Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cancer Sci ; 112(12): 4944-4956, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34533861

ABSTRACT

Diverse metabolic changes are induced by various driver oncogenes during the onset and progression of leukemia. By upregulating glycolysis, cancer cells acquire a proliferative advantage over normal hematopoietic cells; in addition, these changes in energy metabolism contribute to anticancer drug resistance. Because leukemia cells proliferate by consuming glucose as an energy source, an alternative nutrient source is essential when glucose levels in bone marrow are insufficient. We profiled sugar metabolism in leukemia cells and found that mannose is an energy source for glycolysis, the tricarboxylic acid (TCA) cycle, and the pentose phosphate pathway. Leukemia cells express high levels of phosphomannose isomerase (PMI), which mobilizes mannose to glycolysis; consequently, even mannose in the blood can be used as an energy source for glycolysis. Conversely, suppression of PMI expression or a mannose load exceeding the processing capacity of PMI inhibited transcription of genes related to mitochondrial metabolism and the TCA cycle, therefore suppressing the growth of leukemia cells. High PMI expression was also a poor prognostic factor for acute myeloid leukemia. Our findings reveal a new mechanism for glucose starvation resistance in leukemia. Furthermore, the combination of PMI suppression and mannose loading has potential as a novel treatment for driver oncogene-independent leukemia.


Subject(s)
Leukemia/drug therapy , Mannose-6-Phosphate Isomerase/metabolism , Mannose/administration & dosage , Up-Regulation , Animals , Cell Line, Tumor , Citric Acid Cycle/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , Humans , K562 Cells , Leukemia/enzymology , Leukemia/genetics , Leukemia/pathology , Mannose/pharmacology , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mice , Pentose Phosphate Pathway/drug effects , Prognosis , THP-1 Cells , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
2.
Med Oncol ; 38(9): 103, 2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34313879

ABSTRACT

Mannose induces tumor cell apoptosis and inhibits glucose metabolism by accumulating intracellularly as mannose 6-phosphate while the drug sensitivity of tumors is negatively correlated with mannose phosphate isomerase gene (MPI) expression. In this study, we performed a first attempt to explore the relationship between the targeted gene MPI and immune infiltration and genetic and clinical characteristics of head and neck squamous carcinoma (HNSC) using computational algorithms and bioinformatic analysis, and further to verify the co-inhibition effects of mannose with genotoxicity, immune responses, and microbes dysbiosis in oral squamous cell carcinoma (OSCC) in vitro and in vivo. Our results found that patients with lower MPI expression had higher survival rate. The enhancement of MPI expression was in response to DNA damage gene, and ATM inhibitor was verified as a potential drug with a synergistic effect with mannose on HSC-3. In the HNSC, infiltrated immunocytes CD8+ T cell and B cell were the significantly reduced risk cells, while IL-22 and IFN-γ showed negative correlation with MPI. Finally, mannose could reverse immunophenotyping caused by antibiotics in mice, resulting in the decrease of CD8+ T cells and increase of myeloid-derived suppressor cells (MDSCs). In conclusion, the MPI gene showed a significant correlation with immune infiltration and genetic and clinical characteristics of HNSC. The treatment of ATM inhibitor, immune regulating cells of CD8+ T cells and MDSCs, and oral microbiomes in combination with mannose could exhibit co-inhibitory therapeutic effect for OSCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Computational Biology/methods , Lymphocytes, Tumor-Infiltrating/immunology , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannose/pharmacology , Mouth Neoplasms/drug therapy , Animals , Apoptosis , Biomarkers, Tumor/analysis , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Humans , Male , Mannose-6-Phosphate Isomerase/metabolism , Mice , Mice, Inbred BALB C , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Myeloid-Derived Suppressor Cells/immunology , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Article in English | MEDLINE | ID: mdl-30987578

ABSTRACT

BACKGROUND AND OBJECTIVE: The present paper aims to study the inhibition of Candida albicans growth as candidiasis treatment, using seeds of Lepidium sativum as source. METHODS: In vitro assays were carried out on the antifungal activity of three kinds of extracts from L. sativum seeds against four strains of C. albicans, then testing the same phytochemicals on the inhibition of Lipase (LCR). A new in silico study was achieved using molecular docking, with Autodock vina program, to find binding affinity of two important and major lepidine alkaloids (lepidine E and B) towards the four enzymes secreted by C. albicans as target drugs, responsible of vitality and virulence of this yeast cells: Lipase, Serine/threonine phosphatase, Phosphomannose isomerase and Sterol 14-alpha demethylase (CYP51). RESULTS: The results of the microdillution assay show that the hexanic and alkaloidal extracts have an antifungal activity with MICs: 2.25 mg/ml and 4.5mg/ml, respectively. However, Candida rugosa lipase assay gives a remarkable IC50 values for the hexanic extract (1.42± 0.04 mg/ml) followed by 1.7± 0.1 and 2.29 ± 0.09 mg/ml of ethyl acetate and alkaloidal extracts respectively. The molecular docking confirms a significant correlation between C. albicans growth and inhibition of crucial enzymes involved in the invasion mechanism and cellular metabolisms, for the first time there were an interesting and new positive results on binding modes of lepidine E and B on the four studied enzymes. CONCLUSION: Through this work, we propose Lepidine B & E as potent antifungal drugs.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Enzyme Inhibitors/pharmacology , Fungal Proteins/antagonists & inhibitors , Lepidium sativum , Molecular Docking Simulation , Plant Extracts/pharmacology , Seeds , Antifungal Agents/chemistry , Antifungal Agents/isolation & purification , Candida albicans/enzymology , Candida albicans/growth & development , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Lepidium sativum/chemistry , Lipase/antagonists & inhibitors , Lipase/metabolism , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannose-6-Phosphate Isomerase/metabolism , Molecular Targeted Therapy , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Protein Conformation , Seeds/chemistry , Structure-Activity Relationship , Virulence
4.
Dis Model Mech ; 6(1): 95-105, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22899857

ABSTRACT

Individuals with congenital disorders of glycosylation (CDG) have recessive mutations in genes required for protein N-glycosylation, resulting in multi-systemic disease. Despite the well-characterized biochemical consequences in these individuals, the underlying cellular defects that contribute to CDG are not well understood. Synthesis of the lipid-linked oligosaccharide (LLO), which serves as the sugar donor for the N-glycosylation of secretory proteins, requires conversion of fructose-6-phosphate to mannose-6-phosphate via the phosphomannose isomerase (MPI) enzyme. Individuals who are deficient in MPI present with bleeding, diarrhea, edema, gastrointestinal bleeding and liver fibrosis. MPI-CDG patients can be treated with oral mannose supplements, which is converted to mannose-6-phosphate through a minor complementary metabolic pathway, restoring protein glycosylation and ameliorating most symptoms, although liver disease continues to progress. Because Mpi deletion in mice causes early embryonic lethality and thus is difficult to study, we used zebrafish to establish a model of MPI-CDG. We used a morpholino to block mpi mRNA translation and established a concentration that consistently yielded 13% residual Mpi enzyme activity at 4 days post-fertilization (dpf), which is within the range of MPI activity detected in fibroblasts from MPI-CDG patients. Fluorophore-assisted carbohydrate electrophoresis detected decreased LLO and N-glycans in mpi morphants. These deficiencies resulted in 50% embryonic lethality by 4 dpf. Multi-systemic abnormalities, including small eyes, dysmorphic jaws, pericardial edema, a small liver and curled tails, occurred in 82% of the surviving larvae. Importantly, these phenotypes could be rescued with mannose supplementation. Thus, parallel processes in fish and humans contribute to the phenotypes caused by Mpi depletion. Interestingly, mannose was only effective if provided prior to 24 hpf. These data provide insight into treatment efficacy and the broader molecular and developmental abnormalities that contribute to disorders associated with defective protein glycosylation.


Subject(s)
Congenital Disorders of Glycosylation/diet therapy , Congenital Disorders of Glycosylation/enzymology , Mannose-6-Phosphate Isomerase/deficiency , Mannose-6-Phosphate Isomerase/genetics , Mannose/administration & dosage , Animals , Base Sequence , Congenital Disorders of Glycosylation/genetics , Dietary Supplements , Disease Models, Animal , Gene Knockdown Techniques , Humans , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mice , Morpholinos/administration & dosage , Morpholinos/genetics , Mutation , Phenotype , Zebrafish/genetics , Zebrafish/metabolism
5.
Bioorg Med Chem ; 20(4): 1511-20, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22269276

ABSTRACT

In the design of inhibitors of phosphosugar metabolizing enzymes and receptors with therapeutic interest, malonate has been reported in a number of cases as a good and hydrolytically-stable surrogate of the phosphate group, since both functions are dianionic at physiological pH and of comparable size. We have investigated a series of malonate-based mimics of the best known phosphate inhibitors of class II (zinc) fructose-1,6-bis-phosphate aldolases (FBAs) (e.g., from Mycobacterium tuberculosis), type I (zinc) phosphomannose isomerase (PMI) from Escherichia coli, and phosphoglucose isomerase (PGI) from yeast. In the case of FBAs, replacement of one phosphate by one malonate on a bis-phosphorylated inhibitor (1) led to a new compound (4) still showing a strong inhibition (K(i) in the nM range) and class II versus class I selectivity (up to 8×10(4)). Replacement of the other phosphate however strongly affected binding efficiency and selectivity. In the case of PGI and PMI, 5-deoxy-5-malonate-D-arabinonohydroxamic acid (8) yielded a strong decrease in binding affinities when compared to its phosphorylated parent compound 5-phospho-D-arabinonohydroxamic acid (2). Analysis of the deposited 3D structures of the kinetically evaluated enzymes complexed to the phosphate-based inhibitors indicate that malonate could be a good phosphate surrogate only if phosphate is not tightly bound at the enzyme active site, such as in position 7 of compound 1 for FBAs. These observations are of importance for further design of inhibitors of phosphorylated-compounds metabolizing enzymes with therapeutic interest.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Fructose-Bisphosphate Aldolase/antagonists & inhibitors , Glucose-6-Phosphate Isomerase/antagonists & inhibitors , Malonates/chemical synthesis , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Animals , Catalytic Domain , Enzyme Activation/drug effects , Escherichia/enzymology , Humans , Inhibitory Concentration 50 , Malonates/chemistry , Malonates/pharmacology , Models, Biological , Molecular Structure , Yeasts/enzymology
6.
J Biol Chem ; 286(45): 39431-8, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21949237

ABSTRACT

Congenital disorders of glycosylation (CDG) are rare genetic disorders due to impaired glycosylation. The patients with subtypes CDG-Ia and CDG-Ib have mutations in the genes encoding phosphomannomutase 2 (PMM2) and phosphomannose isomerase (MPI or PMI), respectively. PMM2 (mannose 6-phosphate → mannose 1-phosphate) and MPI (mannose 6-phosphate ⇔ fructose 6-phosphate) deficiencies reduce the metabolic flux of mannose 6-phosphate (Man-6-P) into glycosylation, resulting in unoccupied N-glycosylation sites. Both PMM2 and MPI compete for the same substrate, Man-6-P. Daily mannose doses reverse most of the symptoms of MPI-deficient CDG-Ib patients. However, CDG-Ia patients do not benefit from mannose supplementation because >95% Man-6-P is catabolized by MPI. We hypothesized that inhibiting MPI enzymatic activity would provide more Man-6-P for glycosylation and possibly benefit CDG-Ia patients with residual PMM2 activity. Here we show that MLS0315771, a potent MPI inhibitor from the benzoisothiazolone series, diverts Man-6-P toward glycosylation in various cell lines including fibroblasts from CDG-Ia patients and improves N-glycosylation. Finally, we show that MLS0315771 increases mannose metabolic flux toward glycosylation in zebrafish embryos.


Subject(s)
Congenital Disorders of Glycosylation/drug therapy , Congenital Disorders of Glycosylation/enzymology , Enzyme Inhibitors/pharmacology , Fibroblasts/enzymology , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Phosphotransferases (Phosphomutases)/genetics , Animals , Congenital Disorders of Glycosylation/genetics , Enzyme Inhibitors/therapeutic use , Glycosylation/drug effects , HeLa Cells , Humans , Mannose/genetics , Mannose/metabolism , Mannose-6-Phosphate Isomerase/genetics , Mannose-6-Phosphate Isomerase/metabolism , Mannosephosphates/genetics , Mannosephosphates/metabolism , Mutation , Zebrafish/genetics , Zebrafish/metabolism
7.
J Invertebr Pathol ; 108(1): 7-12, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21683706

ABSTRACT

Phosphomannose isomerase (PMI) catalyzes the reversible interconversion of fructose 6-phosphate (Fru-6-P) and mannose 6-phosphate (Man-6-P), providing a link between glycolysis and the mannose metabolic pathway. In this study, we identified pmi gene (Mapmi) from the entomopathogenic fungus, Metarhizium acridum, and analyzed its functions using RNA interference (RNAi). Amending the growth medium with cell stress chemicals significantly reduced growth, conidial production and percent germination in Mapmi-RNAi mutant strain, compared to the wild-type strain. Growth of RNAi mutant was lower than the wild type strain with glucose or fructose as sole carbon source. RNAi mutant exhibited a normal growth phenotype with mannose at low concentrations, while trace or high concentration of mannose was more negatively impacted the growth of RNAi mutant than the wild type strain. Infection with Mapmi-RNAi mutant against Locusta migratoria manilensis (Meyen) led to a significantly reduced virulence compared to infection with the wild-type strain. These results suggest that Mapmi plays essential roles in stress tolerance and pathogenicity of M. acridum.


Subject(s)
Fungal Proteins/genetics , Genes, Fungal , Mannose-6-Phosphate Isomerase/genetics , Metarhizium/genetics , Stress, Physiological , Animals , Cloning, Molecular , Expressed Sequence Tags , Fungal Proteins/antagonists & inhibitors , Gene Expression Regulation, Fungal , Gene Library , Locusta migratoria/microbiology , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Metarhizium/pathogenicity , Metarhizium/physiology , RNA Interference , Spores, Fungal/drug effects , Spores, Fungal/genetics , Spores, Fungal/growth & development , Virulence/genetics
8.
J Phys Chem B ; 115(25): 8304-16, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21650197

ABSTRACT

Using polarizable molecular mechanics, a recent study [de Courcy et al. J. Am. Chem. Soc., 2010, 132, 3312] has compared the relative energy balances of five competing inhibitors of the FAK kinase. It showed that the inclusion of structural water molecules was indispensable for an ordering consistent with the experimental one. This approach is now extended to compare the binding affinities of four active site ligands to the Type I Zn-metalloenzyme phosphomannose isomerase (PMI) from Candida albicans. The first three ones are the PMI substrate ß-D-mannopyranose 6-phosphate (ß-M6P) and two isomers, α-D-mannopyranose 6-phosphate (α-M6P) and ß-D-glucopyranose 6-phosphate (ß-G6P). They have a dianionic 6-phosphate substituent and differ by the relative configuration of the two carbon atoms C1 and C2 of the pyranose ring. The fourth ligand, namely 6-deoxy-6-dicarboxymethyl-ß-D-mannopyranose (ß-6DCM), is a substrate analogue that has the ß-M6P phosphate replaced by the nonhydrolyzable phosphate surrogate malonate. In the energy-minimized structures of all four complexes, one of the ligand hydroxyl groups binds Zn(II) through a water molecule, and the dianionic moiety binds simultaneously to Arg304 and Lys310 at the entrance of the cavity. Comparative energy-balances were performed in which solvation of the complexes and desolvation of PMI and of the ligands are computed using the Langlet-Claverie continuum reaction field procedure. They resulted into a more favorable balance in favor of ß-M6P than α-M6P and ß-G6P, consistent with the experimental results that show ß-M6P to act as a PMI substrate, while α-M6P and ß-G6P are inactive or at best weak inhibitors. However, these energy balances indicated the malonate ligand ß-6DCM to have a much lesser favorable relative complexation energy than the substrate ß-M6P, while it has an experimental 10-fold higher affinity than it on Type I PMI from Saccharomyces cerevisiae. The energy calculations were validated by comparison with parallel ab initio quantum chemistry on model binding sites extracted from the energy-minimized PMI-inhibitor complexes. We sought to improve the models upon including explicit water molecules solvating the dianionic moieties in their ionic bonds with the Arg304 and Lys310 side-chains. Energy-minimization resulted in the formation of three networks of structured waters. The first water of each network binds to one of the three accessible anionic oxygens. The networks extend to PMI residues (Asp17, Glu48, Asp300) remote from the ligand binding site. The final comparative energy balances also took into account ligand desolvation in a box of 64 waters. They now resulted into a large preference in favor of ß-6DCM over ß-M6P. The means to further augment the present model upon including entropy effects and sampling were discussed. Nevertheless a clear-cut conclusion emerging from this as well as our previous study on FAK kinase is that both polarization and charge-transfer contributions are critical elements of the energy balances.


Subject(s)
Fungal Proteins/chemistry , Ligands , Mannose-6-Phosphate Isomerase/chemistry , Mannosephosphates/chemistry , Water/chemistry , Zinc/chemistry , Amino Acid Sequence , Candida albicans/enzymology , Isomerism , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Sequence Alignment , Substrate Specificity , Thermodynamics
9.
J Med Chem ; 54(10): 3661-8, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21539312

ABSTRACT

We report the discovery and validation of a series of benzoisothiazolones as potent inhibitors of phosphomannose isomerase (PMI), an enzyme that converts mannose-6-phosphate (Man-6-P) into fructose-6-phosphate (Fru-6-P) and, more importantly, competes with phosphomannomutase 2 (PMM2) for Man-6-P, diverting this substrate from critical protein glycosylation events. In congenital disorder of glycosylation type Ia, PMM2 activity is compromised; thus, PMI inhibition is a potential strategy for the development of therapeutics. High-throughput screening (HTS) and subsequent chemical optimization led to the identification of a novel class of benzoisothiazolones as potent PMI inhibitors having little or no PMM2 inhibition. Two complementary synthetic routes were developed, enabling the critical structural requirements for activity to be determined, and the compounds were subsequently profiled in biochemical and cellular assays to assess efficacy. The most promising compounds were also profiled for bioavailability parameters, including metabolic stability, plasma stability, and permeability. The pharmacokinetic profile of a representative of this series (compound 19; ML089) was also assessed, demonstrating the potential of this series for in vivo efficacy when dosed orally in disease models.


Subject(s)
Congenital Disorders of Glycosylation/diagnosis , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Thiazoles/chemistry , Administration, Oral , Chemistry, Pharmaceutical/methods , Combinatorial Chemistry Techniques/methods , Congenital Disorders of Glycosylation/genetics , Drug Design , Humans , Inhibitory Concentration 50 , Models, Chemical , Permeability , Structure-Activity Relationship
10.
Proteins ; 79(1): 203-20, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21058398

ABSTRACT

Type I phosphomannose isomerases (PMIs) are zinc-dependent metalloenzymes involved in the reversible isomerization of D-mannose 6-phosphate (M6P) and D-fructose 6-phosphate (F6P). 5-Phospho-D-arabinonohydroxamic acid (5PAH), an inhibitor endowed with nanomolar affinity for yeast (Type I) and Pseudomonas aeruginosa (Type II) PMIs (Roux et al., Biochemistry 2004; 43:2926-2934), strongly inhibits human (Type I) PMI (for which we report an improved expression and purification procedure), as well as Escherichia coli (Type I) PMI. Its K(i) value of 41 nM for human PMI is the lowest value ever reported for an inhibitor of PMI. 5-Phospho-D-arabinonhydrazide, a neutral analogue of the reaction intermediate 1,2-cis-enediol, is about 15 times less efficient at inhibiting both enzymes, in accord with the anionic nature of the postulated high-energy reaction intermediate. Using the polarizable molecular mechanics, sum of interactions between fragments ab initio computed (SIBFA) procedure, computed structures of the complexes between Candida albicans (Type I) PMI and the cyclic substrate ß-D-mannopyranose 6-phosphate (ß-M6P) and between the enzyme and the high-energy intermediate analogue inhibitor 5PAH are reported. Their analysis allows us to identify clearly the nature of each individual active site amino acid and to formulate a hypothesis for the overall mechanism of the reaction catalyzed by Type I PMIs, that is, the ring-opening and isomerization steps, respectively. Following enzyme-catalyzed ring-opening of ß-M6P by zinc-coordinated water and Gln111 ligands, Lys136 is identified as the probable catalytic base involved in proton transfer between the two carbon atoms C1 and C2 of the substrate D-mannose 6-phosphate.


Subject(s)
Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannose-6-Phosphate Isomerase/chemistry , Amino Acid Sequence , Binding, Competitive , Candida albicans/enzymology , Catalytic Domain , Escherichia coli/enzymology , Fructosephosphates/chemistry , Humans , Hydrazines/chemistry , Hydroxamic Acids/chemistry , Kinetics , Mannose-6-Phosphate Isomerase/biosynthesis , Mannosephosphates/chemistry , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Conformation , Sequence Alignment , Sugar Phosphates/chemistry
11.
Bioorg Med Chem ; 17(20): 7100-7, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19783448

ABSTRACT

Non-hydrolyzable d-mannose 6-phosphate analogues in which the phosphate group was replaced by a phosphonomethyl, a dicarboxymethyl, or a carboxymethyl group were synthesized and kinetically evaluated as substrate analogues acting as potential inhibitors of type I phosphomannose isomerases (PMIs) from Saccharomyces cerevisiae and Escherichia coli. While 6-deoxy-6-phosphonomethyl-d-mannose and 6-deoxy-6-carboxymethyl-D-mannose did not inhibit the enzymes significantly, 6-deoxy-6-dicarboxymethyl-D-mannose appeared as a new strong competitive inhibitor of both S. cerevisiae and E. coli PMIs with K(m)/K(i) ratios of 28 and 8, respectively. We thus report the first malonate-based inhibitor of an aldose-ketose isomerase to date. Phosphonomethyl mimics of the 1,2-cis-enediolate high-energy intermediate postulated for the isomerization reaction catalyzed by PMIs were also synthesized but behave as poor inhibitors of PMIs. A polarizable molecular mechanics (SIBFA) study was performed on the complexes of d-mannose 6-phosphate and two of its analogues with PMI from Candida albicans, an enzyme involved in yeast infection homologous to S. cerevisiae and E. coli PMIs. It shows that effective binding to the catalytic site occurs with retention of the Zn(II)-bound water molecule. Thus the binding of the hydroxyl group on C1 of the ligand to Zn(II) should be water-mediated. The kinetic study reported here also suggests the dianionic character of the phosphate surrogate as a likely essential parameter for strong binding of the inhibitor to the enzyme active site.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannosephosphates/chemical synthesis , Mannosephosphates/pharmacology , Uronic Acids/pharmacology , Chromatography, Ion Exchange , Drug Evaluation, Preclinical , Kinetics , Magnetic Resonance Spectroscopy , Mannose-6-Phosphate Isomerase/chemistry , Mannose-6-Phosphate Isomerase/metabolism , Models, Molecular , Saccharomyces cerevisiae/enzymology , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity
12.
FEBS Lett ; 582(23-24): 3479-83, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18801364

ABSTRACT

Phosphomannose isomerase (PMI) plays a pivotal role in biosynthesis of GDP-mannose, an important precursor of many polysaccharides. We demonstrate in this study that Pseudomonas aeruginosa pslB encodes a protein with GDP-mannose pyrophosphorylase/PMI dual activities. The PMI activity is Co2+-dependent and could be inhibited by GDP-mannose in a competitive manner. Furthermore, the activity could be inactivated by 2,3-butanedione suggesting the presence of a catalytic Arg residue. Site-specific mutations at R373, R472, R479, E410, H411, N433 and E458 increase the KM approximately 8-20-fold. The PMI activity of PslB was completely diminished with a R408K or R408A, reflecting the importance of this residue in catalysis. Overall, these results provide a basis for understanding the catalytic mechanism of PMI.


Subject(s)
Bacterial Proteins/metabolism , Mannose-6-Phosphate Isomerase/metabolism , Pseudomonas aeruginosa/enzymology , Amino Acid Sequence , Amino Acid Substitution , Arginine/chemistry , Arginine/genetics , Arginine/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Catalysis , Catalytic Domain , Cobalt/metabolism , DNA Mutational Analysis , Diacetyl/pharmacology , Enzyme Inhibitors/pharmacology , Guanosine Diphosphate Mannose/metabolism , Guanosine Diphosphate Mannose/pharmacology , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannose-6-Phosphate Isomerase/genetics , Molecular Sequence Data , Mutation
13.
J Comput Chem ; 28(5): 938-57, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17253648

ABSTRACT

Type I phosphomannose isomerase (PMI) is a Zn-dependent metalloenzyme involved in the isomerization of D-fructose 6-phosphate to D-mannose 6-phosphate. One of our laboratories has recently designed and synthesized 5-phospho-D-arabinonohydroxamate (5PAH), an inhibitor endowed with a nanomolar affinity for PMI (Roux et al., Biochemistry 2004, 43, 2926). By contrast, the 5-phospho-D-arabinonate (5PAA), in which the hydroxamate moiety is replaced by a carboxylate one, is devoid of inhibitory potency. Subsequent biochemical studies showed that in its PMI complex, 5PAH binds Zn(II) through its hydroxamate moiety rather than through its phosphate. These results have stimulated the present theoretical investigation in which we resort to the SIBFA polarizable molecular mechanics procedure to unravel the structural and energetical aspects of 5PAH and 5PAA binding to a 164-residue model of PMI. Consistent with the experimental results, our theoretical studies indicate that the complexation of PMI by 5PAH is much more favorable than by 5PAA, and that in the 5PAH complex, Zn(II) ligation by hydroxamate is much more favorable than by phosphate. Validations by parallel quantum-chemical computations on model of the recognition site extracted from the PMI-inhibitor complexes, and totaling up to 140 atoms, showed the values of the SIBFA intermolecular interaction energies in such models to be able to reproduce the quantum-chemistry ones with relative errors < 3%. On the basis of the PMI-5PAH SIBFA energy-minimized structure, we report the first hypothesis of a detailed view of the active site of the zinc PMI complexed to the high-energy intermediate analogue inhibitor, which allows us to identify active site residues likely involved in the proton transfer between the two adjacent carbons of the substrates.


Subject(s)
Candida albicans/enzymology , Computer Simulation , Hydroxamic Acids/metabolism , Mannose-6-Phosphate Isomerase/metabolism , Pentosephosphates/metabolism , Quantum Theory , Sugar Phosphates/metabolism , Zinc/metabolism , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Hydroxamic Acids/antagonists & inhibitors , Hydroxamic Acids/chemistry , Isomerism , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannose-6-Phosphate Isomerase/chemistry , Molecular Conformation , Pentosephosphates/antagonists & inhibitors , Pentosephosphates/chemistry , Sugar Phosphates/antagonists & inhibitors , Sugar Phosphates/chemistry , Zinc/chemistry
14.
J Enzyme Inhib Med Chem ; 21(2): 187-92, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16791965

ABSTRACT

D-Sorbitol-6-phosphate 2-dehydrogenase catalyzes the NADH-dependent conversion of D-fructose 6-phosphate to D-sorbitol 6-phosphate and improved production and purification of the enzyme from Escherichia coli is reported. Preliminary inhibition studies of the enzyme revealed 5-phospho-D-arabinonohydroxamic acid and 5-phospho-D-arabinonate as new substrate analogue inhibitors of the F6P catalyzed reduction with IC50 values of (40 +/- 1) microM and (48 +/- 3) microM and corresponding Km/IC50 ratio values of 14 and 12, respectively. Furthermore, we report here the phosphomannose isomerase substrate D-mannose 6-phosphate as the best inhibitor of E. coli D-sorbitol-6-phosphate 2-dehydrogenase yet reported with an IC50 = 7.5 +/- 0.4 microM and corresponding Km/IC50 ratio = about 76.


Subject(s)
Enzyme Inhibitors/pharmacology , Escherichia coli/enzymology , Sugar Alcohol Dehydrogenases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Glucose-6-Phosphate Isomerase/antagonists & inhibitors , Glucose-6-Phosphate Isomerase/metabolism , Hexosephosphates/metabolism , Hexosephosphates/pharmacology , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacology , Kinetics , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannose-6-Phosphate Isomerase/metabolism , Mannosephosphates/metabolism , Mannosephosphates/pharmacology , Pentosephosphates/metabolism , Pentosephosphates/pharmacology , Substrate Specificity , Sugar Alcohol Dehydrogenases/isolation & purification , Sugar Alcohol Dehydrogenases/metabolism , Sugar Phosphates/metabolism , Sugar Phosphates/pharmacology
15.
Anal Chem ; 77(17): 5596-603, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16131071

ABSTRACT

Electrospray ionization Fourier transform ion cyclotron resonance (ESI-FTICR) mass spectrometry was used to study the noncovalent complexation of a metalloenzyme, phosphomannose isomerase (PMI), which catalyzes the interconversion of mannose 6-phosphate and fructose 6-phosphate. The zinc cofactor binding effect and the noncovalent interactions of the holoenzyme with its two natural substrates and two inhibitors, erythrose 4-phosphate and mannitol 1-phosphate, were investigated. Under nondenaturing conditions, the intact zinc-containing monomeric protein ions were reproducibly observed with no dissociation. Molecular ions corresponding to apo-PMI monomer were obtained by dialyzing the holoenzyme against EDTA. The binding/release of the metal ion did not alter the charge-state distributions of the protein to any significant extent, but changed the binding affinity of the substrates by at least 5-fold. Using ESI-FTICR mass spectrometry, the binding stoichiometry and specificity of the enzyme-substrate and enzyme-inhibitor complexes were directly determined. The first time report of the apparent dissociation constant for the isomeric substrates of PMI was measured to be 88.8 microM. The relative dissociation constant of the two inhibitors derived from gas-phase noncovalent complexation was very similar to the relative inhibition constant derived from solution phase kinetics.


Subject(s)
Mannose-6-Phosphate Isomerase/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Spectroscopy, Fourier Transform Infrared/methods , Zinc/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Kinetics , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Molecular Structure , Substrate Specificity , Zinc/chemistry
16.
Biochemistry ; 43(10): 2926-34, 2004 Mar 16.
Article in English | MEDLINE | ID: mdl-15005628

ABSTRACT

The phosphomannose isomerases (PMI) comprise three families of proteins: type I, type II, and type III PMIs. Members of all three families catalyze the reversible isomerization of D-mannose 6-phosphate (M6P) and D-fructose 6-phosphate (F6P) but share little or no sequence identity. Because (1) PMIs are essential for the survival of several microorganisms, including yeasts and bacteria, and (2) the PMI enzymes from several pathogens do not share significant sequence identity to the human protein, PMIs have been considered as potential therapeutic targets. Elucidation of the catalytic and regulatory mechanisms of the different types of PMIs is strongly needed for rational species-specific drug design. To date, inhibition and crystallographic studies of all PMIs are still largely unexplored. As part of our research program on aldose-ketose isomerases, we report in this paper the evaluation of two new inhibitors of type I and type II PMIs from baker's yeast and Pseudomonas aeruginosa, respectively. We found that 5-phospho-D-arabinonohydroxamic acid (5PAH), which is the most potent inhibitor of phosphoglucose isomerase (PGI), is by far the best inhibitor ever reported of both type I and type II PMI-catalyzed isomerization of M6P to F6P. 5PAH, which has an inhibition constant at least 3 orders of magnitude smaller than that of previously reported PMI inhibitors, may be the first high-energy intermediate analogue inhibitor of the enzymes. We also tested the related molecule 5-phospho-D-arabinonate (5PAA), which is a strong competitive inhibitor of PGI, and found that it does not inhibit either PMI. All together, our results are consistent with a catalytic role for the metal cofactor in PMI activity.


Subject(s)
Coenzymes/chemistry , Hydroxamic Acids/chemistry , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannose-6-Phosphate Isomerase/chemistry , Metals/chemistry , Sugar Phosphates/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Binding, Competitive , Catalysis , Enzyme Inhibitors/chemical synthesis , Fructosephosphates/chemistry , Glucose-6-Phosphate Isomerase/antagonists & inhibitors , Glucose-6-Phosphate Isomerase/chemistry , Isomerism , Kinetics , Mannose-6-Phosphate Isomerase/classification , Mannosephosphates/chemistry , Models, Chemical , Pseudomonas aeruginosa/enzymology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/chemistry
17.
J Enzyme Inhib ; 16(3): 287-92, 2001.
Article in English | MEDLINE | ID: mdl-11697049

ABSTRACT

Phosphomannose isomerase (PMI; EC. 5.3.1.8) is an essential metalloenzyme in the early steps of the protein glycosylation pathway in both prokaryotes and eukaryotes. The Cys150 residue (according to Candida albicans PMI numbering) is conserved in the active centre of mammalian and yeast PMI, but not in bacterial species where it is replaced by Asn. Here, the dose- and time-dependent inhibitory effect of the NO-donor S-nitroso-acetyl-penicillamine on the Saccharomyces cerevisiae PMI catalytic activity is reported. The analysis of the X-ray crystal structure of C. albicans PMI and of the molecular model of S. cerevisiae PMI provides a rationale for the low reactivity of Cys150 towards alkylating and nitrosylating agents.


Subject(s)
Enzyme Inhibitors/pharmacology , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , S-Nitroso-N-Acetylpenicillamine/pharmacology , Saccharomyces cerevisiae/enzymology , Binding Sites , Enzyme Inhibitors/metabolism , Mannose-6-Phosphate Isomerase/chemistry , Mannose-6-Phosphate Isomerase/metabolism , Molecular Structure , Protein Structure, Tertiary , S-Nitroso-N-Acetylpenicillamine/metabolism , Time Factors , Zinc/metabolism
18.
J Enzyme Inhib ; 15(6): 517-31, 2000.
Article in English | MEDLINE | ID: mdl-11140608

ABSTRACT

Aminobenzolamide (5-sulfanilylamido-1,3,4-thiadiazole-2-sulfonamide) is a potent inhibitor of the zinc enzyme carbonic anhydrase (CA, EC 4.2.1.1), being at the same time structurally similar to the antimicrobial sulfonamides. Here we report that the reaction of aminobenzolamide with arylsulfonyl isocyanates affords a series of new arylsulfonylureido derivatives which were subsequently used as ligands (in the form of conjugate bases, as sulfonamide anions) for the preparation of metal complexes containing Ag(I) and Zn(II). All the new compounds proved to be very potent inhibitors of CA (isozymes I, II and IV). The newly synthesized complexes, unlike the free ligands, also act as effective antifungal agents against several Aspergillus and Candida spp., some of them showing activities comparable to ketoconazole, with minimum inhibitory concentrations in the range of 1.8-5 microg/mL. The mechanism of antifungal action of these complexes seem to be unconnected with inhibition of lanosterol-14-alpha-demethylase, since the levels of sterols assessed in the fungi cultures were equal in the absence or in the presence of the tested compounds. Probably the new complexes act as inhibitors of phosphomannose isomerase, a key enzyme in the biosynthesis of yeast cell walls.


Subject(s)
Antifungal Agents/pharmacology , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/pharmacology , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Aminobenzoates/chemical synthesis , Aminobenzoates/pharmacology , Animals , Antifungal Agents/chemical synthesis , Aspergillus/drug effects , Aspergillus/growth & development , Aspergillus/metabolism , Benzolamide/chemical synthesis , Benzolamide/pharmacology , Candida/drug effects , Candida/growth & development , Candida/metabolism , Carbonic Anhydrases/drug effects , Carbonic Anhydrases/metabolism , Cattle , Humans , Kinetics , Ligands , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Microbial Sensitivity Tests , Silver/chemistry , Sterols/metabolism , Structure-Activity Relationship , Thiadiazoles/chemical synthesis , Thiadiazoles/pharmacology , Zinc/chemistry
19.
Bioorg Med Chem Lett ; 8(17): 2303-8, 1998 Sep 08.
Article in English | MEDLINE | ID: mdl-9873532

ABSTRACT

Phosphomannose Isomerase (PMI) has been shown by genetic methods to be an essential enzyme in fungal cell wall biosynthesis. The PMI inhibitor AF14049 was discovered as an unanticipated side product from high-throughput library screening against the enzyme from C, albicans. Solid-phase synthetic methods were developed and a series of libraries and discrete analogs synthesized to explore SAR around AF14049.


Subject(s)
Amides/chemical synthesis , Databases as Topic , Enzyme Inhibitors/chemical synthesis , Indans/chemical synthesis , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Animals , Binding Sites , Candida albicans/enzymology , Cell Wall/metabolism , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Indans/chemistry , Indans/pharmacology , Indicators and Reagents , Kinetics , Mannose-6-Phosphate Isomerase/chemistry , Molecular Structure , Saccharomyces cerevisiae/enzymology , Structure-Activity Relationship , Swine
20.
Mol Cell Biochem ; 177(1-2): 183-91, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9450661

ABSTRACT

Phosphomannose isomerase (pmi, EC 5.3.1.8) was purified to homogeneity from a wild strain of Xanthomonas campestris. The apparent molecular weight as determined by SDS-PAGE and Sephadex G-100 Superfine was found to be 58 kDa. The purified enzyme showed a single band on acrylamide gel electrophocusing with pI = 5.25. The optimum pH was 7.0 and the Km for D-mannose-6-phosphate was 2 mM. Pmi can be activated by bivalent cations with the order of Co2+>Zn2+>Mn2+>Ni2+>Ca2+. Addition of low concentration of ZnCl2 (2 x 10[-7] M) in the growth medium resulted in the enhancement of pmi activity to around 2.5 x fold. The half life of pmi, as it was measured by the addition of chloramphenicol, was 110 min, whereas in the medium supplemented with ZnCl2 was 270 min. Chemical modification experiments implied the existence of one histidyl residue located at or near the active site.


Subject(s)
Mannose-6-Phosphate Isomerase/metabolism , Xanthomonas campestris/enzymology , Zinc/pharmacology , Diethyl Pyrocarbonate/pharmacology , Enzyme Activation/drug effects , Enzyme Stability/drug effects , Half-Life , Isoelectric Point , Kinetics , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mannose-6-Phosphate Isomerase/genetics , Mannose-6-Phosphate Isomerase/isolation & purification , Molecular Weight , Reducing Agents/pharmacology , Transcription, Genetic/drug effects , Xanthomonas campestris/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...