Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters








Publication year range
1.
Cancers (Basel) ; 16(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39001459

ABSTRACT

Aldehyde dehydrogenases of the subfamily 1A (ALDH1A) are enzymes necessary for the oxidation of all-trans or 9-cis retinal to retinoic acid (RA). Retinoic acid and its derivatives are important for normal development and maintenance of epithelia, reproduction, memory, and immune function in adults. Moreover, in recent years, it has been demonstrated that ALDH1A members are also expressed and functional in several human cancers where their role is not limited to the synthesis of RA. Here, we review the current knowledge about ALDH1A3, one of the 1A isoforms, in cancers with an emphasis on two of the deadliest tumors that affect humans: glioblastoma multiforme and mesothelioma. In both tumors, ALDH1A3 is considered a negative prognostic factor, and its level correlates with excessive proliferation, chemoresistance, and invasiveness. We also review the recent attempts to develop both ALDH1A3-selective inhibitors for cancer therapy and ALDH1A3-specific fluorescent substrates for fluorescence-guided tumor resection.

2.
Int J Mol Sci ; 24(7)2023 Apr 03.
Article in English | MEDLINE | ID: mdl-37047661

ABSTRACT

Aldehyde dehydrogenase 1A3 (ALDH1A3), one of the three members of the aldehyde dehydrogenase 1A subfamily, has been associated with increased progression and drug resistance in various types of solid tumours. Recently, it has been reported that high ALDH1A3 expression is prognostic of poor survival in patients with malignant pleural mesothelioma (MPM), an asbestos-associated chemoresistant cancer. We treated MPM cells, cultured as multicellular spheroids, with NR6, a potent and highly selective ALDH1A3 inhibitor. Here we report that NR6 treatment caused the accumulation of toxic aldehydes, induced DNA damage, CDKN2A expression and cell growth arrest. We observed that, in CDKN2A proficient cells, NR6 treatment induced IL6 expression, but abolished CXCL8 expression and IL-8 release, preventing both neutrophil recruitment and generation of neutrophil extracellular traps (NETs). Furthermore, we demonstrate that in response to ALDH1A3 inhibition, CDKN2A loss skewed cell fate from senescence to apoptosis. Dissecting the role of ALDH1A3 isoform in MPM cells and tumour microenvironment can open new fronts in the treatment of this cancer.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Humans , Aldehyde Dehydrogenase , Cell Line, Tumor , Enzyme Inhibitors/therapeutic use , Lung Neoplasms/genetics , Mesothelioma/drug therapy , Mesothelioma/genetics , Mesothelioma/metabolism , Neutrophil Infiltration , Pleural Neoplasms/pathology , Spheroids, Cellular/metabolism , Tumor Microenvironment , Retinal Dehydrogenase/metabolism
3.
Commun Biol ; 5(1): 895, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36050388

ABSTRACT

Glioblastoma (GBM) is the most aggressive primary brain tumour for which both effective treatments and efficient tools for an early-stage diagnosis are lacking. Herein, we present curcumin-based fluorescent probes that are able to bind to aldehyde dehydrogenase 1A3 (ALDH1A3), an enzyme overexpressed in glioma stem cells (GSCs) and associated with stemness and invasiveness of GBM. Two compounds are selective versus ALDH1A3, without showing any appreciable interaction with other ALDH1A isoenzymes. Indeed, their fluorescent signal is detectable only in our positive controls in vitro and absent in cells that lack ALDH1A3. Remarkably, in vivo, our Probe selectively accumulate in glioblastoma cells, allowing the identification of the growing tumour mass. The significant specificity of our compounds is the necessary premise for their further development into glioblastoma cells detecting probes to be possibly used during neurosurgical operations.


Subject(s)
Aldehyde Oxidoreductases , Brain Neoplasms , Curcumin , Glioblastoma , Aldehyde Dehydrogenase/chemistry , Aldehyde Dehydrogenase/metabolism , Aldehyde Oxidoreductases/chemistry , Aldehyde Oxidoreductases/metabolism , Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Brain Neoplasms/surgery , Curcumin/metabolism , Curcumin/pharmacology , Early Diagnosis , Fluorescent Dyes/metabolism , Glioblastoma/diagnosis , Glioblastoma/metabolism , Glioblastoma/surgery , Humans , Neoplastic Stem Cells/metabolism
4.
Commun Biol ; 4(1): 1420, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934174

ABSTRACT

Elevated aldehyde dehydrogenase (ALDH) activity correlates with poor outcome for many solid tumors as ALDHs may regulate cell proliferation and chemoresistance of cancer stem cells (CSCs). Accordingly, potent, and selective inhibitors of key ALDH enzymes may represent a novel CSC-directed treatment paradigm for ALDH+ cancer types. Of the many ALDH isoforms, we and others have implicated the elevated expression of ALDH1A3 in mesenchymal glioma stem cells (MES GSCs) as a target for the development of novel therapeutics. To this end, our structure of human ALDH1A3 combined with in silico modeling identifies a selective, active-site inhibitor of ALDH1A3. The lead compound, MCI-INI-3, is a selective competitive inhibitor of human ALDH1A3 and shows poor inhibitory effect on the structurally related isoform ALDH1A1. Mass spectrometry-based cellular thermal shift analysis reveals that ALDH1A3 is the primary binding protein for MCI-INI-3 in MES GSC lysates. The inhibitory effect of MCI-INI-3 on retinoic acid biosynthesis is comparable with that of ALDH1A3 knockout, suggesting that effective inhibition of ALDH1A3 is achieved with MCI-INI-3. Further development is warranted to characterize the role of ALDH1A3 and retinoic acid biosynthesis in glioma stem cell growth and differentiation.


Subject(s)
Aldehyde Oxidoreductases/antagonists & inhibitors , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Tretinoin/metabolism , Humans
5.
Cancers (Basel) ; 13(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478031

ABSTRACT

Aldehyde dehydrogenase 1A3 (ALDH1A3) belongs to an enzymatic superfamily composed by 19 different isoforms, with a scavenger role, involved in the oxidation of a plethora of aldehydes to the respective carboxylic acids, through a NAD+-dependent reaction. Previous clinical studies highlighted the high expression of ALDH1A3 in cancer stem cells (CSCs) correlated to a higher risk of cancer relapses, chemoresistance and a poor clinical outcome. We report on the structural, biochemical, and cellular characterization of NR6, a new selective ALDH1A3 inhibitor derived from an already published ALDH non-selective inhibitor with cytotoxic activity on glioblastoma and colorectal cancer cells. Crystal structure, through X-Ray analysis, showed that NR6 binds a non-conserved tyrosine residue of ALDH1A3 which drives the selectivity towards this isoform, as supported by computational binding simulations. Moreover, NR6 shows anti-metastatic activity in wound healing and invasion assays and induces the downregulation of cancer stem cell markers. Overall, our work confirms the role of ALDH1A3 as an important target in glioblastoma and colorectal cells and propose NR6 as a promising molecule for future preclinical studies.

6.
ACS Med Chem Lett ; 11(5): 963-970, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32435412

ABSTRACT

Members of the aldehyde dehydrogenase 1A family are commonly acknowledged as hallmarks of cancer stem cells, and their overexpression is significantly associated with poor prognosis in different types of malignancies. Accordingly, treatments targeting these enzymes may represent a successful strategy to fight cancer. In this work we describe a novel series of imidazo[1,2-a]pyridines, designed as aldehyde dehydrogenase inhibitors by means of a structure-based optimization of a previously developed lead. The novel compounds were evaluated in vitro for their activity and selectivity against the three isoforms of the ALDH1A family and investigated through crystallization and modeling studies for their ability to interact with the catalytic site of the 1A3 isoform. Compound 3f emerged as the first in class submicromolar competitive inhibitor of the target enzyme.

7.
Biochem Soc Trans ; 48(2): 693-707, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32311017

ABSTRACT

Dehydrogenases are oxidoreductase enzymes that play a variety of fundamental functions in the living organisms and have primary roles in pathogen survival and infection processes as well as in cancer development. We review here a sub-set of NAD-dependent dehydrogenases involved in human diseases and the recent advancements in drug development targeting pathogen-associated NAD-dependent dehydrogenases. We focus also on the molecular aspects of the inhibition process listing the structures of the most relevant molecules targeting this enzyme family. Our aim is to review the most impacting findings regarding the discovery of novel inhibitory compounds targeting the selected NAD-dependent dehydrogenases involved in cancer and infectious diseases.


Subject(s)
Communicable Diseases/drug therapy , Drug Discovery , NAD/chemistry , Neoplasms/drug therapy , Oxidoreductases/metabolism , Aldehyde Oxidoreductases/chemistry , Animals , Antimalarials/therapeutic use , Antitubercular Agents/therapeutic use , Drug Design , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Inhibins/chemistry , Isoenzymes/chemistry , Malaria/drug therapy , Tuberculosis/drug therapy
8.
J Med Chem ; 63(9): 4603-4616, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32223240

ABSTRACT

Glioblastoma multiforme (GBM) is the deadliest form of brain tumor. It is known for its ability to escape the therapeutic options available to date thanks to the presence of a subset of cells endowed with stem-like properties and ability to resist to cytotoxic treatments. As the cytosolic enzyme aldehyde dehydrogenase 1A3 turns out to be overexpressed in these kinds of cells, playing a key role for their vitality, treatments targeting this enzyme may represent a successful strategy to fight GBM. In this work, we describe a novel class of imidazo[1,2-a]pyridine derivatives as aldehyde dehydrogenase 1A3 inhibitors, reporting the evidence of their significance as novel drug candidates for the treatment of GBM. Besides showing an interesting functional profile, in terms of activity against the target enzyme and selectivity toward highly homologous isoenzymes, representative examples of the series also showed a nanomolar to picomolar efficacy against patient-derived GBM stem-like cells, thus proving the concept that targeting aldehyde dehydrogenase might represent a novel and promising way to combat GBM by striking its ability to divide immortally.


Subject(s)
Aldehyde Oxidoreductases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Neoplastic Stem Cells/drug effects , Pyridines/pharmacology , Aldehyde Oxidoreductases/chemistry , Aldehyde Oxidoreductases/metabolism , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Glioblastoma/drug therapy , Humans , Imidazoles/chemical synthesis , Imidazoles/metabolism , Molecular Docking Simulation , Molecular Structure , Protein Binding , Pyridines/chemical synthesis , Pyridines/metabolism , Structure-Activity Relationship
9.
J Biol Chem ; 295(11): 3635-3651, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31988240

ABSTRACT

All cells require sustained intracellular energy flux, which is driven by redox chemistry at the subcellular level. NAD+, its phosphorylated variant NAD(P)+, and its reduced forms NAD(P)/NAD(P)H are all redox cofactors with key roles in energy metabolism and are substrates for several NAD-consuming enzymes (e.g. poly(ADP-ribose) polymerases, sirtuins, and others). The nicotinamide salvage pathway, constituted by nicotinamide mononucleotide adenylyltransferase (NMNAT) and nicotinamide phosphoribosyltransferase (NAMPT), mainly replenishes NAD+ in eukaryotes. However, unlike NMNAT1, NAMPT is not known to be a nuclear protein, prompting the question of how the nuclear NAD+ pool is maintained and how it is replenished upon NAD+ consumption. In the present work, using human and murine cells; immunoprecipitation, pulldown, and surface plasmon resonance assays; and immunofluorescence, small-angle X-ray scattering, and MS-based analyses, we report that GAPDH and NAMPT form a stable complex that is essential for nuclear translocation of NAMPT. This translocation furnishes NMN to replenish NAD+ to compensate for the activation of NAD-consuming enzymes by stressful stimuli induced by exposure to H2O2 or S-nitrosoglutathione and DNA damage inducers. These results indicate that by forming a complex with GAPDH, NAMPT can translocate to the nucleus and thereby sustain the stress-induced NMN/NAD+ salvage pathway.


Subject(s)
Cell Nucleus/enzymology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , NAD/metabolism , Nicotinamide Mononucleotide/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Stress, Physiological , Animals , Cell Line, Tumor , HeLa Cells , Humans , Kinetics , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , NIH 3T3 Cells , Nicotinamide Mononucleotide/chemistry , Nicotinamide Phosphoribosyltransferase/chemistry , Protein Binding , Protein Multimerization , Protein Transport
10.
Sci Rep ; 10(1): 651, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959836

ABSTRACT

Changes in nicotinamide adenine dinucleotide (NAD+) levels that compromise mitochondrial function trigger release of DNA damaging reactive oxygen species. NAD+ levels also affect DNA repair capacity as NAD+ is a substrate for PARP-enzymes (mono/poly-ADP-ribosylation) and sirtuins (deacetylation). The ecto-5'-nucleotidase CD73, an ectoenzyme highly expressed in cancer, is suggested to regulate intracellular NAD+ levels by processing NAD+ and its bio-precursor, nicotinamide mononucleotide (NMN), from tumor microenvironments, thereby enhancing tumor DNA repair capacity and chemotherapy resistance. We therefore investigated whether expression of CD73 impacts intracellular NAD+ content and NAD+-dependent DNA repair capacity. Reduced intracellular NAD+ levels suppressed recruitment of the DNA repair protein XRCC1 to sites of genomic DNA damage and impacted the amount of accumulated DNA damage. Further, decreased NAD+ reduced the capacity to repair DNA damage induced by DNA alkylating agents. Overall, reversal of these outcomes through NAD+ or NMN supplementation was independent of CD73. In opposition to its proposed role in extracellular NAD+ bioprocessing, we found that recombinant human CD73 only poorly processes NMN but not NAD+. A positive correlation between CD73 expression and intracellular NAD+ content could not be made as CD73 knockout human cells were efficient in generating intracellular NAD+ when supplemented with NAD+ or NMN.


Subject(s)
5'-Nucleotidase/metabolism , 5'-Nucleotidase/physiology , DNA Damage , DNA Repair , NAD/metabolism , NAD/physiology , Poly ADP Ribosylation , Poly(ADP-ribose) Polymerases/physiology , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology , 5'-Nucleotidase/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Sirtuins , X-ray Repair Cross Complementing Protein 1/metabolism
11.
PLoS One ; 12(4): e0175815, 2017.
Article in English | MEDLINE | ID: mdl-28419153

ABSTRACT

Mycobacterium smegmatis represents one model for studying the biology of its pathogenic relative Mycobacterium tuberculosis. The structural characterization of a M. tuberculosis ortholog protein can serve as a valid tool for the development of molecules active against the M. tuberculosis target. In this context, we report the biochemical and structural characterization of M. smegmatis phosphoribosylpyrophosphate synthetase (PrsA), the ortholog of M. tuberculosis PrsA, the unique enzyme responsible for the synthesis of phosphoribosylpyrophosphate (PRPP). PRPP is a key metabolite involved in several biosynthetic pathways including those for histidine, tryptophan, nucleotides and decaprenylphosphoryl-arabinose, an essential precursor for the mycobacterial cell wall biosynthesis. Since M. tuberculosis PrsA has been validated as a drug target for the development of antitubercular agents, the data presented here will add to the knowledge of the mycobacterial enzyme and could contribute to the development of M. tuberculosis PrsA inhibitors of potential pharmacological interest.


Subject(s)
Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium smegmatis/enzymology , Ribose-Phosphate Pyrophosphokinase/chemistry , Ribose-Phosphate Pyrophosphokinase/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Catalytic Domain , Crystallography, X-Ray , Drug Discovery , Humans , Models, Molecular , Molecular Targeted Therapy , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium smegmatis/chemistry , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/metabolism , Protein Conformation , Sequence Alignment , Substrate Specificity
12.
Cell Chem Biol ; 24(5): 553-564.e4, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28416276

ABSTRACT

Adenosine 5'-tetraphosphate (Ap4) is a ubiquitous metabolite involved in cell signaling in mammals. Its full physiological significance remains unknown. Here we show that two enzymes committed to NAD biosynthesis, nicotinamide phosphoribosyltransferase (NAMPT) and nicotinate phosphoribosyltransferase (NAPT), can both catalyze the synthesis and degradation of Ap4 through their facultative ATPase activity. We propose a mechanism for this unforeseen additional reaction, and demonstrate its evolutionary conservation in bacterial orthologs of mammalian NAMPT and NAPT. Furthermore, evolutionary distant forms of NAMPT were inhibited in vitro by the FK866 drug but, remarkably, it does not block synthesis of Ap4. In fact, FK866-treated murine cells showed decreased NAD but increased Ap4 levels. Finally, murine cells and plasma with engineered or naturally fluctuating NAMPT levels showed matching Ap4 fluctuations. These results suggest a role of Ap4 in the actions of NAMPT, and prompt to evaluate the role of Ap4 production in the actions of NAMPT inhibitors.


Subject(s)
Adenine Nucleotides/biosynthesis , Adenine Nucleotides/metabolism , Cytokines/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Pentosyltransferases/metabolism , Adenosine Triphosphate/metabolism , Animals , Biocatalysis , Cell Line, Tumor , Evolution, Molecular , Humans , Hydrolysis , Mice
13.
Sci Rep ; 6: 35710, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759097

ABSTRACT

The aldehyde dehydrogenase family 1 member A3 (ALDH1A3) catalyzes the oxidation of retinal to the pleiotropic factor retinoic acid using NAD+. The level of ALDHs enzymatic activity has been used as a cancer stem cell marker and seems to correlate with tumour aggressiveness. Elevated ALDH1A3 expression in mesenchymal glioma stem cells highlights the potential of this isozyme as a prognosis marker and drug target. Here we report the first crystal structure of human ALDH1A3 complexed with NAD+ and the product all-trans retinoic acid (REA). The tetrameric ALDH1A3 folds into a three domain-based architecture highly conserved along the ALDHs family. The structural analysis revealed two different and coupled conformations for NAD+ and REA that we propose to represent two snapshots along the catalytic cycle. Indeed, the isoprenic moiety of REA points either toward the active site cysteine, or moves away adopting the product release conformation. Although ALDH1A3 shares high sequence identity with other members of the ALDH1A family, our structural analysis revealed few peculiar residues in the 1A3 isozyme active site. Our data provide information into the ALDH1As catalytic process and can be used for the structure-based design of selective inhibitors of potential medical interest.


Subject(s)
Aldehyde Oxidoreductases/chemistry , Aldehyde Oxidoreductases/metabolism , NAD/chemistry , NAD/metabolism , Tretinoin/chemistry , Tretinoin/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Prohibitins , Protein Binding , Protein Conformation , Protein Domains , Protein Folding , Protein Multimerization
14.
Biochem J ; 473(2): 123-33, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26512127

ABSTRACT

Mycobacterium tuberculosis O(6)-methylguanine-DNA methyltransferase (MtOGT) contributes to protect the bacterial GC-rich genome against the pro-mutagenic potential of O(6)-methylated guanine in DNA. Several strains of M. tuberculosis found worldwide encode a point-mutated O(6)-methylguanine-DNA methyltransferase (OGT) variant (MtOGT-R37L), which displays an arginine-to-leucine substitution at position 37 of the poorly functionally characterized N-terminal domain of the protein. Although the impact of this mutation on the MtOGT activity has not yet been proved in vivo, we previously demonstrated that a recombinant MtOGT-R37L variant performs a suboptimal alkylated-DNA repair in vitro, suggesting a direct role for the Arg(37)-bearing region in catalysis. The crystal structure of MtOGT complexed with modified DNA solved in the present study reveals details of the protein-protein and protein-DNA interactions occurring during alkylated-DNA binding, and the protein capability also to host unmodified bases inside the active site, in a fully extrahelical conformation. Our data provide the first experimental picture at the atomic level of a possible mode of assembling three adjacent MtOGT monomers on the same monoalkylated dsDNA molecule, and disclose the conformational flexibility of discrete regions of MtOGT, including the Arg(37)-bearing random coil. This peculiar structural plasticity of MtOGT could be instrumental to proper protein clustering at damaged DNA sites, as well as to protein-DNA complexes disassembling on repair.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , DNA Damage/genetics , Mycobacterium tuberculosis/genetics , O(6)-Methylguanine-DNA Methyltransferase/chemistry , O(6)-Methylguanine-DNA Methyltransferase/genetics , Crystallography , Point Mutation/genetics , Protein Structure, Secondary , Protein Structure, Tertiary
15.
FEBS Open Bio ; 5: 419-28, 2015.
Article in English | MEDLINE | ID: mdl-26042198

ABSTRACT

Nicotinic acid phosphoribosyltransferase (EC 2.4.2.11) (NaPRTase) is the rate-limiting enzyme in the three-step Preiss-Handler pathway for the biosynthesis of NAD. The enzyme catalyzes the conversion of nicotinic acid (Na) and 5-phosphoribosyl-1-pyrophosphate (PRPP) to nicotinic acid mononucleotide (NaMN) and pyrophosphate (PPi). Several studies have underlined the importance of NaPRTase for NAD homeostasis in mammals, but no crystallographic data are available for this enzyme from higher eukaryotes. Here, we report the crystal structure of human NaPRTase that was solved by molecular replacement at a resolution of 2.9 Å in its ligand-free form. Our structural data allow the assignment of human NaPRTase to the type II phosphoribosyltransferase subfamily and reveal that the enzyme consists of two domains and functions as a dimer with the active site located at the interface of the monomers. The substrate-binding mode was analyzed by molecular docking simulation and provides hints into the catalytic mechanism. Moreover, structural comparison of human NaPRTase with the other two human type II phosphoribosyltransferases involved in NAD biosynthesis, quinolinate phosphoribosyltransferase and nicotinamide phosphoribosyltransferase, reveals that while the three enzymes share a conserved overall structure, a few distinctive structural traits can be identified. In particular, we show that NaPRTase lacks a tunnel that, in nicotinamide phosphoribosiltransferase, represents the binding site of its potent and selective inhibitor FK866, currently used in clinical trials as an antitumoral agent.

16.
J Bacteriol ; 195(12): 2728-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23564173

ABSTRACT

Mycobacterium tuberculosis displays remarkable genetic stability despite continuous exposure to the hostile environment represented by the host's infected macrophages. Similarly to other organisms, M. tuberculosis possesses multiple systems to counteract the harmful potential of DNA alkylation. In particular, the suicidal enzyme O(6)-methylguanine-DNA methyltransferase (OGT) is responsible for the direct repair of O(6)-alkylguanine in double-stranded DNA and is therefore supposed to play a central role in protecting the mycobacterial genome from the risk of G · C-to-A · T transition mutations. Notably, a number of geographically widely distributed M. tuberculosis strains shows nonsynonymous single-nucleotide polymorphisms in their OGT-encoding gene, leading to amino acid substitutions at position 15 (T15S) or position 37 (R37L) of the N-terminal domain of the corresponding protein. However, the role of these mutations in M. tuberculosis pathogenesis is unknown. We describe here the in vitro characterization of M. tuberculosis OGT (MtOGT) and of two point-mutated versions of the protein mimicking the naturally occurring ones, revealing that both mutated proteins are impaired in their activity as a consequence of their lower affinity for alkylated DNA than the wild-type protein. The analysis of the crystal structures of MtOGT and MtOGT-R37L confirms the high level of structural conservation of members of this protein family and provides clues to an understanding of the molecular bases for the reduced affinity for the natural substrate displayed by mutated MtOGT. Our in vitro results could contribute to validate the inferred participation of mutated OGTs in M. tuberculosis phylogeny and biology.


Subject(s)
Mycobacterium tuberculosis/enzymology , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Amino Acid Sequence , Crystallography, X-Ray , DNA Mutational Analysis , DNA, Bacterial/metabolism , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Point Mutation , Protein Binding , Protein Conformation
17.
J Biomol Struct Dyn ; 30(1): 89-101, 2012.
Article in English | MEDLINE | ID: mdl-22571435

ABSTRACT

Laccases are multicopper oxidases in which substrate oxidation takes place at the type-1 (T1) copper site. The redox potential (E (0)) significantly varies amongst members of the family and is a key parameter for substrate specificity. Despite sharing highly conserved features at the T1 copper site, laccases span a large range of E (0), suggesting that the influence of the metal secondary coordination sphere is important. In silico analysis of structural determinants modulating the E (0) of Rigidoporus lignosus and other fungal laccases indicated that different factors can be considered. First, the length of the T1 copper coordinating histidine bond is observed to be longer in high E (0) laccases than in low E (0) ones. The hydrophobic environment around the T1 copper site appeared as another important structural determinant in modulating the E (0), with a stronger hydrophobic environment correlating with higher E (0). The analysis of hydrogen bonding network (HBN) around the T1-binding pocket revealed that the amino acids building up the metal binding site strongly interact with neighbouring residues and contribute to the stabilization of the protein folds. Changes in these HBNs that modified the Cu1 preferred coordination geometry lead to an increase of E (0). The presence of axial ligands modulates the E (0) of T1 to different extent. Stacking interactions between aromatic residues located in the second coordination shell and the metal ion coordination histidine imidazole ring have also been identified as a factor that modulates the E (0). The electrostatic interactions between the T1 copper site and backbone carbonyl oxygen indicate that Cu1-CO=NH distance is longer in the high E (0) laccases. In short, the in silico study reported herein identifies several structural factors that may influence the E (0) of the examined laccases. Some of these are dependent on the nature of the coordination ligands at the T1 site, but others can be ascribed to the hydrophobic effects, HBNs, axial ligations, stacking and electrostatic interactions, not necessary directly interacting with the copper metal.


Subject(s)
Fungal Proteins/chemistry , Fungi/enzymology , Laccase/chemistry , Models, Molecular , Amino Acids/chemistry , Binding Sites , Fungal Proteins/metabolism , Histidine/analogs & derivatives , Histidine/chemistry , Hydrogen Bonding , Laccase/metabolism , Ligands , Organometallic Compounds/chemistry , Oxidation-Reduction , Protein Binding , Protein Conformation , Static Electricity
18.
Biochem J ; 441(1): 131-41, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21933152

ABSTRACT

Haemophilus influenzae is a major pathogen of the respiratory tract in humans that has developed the capability to exploit host NAD(P) for its nicotinamide dinucleotide requirement. This strategy is organized around a periplasmic enzyme termed NadN (NAD nucleotidase), which plays a central role by degrading NAD into adenosine and NR (nicotinamide riboside), the latter being subsequently internalized by a specific permease. We performed a biochemical and structural investigation on H. influenzae NadN which determined that the enzyme is a Zn2+-dependent 5'-nucleotidase also endowed with NAD(P) pyrophosphatase activity. A 1.3 Å resolution structural analysis revealed a remarkable conformational change that occurs during catalysis between the open and closed forms of the enzyme. NadN showed a broad substrate specificity, recognizing either mono- or di-nucleotide nicotinamides and different adenosine phosphates with a maximal activity on 5'-adenosine monophosphate. Sequence and structural analysis of H. influenzae NadN led us to discover that human CD73 is capable of processing both NAD and NMN, therefore disclosing a possible novel function of human CD73 in systemic NAD metabolism. Our data may prove to be useful for inhibitor design and disclosed unanticipated fascinating evolutionary relationships.


Subject(s)
5'-Nucleotidase/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Haemophilus influenzae/enzymology , Nucleotidases/metabolism , Pyrophosphatases/metabolism , Adenosine Diphosphate , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Binding Sites , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Crystallization , Haemophilus influenzae/genetics , Haemophilus influenzae/metabolism , Humans , Models, Molecular , Molecular Sequence Data , NAD/metabolism , Nicotinamide Mononucleotide/metabolism , Nucleotidases/genetics , Protein Conformation , Pyrophosphatases/genetics , Zinc/chemistry
19.
J Med Chem ; 53(15): 5684-9, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20684605

ABSTRACT

Fluctuations in the brain levels of the neuromodulator kynurenic acid may control cognitive processes and play a causative role in several catastrophic brain diseases. Elimination of the pyridoxal 5'-phosphate dependent enzyme kynurenine aminotransferase II reduces cerebral kynurenic acid synthesis and has procognitive effects. The present description of the crystal structure of human kynurenine aminotransferase II in complex with its potent and specific primary amine-bearing fluoroquinolone inhibitor (S)-(-)-9-(4-aminopiperazin-1-yl)-8-fluoro-3-methyl-6-oxo-2,3-dihydro-6H-1-oxa-3a-azaphenalene-5-carboxylic acid (BFF-122) should facilitate the structure-based development of cognition-enhancing drugs. From a medicinal chemistry perspective our results demonstrate that the issue of inhibitor specificity for highly conserved PLP-dependent enzymes could be successfully addressed.


Subject(s)
Fluoroquinolones/chemistry , Nootropic Agents/chemistry , Piperazines/chemistry , Pyridoxal Phosphate/physiology , Transaminases/chemistry , Adult , Brain/drug effects , Brain/enzymology , Crystallography, X-Ray , Fluoroquinolones/pharmacology , Humans , In Vitro Techniques , Models, Molecular , Nootropic Agents/pharmacology , Piperazines/pharmacology , Protein Conformation , Stereoisomerism , Transaminases/antagonists & inhibitors
20.
J Biomol Struct Dyn ; 27(4): 501-10, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19916571

ABSTRACT

Laccases are polyphenol oxidases which oxidize a broad range of reducing substrates, preferably phenolic compounds, and their use in biotechnological applications is increasing. Recently, the first X-ray structure of active laccase from white rot fungus Rigidoporus lignosus has been reported containing a full complement of copper ions. Comparison among selected fungal laccases of known 3D structure has shown that the Rigidoporus lignosus laccase has a very high similarity with the Trametes versicolor laccase that, being co-crystallized with 2,5-xylidine, shows a well defined binding pocket for the substrate. Global sequence alignment between Rigidoporus lignosus and Trametes versicolor laccases shows 73% of identity but, surprisingly, there is no identity and neither conservative substitutions between the residues composing the loops directly contacting the 2,5-xylidine. Moreover the structural alignment of these two enzymes identifies in these loops a striking structural similarity proposing the question if 2,5- xylidine may bind in same enzyme pocket. Here we report the protein-ligand docking simulation of 3D structure of Rigidoporus -lignosus laccase and 2,5-xylidine. Docking simulation analyses show that spatial conformation of the two 2,5-xylidine binding pockets, despite differences in the residues directly contacting the ligand, may arrange a similar pocket that allows a comparable accommodation of the inhibitor. To validate these results the binding of 2,5-xylidine in the substrate cavity has been confirmed by kinetic competitive experiments.


Subject(s)
Aniline Compounds/chemistry , Aniline Compounds/metabolism , Basidiomycota/enzymology , Laccase/chemistry , Laccase/metabolism , Amino Acid Sequence , Basidiomycota/chemistry , Basidiomycota/genetics , Binding Sites , Binding, Competitive , Catalytic Domain , Computer Simulation , Crystallization , Crystallography, X-Ray , Laccase/genetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL