Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
PLoS Negl Trop Dis ; 15(11): e0009898, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34731172

RESUMO

Given the worldwide burden of neglected tropical diseases, there is ongoing need to develop novel anthelmintic agents to strengthen the pipeline of drugs to combat these burdensome infections. Many diseases caused by parasitic flatworms are treated using the anthelmintic drug praziquantel (PZQ), employed for decades as the key clinical agent to treat schistosomiasis. PZQ activates a flatworm transient receptor potential (TRP) channel within the melastatin family (TRPMPZQ) to mediate sustained Ca2+ influx and worm paralysis. As a druggable target present in many parasitic flatworms, TRPMPZQ is a promising target for a target-based screening campaign with the goal of discovering novel regulators of this channel complex. Here, we have optimized methods to miniaturize a Ca2+-based reporter assay for Schistosoma mansoni TRPMPZQ (Sm.TRPMPZQ) activity enabling a high throughput screening (HTS) approach. This methodology will enable further HTS efforts against Sm.TRPMPZQ as well as other flatworm ion channels. A pilot screen of ~16,000 compounds yielded a novel activator of Sm.TRPMPZQ, and numerous potential blockers. The new activator of Sm.TRPMPZQ represented a distinct chemotype to PZQ, but is a known chemical entity previously identified by phenotypic screening. The fact that a compound prioritized from a phenotypic screening campaign is revealed to act, like PZQ, as an Sm.TRPMPZQ agonist underscores the validity of TRPMPZQ as a druggable target for antischistosomal ligands.


Assuntos
Anti-Helmínticos/farmacologia , Proteínas de Helminto/antagonistas & inibidores , Praziquantel/farmacologia , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/parasitologia , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Animais , Anti-Helmínticos/química , Cálcio/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Humanos , Masculino , Camundongos , Praziquantel/química , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/genética , Esquistossomose mansoni/tratamento farmacológico , Esquistossomose mansoni/metabolismo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
2.
Nutrients ; 13(2)2021 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-33672967

RESUMO

The process of obtaining ascorbic acid (AA) via intestinal absorption and blood circulation is carrier-mediated utilizing the AA transporters SVCT1 and SVCT2, which are expressed in the intestine and brain (SVCT2 in abundance). AA concentration is decreased in Alzheimer's disease (AD), but information regarding the status of intestinal AA uptake in the AD is still lacking. We aimed here to understand how AA homeostasis is modulated in a transgenic mouse model (5xFAD) of AD. AA levels in serum from 5xFAD mice were markedly lower than controls. Expression of oxidative stress response genes (glutathione peroxidase 1 (GPX1) and superoxide dismutase 1 (SOD1)) were significantly increased in AD mice jejunum, and this increase was mitigated by AA supplementation. Uptake of AA in the jejunum was upregulated. This increased AA transport was caused by a marked increase in SVCT1 and SVCT2 protein, mRNA, and heterogeneous nuclear RNA (hnRNA) expression. A significant increase in the expression of HNF1α and specific protein 1 (Sp1), which drive SLC23A1 and SLC23A2 promoter activity, respectively, was observed. Expression of hSVCT interacting proteins GRHPR and CLSTN3 were also increased. SVCT2 protein and mRNA expression in the hippocampus of 5xFAD mice was not altered. Together, these investigations reveal adaptive up-regulation of intestinal AA uptake in the 5xFAD mouse model.


Assuntos
Doença de Alzheimer/metabolismo , Ácido Ascórbico/metabolismo , Jejuno/metabolismo , Transportadores de Sódio Acoplados à Vitamina C/metabolismo , Regulação para Cima/genética , Oxirredutases do Álcool/metabolismo , Animais , Transporte Biológico/genética , Proteínas de Ligação ao Cálcio/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Glutationa Peroxidase/metabolismo , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Hipocampo/metabolismo , Homeostase/genética , Absorção Intestinal/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/genética , RNA Mensageiro/metabolismo , Superóxido Dismutase-1/metabolismo , Glutationa Peroxidase GPX1
3.
Cell Calcium ; 75: 42-52, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30145428

RESUMO

The Ca2+ mobilizing second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) regulates intracellular trafficking events, including translocation of certain enveloped viruses through the endolysosomal system. Targeting NAADP-evoked Ca2+ signaling may therefore be an effective strategy for discovering novel antivirals as well as therapeutics for other disorders. To aid discovery of novel scaffolds that modulate NAADP-evoked Ca2+ signaling in human cells, we have investigated the potential of using the sea urchin egg homogenate system for a screening campaign. Known pharmacological inhibitors of NAADP-evoked Ca2+ release (but not cADPR- or IP3-evoked Ca2+ release) in this invertebrate system strongly correlated with inhibition of MERS-pseudovirus infectivity in a human cell line. A primary screen of 1534 compounds yielded eighteen 'hits' exhibiting >80% inhibition of NAADP-evoked Ca2+ release. A validation pipeline for these candidates yielded seven drugs that inhibited NAADP-evoked Ca2+ release without depleting acidic Ca2+ stores in a human cell line. These candidates displayed a similar penetrance of inhibition in both the sea urchin system and the human cell line, and the extent of inhibition of NAADP-evoked Ca2+ signals correlated well with observed inhibition of infectivity of a Middle East Respiratory syndrome coronavirus (MERS-CoV) pseudovirus. These experiments support the potential of this simple, homogenate system for screening campaigns to discover modulators of NAADP, cADPR and IP3-dependent Ca2+ signaling with potential therapeutic value.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , NADP/análogos & derivados , Óvulo/metabolismo , Ouriços-do-Mar/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , NADP/farmacologia , Óvulo/efeitos dos fármacos , Reprodutibilidade dos Testes , Ouriços-do-Mar/efeitos dos fármacos
4.
Int J Parasitol Drugs Drug Resist ; 6(3): 364-370, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27397763

RESUMO

5-hydroxytryptamine (5-HT) is a key regulator of muscle contraction in parasitic flatworms. In Schistosoma mansoni, the myoexcitatory action of 5-HT is effected through activation of a serotonergic GPCR (Sm.5HTRL), prioritizing pharmacological characterization of this target for anthelmintic drug discovery. Here, we have examined the effects of several aporphine alkaloids on the signaling activity of a heterologously expressed Sm.5HTRL construct using a cAMP biosensor assay. Four structurally related natural products - nuciferine, D-glaucine, boldine and bulbocapnine - were demonstrated to block Sm.5HTRL evoked cAMP generation with the potency of GPCR blockade correlating well with the ability of each drug to inhibit contractility of schistosomule larvae. Nuciferine was also effective at inhibiting both basal and 5-HT evoked motility of adult schistosomes. These data advance our understanding of structure-affinity relationships at Sm.5HTRL, and demonstrate the effectiveness of Sm.5HTRL antagonists as hypomotility-evoking drugs across different parasite life cycle stages.


Assuntos
Anti-Helmínticos/farmacologia , Aporfinas/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/enzimologia , Neurônios Serotoninérgicos/efeitos dos fármacos , Antagonistas da Serotonina/farmacologia , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Locomoção/efeitos dos fármacos
5.
Int J Parasitol Drugs Drug Resist ; 6(3): 356-363, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27397764

RESUMO

The diversity and uniqueness of flatworm G protein coupled receptors (GPCRs) provides impetus for identifying ligands useful as tools for studying flatworm biology, or as therapeutics for treating diseases caused by parasitic flatworm infections. To catalyse this discovery process, technologies optimized for mammalian GPCR high throughput screening need be transposed for screening flatworm GPCRs. Here, we demonstrate the utility of a genetically encoded cAMP biosensor for resolving the properties of an abundantly expressed planarian serotonergic GPCR (S7.1R). Application of this methodology resolved the real time kinetics of GPCR modulation by ligands and demonstrated a marked difference in the kinetic action of antagonists at S7.1R. Notably, bromocriptine caused a protracted inhibition of S7.1R activity in vitro and a protracted paralysis of planarian movement, replicating the effect of S7.1R in vivo RNAi. The lengthy inhibition of function caused by bromocriptine at this abundantly expressed GPCR provides a useful tool to ablate serotonergic signaling in vivo, and is a noteworthy feature for exploitation as an anthelmintic vulnerability.


Assuntos
Anti-Helmínticos/farmacologia , Bromocriptina/farmacologia , Planárias/efeitos dos fármacos , Planárias/enzimologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Neurônios Serotoninérgicos/efeitos dos fármacos , Antagonistas da Serotonina/farmacologia , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Cinética , Locomoção/efeitos dos fármacos
6.
PLoS Pathog ; 12(5): e1005651, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27187180

RESUMO

Schistosomiasis is a tropical parasitic disease afflicting ~200 million people worldwide and current therapy depends on a single drug (praziquantel) which exhibits several non-optimal features. These shortcomings underpin the need for next generation anthelmintics, but the process of validating physiologically relevant targets ('target selection') and pharmacologically profiling them is challenging. Remarkably, even though over a quarter of current human therapeutics target rhodopsin-like G protein coupled receptors (GPCRs), no library screen of a flatworm GPCR has yet been reported. Here, we have pharmacologically profiled a schistosome serotonergic GPCR (Sm.5HTR) implicated as a downstream modulator of PZQ efficacy, in a miniaturized screening assay compatible with high content screening. This approach employs a split luciferase based biosensor sensitive to cellular cAMP levels that resolves the proximal kinetics of GPCR modulation in intact cells. Data evidence a divergent pharmacological signature between the parasitic serotonergic receptor and the closest human GPCR homolog (Hs.5HTR7), supporting the feasibility of optimizing parasitic selective pharmacophores. New ligands, and chemical series, with potency and selectivity for Sm.5HTR over Hs.5HTR7 are identified in vitro and validated for in vivo efficacy against schistosomules and adult worms. Sm.5HTR also displayed a property resembling irreversible inactivation, a phenomenon discovered at Hs.5HTR7, which enhances the appeal of this abundantly expressed parasite GPCR as a target for anthelmintic ligand design. Overall, these data underscore the feasibility of profiling flatworm GPCRs in a high throughput screening format competent to resolve different classes of GPCR modulators. Further, these data underscore the promise of Sm.5HTR as a chemotherapeutically vulnerable node for development of next generation anthelmintics.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Receptores Acoplados a Proteínas G/análise , Schistosoma mansoni/metabolismo , Esquistossomose mansoni , Animais , Western Blotting , Avaliação Pré-Clínica de Medicamentos , Feminino , Células HEK293 , Humanos , Camundongos
7.
Am J Physiol Cell Physiol ; 291(5): C851-9, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16790503

RESUMO

The water-soluble micronutrient thiamine is required for normal tissue growth and development in humans. Thiamine is accumulated into cells through the activity of two cell surface thiamine transporters (hTHTR1 and hTHTR2), which are differentially targeted in polarized tissues. Mutational dysfunction of hTHTR1 is associated with the clinical condition of thiamine-responsive megaloblastic anemia: the symptoms of which are alleviated by thiamine supplementation. Recently, two hTHTR2 mutants (G23V, T422A) have been discovered in clinical kindreds manifesting biotin-responsive basal ganglia disease (BBGD): the symptoms of which are alleviated by biotin administration. Why then does mutation of a specific thiamine transporter isoform precipitate a disorder correctable by exogenous biotin? To investigate the suggestion that hTHTR2 can physiologically function as a biotin transporter, we examined 1) the cell biological basis of hTHTR2 dysfunction associated with the G23V and T422A mutations and 2) the substrate specificity of hTHTR2 and these clinically relevant mutants. We show that the G23V and T422A mutants both abrogate thiamine transport activity rather than targeting of hTHTR2 to the cell surface. Furthermore, biotin accumulation was not detectable in cells overexpressing either the full length hTHTR2 or the clinically relevant hTHTR2 mutants, yet was demonstrable in the same assay using cells overexpressing the human sodium-dependent multivitamin transporter, a known biotin transporter. These results cast doubt on the most parsimonious explanation for the BBGD phenotype, namely that hTHTR2 is a physiological biotin transporter.


Assuntos
Doenças dos Gânglios da Base/genética , Doenças dos Gânglios da Base/metabolismo , Biotina/metabolismo , Biotina/farmacologia , Proteínas de Membrana Transportadoras/metabolismo , Mutação Puntual/genética , Tiamina/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico/efeitos dos fármacos , Células CACO-2 , Sequência Conservada , Cães , Regulação da Expressão Gênica , Ácido Glutâmico/química , Glicosilação , Humanos , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Especificidade por Substrato , Trítio
8.
J Biol Chem ; 281(8): 5233-45, 2006 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-16371350

RESUMO

Humans lack biochemical pathways for thiamine synthesis, so cellular requirements are met via specific carrier-mediated uptake pathways. Two proteins from the solute carrier SLC19A gene family have been identified as human thiamine transporters (hTHTRs), SLC19A1 (hTHTR1) and SLC19A2 (hTHTR2). Both of these transporters are co-expressed but are differentially targeted in polarized cell types that mediate vectorial thiamine transport (e.g. renal and intestinal epithelia). It is important to understand the domain structure of these proteins, namely which regions within the polypeptide sequence are important for physiological delivery to the cell surface, in order to understand the impact of clinically relevant mutations on thiamine transport. Here we have characterized the mechanisms regulating hTHTR2 distribution by using live cell imaging methods that resolve the targeting and trafficking dynamics of full-length hTHTR2, a series of hTHTR2 truncation mutants, as well as chimeras comprising the hTHTR1 and hTHTR2 sequence. We showed the following: (i) that the cytoplasmic COOH-tail of hTHTR2 is not essential for apical targeting in polarized cells; (ii) that delivery of hTHTR2 to the cell surface is critically dependent on the integrity of the transmembrane backbone of the polypeptide so that minimal truncations abrogate cell surface expression of hTHTR2; and (iii) video rate images of hTHTR2-containing intracellular vesicles displayed rapid bi-directional trafficking events to and from the cell surface impaired by microtubule-disrupting but not microfilament-disrupting agents as well as by overexpression of the dynactin subunit dynamitin (p50). Finally, we compared the behavior of hTHTR2 with that of hTHTR1 and the human reduced folate carrier (SLC19A1) to underscore commonalities in the cell surface targeting mechanisms of the entire SLC19A gene family.


Assuntos
Células Epiteliais/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Citoplasma/metabolismo , Análise Mutacional de DNA , Primers do DNA/química , DNA Complementar/metabolismo , Cães , Complexo Dinactina , Citometria de Fluxo , Proteínas de Fluorescência Verde/química , Humanos , Rim/metabolismo , Proteínas Luminescentes/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Microscopia de Vídeo , Proteínas Associadas aos Microtúbulos/química , Microtúbulos/química , Microtúbulos/metabolismo , Família Multigênica , Mutação , Peptídeos/química , Reação em Cadeia da Polimerase , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Tiamina/química , Transfecção
9.
J Biol Chem ; 277(36): 33325-33, 2002 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-12087110

RESUMO

The major pathway for cellular uptake of the water-soluble vitamin folic acid in mammalian cells is via a plasma membrane protein known as the reduced folate carrier (RFC). The molecular determinants that dictate plasma membrane expression of RFC as well as the cellular mechanisms that deliver RFC to the cell surface remain poorly defined. Therefore, we designed a series of fusion proteins of the human RFC (hRFC) with green fluorescent protein to image the targeting and trafficking dynamics of hRFC in living epithelial cells. We show that, in contrast to many other nutrient transporters, the molecular determinants that dictate hRFC plasma membrane expression reside within the hydrophobic backbone of the polypeptide and not within the cytoplasmic NH(2)- or COOH-terminal domains of the protein. Further, the integrity of the hRFC backbone is critical for export of the polypeptide from the endoplasmic reticulum to the cell surface. This trafficking is critically dependent on intact microtubules because microtubule disruption inhibits motility of hRFC-containing vesicles as well as final expression of hRFC in the plasma membrane. For the first time, these data define the mechanisms that control the intracellular trafficking and cell surface localization of hRFC within mammalian epithelia.


Assuntos
Proteínas de Transporte/metabolismo , Células Epiteliais/metabolismo , Proteínas de Membrana Transportadoras , Animais , Transporte Biológico , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Citoesqueleto/metabolismo , DNA Complementar/metabolismo , Cães , Citometria de Fluxo , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Microscopia Confocal , Microscopia de Vídeo , Microtúbulos/metabolismo , Fases de Leitura Aberta , Peptídeos/química , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Proteína Carregadora de Folato Reduzido , Temperatura , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA