Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
1.
J Am Chem Soc ; 145(49): 26883-26889, 2023 12 13.
Article in English | MEDLINE | ID: mdl-38051581

ABSTRACT

(-)-FR901483 (1) isolated from the fungus Cladobotryum sp. No.11231 achieves immunosuppression via nucleic acid biosynthesis inhibition rather than IL-2 production inhibition as accomplished by FK506 and cyclosporin A. Recently, we identified the frz gene cluster for the biosynthesis of 1. It contains frzK, a gene homologous to phosphoribosyl pyrophosphate amidotransferase (PPAT)that catalyzes the initial step of de novo purine biosynthesis. We speculated that frzK encodes a PPAT that escapes inhibition by 1 and functions as a self-resistance enzyme (SRE) for the producing host. Nevertheless, details remained elusive. Here, we report the biochemical and structural analyses of FrzK and its Escherichia coli counterpart, PurF. Recombinantly produced FrzK exhibited PPAT activity, albeit weaker than PurF, but evaded strong inhibition by 1. These results confirmed that the target of 1 is PPAT, and FrzK acts as an SRE by maintaining the de novo purine biosynthetic capability in the presence of 1. To understand how FrzK evades inhibition by 1, we determined the crystal structure of PurF in the complex with 1 and constructed a homology model of FrzK. Sequence and structural analyses of various PPATs identified that many residues unique to FrzK occur near the Flexible Loop that remains disordered when inactive but becomes ordered and covers up the active site upon activation by substrate binding. Kinetic characterizations of mutants of the unique residues revealed that the resistance of FrzK against 1 may be conferred by structurally predisposing the Flexible Loop to the active, closed conformation even in the presence of 1.


Subject(s)
Amidophosphoribosyltransferase , Purines , Amino Acid Sequence , Purines/chemistry , Amidophosphoribosyltransferase/genetics , Amidophosphoribosyltransferase/metabolism , Escherichia coli/metabolism
2.
Genomics ; 114(4): 110424, 2022 07.
Article in English | MEDLINE | ID: mdl-35798250

ABSTRACT

OBJECTIVE: Serine hydroxymethyltransferase 2 (SHMT2) is the first rate-limiting enzyme for serine/glycine biosynthesis and one carbon metabolism. Here, we explore the underlying mechanism of how SHMT2 functions in renal cell carcinoma (RCC) initiation. METHODS: In this study, SHMT2 expression was assessed in RCC tissues. In vitro experiments were performed to investigate the functional role of SHMT2. The detailed mechanisms of SHMT2-mediated PPAT were addressed. RESULTS: Increased SHMT2 facilitated RCC cell proliferation by inducing the G1/S phase transition. And SHMT2 promoted the expression of PPAT. Mechanism dissection revealed that SHMT2 enhanced the m6A modification through the endogenous methyl donor SAM mediated by SHMT2 via serine/glycine one carbon metabolic networks. SHMT2-catalyzed serine/glycine conversion regulated PPAT expression in an m6A-IGF2BP2-dependent manner. SHMT2 promoted RCC cell proliferation by upregulating PPAT expression. CONCLUSIONS: SHMT2 promotes RCC tumorigenesis by increasing PPAT expression. Thus, SHMT2 may be a novel potential therapeutic target for RCC.


Subject(s)
Amidophosphoribosyltransferase , Carcinoma, Renal Cell , Glycine Hydroxymethyltransferase , Kidney Neoplasms , Amidophosphoribosyltransferase/metabolism , Carbon/metabolism , Carcinogenesis/genetics , Carcinoma, Renal Cell/genetics , Cell Proliferation , Cell Transformation, Neoplastic , Glycine/metabolism , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Humans , Kidney Neoplasms/genetics , RNA-Binding Proteins/metabolism , Serine/metabolism
3.
mBio ; 12(2)2021 03 30.
Article in English | MEDLINE | ID: mdl-33785613

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is a sensor that recognizes cytosolic double-stranded RNA derived from microbes to induce host immune response. Viruses, such as herpesviruses, deploy diverse mechanisms to derail RIG-I-dependent innate immune defense. In this study, we discovered that mouse RIG-I is intrinsically resistant to deamidation and evasion by herpes simplex virus 1 (HSV-1). Comparative studies involving human and mouse RIG-I indicate that N495 of human RIG-I dictates species-specific deamidation by HSV-1 UL37. Remarkably, deamidation of the other site, N549, hinges on that of N495, and it is catalyzed by cellular phosphoribosylpyrophosphate amidotransferase (PPAT). Specifically, deamidation of N495 enables RIG-I to interact with PPAT, leading to subsequent deamidation of N549. Collaboration between UL37 and PPAT is required for HSV-1 to evade RIG-I-mediated antiviral immune response. This work identifies an immune regulatory role of PPAT in innate host defense and establishes a sequential deamidation event catalyzed by distinct deamidases in immune evasion.IMPORTANCE Herpesviruses are ubiquitous pathogens in human and establish lifelong persistence despite host immunity. The ability to evade host immune response is pivotal for viral persistence and pathogenesis. In this study, we investigated the evasion, mediated by deamidation, of species-specific RIG-I by herpes simplex virus 1 (HSV-1). Our findings uncovered a collaborative and sequential action between viral deamidase UL37 and a cellular glutamine amidotransferase, phosphoribosylpyrophosphate amidotransferase (PPAT), to inactivate RIG-I and mute antiviral gene expression. PPAT catalyzes the rate-limiting step of the de novo purine synthesis pathway. This work describes a new function of cellular metabolic enzymes in host defense and viral immune evasion.


Subject(s)
Amidophosphoribosyltransferase/metabolism , DEAD Box Protein 58/metabolism , Herpes Simplex/enzymology , Herpesvirus 1, Human/enzymology , Viral Structural Proteins/metabolism , Virus Replication , Amidophosphoribosyltransferase/genetics , Amino Acid Motifs , Animals , DEAD Box Protein 58/chemistry , DEAD Box Protein 58/genetics , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Humans , Mice , Protein Binding , Species Specificity , Viral Structural Proteins/genetics
4.
Nat Commun ; 11(1): 1320, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32184390

ABSTRACT

Glucose metabolism is remodeled in cancer, but the global pattern of cancer-specific metabolic changes remains unclear. Here we show, using the comprehensive measurement of metabolic enzymes by large-scale targeted proteomics, that the metabolism both carbon and nitrogen is altered during the malignant progression of cancer. The fate of glutamine nitrogen is shifted from the anaplerotic pathway into the TCA cycle to nucleotide biosynthesis, with this shift being controlled by glutaminase (GLS1) and phosphoribosyl pyrophosphate amidotransferase (PPAT). Interventions to reduce the PPAT/GLS1 ratio suppresses tumor growth of many types of cancer. A meta-analysis reveals that PPAT shows the strongest correlation with malignancy among all metabolic enzymes, in particular in neuroendocrine cancer including small cell lung cancer (SCLC). PPAT depletion suppresses the growth of SCLC lines. A shift in glutamine fate may thus be required for malignant progression of cancer, with modulation of nitrogen metabolism being a potential approach to SCLC treatment.


Subject(s)
Disease Progression , Glutamine/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Nitrogen/metabolism , Amidophosphoribosyltransferase/metabolism , Animals , Biosynthetic Pathways , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glutaminase/metabolism , Humans , Metabolomics , Mice, Nude , Models, Biological , Molecular Targeted Therapy , Neoplasms/genetics , Prognosis
5.
Am J Hypertens ; 33(12): 1136-1145, 2020 12 31.
Article in English | MEDLINE | ID: mdl-33463674

ABSTRACT

BACKGROUND: There is a diurnal variation in the blood pressure fluctuation of hypertension, and blood pressure fluctuation abnormality is considered to be an independent risk factor for organ damage including cardiovascular complications. In the current study, we tried to identify molecules responsible for blood pressure circadian rhythm formation under the control of the kidney biological clock in hypertension. METHODS: DNA microarray analysis was performed in kidneys from 5-week-old spontaneously hypertensive rats (SHRs)/Izm, stroke-prone SHR rats (SHRSP)/Izm, and Wistar Kyoto (WKY)/Izm rats. To detect variation, mouse tubular epithelial cells (TCMK-1) were stimulated with dexamethasone. We performed immunostaining and western blot analysis in the renal medulla of kidney from 5-week-old WKY rats and SHRs. RESULTS: We extracted 1,032 genes with E-box, a binding sequence for BMAL1 and CLOCK using a Gene Set Enrichment Analysis. In a microarray analysis, we identified 12 genes increased as more than 2-fold in the kidneys of SHRs and SHRSP in comparison to WKY rats. In a periodic regression analysis, phosphoribosyl pyrophosphate amidotransferase (Ppat) and fragile X mental retardation, autosomal homolog 1 (Fxr1) showed circadian rhythm. Immunocytochemistry revealed PPAT-positivity in nuclei and cytoplasm in the tubules, and FXR1-positivity in the cytoplasm of TCMK-1. In 5-week-old WKY rat and SHR kidneys, PPAT was localized in the nucleus and cytoplasm of the proximal and distal tubules, and FXR1 was localized to the cytoplasm of the proximal and distal tubules. CONCLUSIONS: PPAT and FXR1 are pivotal molecules in the control of blood pressure circadian rhythm by the kidney in hypertension.


Subject(s)
ARNTL Transcription Factors/metabolism , Amidophosphoribosyltransferase/metabolism , CLOCK Proteins/metabolism , Circadian Rhythm/genetics , Hypertension/metabolism , Kidney Tubules/metabolism , Kidney/metabolism , RNA-Binding Proteins/metabolism , ARNTL Transcription Factors/genetics , Amidophosphoribosyltransferase/genetics , Animals , Blood Pressure , CLOCK Proteins/genetics , Hypertension/genetics , Kidney Tubules/cytology , Mice , Oligonucleotide Array Sequence Analysis , RNA-Binding Proteins/genetics , Rats , Rats, Inbred SHR , Rats, Inbred WKY
6.
J Proteome Res ; 18(5): 2078-2087, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30964683

ABSTRACT

Purines represent a class of essential metabolites produced by the cell to maintain cellular homeostasis and facilitate cell proliferation. In times of high purine demand, the de novo purine biosynthetic pathway is activated; however, the mechanisms that facilitate this process are largely unknown. One plausible mechanism is through intracellular signaling, which results in enzymes within the pathway becoming post-translationally modified to enhance their individual enzyme activities and the overall pathway metabolic flux. Here, we employ a proteomic strategy to investigate the extent to which de novo purine biosynthetic pathway enzymes are post-translationally modified in 293T cells. We identified 7 post-translational modifications on 135 residues across the 6 human pathway enzymes. We further asked whether there were differences in the post-translational modification state of each pathway enzyme isolated from cells cultured in the presence or absence of purines. Of the 174 assigned modifications, 67% of them were only detected in one experimental growth condition in which a significant number of serine and threonine phosphorylations were noted. A survey of the most-probable kinases responsible for these phosphorylation events uncovered a likely AKT phosphorylation site at residue Thr397 of PPAT, which was only detected in cells under purine-supplemented growth conditions. These data suggest that this modification might alter enzyme activity or modulate its interaction(s) with downstream pathway enzymes. Together, these findings propose a role for post-translational modifications in pathway regulation and activation to meet intracellular purine demand.


Subject(s)
Amidophosphoribosyltransferase/metabolism , Peptide Mapping/methods , Protein Processing, Post-Translational , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Purines/metabolism , Acetylation , Adenylosuccinate Lyase/genetics , Adenylosuccinate Lyase/metabolism , Amidophosphoribosyltransferase/genetics , Amino Acid Sequence , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Peptides/chemical synthesis , Peptides/metabolism , Phosphoribosylglycinamide Formyltransferase/genetics , Phosphoribosylglycinamide Formyltransferase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/metabolism , Signal Transduction , Threonine/metabolism , Ubiquitination
7.
Curr Cancer Drug Targets ; 19(5): 408-416, 2019.
Article in English | MEDLINE | ID: mdl-30479216

ABSTRACT

BACKGROUND: Cancer remains one of the most serious disease worldwide. Robust metabolism is the hallmark of cancer. PPAT (phosphoribosyl pyrophosphate amidotransferase) catalyzes the first committed step of de novo purine biosynthesis. Hence PPAT, the key regulatory spot in De novo purine nucleotide biosynthesis, is an attractive and credible drug target for leukemia and other cancer therapeutics. OBJECTIVE: In the present study, detailed computational analysis has been performed for PPAT protein, the key enzyme in de novo purine biosynthesis which is inhibited by many folate derivatives, hence we aimed to investigate and gauge the inhibitory effect of antifolate derivatives; lomexterol (LTX) methotrexate (LTX), and pipretixin (PTX) with human PPAT to effectively capture and inhibit De novo purine biosynthesis pathway. METHODS: The sequence to structure computational approaches followed by molecular docking experiments was performed to gain insight into the inhibitory mode, binding orientation and binding affinities of selected antifolate derivatives against important structural features of PPAT. RESULTS: Results indicated a strong affinity of antifolate inhibitors for the conserved active site of PPAT molecule encompassing a number of hydrophobic, hydrogen bonding, Vander Waals and electrostatic interactions. CONCLUSION: Conclusively, the strong physical interaction of selected antifolate inhibitors with human PPAT suggests the selective inhibition of De novo purine biosynthesis pathway by antifolate derivatives towards cancer therapeutics.


Subject(s)
Amidophosphoribosyltransferase/chemistry , Amidophosphoribosyltransferase/metabolism , Folic Acid Antagonists/metabolism , Molecular Docking Simulation , Purines/metabolism , Amino Acid Sequence , Computer Simulation , Folic Acid Antagonists/chemistry , Humans , Models, Molecular , Neoplasms/drug therapy , Protein Conformation , Sequence Homology
8.
Nat Chem Biol ; 15(2): 141-150, 2019 02.
Article in English | MEDLINE | ID: mdl-30559427

ABSTRACT

The nucleotide ppGpp is a highly conserved regulatory molecule in bacteria that helps tune growth rate to nutrient availability. Despite decades of study, how ppGpp regulates growth remains poorly understood. Here, we developed and validated a capture-compound mass spectrometry approach that identified >50 putative ppGpp targets in Escherichia coli. These targets control many key cellular processes and include 13 enzymes required for nucleotide synthesis. We demonstrated that ppGpp inhibits the de novo synthesis of all purine nucleotides by directly targeting the enzyme PurF. By solving a structure of PurF bound to ppGpp, we designed a mutation that ablates ppGpp-based regulation, leading to dysregulation of purine-nucleotide synthesis following ppGpp accumulation. Collectively, our results provide new insights into ppGpp-based growth control and a nearly comprehensive set of targets for future exploration. The capture compounds developed should also enable the rapid identification of ppGpp targets in any species, including pathogens.


Subject(s)
Escherichia coli/growth & development , Guanosine Pentaphosphate/biosynthesis , Guanosine Pentaphosphate/physiology , Amidophosphoribosyltransferase/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Guanine Nucleotides/biosynthesis , Guanine Nucleotides/physiology , Guanosine Tetraphosphate , Purines/antagonists & inhibitors , Purines/biosynthesis
9.
Plant Cell Environ ; 39(8): 1767-79, 2016 08.
Article in English | MEDLINE | ID: mdl-27004600

ABSTRACT

Purines are essential molecules formed in a highly regulated pathway in all organisms. In tropical legumes, the nitrogen fixed in the nodules is used to generate ureides through the oxidation of de novo synthesized purines. Glutamine phosphoribosyl pyrophosphate amidotransferase (PRAT) catalyses the first committed step of de novo purine synthesis. In Phaseolus vulgaris there are three genes coding for PRAT. The three full-length sequences, which are intron-less genes, were cloned, and their expression levels were determined under conditions that affect the synthesis of purines. One of the three genes, PvPRAT3, is highly expressed in nodules and protein amount and enzymatic activity in these tissues correlate with nitrogen fixation activity. Inhibition of PvPRAT3 gene expression by RNAi-silencing and subsequent metabolomic analysis of the transformed roots shows that PvPRAT3 is essential for the synthesis of ureides in P. vulgaris nodules.


Subject(s)
Amidophosphoribosyltransferase/metabolism , Nitrogen/metabolism , Phaseolus/enzymology , Root Nodules, Plant/metabolism , Amidophosphoribosyltransferase/genetics , Amino Acid Sequence , Isoenzymes/metabolism , Nitrogen Fixation , Phaseolus/genetics , Sequence Analysis, DNA
10.
Oncotarget ; 6(27): 23445-61, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26140362

ABSTRACT

Cancer cells exhibit altered metabolism including aerobic glycolysis that channels several glycolytic intermediates into de novo purine biosynthetic pathway. We discovered increased expression of phosphoribosyl amidotransferase (PPAT) and phosphoribosylaminoimidazole carboxylase, phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS) enzymes of de novo purine biosynthetic pathway in lung adenocarcinomas. Transcript analyses from next-generation RNA sequencing and gene expression profiling studies suggested that PPAT and PAICS can serve as prognostic biomarkers for aggressive lung adenocarcinoma. Immunohistochemical analysis of PAICS performed on tissue microarrays showed increased expression with disease progression and was significantly associated with poor prognosis. Through gene knockdown and over-expression studies we demonstrate that altering PPAT and PAICS expression modulates pyruvate kinase activity, cell proliferation and invasion. Furthermore we identified genomic amplification and aneuploidy of the divergently transcribed PPAT-PAICS genomic region in a subset of lung cancers. We also present evidence for regulation of both PPAT and PAICS and pyruvate kinase activity by L-glutamine, a co-substrate for PPAT. A glutamine antagonist, 6-Diazo-5-oxo-L-norleucine (DON) blocked glutamine mediated induction of PPAT and PAICS as well as reduced pyruvate kinase activity. In summary, this study reveals the regulatory mechanisms by which purine biosynthetic pathway enzymes PPAT and PAICS, and pyruvate kinase activity is increased and exposes an existing metabolic vulnerability in lung cancer cells that can be explored for pharmacological intervention.


Subject(s)
Adenocarcinoma/metabolism , Amidophosphoribosyltransferase/metabolism , Carboxy-Lyases/metabolism , Lung Neoplasms/metabolism , Peptide Synthases/metabolism , Aged , Aneuploidy , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation , Chickens , Diazooxonorleucine/chemistry , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glutamine/chemistry , Glutamine/metabolism , Humans , Male , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Prognosis , Purines/chemistry
11.
Appl Environ Microbiol ; 81(17): 5761-72, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26070680

ABSTRACT

An outstanding challenge toward efficient production of biofuels and value-added chemicals from plant biomass is the impact that lignocellulose-derived inhibitors have on microbial fermentations. Elucidating the mechanisms that underlie their toxicity is critical for developing strategies to overcome them. Here, using Escherichia coli as a model system, we investigated the metabolic effects and toxicity mechanisms of feruloyl amide and coumaroyl amide, the predominant phenolic compounds in ammonia-pretreated biomass hydrolysates. Using metabolomics, isotope tracers, and biochemical assays, we showed that these two phenolic amides act as potent and fast-acting inhibitors of purine and pyrimidine biosynthetic pathways. Feruloyl or coumaroyl amide exposure leads to (i) a rapid buildup of 5-phosphoribosyl-1-pyrophosphate (PRPP), a key precursor in nucleotide biosynthesis, (ii) a rapid decrease in the levels of pyrimidine biosynthetic intermediates, and (iii) a long-term generalized decrease in nucleotide and deoxynucleotide levels. Tracer experiments using (13)C-labeled sugars and [(15)N]ammonia demonstrated that carbon and nitrogen fluxes into nucleotides and deoxynucleotides are inhibited by these phenolic amides. We found that these effects are mediated via direct inhibition of glutamine amidotransferases that participate in nucleotide biosynthetic pathways. In particular, feruloyl amide is a competitive inhibitor of glutamine PRPP amidotransferase (PurF), which catalyzes the first committed step in de novo purine biosynthesis. Finally, external nucleoside supplementation prevents phenolic amide-mediated growth inhibition by allowing nucleotide biosynthesis via salvage pathways. The results presented here will help in the development of strategies to overcome toxicity of phenolic compounds and facilitate engineering of more efficient microbial producers of biofuels and chemicals.


Subject(s)
Amides/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli/metabolism , Phenol/pharmacology , Purines/biosynthesis , Pyrimidines/biosynthesis , Amidophosphoribosyltransferase/antagonists & inhibitors , Amidophosphoribosyltransferase/genetics , Amidophosphoribosyltransferase/metabolism , Biosynthetic Pathways/drug effects , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
12.
Microb Cell Fact ; 13: 101, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25023436

ABSTRACT

BACKGROUND: Purine nucleotides are essential metabolites for living organisms because they are involved in many important processes, such as nucleic acid synthesis, energy supply, and biosynthesis of several amino acids and riboflavin. Owing to the pivotal roles of purines in cell physiology, the pool of intracellular purine nucleotides must be maintained under strict control, and hence the de novo purine biosynthetic pathway is tightly regulated by transcription repression and inhibition mechanism. Deregulation of purine pathway is essential for this pathway engineering in Bacillus subtilis. RESULTS: Deregulation of purine pathway was attempted to improve purine nucleotides supply, based on a riboflavin producer B. subtilis strain with modification of its rib operon. To eliminate transcription repression, the pur operon repressor PurR and the 5'-UTR of pur operon containing a guanine-sensing riboswitch were disrupted. Quantitative RT-PCR analysis revealed that the relative transcription levels of purine genes were up-regulated about 380 times. Furthermore, site-directed mutagenesis was successfully introduced into PRPP amidotransferase (encoded by purF) to remove feedback inhibition by homologous alignment and analysis. Overexpression of the novel mutant PurF (D293V, K316Q and S400W) significantly increased PRPP amidotransferase activity and triggered a strong refractory effect on purine nucleotides mediated inhibition. Intracellular metabolite target analysis indicated that the purine nucleotides supply in engineered strains was facilitated by a stepwise gene-targeted deregulation. With these genetic manipulations, we managed to enhance the metabolic flow through purine pathway and consequently increased riboflavin production 3-fold (826.52 mg/L) in the purF-VQW mutant strain. CONCLUSIONS: A sequential optimization strategy was applied to deregulate the rib operon and purine pathway of B. subtilis to create genetic diversities and to improve riboflavin production. Based on the deregulation of purine pathway at transcription and metabolic levels, an extended application is recommended for the yield of products, like inosine, guanosine, adenosine and folate which are directly stemming from purine pathway in B. subtilis.


Subject(s)
Bacillus subtilis/metabolism , Biosynthetic Pathways , Purines/metabolism , Riboflavin/biosynthesis , Amidophosphoribosyltransferase/metabolism , Amino Acid Sequence , Bacillus subtilis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Feedback, Physiological , Gene Expression Regulation, Bacterial , Molecular Sequence Data , Mutation/genetics , Nucleotides/metabolism , Operon/genetics , Purines/chemistry , Riboflavin/chemistry , Sequence Alignment , Transcription, Genetic
13.
Gene ; 518(2): 280-6, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23357222

ABSTRACT

The first step of the purine de novo synthesis pathway is catalyzed by amidophosphoribosyltransferase (E.C.2.4.2.14) which is encoded by two Prat genes in D. melanogaster, Prat and Prat2. Prat is a retrogene duplication of Prat2, where each gene has a distinct expression pattern. Prat transcription is restricted to proliferating tissues such as imaginal discs and the female germ line. Three conserved putative DNA replication-related element binding factor (DREF) sites lie upstream of the Prat coding region. These elements are upstream of many genes important in cell proliferation. We have found that DREF binds directly upstream of Prat and that the DRE sites associated with its activity are necessary for Prat expression; furthermore, we have determined that a second cis-acting element is present upstream of the Prat gene. Finally, the genes Distal-less, Mi-2 and dMyc, which influence Dref activity, do not appear to affect Prat transcription.


Subject(s)
Amidophosphoribosyltransferase/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , Transcription Factors/genetics , Transcription, Genetic , Adenosine Triphosphatases/genetics , Amidophosphoribosyltransferase/metabolism , Animals , Autoantigens/genetics , Base Sequence , DNA-Binding Proteins/genetics , Drosophila Proteins/metabolism , Female , Homeodomain Proteins/genetics , Imaginal Discs/metabolism , Purines/biosynthesis , Sequence Alignment , Transcription Factors/metabolism
14.
PLoS One ; 7(10): e48207, 2012.
Article in English | MEDLINE | ID: mdl-23133571

ABSTRACT

Phosphoribosylamine (PRA) is an intermediate in the biosynthetic pathway that is common to thiamine and purines. Glutamine phosphoribosyl pyrophosphate (PRPP) amidotransferase is the product of the purF gene in Salmonella enterica and catalyzes the synthesis of PRA from PRPP and glutamine. Strains lacking PurF require exogenous addition of purines for growth. However, under some growth conditions or with specific secondary mutations these strains grow in the absence of exogenous thiamine. Mutant alleles of hisA, which encodes 1-(5-phosphoribosyl)-5-[(5-phosphoribosylamino) methylideneamino] imidazole-4-carboxamide (ProFAR) isomerase, allowed PurF-independent PRA formation. The alleles of hisA that suppressed the requirement for exogenous thiamine resulted in proteins with reduced enzymatic activity. Data presented here showed that decreased activity of HisA altered metabolite pools and allowed PRA formation from ProFAR. Possible mechanisms of this conversion were proposed. The results herein emphasize the plasticity of the metabolic network and specifically highlight the potential for chemical syntheses to contribute to network robustness.


Subject(s)
Amidophosphoribosyltransferase/genetics , Histidine/metabolism , Salmonella enterica/metabolism , Alleles , Amidophosphoribosyltransferase/metabolism , Chromatography, High Pressure Liquid/methods , DNA/metabolism , Histidine/chemistry , Metabolic Networks and Pathways/physiology , Models, Chemical , Models, Genetic , Mutation , Operon , Purines/metabolism , Thiamine/metabolism
15.
Wei Sheng Wu Xue Bao ; 52(6): 718-25, 2012 Jun 04.
Article in Chinese | MEDLINE | ID: mdl-22934352

ABSTRACT

OBJECTIVE: To study the effects of overexpression of key enzyme genes (prs, purF and guaB) on guanosine production in Bacillus amyloliquefaciens TA208. METHODS: The prs, purF, guaB and prs-purF genes were inserted into constructed expression plasmid PBE43. All these constructed plasmids were electroporated into B. amyloliquefaciens TA208. The transcriptional level of various genes in the resulting strains was tested by real-time quantitative PCR. The activity of inosine 5'-monophosphate dehydrogenase in the resulting strains was detected. Finally, cell growth, glucose consumption and guanosine production of 4 engineering strains along with control strain were examined. RESULTS: The transcriptional analysis showed that overexpression of prs, purF and guaB gene accompanied by their own transcription level up-regulated. Overexpression of prs or purF genes alone slightly down-regulated the transcriptional level of purine operon, but overexpression of guaB gene independently did not disturb the transcription of prs gene and purine operon. Enzyme activity analysis showed that overexpression of prs or purF gene did not change the activity of inosine 5'-monophosphate dehydrogenase and its activity increased by 126% through overexpression of guaB gene. Finally, by fermentation flask test, we found that overexpression of prs and purF gene alone could not promote guanosine accumulation. However, overexpression of guaB gene resulted in an increase in the production of guanosine, which was 20.7% higher than the control strain. The guanosine concentration and the conversion ratio from glucose to guanosine in the host strain containing co-expression plasmid were 14.4% and 6.8% higher than the control strain. CONCLUSION: Overexpression of guaB gene could enhance the guanosine yield in the culture broth. However, for prs and purF gene, only co-expression of them could lead to a significant improvement of guanosine production in B. amyloliquefaciens. It should provide a valuable insight into the construction of industrially important strains for guanosine production by metabolic engineering.


Subject(s)
Amidophosphoribosyltransferase/biosynthesis , Bacillus/enzymology , Bacillus/genetics , Guanosine/metabolism , IMP Dehydrogenase/biosynthesis , Ribose-Phosphate Pyrophosphokinase/biosynthesis , Amidophosphoribosyltransferase/genetics , Amidophosphoribosyltransferase/metabolism , Bacillus/metabolism , Genes, Bacterial , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Metabolic Engineering , Plasmids/genetics , Ribose-Phosphate Pyrophosphokinase/genetics , Ribose-Phosphate Pyrophosphokinase/metabolism , Transfection , Up-Regulation
16.
Mol Plant ; 5(6): 1227-41, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22532604

ABSTRACT

A series of reticulated Arabidopsis thaliana mutants were previously described. All mutants show a reticulate leaf pattern, namely green veins on a pale leaf lamina. They have an aberrant mesophyll structure but an intact layer of bundle sheath cells around the veins. Here, we unravel the function of the previously described reticulated EMS-mutant dov1 (differential development of vascular associated cells 1). By positional cloning, we identified the mutated gene, which encodes glutamine phosphoribosyl pyrophosphate aminotransferase 2 (ATase2), an enzyme catalyzing the first step of purine nucleotide biosynthesis. dov1 is allelic to the previously characterized cia1-2 mutant that was isolated in a screen for mutants with impaired chloroplast protein import. We show that purine-derived total cytokinins are lowered in dov1 and crosses with phytohormone reporter lines revealed differential reporter activity patterns in dov1. Metabolite profiling unraveled that amino acids that are involved in purine biosynthesis are increased in dov1. This study identified the molecular basis of an established mutant line, which has the potential for further investigation of the interaction between metabolism and leaf development.


Subject(s)
Amidophosphoribosyltransferase/genetics , Amidophosphoribosyltransferase/metabolism , Arabidopsis/enzymology , Arabidopsis/genetics , Mutation , Plant Leaves/genetics , Purines/metabolism , Alleles , Arabidopsis/cytology , Arabidopsis/metabolism , Base Sequence , Cell Differentiation , Cloning, Molecular , Cytokinins/metabolism , Mesophyll Cells/cytology , Mesophyll Cells/metabolism , Photosynthesis , Plant Growth Regulators/metabolism
17.
Nucleosides Nucleotides Nucleic Acids ; 30(12): 1140-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22132968

ABSTRACT

5-Phosphoribosyl-1-pyrophosphate (PRPP) is an important regulator of de novo purine synthesis. A method for the measurement of PRPP in erythrocytes was designed, which is based on the determination of [(13)C(5)]glutamate derived from [(13)C(5)]glutamine following the utilization of PRPP by the action of amidophosphoribosyltransferase. The present study describes a gas chromatographic-mass spectrometric method for determination of [(13)C(5)]glutamate using [(13)C(2)]glutamate as an internal standard. The methods involved purification by anion-exchange chromatography using a BondElut SAX and derivatization with isobutyl chlorocarbonate in water-methanol-pyridine. Quantitation was performed by selected ion monitoring of the protonated molecular ions in the chemical ionization mode. The intra-day reproducibility in the amounts of [(13)C(5)]glutamate determined was in good agreement with the actual amounts added in erythrocytes. A linear relationship was found between the amount of PRPP added and the amount of [(13)C(5)]glutamate formed from [(13)C(5)]glutamine using amidophosphoribosyltransferase.


Subject(s)
Amidophosphoribosyltransferase/metabolism , Isotope Labeling/methods , Mass Spectrometry/methods , Phosphoribosyl Pyrophosphate/analysis , Carbon Isotopes , Glutamine/metabolism , Humans , Indicator Dilution Techniques
18.
Fungal Genet Biol ; 48(10): 956-65, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21763446

ABSTRACT

Coniothyrium minitans is an important sclerotial parasite of the fungal phytopathogen, Sclerotinia sclerotiorum. Previously, we constructed a T-DNA insertional library, and screened for many conidiation-deficient mutants from this library. Here, we report a T-DNA insertional mutant ZS-1T21882 that completely lost conidiation. In mutant ZS-1T21882, the T-DNA was integrated into a gene (CmPrat-1) which encodes phosphoribosylamidotransferase (PRAT, EC 2.4.2.14), an enzyme catalyzing the first committed step in de novo purine nucleotide synthesis. Gene replacement and complementation experiments confirmed that phosphoribosylamidotransferase is essential for conidiation of C. minitans. Mutant ZS-1T21882 did not grow on modified Czapek-Dox broth (MCD), but it grew well on MCD amended with IMP or AMP. The conidial production of this mutant was dependent on the dosage of IMP amended. At low concentrations, such as 0.1 mM and 0.25 mM, the mutant produced very few pycnidia, while up to 0.75 mM or higher, the conidiation of this mutant was restored completely. cAMP could not restore the conidiation of mutant ZS-1T21882 when amended into MCD, but could when amended into PDA. Neither GMP nor cGMP could restore the conidiation in MCD or in PDA. Our findings suggest that phosphoribosylamidotransferase is essential for conidiation of C. minitans via adenosine related molecules. Furthermore, when dual cultured with its host, this mutant produced conidia in the host mycelium and on the sclerotia of S. sclerotiorum, but not in dead mycelium or on dead sclerotia, suggesting that C. minitans is likely to able to obtain adenosine or related components from its host during parasitization.


Subject(s)
Adenosine/biosynthesis , Amidophosphoribosyltransferase/metabolism , Ascomycota/enzymology , Spores, Fungal/enzymology , Amidophosphoribosyltransferase/biosynthesis , Amino Acid Sequence , Ascomycota/physiology , Cyclic AMP/genetics , Cyclic AMP/metabolism , DNA, Bacterial/genetics , Molecular Sequence Data , Mutation/genetics , Mutation/physiology , Mycelium/enzymology , Signal Transduction , Spores, Fungal/genetics , Spores, Fungal/physiology
19.
Genetics ; 188(2): 359-67, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21441212

ABSTRACT

The biosynthetic pathways and multiple functions of purine nucleotides are well known. However, the pathways that respond to alterations in purine nucleotide synthesis in vivo in an animal model organism have not been identified. We examined the effects of inhibiting purine de novo synthesis in vivo and in cultured cells of Drosophila melanogaster. The purine de novo synthesis gene ade2 encodes phosphoribosylformylglycinamidine synthase (EC 6.3.5.3). An ade2 deletion, generated by P-element transposon excision, causes lethality in early pupal development, with darkening, or necrosis, of leg and wing imaginal disc tissue upon disc eversion. Together with analysis of a previously isolated weaker allele, ade2(4), and an allele of the Prat gene, which encodes an enzyme for the first step in the pathway, we determined that the lethal arrest and imaginal disc phenotypes involve apoptosis. A transgene expressing the baculovirus caspase inhibitor p35, which suppresses apoptosis caused by other stresses such as DNA damage, suppresses both the imaginal disc tissue darkening and the pupal lethality of all three purine de novo synthesis mutants. Furthermore, we showed the presence of apoptosis at the cellular level in both ade2 and Prat mutants by detecting TUNEL-positive nuclei in wing imaginal discs. Purine de novo synthesis inhibition was also examined in tissue culture by ade2 RNA interference followed by analysis of genome-wide changes in transcript levels. Among the upregulated genes was HtrA2, which encodes an apoptosis effector and is thus a candidate for initiating apoptosis in response to purine depletion.


Subject(s)
Apoptosis , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Purine Nucleotides/biosynthesis , Amidophosphoribosyltransferase/genetics , Amidophosphoribosyltransferase/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Biosynthetic Pathways , Blotting, Western , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Cell Line , Crosses, Genetic , DNA Transposable Elements/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Female , Gene Expression Profiling , High-Temperature Requirement A Serine Peptidase 2 , In Situ Nick-End Labeling , Male , Molecular Sequence Data , Mutagenesis, Insertional , Mutation , Oligonucleotide Array Sequence Analysis , Pupa/genetics , Pupa/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
20.
J Mol Biol ; 395(2): 417-29, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19900465

ABSTRACT

Guanosine 5'-monophosphate synthetase(s) (GMPS) catalyzes the final step of the de novo synthetic pathway of purine nucleotides. GMPS consists of two functional units that are present as domains or subunits: glutamine amidotransferase (GATase) and ATP pyrophosphatase (ATPPase). GATase hydrolyzes glutamine to yield glutamate and ammonia, while ATPPase utilizes ammonia to convert adenyl xanthosine 5'-monophosphate (adenyl-XMP) into guanosine 5'-monophosphate. Here we report the crystal structure of PH-ATPPase (the ATPPase subunit of the two-subunit-type GMPS from the hyperthermophilic archaeon Pyrococcus horikoshii OT3). PH-ATPPase consists of two domains (N-domain and C-domain) and exists as a homodimer in the crystal and in solution. The N-domain contains an ATP-binding platform called P-loop, whereas the C-domain contains the xanthosine 5'-monophosphate (XMP)-binding site and also contributes to homodimerization. We have also demonstrated that PH-GATase (the glutamine amidotransferase subunit of the two-subunit-type GMPS from the hyperthermophilic archaeon P. horikoshii OT3) alone is inactive, and that all substrates of PH-ATPPase except for ammonia (Mg(2+), ATP and XMP) are required to stabilize the active complex of PH-ATPPase and PH-GATase subunits.


Subject(s)
Amidophosphoribosyltransferase/chemistry , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/chemistry , Pyrococcus horikoshii/enzymology , Pyrophosphatases/chemistry , Amidophosphoribosyltransferase/genetics , Amidophosphoribosyltransferase/metabolism , Amino Acid Sequence , Ammonia/pharmacology , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Carbon-Nitrogen Ligases , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits , Pyrococcus horikoshii/genetics , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Structural Homology, Protein , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...