Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 588(7839): 712-716, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33328633

RESUMO

Altered expression of mitochondrial DNA (mtDNA) occurs in ageing and a range of human pathologies (for example, inborn errors of metabolism, neurodegeneration and cancer). Here we describe first-in-class specific inhibitors of mitochondrial transcription (IMTs) that target the human mitochondrial RNA polymerase (POLRMT), which is essential for biogenesis of the oxidative phosphorylation (OXPHOS) system1-6. The IMTs efficiently impair mtDNA transcription in a reconstituted recombinant system and cause a dose-dependent inhibition of mtDNA expression and OXPHOS in cell lines. To verify the cellular target, we performed exome sequencing of mutagenized cells and identified a cluster of amino acid substitutions in POLRMT that cause resistance to IMTs. We obtained a cryo-electron microscopy (cryo-EM) structure of POLRMT bound to an IMT, which further defined the allosteric binding site near the active centre cleft of POLRMT. The growth of cancer cells and the persistence of therapy-resistant cancer stem cells has previously been reported to depend on OXPHOS7-17, and we therefore investigated whether IMTs have anti-tumour effects. Four weeks of oral treatment with an IMT is well-tolerated in mice and does not cause OXPHOS dysfunction or toxicity in normal tissues, despite inducing a strong anti-tumour response in xenografts of human cancer cells. In summary, IMTs provide a potent and specific chemical biology tool to study the role of mtDNA expression in physiology and disease.


Assuntos
Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Microscopia Crioeletrônica , DNA Mitocondrial/efeitos dos fármacos , DNA Mitocondrial/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Estabilidade Enzimática/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Mitocondriais/efeitos dos fármacos , Humanos , Masculino , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Especificidade por Substrato/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Biol Chem ; 292(20): 8158-8173, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28325835

RESUMO

The concentration of free cytosolic Ca2+ and the voltage across the plasma membrane are major determinants of cell function. Ca2+-permeable non-selective cationic channels are known to regulate these parameters, but understanding of these channels remains inadequate. Here we focus on transient receptor potential canonical 4 and 5 proteins (TRPC4 and TRPC5), which assemble as homomers or heteromerize with TRPC1 to form Ca2+-permeable non-selective cationic channels in many mammalian cell types. Multiple roles have been suggested, including in epilepsy, innate fear, pain, and cardiac remodeling, but limitations in tools to probe these channels have restricted progress. A key question is whether we can overcome these limitations and develop tools that are high-quality, reliable, easy to use, and readily accessible for all investigators. Here, through chemical synthesis and studies of native and overexpressed channels by Ca2+ and patch-clamp assays, we describe compound 31, a remarkable small-molecule inhibitor of TRPC1/4/5 channels. Its potency ranged from 9 to 1300 pm, depending on the TRPC1/4/5 subtype and activation mechanism. Other channel types investigated were unaffected, including TRPC3, TRPC6, TRPV1, TRPV4, TRPA1, TRPM2, TRPM8, and store-operated Ca2+ entry mediated by Orai1. These findings suggest identification of an important experimental tool compound, which has much higher potency for inhibiting TRPC1/4/5 channels than previously reported agents, impressive specificity, and graded subtype selectivity within the TRPC1/4/5 channel family. The compound should greatly facilitate future studies of these ion channels. We suggest naming this TRPC1/4/5-inhibitory compound Pico145.


Assuntos
Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cátion TRPC/antagonistas & inibidores , Cálcio/metabolismo , Células HEK293 , Humanos , Proteína ORAI1/antagonistas & inibidores , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo
3.
Biochim Biophys Acta Biomembr ; 1860(5): 1008-1014, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29357287

RESUMO

Ras proteins are oncoproteins which play a pivotal role in cellular signaling pathways. All Ras proteins' signaling strongly depends on their correct localization in the cell membrane. Over 30% of cancers are driven by mutant Ras proteins, and KRas4B is the Ras isoform most frequently mutated. C6-ceramide has been shown to inhibit the growth activity of KRas4B mutated cells. However, the mechanism underlying this inhibition remains elusive. Here, we established a heterogeneous model biomembrane containing C6-ceramide. C6-ceramide incorporation does not disrupt the lipid membrane. Addition of KRas4B leads to drastic changes in the lateral membrane organization of the membrane, however. In contrast to the partitioning behavior in other membranes, KRas4B forms small, monodisperse nanoclusters dispersed in a fluid-like environment, in all likelihood induced by some kind of lipid sorting mechanism. Fluorescence cross-correlation data indicate no direct interaction between C6-ceramide and KRas4B, suggesting that KRas4B essentially recruits other lipids. A FRET-based binding assay reveals that the stability of KRas4B proteins inserted into the membrane containing C6-ceramide is reduced. Based on the combined results obtained, we postulate a molecular mechanism for the inhibition of KRas4B mutated cells' activity through C6-ceramide.


Assuntos
Ceramidas/metabolismo , Bicamadas Lipídicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Ceramidas/química , Transferência Ressonante de Energia de Fluorescência , Humanos , Bicamadas Lipídicas/química , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo , Microscopia de Força Atômica , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas p21(ras)/química , Lipossomas Unilamelares/química , Lipossomas Unilamelares/metabolismo
4.
Biochemistry ; 56(30): 3972-3982, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28681606

RESUMO

Proteins typically interact with multiple binding partners, and often different parts of their surfaces are employed to establish these protein-protein interactions (PPIs). Members of the class of 14-3-3 adapter proteins bind to several hundred other proteins in the cell. Multiple small molecules for the modulation of 14-3-3 PPIs have been disclosed; however, they all target the conserved phosphopeptide binding channel, so that selectivity is difficult to achieve. Here we report on the discovery of two individual secondary binding sites that have been identified by combining nuclear magnetic resonance-based fragment screening and X-ray crystallography. The two pockets that these fragments occupy are part of at least three physiologically relevant and structurally characterized 14-3-3 PPI interfaces, including those with serotonin N-acetyltransferase and plant transcription factor FT. In addition, the high degree of conservation of the two sites implies their relevance for 14-3-3 PPIs. This first identification of secondary sites on 14-3-3 proteins bound by small molecule ligands might facilitate the development of new chemical tool compounds for more selective PPI modulation.


Assuntos
Proteínas 14-3-3/metabolismo , Biomarcadores Tumorais/metabolismo , Exorribonucleases/metabolismo , Modelos Moleculares , Fatores de Transcrição/metabolismo , Proteínas 14-3-3/química , Proteínas 14-3-3/genética , Aciltransferases , Sequência de Aminoácidos , Sítios de Ligação , Biomarcadores Tumorais/química , Biomarcadores Tumorais/genética , Sequência Conservada , Cristalografia por Raios X , Exorribonucleases/química , Exorribonucleases/genética , Deleção de Genes , Humanos , Cinética , Ligantes , Ressonância Magnética Nuclear Biomolecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Biblioteca de Peptídeos , Fosforilação , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética
5.
Chemistry ; 23(25): 6083-6093, 2017 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-27809361

RESUMO

The K-Ras GTPase is a major target in anticancer drug discovery. However, direct interference with signaling by K-Ras has not led to clinically useful drugs yet. Correct localization and signaling by farnesylated K-Ras is regulated by the prenyl binding protein PDEδ. Interfering with binding of PDEδ to K-Ras by means of small molecules provides a novel opportunity to suppress oncogenic signaling. Here we describe the identification and structure-guided development of novel K-Ras-PDEδ inhibitor chemotypes based on pyrrolopyridazinones and pyrazolopyridazinones that bind to the farnesyl binding pocket of PDEδ with low nanomolar affinity. We delineate the structure-property relationship and in vivo pharmacokinetic (PK) and toxicokinetic (Tox) studies for pyrazolopyridazinone-based K-Ras-PDEδ inhibitors. These findings may inspire novel drug discovery efforts aimed at the development of drugs targeting oncogenic Ras.

6.
Biochim Biophys Acta ; 1844(1 Pt A): 156-61, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23707564

RESUMO

Multiple factors have to be optimized in the course of a drug discovery project. Traditionally this includes potency on a single target, eventually specificity as well as the pharmacokinetic, physicochemical and the safety profile. Recently an additional dimension has been added by realizing that the therapeutic outcome of a drug is often determined not only by its activity on a single target but also by its activity profile across a variety of biological targets. To address the polypharmacology of drug candidates many compounds are tested on a set of targets or in phenotypic screens generating a tremendous amount of data. To extract useful information computational methods at the interface of proteomics and cheminformatics are indispensable. This review will focus on some recent developments in this field. This article is part of a Special Issue entitled: Computational Proteomics in the Post-Identification Era. Guest Editors: Martin Eisenacher and Christian Stephan.


Assuntos
Química Farmacêutica , Proteômica , Descoberta de Drogas
7.
Bioorg Med Chem ; 22(18): 5097-109, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24398384

RESUMO

In search of new compounds with strong antiproliferative activity and simple molecular structure, we designed a novel series of agents based on the 2-amino-3-alkoxycarbonyl/cyano-5-arylethylthiophene scaffold. The presence of the ethyl spacer between the 2',5'-dimethoxyphenyl and the 5-position of the thiophene ring, as well as the number and location of methoxy substitutents on the phenyl ring, played a profound role in affecting the antiproliferative activity. Among the synthesized compounds, we identified the 2-amino-3-cyano-[2-(2,5-dimethoxyphenyl)ethyl] thiophene 2c as the most promising derivative against a wide panel of cancer cell lines (IC50=17-130 nM). The antiproliferative activity of this compound appears to correlate well with its ability to inhibit tubulin assembly and the binding of colchicine to tubulin. Moreover 2c, as determined by flow cytometry, strongly induced arrest in the G2/M phase of the cell cycle, and annexin-V and propidium iodide staining indicate that cell death proceeds through an apoptotic mechanism that follows the intrinsic mitochondrial pathway.


Assuntos
Antineoplásicos/farmacologia , Desenho de Fármacos , Tiofenos/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células HeLa , Humanos , Camundongos , Modelos Moleculares , Estrutura Molecular , Polimerização/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade , Tiofenos/síntese química , Tiofenos/química , Tubulina (Proteína)/metabolismo
8.
J Med Chem ; 65(9): 6643-6655, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35486541

RESUMO

Despite the clinical efficacy of epidermal growth factor receptor (EGFR) inhibitors, a subset of patients with non-small cell lung cancer displays insertion mutations in exon20 in EGFR and Her2 with limited treatment options. Here, we present the development and characterization of the novel covalent inhibitors LDC8201 and LDC0496 based on a 1H-pyrrolo[2,3-b]pyridine scaffold. They exhibited intense inhibitory potency toward EGFR and Her2 exon20 insertion mutations as well as selectivity over wild type EGFR and within the kinome. Complex crystal structures with the inhibitors and biochemical and cellular on-target activity document their favorable binding characteristics. Ultimately, we observed tumor shrinkage in mice engrafted with patient-derived EGFR-H773_V774insNPH mutant cells during treatment with LDC8201. Together, these results highlight the potential of covalent pyrrolopyridines as inhibitors to target exon20 insertion mutations.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Mutagênese Insercional , Mutação , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
10.
Bioorg Med Chem Lett ; 20(5): 1485-7, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20153186

RESUMO

Various carboxylic acids, phosphonic acids, sulfonic acids, tetrazoles as well as sulfonylhydantoins were prepared as phosphate mimics of the chiral aminophosphate 1-P to act as agonists on the S1P(1) receptor. It was found that amino phosphonates and amino carboxylates are potent S1P(1) binders. beta-Amino acid 11 could be shown to reversibly reduce blood lymphocyte counts in rats after po administration.


Assuntos
Imunossupressores/química , Isoleucina/análogos & derivados , Fosfatos/química , Propilenoglicóis/química , Receptores de Lisoesfingolipídeo/agonistas , Esfingosina/análogos & derivados , Administração Oral , Aminoácidos/química , Animais , Cloridrato de Fingolimode , Imunossupressores/síntese química , Imunossupressores/farmacologia , Isoleucina/síntese química , Isoleucina/química , Isoleucina/farmacologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Fosforilação , Propilenoglicóis/farmacologia , Ratos , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/química , Esfingosina/farmacologia
11.
Chem Biodivers ; 6(5): 725-38, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19479838

RESUMO

The synthesis of 7-oxasphingosine (3) and 7-oxaceramide (4) was improved by starting from the 4-methoxybenzyl-protected d-galactal 9. The sphingosine analogues 5-7 and 24 were synthesized via the azido alcohol 13. The 7-thiasphingosine 5 is a poorer substrate for both isoforms of sphingosine kinase (SPHK) than sphingosine, but showed a slight preference for SPHK2. The sulfone 6 and the 7-aza compounds 7 and 24 were not phosphorylated by either SPHK1 or SPHK2, and none of 5-7 and 24 activated invariant natural killer T (iNKT) cell clones when presented by human CD1d-transfected antigen-presenting cells (APC) or by plate-bound human CD1d. Only 7 and 24 associated with plate-bound recombinant CD1d prevented stimulation of iNKT cells by alpha-galactosylceramide (alpha-GalCer).


Assuntos
Ceramidas/química , Células T Matadoras Naturais/imunologia , Fosfotransferases (Aceptor do Grupo Álcool)/química , Esfingosina/química , Células Apresentadoras de Antígenos/imunologia , Antígenos CD1d/genética , Antígenos CD1d/imunologia , Ceramidas/síntese química , Ceramidas/farmacologia , Humanos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Esfingosina/síntese química , Esfingosina/farmacologia
12.
Chem Biodivers ; 6(5): 705-24, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19479849

RESUMO

The analogues 7-9 of 7-oxaceramide and 7-oxasphingosine were synthesized from the known azidosphingosine 21. The 1,4-disubstituted 1,2,3-triazole analogues 10-16 of ceramides were synthesized by the click reaction of the known azide 24. None of the analogues 7-15 was active as inhibitor of SPHK type 1 and of acid sphingomyelinase, whereas 16 is a weak inhibitor of SPHK1. Triazoles 10, 11, and 15 did not inhibit ceramide phosphorylation by CerK, and none of 7, 8, and 10-15 activated invariant natural killer T (iNKT) cell clones when presented by human CD1d-transfected antigen-presenting cells (APC) or by plate-bound human CD1d [55]. Triazoles 14 and 15 prevent binding of alpha-galactosylceramide (alpha-GalCer) to plate-bound human CD1d and subsequent T-cell response to alpha-GalCer. Only 15 reduced activation by alpha-GalCer significantly and independently of the cytokine measured.


Assuntos
Ceramidas/química , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Esfingosina/análogos & derivados , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD1d/genética , Antígenos CD1d/imunologia , Ceramidas/síntese química , Humanos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Esfingosina/síntese química , Esfingosina/química , Esfingosina/farmacologia
13.
Chem Biodivers ; 6(10): 1688-715, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19842132

RESUMO

The conformationally based piperidinone sphingosine analogues 7, 8, 15, and 16 were synthesized from allylic alcohol 34 via lactams 31 and 32. The L-arabino diol 7 and the L-ribo diol 8 were transformed into the amino alcohols 17-24. The L-gluco ceramide analogues 43, 46a, and 47, and the L-altro ceramide analogues 51a and 52 were synthesized from either 31 or 32. The L-ribo diols 8 and 16, and the amino alcohols 19 and 20 inhibit sphingosine kinase 1 (SPHK1), while the L-arabino analogues 7, 15, 17, and 18 are inactive. The L-arabino and the L-ribo dimethylamines 21-24, the L-gluco ceramide analogues 43, 46a, and 47, and the L-altro ceramide analogues 51a and 52 did not block SPHK1. Neither the L-arabino diol 7 nor the L-ribo diol 8 inhibited SPHK2 or ceramide kinase. The L-arabino diols 7 and 15 stimulate invariant natural killer T (iNKT) cells when presented by living antigen-presenting cells (APC) and also by plate-bound human CD1d, whereas the L-ribo diols 8 and 16, the L-arabino amino alcohols 17-18, and the dimethylamines 21-22 did not activate iNKT cells. The L-gluco ceramide analogues 43, 46a, and 47 had strongly stimulatory effects on iNKT cells when presented by living APC and also by plate-bound human CD1d, whereas the L-altro ceramide analogue 52 activated only weakly. All activatory compounds induced preferentially the release of pro-inflammatory cytokines, indicating the formation of a stable CD1d--lipid--T-cell receptor complex.


Assuntos
Ceramidas/química , Inibidores Enzimáticos/síntese química , Células T Matadoras Naturais/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Piperidinas/química , Piperidinas/farmacologia , Antígenos CD1d/imunologia , Antígenos CD1d/metabolismo , Ceramidas/metabolismo , Ceramidas/farmacologia , Citocinas/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Metabolismo dos Lipídeos/imunologia , Metabolismo dos Lipídeos/fisiologia , Modelos Químicos , Conformação Molecular , Células T Matadoras Naturais/citologia , Células T Matadoras Naturais/imunologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Piperidinas/síntese química , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo
14.
ChemMedChem ; 14(11): 1074-1078, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-30945468

RESUMO

Despite the availability of hundreds of antibiotic drugs, infectious diseases continue to remain one of the most notorious health issues. In addition, the disparity between the spread of multidrug-resistant pathogens and the development of novel classes of antibiotics exemplify an important unmet medical need that can only be addressed by identifying novel targets. Herein we demonstrate, by the development of the first in vivo active DegS inhibitors based on a pyrazolo[1,5-a]-1,3,5-triazine scaffold, that the serine protease DegS and the cell envelope stress-response pathway σE represent a target for generating antibiotics with a novel mode of action. Moreover, DegS inhibition is synergistic with well-established membrane-perturbing antibiotics, thereby opening promising avenues for rational antibiotic drug design.


Assuntos
Antibacterianos/farmacologia , Proteínas de Escherichia coli/antagonistas & inibidores , Escherichia coli/efeitos dos fármacos , Inibidores de Serina Proteinase/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Relação Dose-Resposta a Droga , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Estrutura Molecular , Inibidores de Serina Proteinase/síntese química , Inibidores de Serina Proteinase/química , Relação Estrutura-Atividade
15.
Stem Cell Reports ; 12(3): 502-517, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30773488

RESUMO

Neuroinflammation is a hallmark of neurological disorders and is accompanied by the production of neurotoxic agents such as nitric oxide. We used stem cell-based phenotypic screening and identified small molecules that directly protected neurons from neuroinflammation-induced degeneration. We demonstrate that inhibition of CDK5 is involved in, but not sufficient for, neuroprotection. Instead, additional inhibition of GSK3ß is required to enhance the neuroprotective effects of CDK5 inhibition, which was confirmed using short hairpin RNA-mediated knockdown of CDK5 and GSK3ß. Quantitative phosphoproteomics and high-content imaging demonstrate that neurite degeneration is mediated by aberrant phosphorylation of multiple microtubule-associated proteins. Finally, we show that our hit compound protects neurons in vivo in zebrafish models of motor neuron degeneration and Alzheimer's disease. Thus, we demonstrate an overlap of CDK5 and GSK3ß in mediating the regulation of the neuronal cytoskeleton and that our hit compound LDC8 represents a promising starting point for neuroprotective drugs.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Citoesqueleto/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Inflamação/metabolismo , Degeneração Neural/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Animais , Citoesqueleto/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Degeneração Neural/tratamento farmacológico , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra/metabolismo
16.
Chem Phys Lipids ; 151(2): 125-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18039471

RESUMO

An efficient, one-pot procedure for the synthesis of ceramide 1-phosphates with varying N-acyl substituents, to serve as tool compounds for analytical and biological investigations, was developed. Sphingosine 1-phosphate was silylated in situ to increase its solubility and to protect the 3-hydroxy functionality and then allowed to react with activated acid derivatives in the presence of diisopropylethylamine. Simultaneous cleavage of the silyl protecting groups and separation from reagents and by-products was achieved by medium pressure chromatography on reversed phase material. Thus, ceramide 1-phosphates with various fatty acid chains and with fluorescent and affinity labels attached to the sphingoid backbone were prepared in good yields.


Assuntos
Ceramidas/síntese química , Lisofosfolipídeos/síntese química , Esfingosina/análogos & derivados , Ceramidas/química , Lisofosfolipídeos/química , Estrutura Molecular , Esfingosina/síntese química , Esfingosina/química
17.
Bioorg Med Chem ; 16(16): 7552-60, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18678497

RESUMO

The potent antiproliferative agent SDZ LAP 977, which has shown efficacy in a clinical proof of concept study in actinic keratosis patients, has been previously demonstrated to block the cell cycle in mitosis. In the present study, we further explored the mode of action: SDZ LAP 977 binds to the "colchicine binding site" on tubulin and, thus, inhibits tubulin polymerization in vitro. Moreover, we established structure-activity relationships for the effect of modifications in the 2,5-dimethoxyphenyl moiety ("ring A") of the molecule on in vitro antiproliferative activity.


Assuntos
Fenóis/química , Fenóis/farmacologia , Sítios de Ligação , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular , Colchicina/metabolismo , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Microscopia de Fluorescência , Microtúbulos/efeitos dos fármacos , Mitose/efeitos dos fármacos , Fenóis/síntese química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Relação Estrutura-Atividade , Tubulina (Proteína)/metabolismo
18.
Sci Immunol ; 3(26)2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143555

RESUMO

The death of a cell is an inevitable part of its biology. During homeostasis, most cells die through apoptosis. If homeostasis is disturbed, cell death can switch to proinflammatory forms of death, such as necroptosis, pyroptosis, or NETosis. We demonstrate that the formation of neutrophil extracellular traps (NETs), a special form of neutrophil cell death that releases chromatin structures to the extracellular space, is dependent on gasdermin D (GSDMD). GSDMD is a pore-forming protein and an executor of pyroptosis. We screened a chemical library and found a small molecule based on the pyrazolo-oxazepine scaffold that efficiently blocks NET formation and GSDMD-mediated pyroptotic cell death in human cells. During NETosis, GSDMD is proteolytically activated by neutrophil proteases and, in turn, affects protease activation and nuclear expansion in a feed-forward loop. In addition to the central role of GSDMD in pyroptosis, we propose that GSDMD also plays an essential function in NETosis.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Morte Celular/fisiologia , Armadilhas Extracelulares/fisiologia , Proteínas de Neoplasias/fisiologia , Neutrófilos/fisiologia , Animais , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos Mutantes , Peptídeo Hidrolases/farmacologia , Proteínas de Ligação a Fosfato
19.
Br J Pharmacol ; 175(5): 830-839, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29247460

RESUMO

BACKGROUND AND PURPOSE: (-)-Englerin A (EA) is a potent cytotoxic agent against renal carcinoma cells. It achieves its effects by activation of transient receptor potential canonical (TRPC)4/TRPC1 heteromeric channels. It is also an agonist at channels formed by the related protein, TRPC5. Here, we sought an EA analogue, which might enable a better understanding of these effects of EA. EXPERIMENTAL APPROACH: An EA analogue, A54, was synthesized by chemical elaboration of EA. The effects of EA and A54 on the activity of human TRPC4 or TRPC5 channels overexpressed on A498 and HEK 293 cells were investigated, firstly, by measuring intracellular Ca2+ and, secondly, current using whole-cell patch clamp recordings. KEY RESULTS: A54 had weak or no agonist activity at endogenous TRPC4/TRPC1 channels in A498 cells or TRPC4 or TRPC5 homomeric channels overexpressed in HEK 293 cells. A54 strongly inhibited EA-mediated activation of TRPC4/TRPC1 or TRPC5 and weakly inhibited activation of TRPC4. Studies of TRPC5 showed that A54 shifted the EA concentration-response curve to the right without changing its slope, consistent with competitive antagonism. In contrast, Gd3+ -activated TRPC5 or sphingosine-1-phosphate-activated TRPC4 channels were not inhibited but potentiated by A54. A54 did not activate TRPC3 channels or affect the activation of these channels by the agonist 1-oleoyl-2-acetyl-sn-glycerol. CONCLUSIONS AND IMPLICATIONS: This study has revealed a new tool compound for EA and TRPC1/4/5 channel research, which could be useful for characterizing endogenous TRPC1/4/5 channels and understanding EA-binding sites and their physiological relevance.


Assuntos
Potenciais da Membrana/fisiologia , Sesquiterpenos de Guaiano/antagonistas & inibidores , Canais de Cátion TRPC/fisiologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Diglicerídeos/farmacologia , Sinergismo Farmacológico , Gadolínio/farmacologia , Humanos , Lisofosfolipídeos/farmacologia , Sesquiterpenos de Guaiano/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia
20.
ACS Chem Biol ; 13(5): 1307-1312, 2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29658704

RESUMO

Covalent modifications of nonactive site lysine residues by small molecule probes has recently evolved into an important strategy for interrogating biological systems. Here, we report the discovery of a class of bioreactive compounds that covalently modify lysine residues in DegS, the rate limiting protease of the essential bacterial outer membrane stress response pathway. These modifications lead to an allosteric activation and allow the identification of novel residues involved in the allosteric activation circuit. These findings were validated by structural analyses via X-ray crystallography and cell-based reporter systems. We anticipate that our findings are not only relevant for a deeper understanding of the structural basis of allosteric activation in DegS and other HtrA serine proteases but also pinpoint an alternative use of covalent small molecules for probing essential biochemical mechanisms.


Assuntos
Lisina/química , Sondas Moleculares/química , Regulação Alostérica , Proteínas de Bactérias/química , Catálise , Cristalografia por Raios X , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA