Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nat Commun ; 14(1): 3548, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37322067

RESUMEN

Lipoic acid is an essential enzyme cofactor in central metabolic pathways. Due to its claimed antioxidant properties, racemic (R/S)-lipoic acid is used as a food supplement but is also investigated as a pharmaceutical in over 180 clinical trials covering a broad range of diseases. Moreover, (R/S)-lipoic acid is an approved drug for the treatment of diabetic neuropathy. However, its mechanism of action remains elusive. Here, we performed chemoproteomics-aided target deconvolution of lipoic acid and its active close analog lipoamide. We find that histone deacetylases HDAC1, HDAC2, HDAC3, HDAC6, HDAC8, and HDAC10 are molecular targets of the reduced form of lipoic acid and lipoamide. Importantly, only the naturally occurring (R)-enantiomer inhibits HDACs at physiologically relevant concentrations and leads to hyperacetylation of HDAC substrates. The inhibition of HDACs by (R)-lipoic acid and lipoamide explain why both compounds prevent stress granule formation in cells and may also provide a molecular rationale for many other phenotypic effects elicited by lipoic acid.


Asunto(s)
Inhibidores de Histona Desacetilasas , Ácido Tióctico , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/química , Ácido Tióctico/farmacología , Histona Desacetilasas/metabolismo , Antioxidantes/farmacología
2.
JCI Insight ; 7(10)2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35439169

RESUMEN

Systemic therapies for pancreatic ductal adenocarcinoma (PDAC) remain unsatisfactory. Clinical prognosis is particularly poor for tumor subtypes with activating aberrations in the MYC pathway, creating an urgent need for novel therapeutic targets. To unbiasedly find MYC-associated epigenetic dependencies, we conducted a drug screen in pancreatic cancer cell lines. Here, we found that protein arginine N-methyltransferase 5 (PRMT5) inhibitors triggered an MYC-associated dependency. In human and murine PDACs, a robust connection of MYC and PRMT5 was detected. By the use of gain- and loss-of-function models, we confirmed the increased efficacy of PRMT5 inhibitors in MYC-deregulated PDACs. Although inhibition of PRMT5 was inducing DNA damage and arresting PDAC cells in the G2/M phase of the cell cycle, apoptotic cell death was executed predominantly in cells with high MYC expression. Experiments in primary patient-derived PDAC models demonstrated the existence of a highly PRMT5 inhibitor-sensitive subtype. Our work suggests developing PRMT5 inhibitor-based therapies for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Detección Precoz del Cáncer , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Epigénesis Genética , Humanos , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Neoplasias Pancreáticas
3.
Nature ; 594(7862): 246-252, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33845483

RESUMEN

The emergence and global spread of SARS-CoV-2 has resulted in the urgent need for an in-depth understanding of molecular functions of viral proteins and their interactions with the host proteome. Several individual omics studies have extended our knowledge of COVID-19 pathophysiology1-10. Integration of such datasets to obtain a holistic view of virus-host interactions and to define the pathogenic properties of SARS-CoV-2 is limited by the heterogeneity of the experimental systems. Here we report a concurrent multi-omics study of SARS-CoV-2 and SARS-CoV. Using state-of-the-art proteomics, we profiled the interactomes of both viruses, as well as their influence on the transcriptome, proteome, ubiquitinome and phosphoproteome of a lung-derived human cell line. Projecting these data onto the global network of cellular interactions revealed crosstalk between the perturbations taking place upon infection with SARS-CoV-2 and SARS-CoV at different levels and enabled identification of distinct and common molecular mechanisms of these closely related coronaviruses. The TGF-ß pathway, known for its involvement in tissue fibrosis, was specifically dysregulated by SARS-CoV-2 ORF8 and autophagy was specifically dysregulated by SARS-CoV-2 ORF3. The extensive dataset (available at https://covinet.innatelab.org ) highlights many hotspots that could be targeted by existing drugs and may be used to guide rational design of virus- and host-directed therapies, which we exemplify by identifying inhibitors of kinases and matrix metalloproteases with potent antiviral effects against SARS-CoV-2.


Asunto(s)
COVID-19/metabolismo , Interacciones Huésped-Patógeno , Proteoma/metabolismo , Proteómica , SARS-CoV-2/patogenicidad , Síndrome Respiratorio Agudo Grave/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Animales , Antivirales/farmacología , Autofagia/efectos de los fármacos , COVID-19/inmunología , COVID-19/virología , Línea Celular , Conjuntos de Datos como Asunto , Evaluación Preclínica de Medicamentos , Interacciones Huésped-Patógeno/inmunología , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Fosforilación , Mapas de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Proteoma/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , SARS-CoV-2/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Síndrome Respiratorio Agudo Grave/virología , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitinación , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Viroporinas/metabolismo
4.
Nat Chem ; 12(2): 145-158, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31844194

RESUMEN

New drugs are desperately needed to combat methicillin-resistant Staphylococcus aureus (MRSA) infections. Here, we report screening commercial kinase inhibitors for antibacterial activity and found the anticancer drug sorafenib as major hit that effectively kills MRSA strains. Varying the key structural features led to the identification of a potent analogue, PK150, that showed antibacterial activity against several pathogenic strains at submicromolar concentrations. Furthermore, this antibiotic eliminated challenging persisters as well as established biofilms. PK150 holds promising therapeutic potential as it did not induce in vitro resistance, and shows oral bioavailability and in vivo efficacy. Analysis of the mode of action using chemical proteomics revealed several targets, which included interference with menaquinone biosynthesis by inhibiting demethylmenaquinone methyltransferase and the stimulation of protein secretion by altering the activity of signal peptidase IB. Reduced endogenous menaquinone levels along with enhanced levels of extracellular proteins of PK150-treated bacteria support this target hypothesis. The associated antibiotic effects, especially the lack of resistance development, probably stem from the compound's polypharmacology.


Asunto(s)
Antibacterianos/uso terapéutico , Benzodioxoles/uso terapéutico , Reposicionamiento de Medicamentos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Sorafenib/análogos & derivados , Sorafenib/uso terapéutico , Animales , Antibacterianos/síntesis química , Antibacterianos/farmacocinética , Autólisis/inducido químicamente , Benzodioxoles/síntesis química , Benzodioxoles/farmacocinética , Biopelículas/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Masculino , Staphylococcus aureus Resistente a Meticilina/fisiología , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Sorafenib/farmacocinética , Relación Estructura-Actividad
5.
J Agric Food Chem ; 65(10): 2147-2154, 2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-28225606

RESUMEN

The interaction of astringent substances with salivary proteins, which results in protein precipitation, is considered a key event in the molecular mechanism underlying the oral sensation of puckering astringency. As the chemical nature of orally active astringents is diverse and the knowledge of their interactions with salivary proteins rather fragmentary, human whole saliva samples were incubated with suprathreshold and isointensity solutions of the astringent polyphenol (-)-epigallocatechin gallate, the multivalent metal salt iron(III) sulfate, the amino-functionalized polysaccharide chitosan, and the basic protein lysozyme. After separation of the precipitated proteins, the proteins affected by the astringents were identified and relatively quantified for the first time by complementary bottom-up and top-down mass spectrometry-based proteomics approaches. Major salivary target proteins, which may be involved in astringency perception, are reported here for each astringent stimulus.


Asunto(s)
Astringentes/metabolismo , Boca/metabolismo , Proteínas y Péptidos Salivales/metabolismo , Adulto , Astringentes/química , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Masculino , Espectrometría de Masas , Proteómica , Proteínas y Péptidos Salivales/química , Gusto , Adulto Joven
6.
ChemMedChem ; 11(18): 2084-94, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27472906

RESUMEN

Despite the considerable interest in protein kinase C-related kinase 1 (PRK1) as a target in cancer research, there is still a lack of PRK1 inhibitors with suitable selectivity profiles and physicochemical properties. To identify new PRK1 inhibitors we applied a virtual screening approach, which combines ensemble docking, minimization of the protein-ligand complex, binding free energy calculations, and application of quantitative structure-activity relationship (QSAR) models for predicting in vitro activity. The developed approach was then applied in a prospective manner to screen available libraries of kinase inhibitors from Selleck and GlaxoSmithKline (GSK). Compounds that showed favorable prediction were then tested in vitro for PRK1 inhibition. Some of the hits were found to inhibit PRK1 in the low-nanomolar range. Three in vitro hits were additionally tested in a mass-spectrometry-based cellular kinase profiling assay to examine selectivity. Our findings show that nanomolar and drug-like inhibitors can be identified by the virtual screening approach presented herein. The identified inhibitors are valuable tools for gaining a better understanding of PRK1 inhibition, and the identified hits can serve as starting points for further chemical optimization.


Asunto(s)
Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Termodinámica , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Humanos , Estructura Molecular , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad Cuantitativa
7.
J Pathol ; 230(4): 410-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23592244

RESUMEN

Chemotherapeutic drugs kill cancer cells, but it is unclear why this happens in responding patients but not in non-responders. Proteomic profiles of patients with oesophageal adenocarcinoma may be helpful in predicting response and selecting more effective treatment strategies. In this study, pretherapeutic oesophageal adenocarcinoma biopsies were analysed for proteomic changes associated with response to chemotherapy by MALDI imaging mass spectrometry. Resulting candidate proteins were identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and investigated for functional relevance in vitro. Clinical impact was validated in pretherapeutic biopsies from an independent patient cohort. Studies on the incidence of these defects in other solid tumours were included. We discovered that clinical response to cisplatin correlated with pre-existing defects in the mitochondrial respiratory chain complexes of cancer cells, caused by loss of specific cytochrome c oxidase (COX) subunits. Knockdown of a COX protein altered chemosensitivity in vitro, increasing the propensity of cancer cells to undergo cell death following cisplatin treatment. In an independent validation, patients with reduced COX protein expression prior to treatment exhibited favourable clinical outcomes to chemotherapy, whereas tumours with unchanged COX expression were chemoresistant. In conclusion, previously undiscovered pre-existing defects in mitochondrial respiratory complexes cause cancer cells to become chemosensitive: mitochondrial defects lower the cells' threshold for undergoing cell death in response to cisplatin. By contrast, cancer cells with intact mitochondrial respiratory complexes are chemoresistant and have a high threshold for cisplatin-induced cell death. This connection between mitochondrial respiration and chemosensitivity is relevant to anticancer therapeutics that target the mitochondrial electron transport chain.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Adenocarcinoma/patología , Anciano , Biomarcadores de Tumor/genética , Biopsia , Línea Celular Tumoral , Quimioterapia Adyuvante , Cromatografía Liquida , Cisplatino/administración & dosificación , Regulación hacia Abajo , Resistencia a Antineoplásicos , Complejo IV de Transporte de Electrones/genética , Neoplasias Esofágicas/enzimología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Fluorouracilo/administración & dosificación , Humanos , Persona de Mediana Edad , Mitocondrias/enzimología , Mitocondrias/patología , Terapia Neoadyuvante , Medicina de Precisión , Proteómica/métodos , Interferencia de ARN , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem , Transfección , Resultado del Tratamiento
8.
J Proteome Res ; 12(4): 1723-31, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23495751

RESUMEN

Kinases are involved in the regulation of many cellular processes and aberrant kinase signaling has been implicated in human disease. As a consequence, kinases are attractive drug targets. Assessing kinase function and drug selectivity in a more physiological context is challenging and often hampered by the generally low expression level of kinases and the extensive post-translation modification in vivo. Kinase drug selectivity screens by chemical proteomics have gained attention because they allow the profiling of hundreds of kinases against one drug at the same time. Here, we directly compared two such methods, notably, immobilized broad spectrum kinase inhibitors (kinobeads) and active site labeling using desthiobiotin-ATP and -ADP probes. Affinity purification of ∼ 100 kinases by either kinobeads or ATP/ADP probes was readily achieved using 1 mg of cellular protein. Bioinformatic analysis revealed a high degree of complementarity of the two techniques. Kinobeads covered the Tyrosine Kinase family particularly well and ATP probes enriched higher numbers of STE family kinases. A consecutive combination of both enrichment strategies therefore allowed for the coverage of a larger part of the kinome than any one technique alone. While kinobeads are very selective for kinases, the ATP/ADP probes also enriched a large number of other nucleotide binding proteins. Both methods were applied to the selectivity profiling of the small molecular Aurora kinase inhibitor tozasertib in K562 cells. Our data confirmed Aurora A, B, and BCR-ABL as the main targets of tozasertib and identified TNK1, STK2, RPS6KA1, and RPS6KA3 as submicromolar off targets.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteómica/métodos , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Aurora Quinasas/antagonistas & inhibidores , Aurora Quinasas/metabolismo , Proteínas Fetales/metabolismo , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Células K562/efectos de los fármacos , Piperazinas/metabolismo , Piperazinas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
9.
Chem Biol ; 19(1): 72-84, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22284356

RESUMEN

Preclinical stages in the drug discovery process require a multitude of biochemical and genetic assays in order to characterize the effects of drug candidates on cellular systems and model organisms. Early attempts to apply unbiased proteomic techniques to the identification of protein targets and off-targets as well as to elucidate the mode of action of candidate drug molecules suffered from a striking discrepancy between scientific expectations and what the technology was able to deliver at the time. Dramatic technological improvements in mass spectrometry-based proteomic and chemoproteomic strategies have radically changed this situation. This review, therefore, highlights proteomic approaches suitable for preclinical drug discovery illustrated by recent success stories.


Asunto(s)
Espectrometría de Masas , Proteómica , Animales , Biomarcadores/metabolismo , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Preparaciones Farmacéuticas/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas/metabolismo
10.
J Biol Chem ; 279(13): 12804-11, 2004 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-14676191

RESUMEN

The polarization of eukaryotic cells is controlled by the concerted activities of asymmetrically localized proteins. The PAR proteins, first identified in Caenorhabditis elegans, are common regulators of cell polarity conserved from nematode and flies to man. However, little is known about the molecular mechanisms by which these proteins and protein complexes establish cell polarity in mammals. We have mapped multiprotein complexes formed around the putative human Par orthologs MARK4 (microtubule-associated protein/microtubule affinity-regulating kinase 4) (Par-1), Par-3, LKB1 (Par-4), 14-3-3zeta and eta (Par-5), Par-6a, -b, -c, and PKClambda (PKC3). We employed a proteomic approach comprising tandem affinity purification (TAP) of protein complexes from cultured cells and protein sequencing by tandem mass spectrometry. From these data we constructed a highly interconnected protein network consisting of three core complex "modules" formed around MARK4 (Par-1), Par-3.Par-6, and LKB1 (Par-4). The network confirms most previously reported interactions. In addition we identified more than 50 novel interactors, some of which, like the 14-3-3 phospho-protein scaffolds, occur in more than one distinct complex. We demonstrate that the complex formation between LKB1.Par-4, PAPK, and Mo25 results in the translocation of LKB1 from the nucleus to the cytoplasm and to tight junctions and show that the LKB1 complex may activate MARKs, which are known to introduce 14-3-3 binding sites into several substrates. Our findings suggest co-regulation and/or signaling events between the distinct Par complexes and provide a basis for further elucidation of the molecular mechanisms that govern cell polarity.


Asunto(s)
Proteínas Serina-Treonina Quinasas/química , Proteoma , Proteínas 14-3-3 , Quinasas de la Proteína-Quinasa Activada por el AMP , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas de Caenorhabditis elegans/química , Línea Celular , Núcleo Celular/metabolismo , Clonación Molecular , Citoplasma/metabolismo , ADN Complementario/metabolismo , Perros , Humanos , Isoenzimas , Espectrometría de Masas , Microscopía Fluorescente , Modelos Biológicos , Datos de Secuencia Molecular , Pruebas de Precipitina , Unión Proteica , Proteína Quinasa C/química , Transporte de Proteínas , Proteínas/química , Homología de Secuencia de Aminoácido , Tirosina 3-Monooxigenasa/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA