Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 158(3): 534-48, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25018104

RESUMEN

Depending on endoplasmic reticulum (ER) stress levels, the ER transmembrane multidomain protein IRE1α promotes either adaptation or apoptosis. Unfolded ER proteins cause IRE1α lumenal domain homo-oligomerization, inducing trans autophosphorylation that further drives homo-oligomerization of its cytosolic kinase/endoribonuclease (RNase) domains to activate mRNA splicing of adaptive XBP1 transcription factor. However, under high/chronic ER stress, IRE1α surpasses an oligomerization threshold that expands RNase substrate repertoire to many ER-localized mRNAs, leading to apoptosis. To modulate these effects, we developed ATP-competitive IRE1α Kinase-Inhibiting RNase Attenuators-KIRAs-that allosterically inhibit IRE1α's RNase by breaking oligomers. One optimized KIRA, KIRA6, inhibits IRE1α in vivo and promotes cell survival under ER stress. Intravitreally, KIRA6 preserves photoreceptor functional viability in rat models of ER stress-induced retinal degeneration. Systemically, KIRA6 preserves pancreatic ß cells, increases insulin, and reduces hyperglycemia in Akita diabetic mice. Thus, IRE1α powerfully controls cell fate but can itself be controlled with small molecules to reduce cell degeneration.


Asunto(s)
Estrés del Retículo Endoplásmico , Endorribonucleasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Regulación Alostérica , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Endorribonucleasas/química , Endorribonucleasas/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Retina/metabolismo , Ribonucleasas/antagonistas & inhibidores
2.
Nat Chem Biol ; 17(11): 1148-1156, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34556859

RESUMEN

The unfolded protein response (UPR) homeostatically matches endoplasmic reticulum (ER) protein-folding capacity to cellular secretory needs. However, under high or chronic ER stress, the UPR triggers apoptosis. This cell fate dichotomy is promoted by differential activation of the ER transmembrane kinase/endoribonuclease (RNase) IRE1α. We previously found that the RNase of IRE1α can be either fully activated or inactivated by ATP-competitive kinase inhibitors. Here we developed kinase inhibitors, partial antagonists of IRE1α RNase (PAIRs), that partially antagonize the IRE1α RNase at full occupancy. Biochemical and structural studies show that PAIRs promote partial RNase antagonism by intermediately displacing the helix αC in the IRE1α kinase domain. In insulin-producing ß-cells, PAIRs permit adaptive splicing of Xbp1 mRNA while quelling destructive ER mRNA endonucleolytic decay and apoptosis. By preserving Xbp1 mRNA splicing, PAIRs allow B cells to differentiate into immunoglobulin-producing plasma cells. Thus, an intermediate RNase-inhibitory 'sweet spot', achieved by PAIR-bound IRE1α, captures a desirable conformation for drugging this master UPR sensor/effector.


Asunto(s)
Adenosina Trifosfato/farmacología , Endorribonucleasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/química , Endorribonucleasas/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Desplegamiento Proteico/efectos de los fármacos
3.
Nat Chem Biol ; 8(12): 982-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23086298

RESUMEN

Under endoplasmic reticulum stress, unfolded protein accumulation leads to activation of the endoplasmic reticulum transmembrane kinase/endoRNase (RNase) IRE1α. IRE1α oligomerizes, autophosphorylates and initiates splicing of XBP1 mRNA, thus triggering the unfolded protein response (UPR). Here we show that IRE1α's kinase-controlled RNase can be regulated in two distinct modes with kinase inhibitors: one class of ligands occupies IRE1α's kinase ATP-binding site to activate RNase-mediated XBP1 mRNA splicing even without upstream endoplasmic reticulum stress, whereas a second class can inhibit the RNase through the same ATP-binding site, even under endoplasmic reticulum stress. Thus, alternative kinase conformations stabilized by distinct classes of ATP-competitive inhibitors can cause allosteric switching of IRE1α's RNase--either on or off. As dysregulation of the UPR has been implicated in a variety of cell degenerative and neoplastic disorders, small-molecule control over IRE1α should advance efforts to understand the UPR's role in pathophysiology and to develop drugs for endoplasmic reticulum stress-related diseases.


Asunto(s)
Endorribonucleasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Catálisis , Células Cultivadas , Reactivos de Enlaces Cruzados , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Conformación Molecular , Mutación/genética , Mutación/fisiología , Fosforilación , Empalme del ARN/efectos de los fármacos , Factores de Transcripción del Factor Regulador X , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteína 1 de Unión a la X-Box
4.
bioRxiv ; 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-37461469

RESUMEN

Purpose: Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Non-specific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small molecule preclinical candidate, BKIDC-1553, with antiglycolytic activity. Experimental design: We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell cycle, metabolic and enzymatic assays were used to demonstrate their mechanism of action. A human PDX model implanted in mice and a human organoid were studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. Results: We demonstrate a new class of small molecule inhibitors where antiglycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. Conclusion: This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer.

5.
Mol Cancer Ther ; 23(7): 973-994, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38507737

RESUMEN

Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Nonspecific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small-molecule preclinical candidate, BKIDC-1553, with antiglycolytic activity. We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell-cycle, metabolic, and enzymatic assays were used to demonstrate their mechanism of action. A human patient-derived xenograft model implanted in mice and a human organoid were studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. We demonstrate a new class of small-molecule inhibitors where antiglycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer.


Asunto(s)
Proliferación Celular , Glucólisis , Ensayos Antitumor por Modelo de Xenoinjerto , Masculino , Humanos , Animales , Ratones , Glucólisis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
6.
Chembiochem ; 14(2): 209-16, 2013 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-23319368

RESUMEN

Protein kinases are essential enzymes for cellular signaling, and are often regulated by participation in protein complexes. The mitogen-activated protein kinase (MAPK) p38 is involved in multiple pathways, and its regulation depends on its interactions with other signaling proteins. However, the identification of p38-interacting proteins is challenging. For this reason, we have developed label transfer reagents (LTRs) that allow labeling of p38 signaling complexes. These LTRs leverage the potency and selectivity of known p38 inhibitors to place a photo-crosslinker and tag in the vicinity of p38 and its binding partners. Upon UV irradiation, proteins that are in close proximity to p38 are covalently crosslinked, and labeled proteins are detected and/or purified with an orthogonal chemical handle. Here we demonstrate that p38-selective LTRs selectively label a diversity of p38 binding partners, including substrates, activators, and inactivators. Furthermore, these LTRs can be used in immunoprecipitations to provide low-resolution structural information on p38-containing complexes.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Humanos , Indicadores y Reactivos , Modelos Moleculares , Transducción de Señal , Coloración y Etiquetado , Rayos Ultravioleta , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/aislamiento & purificación
7.
ACS Chem Biol ; 18(2): 431-440, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36724382

RESUMEN

Cyclin-dependent kinases (CDKs) are key mediators of cell proliferation and have been a subject of oncology drug discovery efforts for over two decades. Several CDK and activator cyclin family members have been implicated in regulating the cell division cycle. While it is thought that there are canonical CDK-cyclin pairing preferences, the extent of selectivity is unclear, and increasing evidence suggests that the cell-cycle CDKs can be activated by a pool of available cyclins. The molecular details of CDK-cyclin specificity are not completely understood despite their importance for understanding cancer cell cycles and for pharmacological inhibition of cancer proliferation. We report here a biolayer interferometry assay that allows for facile quantification of CDK binding interactions with their cyclin activators. We applied this assay to measure the impact of Cdk2 inhibitors on Cyclin A (CycA) association and dissociation kinetics. We found that Type I inhibitors increase the affinity between Cdk2 and CycA by virtue of a slowed cyclin dissociation rate. In contrast, Type II inhibitors and other small-molecule Cdk2 binders have distinct effects on the CycA association and dissociation processes to decrease affinity. We propose that the differential impact of small molecules on the cyclin binding kinetics arises from the plasticity of the Cdk2 active site as the kinase transitions between active, intermediate, and inactive states.


Asunto(s)
Quinasas CDC2-CDC28 , Quinasas Ciclina-Dependientes , Quinasas Ciclina-Dependientes/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas CDC2-CDC28/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Ciclinas/metabolismo , Fosforilación , Quinasa 4 Dependiente de la Ciclina/metabolismo
8.
J Am Chem Soc ; 134(46): 19017-25, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23088519

RESUMEN

Protein kinases are key components of most mammalian signal transduction networks and are therapeutically relevant drug targets. Efforts to study protein kinase function would benefit from new technologies that are able to profile kinases in complex proteomes. Here, we describe active site-directed probes for profiling kinases in whole cell extracts and live cells. These probes contain general ligands that stabilize a specific inactive conformation of the ATP-binding sites of protein kinases, as well as trifluoromethylphenyl diazirine and alkyne moieties that allow covalent modification and enrichment of kinases, respectively. A diverse group of serine/threonine and tyrosine kinases were identified as specific targets of these probes in whole cell extracts. In addition, a number of kinase targets were selectively labeled in live cells. Our chemical proteomics approach should be valuable for interrogating protein kinase active sites in physiologically relevant environments.


Asunto(s)
Etiquetas de Fotoafinidad , Inhibidores de Proteínas Quinasas/química , Adenosina Trifosfato/química , Modelos Moleculares , Proteómica
9.
J Am Chem Soc ; 131(19): 6686-8, 2009 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-19391594

RESUMEN

We report a new chemical genetic method for creating bivalent ligands of protein kinases. The kinase inhibitors that are generated with this methodology consist of two components: (1) a synthetic, small molecule that targets the ATP-binding cleft and (2) a peptidic ligand that enhances selectivity between kinases by targeting a secondary binding domain. A key feature of these bivalent inhibitors is that they are assembled on a protein scaffold with a chemoselective protein labeling technique. The utility of this methodology is demonstrated through the generation of a panel of protein-small molecule conjugates that simultaneously target the SH1 and SH3 domains of the closely related tyrosine kinases Src and Abl. The assembled bivalent ligands are significantly more potent inhibitors of Src and Abl than either modular component alone. Importantly, these protein-small molecule conjugates show a high degree of selectivity for their intended kinase target.


Asunto(s)
Guanina/análogos & derivados , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/química , Proteínas/genética , Dominios Homologos src , Secuencia de Aminoácidos , Sitios de Unión , Guanina/química , Ligandos , Proteínas/química
10.
ACS Chem Biol ; 8(4): 691-9, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23305300

RESUMEN

Bioorthogonal ligation methods that allow the selective conjugation of fluorophores or biotin to proteins and small molecule probes that contain inert chemical handles are an important component of many chemical proteomic strategies. Here, we present a new catch-and-release enrichment strategy that utilizes a hexylchloride group as a bioorthogonal chemical handle. Proteins and small molecules that contain a hexylchloride tag can be efficiently captured by an immobilized version of the self-labeling protein HaloTag. Furthermore, by using a HaloTag fusion protein that contains a protease cleavage site, captured proteins can be selectively eluted under mild conditions. We demonstrate the utility of the hexylchloride-based catch-and-release strategy by enriching protein kinases that are covalently and noncovalently bound to ATP-binding site-directed probes from mammalian cell lysates. Our catch-and-release system creates new possibilities for profiling enzyme families and for the identification of the cellular targets of bioactive small molecules.


Asunto(s)
Cloruros/química , Proteómica , Células HeLa , Humanos
11.
ACS Chem Biol ; 8(12): 2734-43, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24106839

RESUMEN

Over the past decade, an increasingly diverse array of potent and selective inhibitors that target the ATP-binding sites of protein kinases have been developed. Many of these inhibitors, like the clinically approved drug imatinib (Gleevec), stabilize a specific catalytically inactive ATP-binding site conformation of their kinases targets. Imatinib is notable in that it is highly selective for its kinase target, Abl, over other closely related tyrosine kinases, such as Src. In addition, imatinib is highly sensitive to the phosphorylation state of Abl's activation loop, which is believed to be a general characteristic of all inhibitors that stabilize a similar inactive ATP-binding site conformation. In this report, we perform a systematic analysis of a diverse series of ATP-competitive inhibitors that stabilize a similar inactive ATP-binding site conformation as imatinib with the tyrosine kinases Src and Abl. In contrast to imatinib, many of these inhibitors have very similar potencies against Src and Abl. Furthermore, only a subset of this class of inhibitors is sensitive to the phosphorylation state of the activation loop of these kinases. In attempting to explain this observation, we have uncovered an unexpected correlation between Abl's activation loop and another flexible active site feature, called the phosphate-binding loop (p-loop). These studies shed light on how imatinib is able to obtain its high target selectivity and reveal how the conformational preference of flexible active site regions can vary between closely related kinases.


Asunto(s)
Antineoplásicos/química , Benzamidas/química , Piperazinas/química , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-abl/química , Pirimidinas/química , Familia-src Quinasas/química , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Sitios de Unión , Activación Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Mesilato de Imatinib , Cinética , Ligandos , Simulación del Acoplamiento Molecular , Mutación , Fosfatos/química , Fosfatos/metabolismo , Fosforilación , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
12.
ACS Chem Biol ; 7(3): 487-95, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22148755

RESUMEN

The identification of potent and selective modulators of protein kinase function remains a challenge, and new strategies are needed for generating these useful ligands. Here, we describe the generation of bivalent inhibitors of three unrelated protein kinases: the CAMK family kinase Pim1, the mitogen-activated protein kinase (MAPK) p38α, and the receptor tyrosine kinase (RTK) epidermal growth factor receptor (EGFR). These bivalent inhibitors consist of an ATP-competitive inhibitor that is covalently tethered to an engineered form of the self-labeling protein O(6)-alkylguanine-DNA alkyltransferase (SNAP-tag). In each example, SNAP-tag is fused to a peptide ligand that binds to a signaling interaction site of the kinase being targeted. These interactions increase the overall selectivity and potency of the bivalent inhibitors that were generated. The ability to exploit disparate binding sites in diverse kinases points to the generality of the method described. Finally, we demonstrate that ATP-competitive inhibitors that are conjugated to the bio-orthogonal tag O(4)-benzyl-2-chloro-6-aminopyrimidine (CLP) are cell-permeable. The selective labeling of SNAP-tag with CLP conjugates allows the rapid assembly of bivalent inhibitors in living cells.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Adenosina Trifosfato/química , Animales , Sitios de Unión/efectos de los fármacos , Células COS , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas/química , Relación Estructura-Actividad
13.
J Med Chem ; 55(5): 2416-26, 2012 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-22320388

RESUMEN

Toxoplasmosis is a disease of prominent health concern that is caused by the protozoan parasite Toxoplasma gondii. Proliferation of T. gondii is dependent on its ability to invade host cells, which is mediated in part by calcium-dependent protein kinase 1 (CDPK1). We have developed ATP competitive inhibitors of TgCDPK1 that block invasion of parasites into host cells, preventing their proliferation. The presence of a unique glycine gatekeeper residue in TgCDPK1 permits selective inhibition of the parasite enzyme over human kinases. These potent TgCDPK1 inhibitors do not inhibit the growth of human cell lines and represent promising candidates as toxoplasmosis therapeutics.


Asunto(s)
Coccidiostáticos/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/metabolismo , Proteínas Protozoarias/antagonistas & inhibidores , Pirazoles/síntesis química , Pirimidinas/síntesis química , Toxoplasma/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Coccidiostáticos/química , Coccidiostáticos/farmacología , Cristalografía por Rayos X , Resistencia a Medicamentos , Pruebas de Enzimas , Humanos , Modelos Moleculares , Estructura Molecular , Naftalenos/síntesis química , Naftalenos/química , Naftalenos/farmacología , Piperidinas/síntesis química , Piperidinas/química , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Protozoarias/metabolismo , Pirazoles/química , Pirazoles/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Relación Estructura-Actividad , Toxoplasma/enzimología
14.
J Med Chem ; 55(6): 2803-10, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22369268

RESUMEN

Diseases caused by the apicomplexan protozoans Toxoplasma gondii and Cryptosporidium parvum are a major health concern. The life cycle of these parasites is regulated by a family of calcium-dependent protein kinases (CDPKs) that have no direct homologues in the human host. Fortuitously, CDPK1 from both parasites contains a rare glycine gatekeeper residue adjacent to the ATP-binding pocket. This has allowed creation of a series of C3-substituted pyrazolopyrimidine compounds that are potent inhibitors selective for CDPK1 over a panel of human kinases. Here we demonstrate that selectivity is further enhanced by modification of the scaffold at the C1 position. The explanation for this unexpected result is provided by crystal structures of the inhibitors bound to CDPK1 and the human kinase c-SRC. Furthermore, the insight gained from these studies was applied to transform an alternative ATP-competitive scaffold lacking potency and selectivity for CDPK1 into a low nanomolar inhibitor of this enzyme with no activity against SRC.


Asunto(s)
Antiprotozoarios/síntesis química , Bencimidazoles/síntesis química , Calcio/fisiología , Cryptosporidium parvum/enzimología , Modelos Moleculares , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/química , Proteínas Protozoarias/antagonistas & inhibidores , Toxoplasma/enzimología , Antiprotozoarios/química , Bencimidazoles/química , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Protozoarias/química , Relación Estructura-Actividad
15.
Mol Biosyst ; 7(2): 447-56, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21060940

RESUMEN

We recently reported a chemical genetic method for generating bivalent inhibitors of protein kinases. This method relies on the use of the DNA repair enzyme O(6)-alkylguanine-DNA alkyltransferase (AGT) to display an ATP-competitive inhibitor and a ligand that targets a secondary binding domain. With this method potent and selective inhibitors of the tyrosine kinases SRC and ABL were identified. Here, we dissect the molecular determinants of the potency and selectivity of these bivalent ligands. Systematic analysis of ATP-competitive inhibitors with varying linker lengths revealed that SRC and ABL have differential sensitivities to ligand presentation. Generation of bivalent constructs that contain ligands with differential affinities for the ATP-binding sites and SH3 domains of SRC and ABL demonstrated the modular nature of inhibitors based on the AGT scaffold. Furthermore, these studies revealed that the interaction between the SH3 domain ligand and the kinase SH3 domain is the major selectivity determinant amongst closely-related tyrosine kinases. Finally, the potency of bivalent inhibitors against distinct phospho-isoforms of SRC was determined. Overall, these results provide insight into how individual ligands can be modified to provide more potent and selective bivalent inhibitors of protein kinases.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Unión Competitiva , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Fosforilación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Dominios Homologos src , Familia-src Quinasas/química
16.
ACS Med Chem Lett ; 1(7): 331-335, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21116453

RESUMEN

The protozoans Cryptosporidium parvum and Toxoplasma gondii are parasites of major health concern to humans. Both parasites contain a group of calcium-dependent protein kinases (CDPKs), which are found in plants and ciliates but not in humans or fungi. Here we describe a series of potent inhibitors that target CDPK1 in C. parvum (CpCDPK1) and T. gondii (TgCDPK1). These inhibitors are highly selective for CpCDPK1 and TgCDPK1 over the mammalian kinases SRC and ABL. Furthermore, they are able to block an early stage of C. parvum invasion of HCT-8 host cells, which is similar to their effects on T. gondii invasion of human fibroblasts.

17.
Mol Biosyst ; 4(6): 542-50, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18493651

RESUMEN

Immobilized kinase inhibitors have emerged as powerful reagents for the determination of kinase inhibitor selectivity and for the enrichment of protein targets from cellular lysates. Here, we report the design and synthesis of a set of "clickable" 4-anilinoquinazoline kinase inhibitors. We demonstrate that the attachment of a flexible tether that contains a bio-orthogonal azide functionality does not adversely affect the potency or selectivity of these inhibitors. Furthermore, we demonstrate the utility of these inhibitors through the generation of an affinity matrix for the enrichment of interacting proteins from cellular lysates.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinazolinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Familia-src Quinasas/antagonistas & inhibidores , Aurora Quinasas , Sitios de Unión , Ciclización , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Quinazolinas/síntesis química , Quinazolinas/química , Estereoisomerismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda