Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Neurobiol Dis ; 160: 105515, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34571136

RESUMO

Brain inclusions mainly composed of misfolded and aggregated TAR DNA binding protein 43 (TDP-43), are characteristic hallmarks of amyotrophic lateral sclerosis (ALS). Irrespective of the role played by the inclusions, their reduction represents an important therapeutic pathway that is worth exploring. Their removal can either lead to the recovery of TDP-43 function by removing the self-templating conformers that sequester the protein in the inclusions, and/or eliminate any potential intrinsic toxicity of the aggregates. The search for curative therapies has been hampered by the lack of ALS models for use in high-throughput screening. We adapted, optimised, and extensively characterised our previous ALS cellular model for such use. The model demonstrated efficient aggregation of endogenous TDP-43, and concomitant loss of its splicing regulation function. We provided a proof-of-principle for its eventual use in high-throughput screening using compounds of the tricyclic family and showed that recovery of TDP-43 function can be achieved by the enhanced removal of TDP-43 aggregates by these compounds. We observed that the degradation of the aggregates occurs independent of the autophagy pathway beyond autophagosome-lysosome fusion, but requires a functional proteasome pathway. The in vivo translational effect of the cellular model was tested with two of these compounds in a Drosophila model expressing a construct analogous to the cellular model, where thioridazine significantly improved the locomotive defect. Our findings have important implications as thioridazine cleared TDP-43 aggregates and recovered TDP-43 functionality. This study also highlights the importance of a two-stage, in vitro and in vivo model system to cross-check the search for small molecules that can clear TDP-43 aggregates in TDP-43 proteinopathies.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Proteínas de Ligação a DNA/metabolismo , Antagonistas de Dopamina/uso terapêutico , Proteínas de Drosophila/metabolismo , Agregação Patológica de Proteínas/tratamento farmacológico , Tioridazina/uso terapêutico , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Antagonistas de Dopamina/farmacologia , Drosophila , Humanos , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Tioridazina/farmacologia
2.
Bioorg Med Chem Lett ; 29(19): 126610, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31471167

RESUMO

Focussed studies on imidazopyridine inhibitors of Plasmodium falciparum cyclic GMP-dependent protein kinase (PfPKG) have significantly advanced the series towards desirable in vitro property space. LLE-based approaches towards combining improvements in cell potency, key physicochemical parameters and structural novelty are described, and a structure-based design hypothesis relating to substituent regiochemistry has directed efforts towards key examples with well-balanced potency, ADME and kinase selectivity profiles.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Imidazóis/química , Malária/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/química , Compostos Bicíclicos Heterocíclicos com Pontes/química , Humanos , Malária/enzimologia , Malária/parasitologia , Modelos Moleculares , Simulação de Acoplamento Molecular , Plasmodium falciparum/enzimologia , Conformação Proteica , Inibidores de Proteínas Quinases/química
3.
Bioorg Med Chem Lett ; 29(3): 509-514, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30553738

RESUMO

Development of a class of bicyclic inhibitors of the Plasmodium falciparum cyclic GMP-dependent protein kinase (PfPKG), starting from known compounds with activity against a related parasite PKG orthologue, is reported. Examination of key sub-structural elements led to new compounds with good levels of inhibitory activity against the recombinant kinase and in vitro activity against the parasite. Key examples were shown to possess encouraging in vitro ADME properties, and computational analysis provided valuable insight into the origins of the observed activity profiles.


Assuntos
Antimaláricos/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Imidazóis/farmacologia , Plasmodium falciparum/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Antimaláricos/síntese química , Antimaláricos/química , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Imidazóis/síntese química , Imidazóis/química , Ligantes , Estrutura Molecular , Testes de Sensibilidade Parasitária , Plasmodium falciparum/enzimologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Piridinas/síntese química , Piridinas/química , Relação Estrutura-Atividade
4.
Drug Discov Today ; 2020 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-32920060

RESUMO

Here, we describe a novel workflow combining informatic and experimental approaches to enable evidence-based prioritising of targets from large sets in parallel. High-throughput protein production and biophysical fragment screening is used to identify those targets that are tractable and ligandable. As proof of concept we have applied this to a set of antibacterial targets comprising 146 essential genes. Of these targets, 51 were selected and 38 delivered results that allowed us to rank them by ligandability. The data obtained against these derisked targets have enabled rapid progression into structurally enabled drug discovery projects, demonstrating the practical value of the fragment-based target screening workflow.

5.
Nat Commun ; 8(1): 430, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874661

RESUMO

To combat drug resistance, new chemical entities are urgently required for use in next generation anti-malarial combinations. We report here the results of a medicinal chemistry programme focused on an imidazopyridine series targeting the Plasmodium falciparum cyclic GMP-dependent protein kinase (PfPKG). The most potent compound (ML10) has an IC50 of 160 pM in a PfPKG kinase assay and inhibits P. falciparum blood stage proliferation in vitro with an EC50 of 2.1 nM. Oral dosing renders blood stage parasitaemia undetectable in vivo using a P. falciparum SCID mouse model. The series targets both merozoite egress and erythrocyte invasion, but crucially, also blocks transmission of mature P. falciparum gametocytes to Anopheles stephensi mosquitoes. A co-crystal structure of PvPKG bound to ML10, reveals intimate molecular contacts that explain the high levels of potency and selectivity we have measured. The properties of this series warrant consideration for further development to produce an antimalarial drug.Protein kinases are promising drug targets for treatment of malaria. Here, starting with a medicinal chemistry approach, Baker et al. generate an imidazopyridine that selectively targets Plasmodium falciparum PKG, inhibits blood stage parasite growth in vitro and in mice and blocks transmission to mosquitoes.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Imidazóis/uso terapêutico , Malária/enzimologia , Malária/transmissão , Piridinas/uso terapêutico , Animais , Linhagem Celular , Cristalografia por Raios X , Culicidae , Proteínas Quinases Dependentes de GMP Cíclico/química , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imidazóis/farmacologia , Estágios do Ciclo de Vida/efeitos dos fármacos , Malária/tratamento farmacológico , Camundongos Endogâmicos BALB C , Modelos Moleculares , Plasmodium chabaudi/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/farmacologia , Resultado do Tratamento
7.
J Biomol Screen ; 18(5): 610-20, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23427046

RESUMO

Ribonuclease H2 (RNase H2) is a nuclease that specifically hydrolyzes RNA residues in RNA-DNA hybrids. Mutations in the RNase H2 enzyme complex have been identified in the genetic disorder Aicardi-Goutières syndrome (AGS), which has similarities to the autoimmune disease systemic lupus eryrthrematosis (SLE). The RNase H2 enzyme has also been recently implicated as a key genome surveillance enzyme. Therefore, small-molecule modulators of RNase H2 activity may have utility in therapeutics and as tools to investigate the cellular functions of RNase H2. A fluorescent quench assay, measuring cleavage of an RNA-DNA duplex substrate by recombinant RNase H2, was developed into a high-throughput format and used to screen a 48 560 compound library. A hit validation strategy was subsequently employed, leading to the identification of two novel inhibitor compounds with in vitro nanomolar range inhibition of RNase H2 activity and >100-fold selectivity compared with RNase H type 1. These compounds are the first small-molecule inhibitors of RNase H2 to be reported. They and their derivatives should provide the basis for the development of tool compounds investigating the cellular functions of the RNase H2 enzyme, and, potentially, for pharmacological manipulation of nucleic acid-mediated immune responses.


Assuntos
Inibidores Enzimáticos/isolamento & purificação , Ensaios de Triagem em Larga Escala/métodos , Ribonuclease H/antagonistas & inibidores , Sobrevivência Celular/efeitos dos fármacos , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Transferência Ressonante de Energia de Fluorescência/métodos , Células HeLa , Humanos , Microscopia de Fluorescência , Modelos Biológicos , Bibliotecas de Moléculas Pequenas/análise , Estudos de Validação como Assunto
8.
J Biomol Screen ; 18(5): 599-609, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23396314

RESUMO

A variety of G-protein-coupled receptor (GPCR) screening technologies have successfully partnered a number of GPCRs with their cognate ligands. GPCR-mediated ß-arrestin recruitment is now recognized as a distinct intracellular signaling pathway, and ligand-receptor interactions may show a bias toward ß-arrestin over classical GPCR signaling pathways. We hypothesized that the failure to identify native ligands for the remaining orphan GPCRs may be a consequence of biased ß-arrestin signaling. To investigate this, we assembled 10 500 candidate ligands and screened 82 GPCRs using PathHunter ß-arrestin recruitment technology. High-quality screening assays were validated by the inclusion of liganded receptors and the detection and confirmation of these established ligand-receptor pairings. We describe a candidate endogenous orphan GPCR ligand and a number of novel surrogate ligands. However, for the majority of orphan receptors studied, measurement of ß-arrestin recruitment did not lead to the identification of cognate ligands from our screening sets. ß-Arrestin recruitment represents a robust GPCR screening technology, and ligand-biased signaling is emerging as a therapeutically exploitable feature of GPCR biology. The identification of cognate ligands for the orphan GPCRs and the extent to which receptors may exist to preferentially signal through ß-arrestin in response to their native ligand remain to be determined.


Assuntos
Arrestinas/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Receptores Acoplados a Proteínas G/agonistas , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Descoberta de Drogas/métodos , Células HEK293 , Humanos , Ligantes , Ligação Proteica/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Saccharomyces cerevisiae , Bibliotecas de Moléculas Pequenas/análise , beta-Arrestinas
9.
J Biomol Screen ; 16(7): 734-43, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21602486

RESUMO

Nicotinamide nucleotide transhydrogenase (NNT) mutant mice show glucose intolerance with impaired insulin secretion during glucose tolerance tests. Uncoupling of the ß cell mitochondrial metabolism due to such mutations makes NNT a novel target for therapeutics in the treatment of pathologies such as type 2 diabetes. The authors propose that increasing NNT activity would help reduce deleterious buildup of reactive oxygen species in the inner mitochondrial matrix. They have expressed human Nnt cDNA for the first time in Saccharomyces cerevisiae, and transhydrogenase activity in mitochondria isolated from these cells is six times greater than is seen in wild-type mitochondria. The same mitochondria have partially uncoupled respiration, and the cells have slower growth rates compared to cells that do not express NNT. The authors have used NNT's role as a redox-driven proton pump to develop a robust fluorimetric assay in permeabilized yeast. Screening in parallel a library of known pharmacologically active compounds (National Institute of Neurological Disorders and Stroke collection) against NNT ± cells, they demonstrate a robust and reproducible assay suitable for expansion into larger and more diverse compound sets. The identification of NNT activators may help in the elucidation of the role of NNT in mammalian cells and assessing its potential as a therapeutic target for insulin secretion disorders.


Assuntos
Ensaios de Triagem em Larga Escala , NADP Trans-Hidrogenases/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Ativação Enzimática/efeitos dos fármacos , Regulação Fúngica da Expressão Gênica , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , NADP Trans-Hidrogenases/genética , Reprodutibilidade dos Testes , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Bibliotecas de Moléculas Pequenas/farmacologia
10.
Tuberculosis (Edinb) ; 91(4): 277-86, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21482481

RESUMO

PknB is an essential serine/threonine kinase of Mycobacterium tuberculosis with possible roles in a number of signalling pathways involved in cell division and metabolism. We screened a library of >50,000 compounds for inhibitors of the in vitro phosphorylation of GarA (Rv1827) by PknB and identified a number of inhibitors. A program of synthetic medicinal chemistry was subsequently conducted around one class of inhibitors and was successful in generating ATP competitive inhibitors with potency in the nanomolar range. Compounds in this class showed cross-reactivity with the related M. tuberculosis kinase, PknF, but not with PknG in an in vitro autophosphorylation assay. These synthesised inhibitors were able to prevent the growth of M. tuberculosis in an Alamar blue assay and in an intracellular model of infection, but only in the micromolar range. We attempted to determine if cell wall permeability was an explanation for the discrepancy between the potent in vitro compared with relatively poor in vivo activity, but found no evidence that the activity of the inhibitors could be improved by weakening the cell wall. Despite a number of drug discovery efforts attempting to develop inhibitors against PknB, it is yet to be reported that any such inhibitors prevent mycobacterial growth at submicromolar concentrations.


Assuntos
Antibacterianos/farmacologia , Macrófagos/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Tuberculose/tratamento farmacológico , Antibacterianos/metabolismo , Divisão Celular , Células Cultivadas , Descoberta de Drogas , Humanos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tuberculose/metabolismo
11.
Cell ; 131(6): 1072-83, 2007 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-18083098

RESUMO

The most virulent form of malaria is caused by waves of replication of blood stages of the protozoan pathogen Plasmodium falciparum. The parasite divides within an intraerythrocytic parasitophorous vacuole until rupture of the vacuole and host-cell membranes releases merozoites that invade fresh erythrocytes to repeat the cycle. Despite the importance of merozoite egress for disease progression, none of the molecular factors involved are known. We report that, just prior to egress, an essential serine protease called PfSUB1 is discharged from previously unrecognized parasite organelles (termed exonemes) into the parasitophorous vacuole space. There, PfSUB1 mediates the proteolytic maturation of at least two essential members of another enzyme family called SERA. Pharmacological blockade of PfSUB1 inhibits egress and ablates the invasive capacity of released merozoites. Our findings reveal the presence in the malarial parasitophorous vacuole of a regulated, PfSUB1-mediated proteolytic processing event required for release of viable parasites from the host erythrocyte.


Assuntos
Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Malária/parasitologia , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/fisiologia , Subtilisinas/fisiologia , Animais , Antígenos de Protozoários/metabolismo , Antígenos de Protozoários/fisiologia , Estágios do Ciclo de Vida , Malária/sangue , Modelos Biológicos , Plasmodium falciparum/patogenicidade , Plasmodium falciparum/ultraestrutura , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/isolamento & purificação , Proteínas de Protozoários/metabolismo , Esporozoítos/fisiologia , Subtilisinas/antagonistas & inibidores , Subtilisinas/isolamento & purificação , Subtilisinas/metabolismo , Vacúolos/parasitologia
12.
J Biol Chem ; 278(48): 47670-7, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-13679371

RESUMO

Malarial merozoites invade erythrocytes; and as an essential step in this invasion process, the 42-kDa fragment of Plasmodium falciparum merozoite surface protein-1 (MSP142) is further cleaved to a 33-kDa N-terminal polypeptide (MSP133) and an 19-kDa C-terminal fragment (MSP119) in a secondary processing step. Suramin was shown to inhibit both merozoite invasion and MSP142 proteolytic cleavage. This polysulfonated naphthylurea bound directly to recombinant P. falciparum MSP142 (Kd = 0.2 microM) and to Plasmodium vivax MSP142 (Kd = 0.3 microM) as measured by fluorescence enhancement in the presence of the protein and by isothermal titration calorimetry. Suramin bound only slightly less tightly to the P. vivax MSP133 (Kd = 1.5 microM) secondary processing product (fluorescence measurements), but very weakly to MSP119 (Kd approximately 15 mM) (NMR measurements). Several residues in MSP119 were implicated in the interaction with suramin using NMR measurements. A series of symmetrical suramin analogues that differ in the number of aromatic rings and substitution patterns of the terminal naphthylamine groups was examined in invasion and processing assays. Two classes of analogue with either two or four bridging rings were found to be active in both assays, whereas two other classes without bridging rings were inactive. We propose that suramin and related compounds inhibit erythrocyte invasion by binding to MSP1 and by preventing its cleavage by the secondary processing protease. The results indicate that enzymatic events during invasion are suitable targets for drug development and validate the novel concept of an inhibitor binding to a macromolecular substrate to prevent its proteolysis by a protease.


Assuntos
Proteína 1 de Superfície de Merozoito/metabolismo , Plasmodium falciparum/metabolismo , Suramina/análogos & derivados , Suramina/química , 2-Naftilamina/química , Animais , Antiprotozoários/farmacologia , Western Blotting , Calorimetria , Relação Dose-Resposta a Droga , Endopeptidases/química , Cinética , Espectroscopia de Ressonância Magnética , Modelos Biológicos , Modelos Químicos , Modelos Moleculares , Plasmodium vivax , Ligação Proteica , Espectrometria de Fluorescência , Temperatura , Ureia/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA