists/pharmacology , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/pharmacology , Base Sequence , Binding Sites/genetics , Codon/drug effects , Humans , Molecular Sequence Data , Protein Binding/genetics , RNA/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/pharmacology , Tetrahydrofolate Dehydrogenase/drug effects
20.
Protein Eng ; 10(1): 23-30, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9051730

ABSTRACT

Orientations of the deazapterin ring and the conformational preferences of groups appended to the deazapterin ring in a set of 8-substituted deazapterin cations docked into the dihydrofolate reductase (DHFR) binding site have been investigated using a methodology based on the simulated annealing technique within molecular dynamics (MD) simulations. Of five possible binding pockets for the 8-substituents, identified from a preliminary manual docking study, one has been definitively eliminated after an analysis of MD trajectories, while another remains uncertain. Using a new method based on standard thermodynamic cycles and a linear approximation of polar and non-polar free energy contributions from MD averages, binding affinities of the different ligands in each binding site have been correlated with experimental dissociation constants. The study has provided insights into structure-activity relationships for use in the design of modified inhibitors of DHFR.


Subject(s)
Models, Molecular , Pterins/chemistry , Tetrahydrofolate Dehydrogenase/drug effects , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive , Crystallography, X-Ray , Folic Acid Antagonists/chemistry , Ligands , Protein Binding/physiology , Protein Conformation , Regression Analysis , Structure-Activity Relationship , Temperature , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/pharmacology , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL