Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Biomol Struct Dyn ; 41(24): 14832-14848, 2023.
Article in English | MEDLINE | ID: mdl-36866624

ABSTRACT

Onchocerciasis is a vector-borne disease caused by the filarial nematode Onchocerca volvulus, which is responsible for most of the visual impairments recorded in Africa, Asia and the Americas. It is known that O. volvulus has similar molecular and biological characteristics as Onchocerca ochengi in cattle. This study was designed to screen for immunogenic epitopes and binding pockets of O. ochengi IMPDH and GMPR ligands using immunoinformatic approaches. In this study, a total of 23 B cell epitopes for IMPDH and 7 B cell epitopes for GMPR were predicted using ABCpred tool, Bepipred 2.0 and Kolaskar and Tongaonkar methods. The CD4+ Th computational results showed 16 antigenic epitopes from IMPDH with strong binding affinity for DRB1_0301, DRB3_0101, DRB1_0103 and DRB1_1501 MHC II alleles while 8 antigenic epitopes from GMPR were predicted to bind DRB1_0101 and DRB1_0401 MHC II alleles, respectively. For the CD8+ CTLs analysis, 8 antigenic epitopes from IMPDH showed strong binding affinity to human leukocyte antigen HLA-A*26:01, HLA-A*03:01, HLA-A*24:02 and HLA-A*01:01 MHC I alleles while 2 antigenic epitopes from GMPR showed strong binding affinity to HLA-A*01:01 allele, respectively. The immunogenic B cell and T cell epitopes were further evaluated for antigenicity, non-alllergernicity, toxicity, IFN-gamma, IL4 and IL10. The docking score revealed favorable binding free energy with IMP and MYD scoring the highest binding affinity at -6.6 kcal/mol with IMPDH and -8.3 kcal/mol with GMPR. This study provides valuable insight on IMPDH and GMPR as potential drug targets and for the development of multiple epitope vaccine candidates.Communicated by Ramaswamy H. Sarma.


Subject(s)
Onchocerca , Vaccines , Humans , Animals , Cattle , Onchocerca/metabolism , Immunoinformatics , GMP Reductase/chemistry , GMP Reductase/metabolism , IMP Dehydrogenase/chemistry , IMP Dehydrogenase/metabolism , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Guanosine , Inosine , HLA-A Antigens
2.
Food Chem ; 401: 134145, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36088713

ABSTRACT

Endogenous hydrocortisone causes detrimental effects on public health and domestic animal products, but the potential mechanisms remain elusive. Hydrocortisone was detected from seventy-two Guanzhong-Black pigs in three replicates (216 samples) (0.00 ± 46.38 µg kg-1), indicating the existence of endogenous hydrocortisone. Herein, we investigated the effects of hydrocortisone on the metabolic signatures in pork via integrative metabolomics and proteomics by UHPLC-Q-Orbitrap HRMS. Animal-derived foods under hydrocortisone-bioaccumulation cause metabolic perturbation by regulating glutamine synthetase expression at the transcriptional level contributed to hydrocortisone-induced toxicity and accelerating the down-regulation of essential amino acids (l-Histidine 10.74-6.48 mg kg-1, l-Phenylalanine 3.70-1.57 mg kg-1), purine nucleotides (GMP 40.29-5.00 µg kg-1, etc) that provide nutritional value in a positive feedback loop. Protein-metabolite interactions suggesting that hydrocortisone enhanced nitric oxide synthase and GMP reductase expression (LOQ 3.24-63.39 µg kg-1), and affected meat flavor perception, eventually lead to the decline of nutritional value and flesh quality of pork.


Subject(s)
Histidine , Hydrocortisone , Swine , Animals , Chromatography, High Pressure Liquid , GMP Reductase , Glutamate-Ammonia Ligase , Purine Nucleotides , Phenylalanine
3.
FEBS J ; 289(18): 5571-5598, 2022 09.
Article in English | MEDLINE | ID: mdl-35338694

ABSTRACT

Mycobacteria express enzymes from both the de novo and purine-salvage pathways. However, the regulation of these processes and the roles of individual metabolic enzymes have not been sufficiently detailed. Both Mycobacterium tuberculosis (Mtb) and Mycobacterium smegmatis (Msm) possess three guaB genes, but information is only available on guaB2, which encodes an essential inosine 5'-monophosphate dehydrogenase (IMPDH) involved in de novo purine biosynthesis. This study shows that guaB1, annotated in databases as a putative IMPDH, encodes a guanosine 5'-monophosphate reductase (GMPR), which recycles guanosine monophosphate to inosine monophosphate within the purine-salvage pathway and contains a cystathionine-ß-synthase domain (CBS), which is essential for enzyme activity. GMPR activity is allosterically regulated by the ATP/GTP ratio in a pH-dependent manner. Bioinformatic analysis has indicated the presence of GMPRs containing CBS domains across the entire Actinobacteria phylum.


Subject(s)
Cystathionine , Mycobacterium tuberculosis , Adenosine Triphosphate , Cystathionine beta-Synthase/genetics , GMP Reductase/genetics , GMP Reductase/metabolism , Guanosine Monophosphate/metabolism , Guanosine Triphosphate , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Inosine , Inosine Monophosphate/metabolism , Mycobacterium tuberculosis/metabolism
4.
Cell Chem Biol ; 29(6): 970-984.e6, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35148834

ABSTRACT

Signal transduction pathways post-translationally regulating nucleotide metabolism remain largely unknown. Guanosine monophosphate reductase (GMPR) is a nucleotide metabolism enzyme that decreases GTP pools by converting GMP to IMP. We observed that phosphorylation of GMPR at Tyr267 is critical for its activity and found that this phosphorylation by ephrin receptor tyrosine kinase EPHA4 decreases GTP pools in cell protrusions and levels of GTP-bound RAC1. EPHs possess oncogenic and tumor-suppressor activities, although the mechanisms underlying switches between these two modes are poorly understood. We demonstrated that GMPR plays a key role in EPHA4-mediated RAC1 suppression. This supersedes GMPR-independent activation of RAC1 by EPHA4, resulting in a negative overall effect on melanoma cell invasion and tumorigenicity. Accordingly, EPHA4 levels increase during melanoma progression and inversely correlate with GMPR levels in individual melanoma tumors. Therefore, phosphorylation of GMPR at Tyr267 is a metabolic signal transduction switch controlling GTP biosynthesis and transformed phenotypes.


Subject(s)
Melanoma , Receptor, EphA4/metabolism , GMP Reductase/genetics , GMP Reductase/metabolism , Guanosine Triphosphate/metabolism , Humans , Melanoma/metabolism , Nucleotides/metabolism , Phosphorylation
5.
Proteins ; 90(1): 200-217, 2022 01.
Article in English | MEDLINE | ID: mdl-34368983

ABSTRACT

Human GMP reductase (hGMPR) enzyme is involved in a cellular metabolic pathway, converting GMP into IMP, and also it is an important target for anti-leukemic agents. Present computational investigations explain dynamical behavior of water molecules during the conformational transition process from GMP to IMP using molecular dynamics simulations. Residues at substrate-binding site of cancerous protein (PDB Id. 2C6Q) are mostly more dynamic in nature than the normal protein (PDB Id. 2BLE). Nineteen conserved water molecules are identified at the GMP/IMP binding site and are classified as (i) conserved stable dynamic and (ii) infrequent dynamic. Water molecules W11, W14, and W16 are classified as conserved stable dynamic due to their immobile character, whereas remaining water molecules (W1, W2, W3, W4, W5, W7, W8, W9, W10, W12, W13, W15, W17, W18, and W19) are infrequent with dynamic nature. Entrance or displacement of these infrequent water molecules at GMP/IMP sites may occur due to forward and backward movement of reference residues involving ligands. Four water molecules of hGMPR-I and nine water molecules of hGMPR-II are observed in repetitive transitions from GMP to IMP pathway, which indicates discrimination between two isoforms of hGMPRs. Water molecules in cancerous protein are more dynamic and unstable compared to normal protein. These water molecules execute rare dynamical events at GMP binding site and could assist in detailed understanding of conformational transitions that influence the hGMPR's biological functionality. The present study should be of interest to the experimental community engaged in leukemia research and drug discovery for CML cancer.


Subject(s)
GMP Reductase , Guanosine Monophosphate , Water , Humans , GMP Reductase/chemistry , GMP Reductase/metabolism , Guanosine Monophosphate/chemistry , Guanosine Monophosphate/metabolism , Molecular Dynamics Simulation , Protein Conformation , Thermodynamics , Water/chemistry
6.
J Phys Chem B ; 125(5): 1351-1362, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33369428

ABSTRACT

Human guanosine monophosphate reductase (hGMPR) enzyme maintains the intracellular balance between adenine and guanine nucleotide pools, and it is an excellent target for the design of isoform-specific antileukemic agents. In the present study, we have investigated solvation properties of substrate GMP or product inosine-5'-monophosphate (IMP)-binding pocket of hGMPR by employing molecular dynamics simulations on conformations A (substrate GMP), B [substrate GMP with cofactor nicotinamide adenine dinucleotide phosphate (NDP)], C (product IMP with cofactor NDP), and D (product IMP). Nineteen water sites are identified precisely; they are responsible for the catalytic activity of this site, control structural and dynamical integrity, and electronic consequences of GMP or IMP in the binding site of hGMPR. The water sites of category-1 (W1, W4, W5, W6, W13, and W15) in normal protein and category-2 (W2, W3, W7, W8, W10, W17, and W18) in cancerous protein are unique and stabilize the guanosine or inosine group of GMP or IMP for participation in the enzymatic reaction, whereas the remaining water centers either stabilize pentose sugar ribose or the phosphate group of GMP or IMP. Furthermore, water sites of category-4 (W11, W14, and W16) appear to be conserved in all conformations during the entire simulation. The GMP-binding site in cancerous protein 2C6Q is significantly expanded, and its dynamics are very different from normal protein 2BLE. Furthermore, unique interactions of GMP(N1)···W2···Asp129/Asn158, IMP(N1)···W3···Glu289, and IMP(O6)···W10···Ser270 might be used in a water mimic drug design for hGMPR-II. In this context, water finding probability, relative interaction energy (J) associated with water site W, entropy, and topologies of these three water sites are thermodynamically acceptable for the water displacement method by the modified ligand. Hence, their positions in the catalytic pocket may also facilitate future drug discovery for chronic myelogenous leukemia by the design of appropriately oriented chemical groups that may displace these water molecules to mimic their structural, electronic, and thermodynamic properties.


Subject(s)
Molecular Dynamics Simulation , Water , Binding Sites , GMP Reductase/metabolism , Humans , Kinetics
7.
Biochemistry ; 59(25): 2359-2370, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32479091

ABSTRACT

The remarkable power and specificity of enzyme catalysis rely on the dynamic alignment of the enzyme, substrates, and cofactors, yet the role of dynamics has usually been approached from the perspective of the protein. We have been using an underappreciated NMR technique, subtesla high-resolution field cycling 31P NMR relaxometry, to investigate the dynamics of the enzyme-bound substrates and cofactor on guanosine-5'-monophosphate reductase (GMPR). GMPR forms two dead end, yet catalytically competent, complexes that mimic distinct steps in the catalytic cycle: E·IMP·NADP+ undergoes a partial hydride transfer reaction, while E·GMP·NADP+ undergoes a partial deamination reaction. A different cofactor conformation is required for each partial reaction. Here we report the effects of mutations designed to perturb cofactor conformation and ammonia binding with the goal of identifying the structural features that contribute to the distinct dynamic signatures of the hydride transfer and deamination complexes. These experiments suggest that Asp129 is a central cog in a dynamic network required for both hydride transfer and deamination. In contrast, Lys77 modulates the conformation and mobility of substrates and cofactors in a reaction-specific manner. Thr105 and Tyr318 are part of a deamination-specific dynamic network that includes the 2'-OH of GMP. These residues have comparatively little effect on the dynamic properties of the hydride transfer complex. These results further illustrate the potential of high-resolution field cycling NMR relaxometry for the investigation of ligand dynamics. In addition, exchange experiments indicate that NH3/NH4+ has a high affinity for the deamination complex but a low affinity for the hydride transfer complex, suggesting that the movement of ammonia may gate the cofactor conformational change. Collectively, these experiments reinforce the view that the enzyme, substrates, and cofactor are linked in intricate, reaction-specific, dynamic networks and demonstrate that distal portions of the substrates and cofactors are critical features in these networks.


Subject(s)
Coenzymes , GMP Reductase , NADP , Humans , Ammonia/metabolism , Biocatalysis , Coenzymes/chemistry , Coenzymes/metabolism , GMP Reductase/genetics , GMP Reductase/metabolism , Guanosine Monophosphate/chemistry , Kinetics , Molecular Conformation , Mutation , NADP/chemistry , NADP/metabolism , Protein Binding
8.
Nat Commun ; 11(1): 1837, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32296055

ABSTRACT

Guanosine 5'-monophosphate reductase (GMPR) is involved in the purine salvage pathway and is conserved throughout evolution. Nonetheless, the GMPR of Trypanosoma brucei (TbGMPR) includes a unique structure known as the cystathionine-ß-synthase (CBS) domain, though the role of this domain is not fully understood. Here, we show that guanine and adenine nucleotides exert positive and negative effects, respectively, on TbGMPR activity by binding allosterically to the CBS domain. The present structural analyses revealed that TbGMPR forms an octamer that shows a transition between relaxed and twisted conformations in the absence and presence of guanine nucleotides, respectively, whereas the TbGMPR octamer dissociates into two tetramers when ATP is available instead of guanine nucleotides. These findings demonstrate that the CBS domain plays a key role in the allosteric regulation of TbGMPR by facilitating the transition of its oligomeric state depending on ligand nucleotide availability.


Subject(s)
Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/metabolism , GMP Reductase/chemistry , GMP Reductase/metabolism , Trypanosoma brucei brucei/enzymology , Allosteric Regulation , Crystallography, X-Ray , Kinetics , Protein Domains , Protein Multimerization , Protein Structure, Secondary
9.
Clin Genet ; 97(2): 276-286, 2020 02.
Article in English | MEDLINE | ID: mdl-31600844

ABSTRACT

Autosomal dominant progressive external ophthalmoplegia (adPEO) is a late-onset, Mendelian mitochondrial disorder characterised by paresis of the extraocular muscles, ptosis, and skeletal-muscle restricted multiple mitochondrial DNA (mtDNA) deletions. Although dominantly inherited, pathogenic variants in POLG, TWNK and RRM2B are among the most common genetic defects of adPEO, identification of novel candidate genes and the underlying pathomechanisms remains challenging. We report the clinical, genetic and molecular investigations of a patient who presented in the seventh decade of life with PEO. Oxidative histochemistry revealed cytochrome c oxidase-deficient fibres and occasional ragged red fibres showing subsarcolemmal mitochondrial accumulation in skeletal muscle, while molecular studies identified the presence of multiple mtDNA deletions. Negative candidate screening of known nuclear genes associated with PEO prompted diagnostic exome sequencing, leading to the prioritisation of a novel heterozygous c.547G>C variant in GMPR (NM_006877.3) encoding guanosine monophosphate reductase, a cytosolic enzyme required for maintaining the cellular balance of adenine and guanine nucleotides. We show that the novel c.547G>C variant causes aberrant splicing, decreased GMPR protein levels in patient skeletal muscle, proliferating and quiescent cells, and is associated with subtle changes in nucleotide homeostasis protein levels and evidence of disturbed mtDNA maintenance in skeletal muscle. Despite confirmation of GMPR deficiency, demonstrating marked defects of mtDNA replication or nucleotide homeostasis in patient cells proved challenging. Our study proposes that GMPR is the 19th locus for PEO and highlights the complexities of uncovering disease mechanisms in late-onset PEO phenotypes.


Subject(s)
DNA, Mitochondrial/genetics , GMP Reductase/genetics , Late Onset Disorders/genetics , Muscle, Skeletal/enzymology , Ophthalmoplegia/genetics , Adenine/metabolism , Aged , Cells, Cultured , Cytochrome-c Oxidase Deficiency/metabolism , DNA Replication , DNA, Mitochondrial/metabolism , Female , Fibroblasts/enzymology , GMP Reductase/deficiency , GMP Reductase/metabolism , Guanine/metabolism , HEK293 Cells , HeLa Cells , Heterozygote , Humans , Late Onset Disorders/metabolism , Late Onset Disorders/pathology , Muscle, Skeletal/pathology , Ophthalmoplegia/enzymology , Ophthalmoplegia/physiopathology , Oxidative Phosphorylation , RNA Splicing , Sequence Deletion , Exome Sequencing
10.
Appl Environ Microbiol ; 84(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30217843

ABSTRACT

Purine nucleoside antibiotic pairs, concomitantly produced by a single strain, are an important group of microbial natural products. Here, we report a target-directed genome mining approach to elucidate the biosynthesis of the purine nucleoside antibiotic pair aristeromycin (ARM) and coformycin (COF) in Micromonospora haikouensis DSM 45626 (a new producer for ARM and COF) and Streptomyces citricolor NBRC 13005 (a new COF producer). We also provide biochemical data that MacI and MacT function as unusual phosphorylases, catalyzing an irreversible reaction for the tailoring assembly of neplanocin A (NEP-A) and ARM. Moreover, we demonstrate that MacQ is shown to be an adenosine-specific deaminase, likely relieving the potential "excess adenosine" for producing cells. Finally, we report that MacR, an annotated IMP dehydrogenase, is actually an NADPH-dependent GMP reductase, which potentially plays a salvage role for the efficient supply of the precursor pool. Hence, these findings illustrate a fine-tuned pathway for the biosynthesis of ARM and also open the way for the rational search for purine antibiotic pairs.IMPORTANCE ARM and COF are well known for their prominent biological activities and unusual chemical structures; however, the logic of their biosynthesis has long been poorly understood. Actually, the new insights into the ARM and COF pathway will not only enrich the biochemical repertoire for interesting enzymatic reactions but may also lay a solid foundation for the combinatorial biosynthesis of this group of antibiotics via a target-directed genome mining strategy.


Subject(s)
Actinobacteria/metabolism , Adenosine/analogs & derivatives , Anti-Bacterial Agents/metabolism , Coformycin/biosynthesis , Purine Nucleosides/biosynthesis , Actinobacteria/genetics , Adenosine/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biosynthetic Pathways , GMP Reductase/genetics , GMP Reductase/metabolism
11.
Biochemistry ; 57(22): 3146-3154, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29547266

ABSTRACT

The ability of enzymes to modulate the dynamics of bound substrates and cofactors is a critical feature of catalysis, but the role of dynamics has largely been approached from the perspective of the protein. Here, we use an underappreciated NMR technique, subtesla high-resolution field-cycling 31P NMR relaxometry, to interrogate the dynamics of enzyme bound substrates and cofactors in guanosine-5'-monophosphate reductase (GMPR). These experiments reveal distinct binding modes and dynamic profiles associated with the 31P nuclei in the Michaelis complexes for the deamination and hydride transfer steps of the catalytic cycle. Importantly, the substrate is constrained and the cofactor is more dynamic in the deamination complex E·GMP·NADP+, whereas the substrate is more dynamic and the cofactor is constrained in the hydride transfer complex E·IMP·NADP+. The presence of D2O perturbed the relaxation of the 31P nuclei in E·IMP·NADP+ but not in E·GMP·NADP+, providing further evidence of distinct binding modes with different dynamic properties. dIMP and dGMP are poor substrates, and the dynamics of the cofactor complexes of dGMP/dIMP are disregulated relative to GMP/IMP. The substrate 2'-OH interacts with Asp219, and mutation of Asp219 to Ala decreases the value of Vmax by a factor of 30. Counterintuitively, loss of Asp219 makes both substrates and cofactors less dynamic. These observations suggest that the interactions between the substrate 2'-OH and Asp219 coordinate the dynamic properties of the Michaelis complexes, and these dynamics are important for progression through the catalytic cycle.


Subject(s)
GMP Reductase/chemistry , GMP Reductase/physiology , Magnetic Resonance Spectroscopy/methods , Binding Sites , Catalysis , Guanosine/metabolism , Kinetics , Magnetic Resonance Imaging , Models, Molecular , NADP/metabolism , Protein Binding
12.
Sci Rep ; 8(1): 2759, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426890

ABSTRACT

Alzheimer's disease (AD) is a severe neurodegenerative disorder for which identification of differentially expressed genes is one way to find new therapeutic targets. Here, we conducted analysis to identify age-independent, AD-specific genes. We found that the MET, WIF1, and NPTX2 genes are downregulated in AD. WIF1 and MET are implicated in Wnt and MET signaling and regulate GSK3ß activity and are thus linked with AD. Importantly, we found that the GMPR gene exhibited a gradual increase in AD progression. A logistic model based on GMPR has good ability to classify AD cases. GMPR's product GMPR1 is in the AMPK and adenosine receptor pathways and is thus associated with Tau phosphorylation in AD. This allows GMPR1 to be a therapeutic target. Therefore, we screened five possible inhibitors to GMPR1 by docking GMPR1 with 1,174 approved drugs. Among them, lumacaftor is ideal. We then tested the effects of lumacaftor on AD model mice. After 20 days of oral administration, we observed that ß-Amyloid accumulation was slowed down, and phosphorylation of Tau was almost eliminated in the treated mice. We highlight the elevated expression level of GMPR in AD and propose a therapeutic strategy of inhibiting GMPR1 with lumacaftor.


Subject(s)
Alzheimer Disease/drug therapy , Aminopyridines/therapeutic use , Amyloid beta-Peptides/metabolism , Benzodioxoles/therapeutic use , Enzyme Inhibitors/therapeutic use , GMP Reductase/antagonists & inhibitors , Molecular Targeted Therapy , tau Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Databases, Genetic , Disease Models, Animal , GMP Reductase/genetics , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Wnt Signaling Pathway/genetics
13.
Parasitol Int ; 66(5): 537-544, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28366788

ABSTRACT

Trypanosoma congolense is one of the most prevalent pathogens which causes trypanosomosis in African animals, resulting in a significant economic loss. In its life cycle, T. congolense is incapable of synthesizing purine nucleotides via a de novo pathway, and thus relies on a salvage pathway to survive. In this study, we identified a gene from T. congolense, TcIL3000_5_1940, as a guanosine 5'-monophosphate reductase (GMPR), an enzyme that modulates the concentration of intracellular guanosine in the pathogen. The recombinant protein was expressed in Escherichia coli, and the gene product was enzymatically confirmed as a unique GMPR, designated as rTcGMPR. This enzyme was constitutively expressed in glycosomes at all of the parasite's developmental stages similar to other purine nucleotide metabolic enzymes. Mycophenolic acid (MPA) was found to inhibit rTcGMPR activity. Hence, it is a potential lead compound for the design of trypanocidal agents, specifically GMPR inhibitor.


Subject(s)
GMP Reductase/antagonists & inhibitors , GMP Reductase/genetics , Trypanocidal Agents/pharmacology , Trypanosoma congolense/drug effects , Trypanosoma congolense/enzymology , Animals , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , GMP Reductase/isolation & purification , Guanosine/metabolism , Mycophenolic Acid/pharmacology , Purines/metabolism , Recombinant Proteins/metabolism , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology
14.
Oncogene ; 36(1): 84-96, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27181209

ABSTRACT

Melanoma progression is associated with increased invasion and, often, decreased levels of microphthalmia-associated transcription factor (MITF). Accordingly, downregulation of MITF induces invasion in melanoma cells; however, little is known about the underlying mechanisms. Here, we report for the first time that depletion of MITF results in elevation of intracellular GTP levels and increased amounts of active (GTP-bound) RAC1, RHO-A and RHO-C. Concomitantly, MITF-depleted cells display larger number of invadopodia and increased invasion. We further demonstrate that the gene for guanosine monophosphate reductase (GMPR) is a direct MITF target, and that the partial repression of GMPR accounts mostly for the above phenotypes in MITF-depleted cells. Reciprocally, transactivation of GMPR is required for MITF-dependent suppression of melanoma cell invasion, tumorigenicity and lung colonization. Moreover, loss of GMPR accompanies downregulation of MITF in vemurafenib-resistant BRAFV600E-melanoma cells and underlies the increased invasion in these cells. Our data uncover novel mechanisms linking MITF-dependent inhibition of invasion to suppression of guanylate metabolism.


Subject(s)
Guanosine Triphosphate/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Ectopic Gene Expression , Extracellular Matrix/metabolism , Female , GMP Reductase/genetics , GMP Reductase/metabolism , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Intracellular Space/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/genetics , rho GTP-Binding Proteins/metabolism
15.
J Biol Chem ; 291(44): 22988-22998, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27613871

ABSTRACT

Guanosine-5'-monophosphate reductase (GMPR) catalyzes the reduction of GMP to IMP and ammonia with concomitant oxidation of NADPH. Here we investigated the structure and dynamics of enzyme-bound substrates and cofactors by measuring 31P relaxation rates over a large magnetic field range using high resolution field cycling NMR relaxometry. Surprisingly, these experiments reveal differences in the low field relaxation profiles for the monophosphate of GMP compared with IMP in their respective NADP+ complexes. These complexes undergo partial reactions that mimic different steps in the overall catalytic cycle. The relaxation profiles indicate that the substrate monophosphates have distinct interactions in E·IMP·NADP+ and E·GMP·NADP+ complexes. These findings were not anticipated by x-ray crystal structures, which show identical interactions for the monophosphates of GMP and IMP in several inert complexes. In addition, the motion of the cofactor is enhanced in the E·GMP·NADP+ complex. Last, the motions of the substrate and cofactor are coordinately regulated; the cofactor has faster local motions than GMP in the deamination complex but is more constrained than IMP in that complex, leading to hydride transfer. These results show that field cycling can be used to investigate the dynamics of protein-bound ligands and provide new insights into how portions of the substrate remote from the site of chemical transformation promote catalysis.


Subject(s)
Coenzymes/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , GMP Reductase/chemistry , Biocatalysis , Coenzymes/metabolism , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , GMP Reductase/genetics , GMP Reductase/metabolism , Guanine Nucleotides/chemistry , Guanine Nucleotides/metabolism , Inosine Monophosphate/chemistry , Inosine Monophosphate/metabolism , Kinetics , Magnetic Resonance Spectroscopy , NADP/chemistry , NADP/metabolism , Protein Binding
16.
Mol Biochem Parasitol ; 208(2): 74-83, 2016 08.
Article in English | MEDLINE | ID: mdl-27343371

ABSTRACT

Purine acquisition is an essential nutritional process for Leishmania. Although purine salvage into adenylate nucleotides has been investigated in detail, little attention has been focused on the guanylate branch of the purine pathway. To characterize guanylate nucleotide metabolism in Leishmania and create a cell culture model in which the pathways for adenylate and guanylate nucleotide synthesis can be genetically uncoupled for functional studies in intact cells, we created and characterized null mutants of L. donovani that were deficient in either GMP reductase alone (Δgmpr) or in both GMP reductase and its paralog IMP dehydrogenase (Δgmpr/Δimpdh). Whereas wild type parasites were capable of utilizing virtually any purine nucleobase/nucleoside, the Δgmpr and Δgmpr/Δimpdh null lines exhibited highly restricted growth phenotypes. The Δgmpr single mutant could not grow in xanthine, guanine, or their corresponding nucleosides, while no purine on its own could support the growth of Δgmpr/Δimpdh cells. Permissive growth conditions for the Δgmpr/Δimpdh necessitated both xanthine, guanine, or the corresponding nucleosides, and additionally, a second purine that could serve as a source for adenylate nucleotide synthesis. Interestingly, GMPR, like its paralog IMPDH, is compartmentalized to the leishmanial glycosome, a process mediated by its COOH-terminal peroxisomal targeting signal. The restricted growth phenotypes displayed by the L. donovani Δgmpr and Δgmpr/Δimpdh null mutants confirms the importance of GMPR in the purine interconversion processes of this parasite.


Subject(s)
Adenosine Monophosphate/metabolism , GMP Reductase/genetics , GMP Reductase/metabolism , Guanosine Monophosphate/metabolism , Leishmania donovani/genetics , Leishmania donovani/metabolism , Gene Knockdown Techniques , Genotype , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Leishmania donovani/growth & development , Mutation , Phenotype , Protein Transport , Purines/metabolism , RNA Interference
17.
Mol Microbiol ; 100(5): 824-40, 2016 06.
Article in English | MEDLINE | ID: mdl-26853689

ABSTRACT

The Leishmania guanosine 5'-monophosphate reductase (GMPR) and inosine 5'-monophosphate dehydrogenase (IMPDH) are purine metabolic enzymes that function maintaining the cellular adenylate and guanylate nucleotide. Interestingly, both enzymes contain a cystathionine-ß-synthase domain (CBS). To investigate this metabolic regulation, the Leishmania GMPR was cloned and shown to be sufficient to complement the guaC (GMPR), but not the guaB (IMPDH), mutation in Escherichia coli. Kinetic studies confirmed that the Leishmania GMPR catalyzed a strict NADPH-dependent reductive deamination of GMP to produce IMP. Addition of GTP or high levels of GMP induced a marked increase in activity without altering the Km values for the substrates. In contrast, the binding of ATP decreased the GMPR activity and increased the GMP Km value 10-fold. These kinetic changes were correlated with changes in the GMPR quaternary structure, induced by the binding of GMP, GTP, or ATP to the GMPR CBS domain. The capacity of these CBS domains to mediate the catalytic activity of the IMPDH and GMPR provides a regulatory mechanism for balancing the intracellular adenylate and guanylate pools.


Subject(s)
Adenosine Triphosphate/metabolism , Cystathionine beta-Synthase/genetics , GMP Reductase/genetics , Gene Expression Regulation , IMP Dehydrogenase/genetics , Leishmania donovani/enzymology , Leishmania major/enzymology , Catalysis , Escherichia coli/genetics , GMP Reductase/isolation & purification , GMP Reductase/metabolism , Genetic Complementation Test , Guanosine Monophosphate/metabolism , IMP Dehydrogenase/metabolism , Kinetics , Leishmania donovani/drug effects , Leishmania donovani/genetics , Leishmania major/drug effects , Leishmania major/genetics , Models, Molecular , NADP/metabolism , Nucleotides/metabolism
18.
PLoS Negl Trop Dis ; 10(1): e0004339, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26731263

ABSTRACT

The metabolic pathway of purine nucleotides in parasitic protozoa is a potent drug target for treatment of parasitemia. Guanosine 5'-monophosphate reductase (GMPR), which catalyzes the deamination of guanosine 5'-monophosphate (GMP) to inosine 5'-monophosphate (IMP), plays an important role in the interconversion of purine nucleotides to maintain the intracellular balance of their concentration. However, only a few studies on protozoan GMPR have been reported at present. Herein, we identified the GMPR in Trypanosoma brucei, a causative protozoan parasite of African trypanosomiasis, and found that the GMPR proteins were consistently localized to glycosomes in T. brucei bloodstream forms. We characterized its recombinant protein to investigate the enzymatic differences between GMPRs of T. brucei and its host animals. T. brucei GMPR was distinct in having an insertion of a tandem repeat of the cystathionine ß-synthase (CBS) domain, which was absent in mammalian and bacterial GMPRs. The recombinant protein of T. brucei GMPR catalyzed the conversion of GMP to IMP in the presence of NADPH, and showed apparent affinities for both GMP and NADPH different from those of its mammalian counterparts. Interestingly, the addition of monovalent cations such as K+ and NH4+ to the enzymatic reaction increased the GMPR activity of T. brucei, whereas none of the mammalian GMPR's was affected by these cations. The monophosphate form of the purine nucleoside analog ribavirin inhibited T. brucei GMPR activity, though mammalian GMPRs showed no or only a little inhibition by it. These results suggest that the mechanism of the GMPR reaction in T. brucei is distinct from that in the host organisms. Finally, we demonstrated the inhibitory effect of ribavirin on the proliferation of trypanosomes in a dose-dependent manner, suggesting the availability of ribavirin to develop a new therapeutic agent against African trypanosomiasis.


Subject(s)
GMP Reductase/metabolism , Trypanosoma brucei brucei/enzymology , Amino Acid Sequence , Animals , Antimetabolites/pharmacology , GMP Reductase/genetics , Gene Expression Regulation, Enzymologic , Hydrogen-Ion Concentration , Molecular Sequence Data , Recombinant Proteins , Ribavirin/pharmacology , Species Specificity , Temperature , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism
20.
Cell Rep ; 5(2): 493-507, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24139804

ABSTRACT

Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.


Subject(s)
GMP Reductase/metabolism , Melanoma/enzymology , Purine Nucleosides/biosynthesis , Animals , Cell Line, Tumor , Cell Movement , Extracellular Matrix/metabolism , GMP Reductase/antagonists & inhibitors , GMP Reductase/genetics , Guanosine Triphosphate/metabolism , HCT116 Cells , Humans , IMP Dehydrogenase/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Transplantation, Heterologous , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...