Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Biol Chem ; 298(7): 102119, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35691342

RESUMO

The metal-dependent M17 aminopeptidases are conserved throughout all kingdoms of life. This large enzyme family is characterized by a conserved binuclear metal center and a distinctive homohexameric arrangement. Recently, we showed that hexamer formation in Plasmodium M17 aminopeptidases was controlled by the metal ion environment, although the functional necessity for hexamer formation is still unclear. To further understand the mechanistic role of the hexameric assembly, here we undertook an investigation of the structure and dynamics of the M17 aminopeptidase from Plasmodium falciparum, PfA-M17. We describe a novel structure of PfA-M17, which shows that the active sites of each trimer are linked by a dynamic loop, and loop movement is coupled with a drastic rearrangement of the binuclear metal center and substrate-binding pocket, rendering the protein inactive. Molecular dynamics simulations and biochemical analyses of PfA-M17 variants demonstrated that this rearrangement is inherent to PfA-M17, and that the transition between the active and inactive states is metal dependent and part of a dynamic regulatory mechanism. Key to the mechanism is a remodeling of the binuclear metal center, which occurs in response to a signal from the neighboring active site and serves to moderate the rate of proteolysis under different environmental conditions. In conclusion, this work identifies a precise mechanism by which oligomerization contributes to PfA-M17 function. Furthermore, it describes a novel role for metal cofactors in the regulation of enzymes, with implications for the wide range of metalloenzymes that operate via a two-metal ion catalytic center, including DNA processing enzymes and metalloproteases.


Assuntos
Aminopeptidases , Plasmodium falciparum/enzimologia , Aminopeptidases/química , Aminopeptidases/metabolismo , Domínio Catalítico , Metais/metabolismo , Plasmodium falciparum/metabolismo
2.
J Biol Chem ; 296: 100173, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33303633

RESUMO

M17 leucyl aminopeptidases are metal-dependent exopeptidases that rely on oligomerization to diversify their functional roles. The M17 aminopeptidases from Plasmodium falciparum (PfA-M17) and Plasmodium vivax (Pv-M17) function as catalytically active hexamers to generate free amino acids from human hemoglobin and are drug targets for the design of novel antimalarial agents. However, the molecular basis for oligomeric assembly is not fully understood. In this study, we found that the active site metal ions essential for catalytic activity have a secondary structural role mediating the formation of active hexamers. We found that PfA-M17 and Pv-M17 exist in a metal-dependent dynamic equilibrium between active hexameric species and smaller inactive species that can be controlled by manipulating the identity and concentration of metals available. Mutation of residues involved in metal ion binding impaired catalytic activity and the formation of active hexamers. Structural resolution of Pv-M17 by cryoelectron microscopy and X-ray crystallography together with solution studies revealed that PfA-M17 and Pv-M17 bind metal ions and substrates in a conserved fashion, although Pv-M17 forms the active hexamer more readily and processes substrates faster than PfA-M17. On the basis of these studies, we propose a dynamic equilibrium between monomer ↔ dimer ↔ tetramer ↔ hexamer, which becomes directional toward the large oligomeric states with the addition of metal ions. This sophisticated metal-dependent dynamic equilibrium may apply to other M17 aminopeptidases and underpin the moonlighting capabilities of this enzyme family.


Assuntos
Aminopeptidases/química , Manganês/química , Plasmodium falciparum/enzimologia , Plasmodium vivax/enzimologia , Multimerização Proteica , Proteínas de Protozoários/química , Aminopeptidases/genética , Aminopeptidases/metabolismo , Domínio Catalítico , Cátions Bivalentes , Clonagem Molecular , Cobalto/química , Cobalto/metabolismo , Microscopia Crioeletrônica , Cristalografia por Raios X , Dipeptídeos/química , Dipeptídeos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Cinética , Magnésio/química , Magnésio/metabolismo , Manganês/metabolismo , Modelos Moleculares , Mutação , Plasmodium falciparum/genética , Plasmodium vivax/genética , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Zinco/química , Zinco/metabolismo
3.
Mol Microbiol ; 116(2): 397-415, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33756056

RESUMO

Endolysin enzymes from bacteriophage cause bacterial lysis by degrading the peptidoglycan cell wall. The streptococcal C1 phage endolysin PlyC, is the most potent endolysin described to date and can rapidly lyse group A, C, and E streptococci. PlyC is known to bind the Group A streptococcal cell wall, but the specific molecular target or the binding site within PlyC remain uncharacterized. Here we report for the first time, that the polyrhamnose backbone of the Group A streptococcal cell wall is the binding target of PlyC. We have also characterized the putative rhamnose binding groove of PlyC and found four key residues that were critical to either the folding or the cell wall binding action of PlyC. Based on our results, we suggest that the interaction between PlyC and the cell wall may not be a high-affinity interaction as previously proposed, but rather a high avidity one, allowing for PlyC's remarkable lytic activity. Resistance to our current antibiotics is reaching crisis levels and there is an urgent need to develop the antibacterial agents with new modes of action. A detailed understanding of this potent endolysin may facilitate future developments of PlyC as a tool against the rise of antibiotic resistance.


Assuntos
Bacteriófagos/metabolismo , Endopeptidases/metabolismo , Peptidoglicano/metabolismo , Ramnose/metabolismo , Streptococcus pyogenes/virologia , Bacteriófagos/genética , Sítios de Ligação/fisiologia , Membrana Celular/metabolismo , Parede Celular/metabolismo , Endopeptidases/genética , Simulação de Acoplamento Molecular , Ligação Proteica/fisiologia , Streptococcus pyogenes/metabolismo
4.
Biochem J ; 478(13): 2697-2713, 2021 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-34133730

RESUMO

During malarial infection, Plasmodium parasites digest human hemoglobin to obtain free amino acids for protein production and maintenance of osmotic pressure. The Plasmodium M1 and M17 aminopeptidases are both postulated to have an essential role in the terminal stages of the hemoglobin digestion process and are validated drug targets for the design of new dual-target anti-malarial compounds. In this study, we profiled the substrate specificity fingerprints and kinetic behaviors of M1 and M17 aminopeptidases from Plasmodium falciparum and Plasmodium vivax, and the mouse model species, Plasmodium berghei. We found that although the Plasmodium M1 aminopeptidases share a largely similar, broad specificity at the P1 position, the P. falciparum M1 displays the greatest diversity in specificity and P. berghei M1 showing a preference for charged P1 residues. In contrast, the Plasmodium M17 aminopeptidases share a highly conserved preference for hydrophobic residues at the P1 position. The aminopeptidases also demonstrated intra-peptide sequence specificity, particularly the M1 aminopeptidases, which showed a definitive preference for peptides with fewer negatively charged intrapeptide residues. Overall, the P. vivax and P. berghei enzymes had a faster substrate turnover rate than the P. falciparum enzymes, which we postulate is due to subtle differences in structural dynamicity. Together, these results build a kinetic profile that allows us to better understand the catalytic nuances of the M1 and M17 aminopeptidases from different Plasmodium species.


Assuntos
Aminopeptidases/metabolismo , Peptídeos/metabolismo , Plasmodium/enzimologia , Proteínas de Protozoários/metabolismo , Aminopeptidases/classificação , Aminopeptidases/genética , Animais , Biocatálise/efeitos dos fármacos , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Leucina/análogos & derivados , Leucina/farmacologia , Malária/parasitologia , Camundongos , Plasmodium/genética , Plasmodium/fisiologia , Plasmodium berghei/enzimologia , Plasmodium berghei/genética , Plasmodium falciparum/enzimologia , Plasmodium falciparum/genética , Plasmodium vivax/enzimologia , Plasmodium vivax/genética , Inibidores de Proteases/farmacologia , Proteínas de Protozoários/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Especificidade por Substrato
5.
Biochem J ; 477(19): 3819-3832, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-32926129

RESUMO

Toxoplasmosis is a parasitic disease caused by infection with Toxoplasma gondii that currently has few therapeutic options. The M1 aminopeptidase enzymes have been shown to be attractive targets for anti-parasitic agents and/or vaccine candidates, suggesting potential to re-purpose inhibitors between parasite M1 aminopeptidase targets. The M1 aminopeptidase TgAPN2 has been suggested to be a potential new drug target for toxoplasmosis. Here we investigate the structure and function of TgAPN2, a homologue of the antimalarial drug target PfA-M1, and evaluate the capacity to use inhibitors that target PfA-M1 against TgAPN2. The results show that despite a similar overall fold, the TgAPN2 has a unique substrate specificity and inhibition profile. Sequence and structure differences are investigated and show how comparative structure-activity relationships may provide a route to obtaining potent inhibitors of TgAPN2.


Assuntos
Aminopeptidases/química , Proteínas de Protozoários/química , Toxoplasma/enzimologia , Cristalografia por Raios X
6.
Proteins ; 85(5): 945-950, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27936485

RESUMO

The cytochrome P450 monooxygenases (P450s) catalyze a vast array of oxygenation reactions that can be useful in biocatalytic applications. CYP101J2 from Sphingobium yanoikuyae is a P450 that catalyzes the hydroxylation of 1,8-cineole. Here we report the crystallization and X-ray structure elucidation of recombinant CYP101J2 to 1.8 Å resolution. The CYP101J2 structure shows the canonical P450-fold and has an open conformation in the absence of substrate. Analysis of the structure revealed that CYP101J2, in the absence of substrate, forms a well-ordered substrate-binding channel that suggests a unique form of substrate guidance in comparison to other bacterial 1,8-cineole-hydroxylating P450 enzymes. Proteins 2017; 85:945-950. © 2016 Wiley Periodicals, Inc.


Assuntos
Proteínas de Bactérias/química , Cicloexanóis/química , Sistema Enzimático do Citocromo P-450/química , Monoterpenos/química , Sphingomonadaceae/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Clonagem Molecular , Cristalografia por Raios X , Cicloexanóis/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Eucaliptol , Expressão Gênica , Hidroxilação , Modelos Moleculares , Monoterpenos/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sphingomonadaceae/enzimologia , Especificidade por Substrato
7.
Biochim Biophys Acta Proteins Proteom ; 1865(11 Pt A): 1336-1347, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28844738

RESUMO

Immunoglobulin E (IgE) is the antibody that plays a central role in the mechanisms of allergic diseases such as asthma. Interactions with its receptors, FcεRI on mast cells and CD23 on B cells, are mediated by the Fc region, a dimer of the Cε2, Cε3 and Cε4 domains. A sub-fragment lacking the Cε2 domains, Fcε3-4, also binds to both receptors, although receptor binding almost exclusively involves the Cε3 domains. This domain also contains the N-linked glycosylation site conserved in other isotypes. We report here the crystal structures of IgE-Fc and Fcε3-4 at the highest resolutions yet determined, 1.75Šand 2.0Šrespectively, revealing unprecedented detail regarding the carbohydrate and its interactions with protein domains. Analysis of the crystallographic B-factors of these, together with all earlier IgE-Fc and Fcε3-4 structures, shows that the Cε3 domains exhibit the greatest intrinsic flexibility and quaternary structural variation within IgE-Fc. Intriguingly, both well-ordered carbohydrate and disordered polypeptide can be seen within the same Cε3 domain. A simplified method for comparing the quaternary structures of the Cε3 domains in free and receptor-bound IgE-Fc structures is presented, which clearly delineates the FcεRI and CD23 bound states. Importantly, differential scanning fluorimetric analysis of IgE-Fc and Fcε3-4 identifies Cε3 as the domain most susceptible to thermally-induced unfolding, and responsible for the characteristically low melting temperature of IgE.


Assuntos
Imunoglobulina E/química , Fragmentos Fc das Imunoglobulinas/química , Receptores de IgE/química , Motivos de Aminoácidos , Sítios de Ligação , Sequência de Carboidratos , Cristalografia por Raios X , Expressão Gênica , Glicosilação , Humanos , Imunoglobulina E/genética , Imunoglobulina E/imunologia , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Modelos Moleculares , Transição de Fase , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Estabilidade Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Desdobramento de Proteína , Receptores de IgE/genética , Receptores de IgE/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Temperatura
8.
Biochem J ; 473(19): 3189-204, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27462122

RESUMO

Malaria is one of the world's most prevalent parasitic diseases, with over 200 million cases annually. Alarmingly, the spread of drug-resistant parasites threatens the effectiveness of current antimalarials and has made the development of novel therapeutic strategies a global health priority. Malaria parasites have a complicated lifecycle, involving an asymptomatic 'liver stage' and a symptomatic 'blood stage'. During the blood stage, the parasites utilise a proteolytic cascade to digest host hemoglobin, which produces free amino acids absolutely necessary for parasite growth and reproduction. The enzymes required for hemoglobin digestion are therefore attractive therapeutic targets. The final step of the cascade is catalyzed by several metalloaminopeptidases, including aminopeptidase P (APP). We developed a novel platform to examine the substrate fingerprint of APP from Plasmodium falciparum (PfAPP) and to show that it can catalyze the removal of any residue immediately prior to a proline. Further, we have determined the crystal structure of PfAPP and present the first examination of the 3D structure of this essential malarial enzyme. Together, these analyses provide insights into potential mechanisms of inhibition that could be used to develop novel antimalarial therapeutics.


Assuntos
Aminopeptidases/metabolismo , Plasmodium falciparum/enzimologia , Aminopeptidases/química , Animais , Cristalografia por Raios X , Dimerização , Escherichia coli/enzimologia , Modelos Moleculares , Conformação Proteica , Proteólise , Especificidade por Substrato
9.
Proteins ; 83(4): 789-95, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25645579

RESUMO

New anti-malarial treatments are desperately required to face the spread of drug resistant parasites. Inhibition of metalloaminopeptidases, PfA-M1 and PfA-M17, is a validated therapeutic strategy for treatment of Plasmodium falciparum malaria. Here, we describe the crystal structures of PfA-M1 and PfA-M17 bound to chemotherapeutic agent Tosedostat. The inhibitor occupies the enzymes' putative product egress channels in addition to the substrate binding pockets; however, adopts different binding poses when bound to PfA-M1 and PfA-M17. These findings will be valuable for the continued development of selective inhibitors of PfA-M1 and PfA-M17.


Assuntos
Antimaláricos/química , Glicina/análogos & derivados , Ácidos Hidroxâmicos/química , Plasmodium falciparum , Proteínas de Protozoários/química , Aminopeptidases/antagonistas & inibidores , Antimaláricos/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Glicina/química , Glicina/metabolismo , Ácidos Hidroxâmicos/metabolismo , Modelos Moleculares , Proteínas de Protozoários/metabolismo
10.
Biochem J ; 461(3): 349-69, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25008945

RESUMO

Despite a century of control and eradication campaigns, malaria remains one of the world's most devastating diseases. Our once-powerful therapeutic weapons are losing the war against the Plasmodium parasite, whose ability to rapidly develop and spread drug resistance hamper past and present malaria-control efforts. Finding new and effective treatments for malaria is now a top global health priority, fuelling an increase in funding and promoting open-source collaborations between researchers and pharmaceutical consortia around the world. The result of this is rapid advances in drug discovery approaches and technologies, with three major methods for antimalarial drug development emerging: (i) chemistry-based, (ii) target-based, and (iii) cell-based. Common to all three of these approaches is the unique ability of structural biology to inform and accelerate drug development. Where possible, SBDD (structure-based drug discovery) is a foundation for antimalarial drug development programmes, and has been invaluable to the development of a number of current pre-clinical and clinical candidates. However, as we expand our understanding of the malarial life cycle and mechanisms of resistance development, SBDD as a field must continue to evolve in order to develop compounds that adhere to the ideal characteristics for novel antimalarial therapeutics and to avoid high attrition rates pre- and post-clinic. In the present review, we aim to examine the contribution that SBDD has made to current antimalarial drug development efforts, covering hit discovery to lead optimization and prevention of parasite resistance. Finally, the potential for structural biology, particularly high-throughput structural genomics programmes, to identify future targets for drug discovery are discussed.


Assuntos
Antimaláricos/farmacologia , Descoberta de Drogas , Modelos Biológicos , Plasmodium falciparum/efeitos dos fármacos , Animais , Antimaláricos/química , Resistência a Múltiplos Medicamentos , Humanos , Estágios do Ciclo de Vida/efeitos dos fármacos , Estrutura Molecular , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo
11.
Res Sq ; 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38746424

RESUMO

New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum ( Pf A-M1) and Plasmodium vivax ( Pv A-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets Pf A-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on Pf A-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of Pf A-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.

12.
mBio ; 15(6): e0096624, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38717141

RESUMO

To combat the global burden of malaria, development of new drugs to replace or complement current therapies is urgently required. Here, we show that the compound MMV1557817 is a selective, nanomolar inhibitor of both Plasmodium falciparum and Plasmodium vivax aminopeptidases M1 and M17, leading to inhibition of end-stage hemoglobin digestion in asexual parasites. MMV1557817 can kill sexual-stage P. falciparum, is active against murine malaria, and does not show any shift in activity against a panel of parasites resistant to other antimalarials. MMV1557817-resistant P. falciparum exhibited a slow growth rate that was quickly outcompeted by wild-type parasites and were sensitized to the current clinical drug, artemisinin. Overall, these results confirm MMV1557817 as a lead compound for further drug development and highlights the potential of dual inhibition of M1 and M17 as an effective multi-species drug-targeting strategy.IMPORTANCEEach year, malaria infects approximately 240 million people and causes over 600,000 deaths, mostly in children under 5 years of age. For the past decade, artemisinin-based combination therapies have been recommended by the World Health Organization as the standard malaria treatment worldwide. Their widespread use has led to the development of artemisinin resistance in the form of delayed parasite clearance, alongside the rise of partner drug resistance. There is an urgent need to develop and deploy new antimalarial agents with novel targets and mechanisms of action. Here, we report a new and potent antimalarial compound, known as MMV1557817, and show that it targets multiple stages of the malaria parasite lifecycle, is active in a preliminary mouse malaria model, and has a novel mechanism of action. Excitingly, resistance to MMV15578117 appears to be self-limiting, suggesting that development of the compound may provide a new class of antimalarial.


Assuntos
Aminopeptidases , Antimaláricos , Plasmodium falciparum , Plasmodium vivax , Antimaláricos/farmacologia , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/enzimologia , Animais , Camundongos , Plasmodium vivax/efeitos dos fármacos , Plasmodium vivax/enzimologia , Aminopeptidases/antagonistas & inibidores , Aminopeptidases/metabolismo , Resistência a Medicamentos , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Feminino
13.
Eur J Med Chem ; 248: 115051, 2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36634455

RESUMO

Malaria remains a global health threat and growing resistance to artemisinin-based therapies calls for therapeutic agents with novel mechanisms of action. The Plasmodium spp M1 and M17 metalloaminopeptidases have been identified as attractive new antimalarial drug targets as inhibition of these enzymes results in antiplasmodial activity. Previously identified novel hydroxamic acid 2 as a moderate inhibitor of PfA-M1 and PfA-M17 and a potent inhibitor of P. falciparum. This study has sought to improve the enzymatic inhibitory properties in addition to increasing the drug-likeness of this scaffold by introducing polar moieties into the S1' region of the active site. Structural biology studies on the co-crystallised structures of potent dual-inhibitor 9aa bound to PfA-M1 and PfA-M17 have revealed that there are few direct interactions between the inhibitor and the S1' domain of these enzymes. Structure-based compound design led to the identification of a variety of novel hydroxamic acids that show improved inhibitory activity against PfA-M1 and PfA-M17, in addition to displaying antiplasmodial activity. Notably, compounds with substitutions on the aniline ring resulted in a loss of potency (Ki > 500 nM) toward PfA-M1 and PfA-M17. ioisosteric replacement of the S1-region biaryl ring system with a bromophenyl moiety resulted in increased potency compared to parent 9aa. Elaboration of 9aa to bioisosterically replace the S1 moiety with an aryl bromide, combined with substituted anilines has resulted in potent selective PfA-M1 inhibitors which show strong activity against Pf-3D7, with meta- and para-fluoroaniline groups of 15ag and 15ah forming hydrogen-bonds with residues within the active site. These findings establish the importance of the previously under-utilised S1' domain and will aid the design of future PfA-M1 and PfA-M17 inhibitors.


Assuntos
Antimaláricos , Malária Falciparum , Plasmodium , Humanos , Plasmodium falciparum , Aminopeptidases , Antimaláricos/química , Malária Falciparum/tratamento farmacológico
14.
Biochem J ; 431(1): 51-61, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20642456

RESUMO

CNS (central nervous system) adrenaline (epinephrine) is implicated in a wide range of physiological and pathological conditions. PNMT (phenylethanolamine N-methyltransferase) catalyses the final step in the biosynthesis of adrenaline, the conversion of noradrenaline (norepinephrine) to adrenaline by methylation. To help elucidate the role of CNS adrenaline, and to develop potential drug leads, potent, selective and CNS-active inhibitors are required. The fragment screening approach has advantages over other lead discovery methods including high hit rates, more efficient hits and the ability to sample chemical diversity more easily. In the present study we applied fragment-based screening approaches to the enzyme PNMT. We used crystallography as the primary screen and identified 12 hits from a small commercial library of 384 drug-like fragments. The hits include nine chemicals with two fused rings and three single-ring chemical systems. Eight of the hits come from three chemical classes: benzimidazoles (a known class of PNMT inhibitor), purines and quinolines. Nine of the hits have measurable binding affinities (~5-700 µM) as determined by isothermal titration calorimetry and all nine have ligand efficiencies of 0.39 kcal/mol per heavy atom or better (1 kcal≈4.184 kJ). We synthesized five elaborated benzimidazole compounds and characterized their binding to PNMT, showing for the first time how this class of inhibitors interact with the noradrenaline-binding site. Finally, we performed a pilot study with PNMT for fragment-based screening by MS showing that this approach could be used as a fast and efficient first-pass screening method prior to characterization of binding mode and affinity of hits.


Assuntos
Inibidores Enzimáticos/química , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Feniletanolamina N-Metiltransferase/química , Benzimidazóis/química , Sítios de Ligação , Calorimetria , Cristalografia por Raios X , Cinética , Ligantes , Espectrometria de Massas , Modelos Moleculares , Feniletanolamina N-Metiltransferase/metabolismo
15.
ChemMedChem ; 16(1): 234-249, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-32945135

RESUMO

Aminopeptidase N (APN/CD13) is a zinc-dependent ubiquitous transmembrane ectoenzyme that is widely present in different types of cells. APN is one of the most extensively studied metalloaminopeptidases as an anti-cancer target due to its significant role in the regulation of metastasis and angiogenesis. Previously, we identified a potent and selective APN inhibitor, N-(2-(Hydroxyamino)-2-oxo-1-(3',4',5'-trifluoro-[1,1'-biphenyl]-4-yl)ethyl)-4-(methylsulfonamido)benzamide (3). Herein, we report the further modifications performed to explore SAR around the S1 subsite of APN and to improve the physicochemical properties. A series of hydroxamic acid analogues were synthesised, and the pharmacological activities were evaluated in vitro. N-(1-(3'-Fluoro-[1,1'-biphenyl]-4-yl)-2-(hydroxyamino)-2-oxoethyl)-4-(methylsulfonamido)benzamide (6 f) was found to display an extremely potent inhibitory activity in the sub-nanomolar range.


Assuntos
Antígenos CD13/antagonistas & inibidores , Ácidos Hidroxâmicos/química , Sítios de Ligação , Antígenos CD13/metabolismo , Desenho de Fármacos , Humanos , Ácidos Hidroxâmicos/metabolismo , Cinética , Relação Estrutura-Atividade
16.
Biochem J ; 422(3): 463-71, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19570037

RESUMO

Substrate specificity is critically important for enzyme catalysis. In the adrenaline-synthesizing enzyme PNMT (phenylethanolamine N-methyltransferase), minor changes in substituents can convert substrates into inhibitors. Here we report the crystal structures of six human PNMT complexes, including the first structure of the enzyme in complex with its physiological ligand R-noradrenaline. Determining this structure required rapid soak methods because of the tendency for noradrenaline to oxidize. Comparison of the PNMT-noradrenaline complex with the previously determined PNMT-p-octopamine complex demonstrates that these two substrates form almost equivalent interactions with the enzyme and show that p-octopamine is a valid model substrate for PNMT. The crystal structures illustrate the adaptability of the PNMT substrate binding site in accepting multi-fused ring systems, such as substituted norbornene, as well as noradrenochrome, the oxidation product of noradrenaline. These results explain why only a subset of ligands recognized by PNMT are methylated by the enzyme; bulky substituents dictate the binding orientation of the ligand and can thereby place the acceptor amine too far from the donor methyl group for methylation to occur. We also show how the critical Glu(185) catalytic residue can be replaced by aspartic acid with a loss of only 10-fold in catalytic efficiency. This is because protein backbone movements place the Asp(185) carboxylate almost coincident with the carboxylate of Glu(185). Conversely, replacement of Glu(185) by glutamine reduces catalytic efficiency almost 300-fold, not only because of the loss of charge, but also because the variant residue does not adopt the same conformation as Glu(185).


Assuntos
Epinefrina/biossíntese , Norepinefrina/metabolismo , Feniletanolamina N-Metiltransferase/química , Feniletanolamina N-Metiltransferase/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Humanos , Ligação Proteica , Especificidade por Substrato
17.
Biochimie ; 166: 38-51, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30654132

RESUMO

The family of M17 aminopeptidases (alias 'leucine aminopeptidases', M17-LAPs) utilize a highly conserved hexameric structure and a binuclear metal center to selectively remove N-terminal amino acids from short peptides. However, M17-LAPs are responsible for a wide variety of functions that are seemingly unrelated to proteolysis. Herein, we aimed to investigate the myriad of functions attributed to M17. Further, we attempted to differentiate between the different molecular mechanisms that allow the conserved hexameric structure of an M17-LAP to mediate such diverse functions. We have provided an overview of research that identifies precise physiological roles of M17-LAPs, and the distinct mechanisms by which the enzymes moderate those roles. The review shows that the conserved hexameric structure of the M17-LAPs has an extraordinary capability to moderate different molecular mechanisms. We have broadly categorized these mechanisms as 'aminopeptidase-based', which include the characteristic proteolysis reactions, and 'association-driven', which involves moderation of the molecule's macromolecular assembly and higher order complexation events. The different molecular mechanisms are capable of eliciting very different cellular outcomes, and must be regarded as distinct when the physiological roles of this large and important family are considered.


Assuntos
Bactérias/enzimologia , Eucariotos/enzimologia , Leucil Aminopeptidase/química , Leucil Aminopeptidase/fisiologia , Animais , Domínio Catalítico , Humanos , Metais/metabolismo , Modelos Moleculares , Especificidade por Substrato
18.
Nat Microbiol ; 4(12): 2237-2245, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31406331

RESUMO

Spore-forming bacteria encompass a diverse range of genera and species, including important human and animal pathogens, and food contaminants. Clostridioides difficile is one such bacterium and is a global health threat because it is the leading cause of antibiotic-associated diarrhoea in hospitals. A crucial mediator of C. difficile disease initiation, dissemination and re-infection is the formation of spores that are resistant to current therapeutics, which do not target sporulation. Here, we show that cephamycin antibiotics inhibit C. difficile sporulation by targeting spore-specific penicillin-binding proteins. Using a mouse disease model, we show that combined treatment with the current standard-of-care antibiotic, vancomycin, and a cephamycin prevents disease recurrence. Cephamycins were found to have broad applicability as an anti-sporulation strategy, as they inhibited sporulation in other spore-forming pathogens, including the food contaminant Bacillus cereus. This study could directly and immediately affect treatment of C. difficile infection and advance drug development to control other important spore-forming bacteria that are problematic in the food industry (B. cereus), are potential bioterrorism agents (Bacillus anthracis) and cause other animal and human infections.


Assuntos
Antibacterianos/farmacologia , Cefamicinas/farmacologia , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/prevenção & controle , Animais , Toxinas Bacterianas/genética , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Clostridioides difficile/genética , Clostridioides difficile/crescimento & desenvolvimento , Infecções por Clostridium/microbiologia , Modelos Animais de Doenças , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Ligação às Penicilinas/efeitos dos fármacos , Proteínas de Ligação às Penicilinas/genética , Esporos Bacterianos/efeitos dos fármacos , Vancomicina/farmacologia , Células Vero/efeitos dos fármacos
19.
ChemMedChem ; 14(5): 603-612, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30653832

RESUMO

Apical membrane antigen 1 (AMA1) is essential for the invasion of host cells by malaria parasites. Several small-molecule ligands have been shown to bind to a conserved hydrophobic cleft in Plasmodium falciparum AMA1. However, a lack of detailed structural information on the binding pose of these molecules has hindered their further optimisation as inhibitors. We have developed a spin-labelled peptide based on RON2, the native binding partner of AMA1, to probe the binding sites of compounds on PfAMA1. The crystal structure of this peptide bound to PfAMA1 shows that it binds at one end of the hydrophobic groove, leaving much of the binding site unoccupied and allowing fragment hits to bind without interference. In paramagnetic relaxation enhancement (PRE)-based NMR screening, the 1 H relaxation rates of compounds binding close to the probe were enhanced. Compounds experienced different degrees of PRE as a result of their different orientations relative to the spin label while bound to AMA1. Thus, PRE-derived distance constraints can be used to identify binding sites and guide further hit optimisation.


Assuntos
Espectroscopia de Ressonância Magnética/métodos , Proteínas de Membrana/antagonistas & inibidores , Sondas Moleculares/química , Peptídeos/química , Proteínas de Protozoários/antagonistas & inibidores , Sequência de Aminoácidos , Antígenos de Protozoários , Benzimidazóis/química , Sítios de Ligação , Membrana Celular/metabolismo , Cristalografia por Raios X , Furanos/química , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Modelos Moleculares , Sondas Moleculares/metabolismo , Estrutura Molecular , Peptídeos/metabolismo , Ligação Proteica , Pirazóis/química , Pirimidinas/química , Pirróis/química , Quinazolinonas/química , Relação Estrutura-Atividade , Sulfonamidas/química
20.
J Med Chem ; 62(2): 622-640, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30537832

RESUMO

There is an urgent clinical need for antimalarial compounds that target malaria caused by both Plasmodium falciparum and Plasmodium vivax. The M1 and M17 metalloexopeptidases play key roles in Plasmodium hemoglobin digestion and are validated drug targets. We used a multitarget strategy to rationally design inhibitors capable of potent inhibition of the M1 and M17 aminopeptidases from both P. falciparum ( Pf-M1 and Pf-M17) and P. vivax ( Pv-M1 and Pv-M17). The novel chemical series contains a hydroxamic acid zinc binding group to coordinate catalytic zinc ion/s, and a variety of hydrophobic groups to probe the S1' pockets of the four target enzymes. Structural characterization by cocrystallization showed that selected compounds utilize new and unexpected binding modes; most notably, compounds substituted with bulky hydrophobic substituents displace the Pf-M17 catalytic zinc ion. Excitingly, key compounds of the series potently inhibit all four molecular targets and show antimalarial activity comparable to current clinical candidates.


Assuntos
Aminopeptidases/antagonistas & inibidores , Antimaláricos/química , Ácidos Hidroxâmicos/química , Plasmodium/enzimologia , Inibidores de Proteases/química , Proteínas de Protozoários/antagonistas & inibidores , Aminopeptidases/metabolismo , Antimaláricos/metabolismo , Antimaláricos/farmacologia , Sítios de Ligação , Domínio Catalítico , Sobrevivência Celular/efeitos dos fármacos , Resistência a Medicamentos/efeitos dos fármacos , Células HEK293 , Humanos , Ácidos Hidroxâmicos/metabolismo , Ácidos Hidroxâmicos/farmacologia , Simulação de Acoplamento Molecular , Plasmodium/efeitos dos fármacos , Inibidores de Proteases/metabolismo , Inibidores de Proteases/farmacologia , Proteínas de Protozoários/metabolismo , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA