Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 317
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(21): 3950-3965.e25, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36170854

RESUMEN

The G protein-coupled receptor cascade leading to production of the second messenger cAMP is replete with pharmacologically targetable proteins, with the exception of the Gα subunit, Gαs. GTPases remain largely undruggable given the difficulty of displacing high-affinity guanine nucleotides and the lack of other drug binding sites. We explored a chemical library of 1012 cyclic peptides to expand the chemical search for inhibitors of this enzyme class. We identified two macrocyclic peptides, GN13 and GD20, that antagonize the active and inactive states of Gαs, respectively. Both macrocyclic peptides fine-tune Gαs activity with high nucleotide-binding-state selectivity and G protein class-specificity. Co-crystal structures reveal that GN13 and GD20 distinguish the conformational differences within the switch II/α3 pocket. Cell-permeable analogs of GN13 and GD20 modulate Gαs/Gßγ signaling in cells through binding to crystallographically defined pockets. The discovery of cyclic peptide inhibitors targeting Gαs provides a path for further development of state-dependent GTPase inhibitors.


Asunto(s)
Péptidos , Receptores Acoplados a Proteínas G , GTP Fosfohidrolasas , Nucleótidos de Guanina , Nucleótidos , Péptidos/química , Péptidos Cíclicos/farmacología
2.
Cell ; 173(5): 1254-1264.e11, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29628140

RESUMEN

The single most frequent cancer-causing mutation across all heterotrimeric G proteins is R201C in Gαs. The current model explaining the gain-of-function activity of the R201 mutations is through the loss of GTPase activity and resulting inability to switch off to the GDP state. Here, we find that the R201C mutation can bypass the need for GTP binding by directly activating GDP-bound Gαs through stabilization of an intramolecular hydrogen bond network. Having found that a gain-of-function mutation can convert GDP into an activator, we postulated that a reciprocal mutation might disrupt the normal role of GTP. Indeed, we found R228C, a loss-of-function mutation in Gαs that causes pseudohypoparathyroidism type 1a (PHP-Ia), compromised the adenylyl cyclase-activating activity of Gαs bound to a non-hydrolyzable GTP analog. These findings show that disease-causing mutations in Gαs can subvert the canonical roles of GDP and GTP, providing new insights into the regulation mechanism of G proteins.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Adenilil Ciclasas/química , Adenilil Ciclasas/metabolismo , Cristalografía por Rayos X , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Enlace de Hidrógeno , Mutagénesis Sitio-Dirigida , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
3.
Nature ; 620(7972): 163-171, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37495694

RESUMEN

An outstanding mystery in biology is why some species, such as the axolotl, can regenerate tissues whereas mammals cannot1. Here, we demonstrate that rapid activation of protein synthesis is a unique feature of the injury response critical for limb regeneration in the axolotl (Ambystoma mexicanum). By applying polysome sequencing, we identify hundreds of transcripts, including antioxidants and ribosome components that are selectively activated at the level of translation from pre-existing messenger RNAs in response to injury. By contrast, protein synthesis is not activated in response to non-regenerative digit amputation in the mouse. We identify the mTORC1 pathway as a key upstream signal that mediates tissue regeneration and translational control in the axolotl. We discover unique expansions in mTOR protein sequence among urodele amphibians. By engineering an axolotl mTOR (axmTOR) in human cells, we show that these changes create a hypersensitive kinase that allows axolotls to maintain this pathway in a highly labile state primed for rapid activation. This change renders axolotl mTOR more sensitive to nutrient sensing, and inhibition of amino acid transport is sufficient to inhibit tissue regeneration. Together, these findings highlight the unanticipated impact of the translatome on orchestrating the early steps of wound healing in a highly regenerative species and provide a missing link in our understanding of vertebrate regenerative potential.


Asunto(s)
Ambystoma mexicanum , Evolución Biológica , Biosíntesis de Proteínas , Regeneración , Serina-Treonina Quinasas TOR , Animales , Humanos , Ratones , Ambystoma mexicanum/fisiología , Secuencia de Aminoácidos , Extremidades/fisiología , Regeneración/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Cicatrización de Heridas , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Especificidad de la Especie , Antioxidantes/metabolismo , Nutrientes/metabolismo , Polirribosomas/genética , Polirribosomas/metabolismo
4.
Cell ; 154(4): 737-47, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23953109

RESUMEN

Mitochondria have long been implicated in the pathogenesis of Parkinson's disease (PD). Mutations in the mitochondrial kinase PINK1 that reduce kinase activity are associated with mitochondrial defects and result in an autosomal-recessive form of early-onset PD. Therapeutic approaches for enhancing the activity of PINK1 have not been considered because no allosteric regulatory sites for PINK1 are known. Here, we show that an alternative strategy, a neo-substrate approach involving the ATP analog kinetin triphosphate (KTP), can be used to increase the activity of both PD-related mutant PINK1(G309D) and PINK1(WT). Moreover, we show that application of the KTP precursor kinetin to cells results in biologically significant increases in PINK1 activity, manifest as higher levels of Parkin recruitment to depolarized mitochondria, reduced mitochondrial motility in axons, and lower levels of apoptosis. Discovery of neo-substrates for kinases could provide a heretofore-unappreciated modality for regulating kinase activity.


Asunto(s)
Mitocondrias/metabolismo , Enfermedad de Parkinson/patología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Adenosina Trifosfato/análogos & derivados , Secuencia de Aminoácidos , Animales , Apoptosis , Axones/metabolismo , Línea Celular , Células Cultivadas , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Cinetina/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/metabolismo , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/genética , Fosforilación , Proteínas Quinasas/química , Ratas , Alineación de Secuencia , Ubiquitina-Proteína Ligasas/metabolismo , Proteína bcl-X/metabolismo
5.
Nature ; 609(7928): 822-828, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36104566

RESUMEN

On-target-off-tissue drug engagement is an important source of adverse effects that constrains the therapeutic window of drug candidates1,2. In diseases of the central nervous system, drugs with brain-restricted pharmacology are highly desirable. Here we report a strategy to achieve inhibition of mammalian target of rapamycin (mTOR) while sparing mTOR activity elsewhere through the use of the brain-permeable mTOR inhibitor RapaLink-1 and the brain-impermeable FKBP12 ligand RapaBlock. We show that this drug combination mitigates the systemic effects of mTOR inhibitors but retains the efficacy of RapaLink-1 in glioblastoma xenografts. We further present a general method to design cell-permeable, FKBP12-dependent kinase inhibitors from known drug scaffolds. These inhibitors are sensitive to deactivation by RapaBlock, enabling the brain-restricted inhibition of their respective kinase targets.


Asunto(s)
Encéfalo , Inhibidores mTOR , Sirolimus , Serina-Treonina Quinasas TOR , Humanos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Quimioterapia Combinada , Glioblastoma/tratamiento farmacológico , Ligandos , Inhibidores mTOR/metabolismo , Inhibidores mTOR/farmacocinética , Inhibidores mTOR/farmacología , Sirolimus/análogos & derivados , Proteína 1A de Unión a Tacrolimus/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Annu Rev Biochem ; 80: 769-95, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21548788

RESUMEN

Kinases are highly regulated enzymes with diverse mechanisms controlling their catalytic output. Over time, chemical discovery efforts for kinases have produced ATP-competitive compounds, allosteric regulators, irreversible binders, and highly specific inhibitors. These distinct classes of small molecules have revealed many novel aspects about kinase-mediated signaling, and some have progressed from simple tool compounds into clinically validated therapeutics. This review explores several small-molecule inhibitors for kinases highlighting elaborate mechanisms by which kinase function is modulated. A complete surprise of targeted kinase drug discovery has been the finding of ATP-competitive inhibitors that behave as agonists, rather than antagonists, of their direct kinase target. These studies hint at a connection between ATP-binding site occupancy and networks of communication that are independent of kinase catalysis. Indeed, kinase inhibitors that induce changes in protein localization, protein-protein interactions, and even enhancement of catalytic activity of the target kinase have been found. The relevance of these findings to the therapeutic efficacy of kinase inhibitors and to the future identification of new classes of drug targets is discussed.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Biología Computacional , Descubrimiento de Drogas , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutación , Neoplasias/enzimología , Neoplasias/genética , Conformación Proteica , Proteínas Quinasas/genética , Alineación de Secuencia
7.
Nat Immunol ; 15(2): 186-94, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24317039

RESUMEN

Signaling via the T cell antigen receptor (TCR) is initiated by Src-family kinases (SFKs). To understand how the kinase Csk, a negative regulator of SFKs, controls the basal state and the initiation of TCR signaling, we generated mice that express a Csk variant sensitive to an analog of the common kinase inhibitor PP1 (Csk(AS)). Inhibition of Csk(AS) in thymocytes, without engagement of the TCR, induced potent activation of SFKs and proximal TCR signaling up to phospholipase C-γ1 (PLC-γ1). Unexpectedly, increases in inositol phosphates, intracellular calcium and phosphorylation of the kinase Erk were impaired. Altering the actin cytoskeleton pharmacologically or providing costimulation via CD28 'rescued' those defects. Thus, Csk has a critical role in preventing TCR signaling. However, our studies also revealed a requirement for actin remodeling, initiated by costimulation, for full TCR signaling.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Proteínas Mutantes/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Timocitos/inmunología , Familia-src Quinasas/metabolismo , Animales , Antígenos CD28/inmunología , Proteína Tirosina Quinasa CSK , Células Cultivadas , Citocalasina D/administración & dosificación , Citoesqueleto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Mutantes/genética , Polimerizacion/efectos de los fármacos , Ingeniería de Proteínas , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Timocitos/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
8.
Nat Immunol ; 15(7): 687-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24908390

RESUMEN

The catalytic activity of Zap70 is crucial for T cell antigen receptor (TCR) signaling, but the quantitative and temporal requirements for its function in thymocyte development are not known. Using a chemical-genetic system to selectively and reversibly inhibit Zap70 catalytic activity in a model of synchronized thymic selection, we showed that CD4(+)CD8(+) thymocytes integrate multiple, transient, Zap70-dependent signals over more than 36 h to reach a cumulative threshold for positive selection, whereas 1 h of signaling was sufficient for negative selection. Titration of Zap70 activity resulted in graded reductions in positive and negative selection but did not decrease the cumulative TCR signals integrated by positively selected OT-I cells, which revealed heterogeneity, even among CD4(+)CD8(+) thymocytes expressing identical TCRs undergoing positive selection.


Asunto(s)
Linfocitos T/fisiología , Proteína Tirosina Quinasa ZAP-70/fisiología , Animales , Calcio/metabolismo , Catálisis , Diferenciación Celular , Péptidos y Proteínas de Señalización Intracelular/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal , Quinasa Syk
9.
Nat Chem Biol ; 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38443470

RESUMEN

K-Ras is the most commonly mutated oncogene in human cancer. The recently approved non-small cell lung cancer drugs sotorasib and adagrasib covalently capture an acquired cysteine in K-Ras-G12C mutation and lock it in a signaling-incompetent state. However, covalent inhibition of G12D, the most frequent K-Ras mutation particularly prevalent in pancreatic ductal adenocarcinoma, has remained elusive due to the lack of aspartate-targeting chemistry. Here we present a set of malolactone-based electrophiles that exploit ring strain to crosslink K-Ras-G12D at the mutant aspartate to form stable covalent complexes. Structural insights from X-ray crystallography and exploitation of the stereoelectronic requirements for attack of the electrophile allowed development of a substituted malolactone that resisted attack by aqueous buffer but rapidly crosslinked with the aspartate-12 of K-Ras in both GDP and GTP state. The GTP-state targeting allowed effective suppression of downstream signaling, and selective inhibition of K-Ras-G12D-driven cancer cell proliferation in vitro and xenograft growth in mice.

10.
Mol Cell Proteomics ; 22(4): 100522, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36863607

RESUMEN

PKC epsilon (PKCε) plays important roles in behavioral responses to alcohol and in anxiety-like behavior in rodents, making it a potential drug target for reducing alcohol consumption and anxiety. Identifying signals downstream of PKCε could reveal additional targets and strategies for interfering with PKCε signaling. We used a chemical genetic screen combined with mass spectrometry to identify direct substrates of PKCε in mouse brain and validated findings for 39 of them using peptide arrays and in vitro kinase assays. Prioritizing substrates with several public databases such as LINCS-L1000, STRING, GeneFriends, and GeneMAINA predicted interactions between these putative substrates and PKCε and identified substrates associated with alcohol-related behaviors, actions of benzodiazepines, and chronic stress. The 39 substrates could be broadly classified in three functional categories: cytoskeletal regulation, morphogenesis, and synaptic function. These results provide a list of brain PKCε substrates, many of which are novel, for future investigation to determine the role of PKCε signaling in alcohol responses, anxiety, responses to stress, and other related behaviors.


Asunto(s)
Proteína Quinasa C-epsilon , Transducción de Señal , Ratones , Animales , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo , Etanol , Consumo de Bebidas Alcohólicas/genética , Encéfalo/metabolismo
11.
Proc Natl Acad Sci U S A ; 119(38): e2204083119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36095197

RESUMEN

Mammalian target of rapamycin (mTOR) is a highly conserved eukaryotic protein kinase that coordinates cell growth and metabolism, and plays a critical role in cancer, immunity, and aging. It remains unclear how mTOR signaling in individual tissues contributes to whole-organism processes because mTOR inhibitors, like the natural product rapamycin, are administered systemically and target multiple tissues simultaneously. We developed a chemical-genetic system, termed selecTOR, that restricts the activity of a rapamycin analog to specific cell populations through targeted expression of a mutant FKBP12 protein. This analog has reduced affinity for its obligate binding partner FKBP12, which reduces its ability to inhibit mTOR in wild-type cells and tissues. Expression of the mutant FKBP12, which contains an expanded binding pocket, rescues the activity of this rapamycin analog. Using this system, we show that selective mTOR inhibition can be achieved in Saccharomyces cerevisiae and human cells, and we validate the utility of our system in an intact metazoan model organism by identifying the tissues responsible for a rapamycin-induced developmental delay in Drosophila.


Asunto(s)
Inhibidores de Proteínas Quinasas , Sirolimus , Serina-Treonina Quinasas TOR , Humanos , Especificidad de Órganos , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Sirolimus/análogos & derivados , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Proteína 1A de Unión a Tacrolimus/genética , Proteína 1A de Unión a Tacrolimus/metabolismo
12.
Nat Chem Biol ; 18(11): 1177-1183, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35864332

RESUMEN

Drugs that directly impede the function of driver oncogenes offer exceptional efficacy and a therapeutic window. The recently approved mutant selective small-molecule cysteine-reactive covalent inhibitor of the G12C mutant of K-Ras, sotorasib, provides a case in point. KRAS is the most frequently mutated proto-oncogene in human cancer, yet despite success targeting the G12C allele, targeted therapy for other hotspot mutants of KRAS has not been described. Here we report the discovery of small molecules that covalently target a G12S somatic mutation in K-Ras and suppress its oncogenic signaling. We show that these molecules are active in cells expressing K-Ras(G12S) but spare the wild-type protein. Our results provide a path to targeting a second somatic mutation in the oncogene KRAS by overcoming the weak nucleophilicity of an acquired serine residue. The chemistry we describe may serve as a basis for the selective targeting of other unactivated serines.


Asunto(s)
Cisteína , Serina , Humanos , Cisteína/metabolismo , Serina/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Mutación , Oncogenes , Acilación
13.
Nat Chem Biol ; 18(6): 596-604, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35314814

RESUMEN

Current small-molecule inhibitors of KRAS(G12C) bind irreversibly in the switch-II pocket (SII-P), exploiting the strong nucleophilicity of the acquired cysteine as well as the preponderance of the GDP-bound form of this mutant. Nevertheless, many oncogenic KRAS mutants lack these two features, and it remains unknown whether targeting the SII-P is a practical therapeutic approach for KRAS mutants beyond G12C. Here we use NMR spectroscopy and a cellular KRAS engagement assay to address this question by examining a collection of SII-P ligands from the literature and from our own laboratory. We show that the SII-Ps of many KRAS hotspot (G12, G13, Q61) mutants are accessible using noncovalent ligands, and that this accessibility is not necessarily coupled to the GDP state of KRAS. The results we describe here emphasize the SII-P as a privileged drug-binding site on KRAS and unveil new therapeutic opportunities in RAS-driven cancer.


Asunto(s)
Mieloma Múltiple , Proteínas Proto-Oncogénicas p21(ras) , Humanos , Ligandos , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética
14.
Cell ; 136(1): 50-61, 2009 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-19135888

RESUMEN

Elongation of telomeres by telomerase replenishes the loss of terminal telomeric DNA repeats during each cell cycle. In budding yeast, Cdc13 plays an essential role in telomere length homeostasis, partly through its interactions with both the telomerase complex and the competing Stn1-Ten1 complex. Previous studies in yeast have shown that telomere elongation by telomerase is cell cycle dependent, but the mechanism underlying this dependence is unclear. In S. cerevisiae, a single cyclin-dependent kinase Cdk1 (Cdc28) coordinates the serial events required for the cell division cycle, but no Cdk1 substrate has been identified among telomerase and telomere-associated factors. Here we show that Cdk1-dependent phosphorylation of Cdc13 is essential for efficient recruitment of the yeast telomerase complex to telomeres by favoring the interaction of Cdc13 with Est1 rather than the competing Stn1-Ten1 complex. These results provide a direct mechanistic link between coordination of telomere elongation and cell-cycle progression in vivo.


Asunto(s)
Proteína Quinasa CDC28 de Saccharomyces cerevisiae/metabolismo , Ciclo Celular , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Fosforilación , Telomerasa/metabolismo
15.
Cell ; 136(5): 952-63, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19269370

RESUMEN

Reversible protein phosphorylation is a signaling mechanism involved in all cellular processes. To create a systems view of the signaling apparatus in budding yeast, we generated an epistatic miniarray profile (E-MAP) comprised of 100,000 pairwise, quantitative genetic interactions, including virtually all protein and small-molecule kinases and phosphatases as well as key cellular regulators. Quantitative genetic interaction mapping reveals factors working in compensatory pathways (negative genetic interactions) or those operating in linear pathways (positive genetic interactions). We found an enrichment of positive genetic interactions between kinases, phosphatases, and their substrates. In addition, we assembled a higher-order map from sets of three genes that display strong interactions with one another: triplets enriched for functional connectivity. The resulting network view provides insights into signaling pathway regulation and reveals a link between the cell-cycle kinase, Cak1, the Fus3 MAP kinase, and a pathway that regulates chromatin integrity during transcription by RNA polymerase II.


Asunto(s)
Fosforilación , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Acetilación , Histonas/metabolismo , Proteínas Quinasas/metabolismo
16.
Genes Dev ; 30(1): 117-31, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26728557

RESUMEN

The transcription cycle of RNA polymerase II (Pol II) is regulated at discrete transition points by cyclin-dependent kinases (CDKs). Positive transcription elongation factor b (P-TEFb), a complex of Cdk9 and cyclin T1, promotes release of paused Pol II into elongation, but the precise mechanisms and targets of Cdk9 action remain largely unknown. Here, by a chemical genetic strategy, we identified ∼ 100 putative substrates of human P-TEFb, which were enriched for proteins implicated in transcription and RNA catabolism. Among the RNA processing factors phosphorylated by Cdk9 was the 5'-to-3' "torpedo" exoribonuclease Xrn2, required in transcription termination by Pol II, which we validated as a bona fide P-TEFb substrate in vivo and in vitro. Phosphorylation by Cdk9 or phosphomimetic substitution of its target residue, Thr439, enhanced enzymatic activity of Xrn2 on synthetic substrates in vitro. Conversely, inhibition or depletion of Cdk9 or mutation of Xrn2-Thr439 to a nonphosphorylatable Ala residue caused phenotypes consistent with inefficient termination in human cells: impaired Xrn2 chromatin localization and increased readthrough transcription of endogenous genes. Therefore, in addition to its role in elongation, P-TEFb regulates termination by promoting chromatin recruitment and activation of a cotranscriptional RNA processing enzyme, Xrn2.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Regulación de la Expresión Génica/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Cromatina/metabolismo , Activación Enzimática/genética , Pruebas Genéticas , Células HCT116 , Humanos , Fosforilación , Unión Proteica
17.
Anal Chem ; 95(50): 18344-18351, 2023 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-38060502

RESUMEN

Protein properties and interactions have been widely investigated by using external labels. However, the micromolar sensitivity of the current dyes limits their applicability due to the high material consumption and assay cost. In response to this challenge, we synthesized a series of cyanine5 (Cy5) dye-based quencher molecules to develop an external dye technique to probe proteins at the nanomolar protein level in a high-throughput one-step assay format. Several families of Cy5 dye-based quenchers with ring and/or side-chain modifications were designed and synthesized by introducing organic small molecules or peptides. Our results showed that steric hindrance and electrostatic interactions are more important than hydrophobicity in the interaction between the luminescent negatively charged europium-chelate-labeled peptide (Eu-probe) and the quencher molecules. The presence of substituents on the quencher indolenine rings reduces their quenching property, whereas the increased positive charge on the indolenine side chain improved the interaction between the quenchers and the luminescent compound. The designed quencher structures entirely altered the dynamics of the Eu-probe (protein-probe) for studying protein stability and interactions, as we were able to reduce the quencher concentration 100-fold. Moreover, the new quencher molecules allowed us to conduct the experiments using neutral buffer conditions, known as the peptide-probe assay. These improvements enabled us to apply the method in a one-step format for nanomolar protein-ligand interaction and protein profiling studies instead of the previously developed two-step protocol. These improvements provide a faster and simpler method with lower material consumption.


Asunto(s)
Colorantes , Péptidos , Carbocianinas/química , Péptidos/química , Luminiscencia , Colorantes Fluorescentes/química
18.
J Am Chem Soc ; 144(35): 15916-15921, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-36001446

RESUMEN

KRAS mutations are one of the most common oncogenic drivers in human cancer. While small molecule inhibitors for the G12C mutant have been successfully developed, allele-specific inhibition for other KRAS hotspot mutants remains challenging. Here we report the discovery of covalent chemical ligands for the common oncogenic mutant K-Ras(G12R). These ligands bind in the Switch II pocket and irreversibly react with the mutant arginine residue. An X-ray crystal structure reveals an imidazolium condensation product formed between the α,ß-diketoamide ligand and the ε- and η-nitrogens of arginine 12. Our results show that arginine residues can be selectively targeted with small molecule electrophiles despite their weak nucleophilicity and provide the basis for the development of mutant-specific therapies for K-Ras(G12R)-driven cancer.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Arginina , Genes ras , Humanos , Ligandos , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética
19.
Nat Immunol ; 11(12): 1085-92, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21037577

RESUMEN

To investigate the role of the kinase Zap70 in T cells, we generated mice expressing a Zap70 mutant whose catalytic activity can be selectively blocked by a small-molecule inhibitor. We found that conventional naive, effector and memory T cells were dependent on the kinase activity of Zap70 for their activation, which demonstrated a nonredundant role for Zap70 in signals induced by the T cell antigen receptor (TCR). In contrast, the catalytic activity of Zap70 was not required for activation of the GTPase Rap1 and inside-out signals that promote integrin adhesion. This Zap70 kinase-independent pathway was sufficient for the suppressive activity of regulatory T cells (T(reg) cells), which was unperturbed by inhibition of the catalytic activity of Zap70. Our results indicate Zap70 is a likely therapeutic target.


Asunto(s)
Biocatálisis , Activación de Linfocitos/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/enzimología , Proteína Tirosina Quinasa ZAP-70/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Separación Celular , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Immunoblotting , Inmunoprecipitación , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Transgénicos , Pirazoles/química , Pirazoles/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
20.
Nature ; 534(7606): 272-6, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27279227

RESUMEN

Precision medicines exert selective pressure on tumour cells that leads to the preferential growth of resistant subpopulations, necessitating the development of next-generation therapies to treat the evolving cancer. The PIK3CA-AKT-mTOR pathway is one of the most commonly activated pathways in human cancers, which has led to the development of small-molecule inhibitors that target various nodes in the pathway. Among these agents, first-generation mTOR inhibitors (rapalogs) have caused responses in 'N-of-1' cases, and second-generation mTOR kinase inhibitors (TORKi) are currently in clinical trials. Here we sought to delineate the likely resistance mechanisms to existing mTOR inhibitors in human cell lines, as a guide for next-generation therapies. The mechanism of resistance to the TORKi was unusual in that intrinsic kinase activity of mTOR was increased, rather than a direct active-site mutation interfering with drug binding. Indeed, identical drug-resistant mutations have been also identified in drug-naive patients, suggesting that tumours with activating MTOR mutations will be intrinsically resistant to second-generation mTOR inhibitors. We report the development of a new class of mTOR inhibitors that overcomes resistance to existing first- and second-generation inhibitors. The third-generation mTOR inhibitor exploits the unique juxtaposition of two drug-binding pockets to create a bivalent interaction that allows inhibition of these resistant mutants.


Asunto(s)
Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/genética , Mutación/genética , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Animales , Sitios de Unión/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Ratones , Mutación/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteínas Quinasas/clasificación , Estructura Terciaria de Proteína/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/química , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA