Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cell ; 184(4): 983-999.e24, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33606986

RESUMEN

Interleukin-12 (IL-12) and IL-23 are heterodimeric cytokines that are produced by antigen-presenting cells to regulate the activation and differentiation of lymphocytes, and they share IL-12Rß1 as a receptor signaling subunit. We present a crystal structure of the quaternary IL-23 (IL-23p19/p40)/IL-23R/IL-12Rß1 complex, together with cryoelectron microscopy (cryo-EM) maps of the complete IL-12 (IL-12p35/p40)/IL-12Rß2/IL-12Rß1 and IL-23 receptor (IL-23R) complexes, which reveal "non-canonical" topologies where IL-12Rß1 directly engages the common p40 subunit. We targeted the shared IL-12Rß1/p40 interface to design a panel of IL-12 partial agonists that preserved interferon gamma (IFNγ) induction by CD8+ T cells but impaired cytokine production from natural killer (NK) cells in vitro. These cell-biased properties were recapitulated in vivo, where IL-12 partial agonists elicited anti-tumor immunity to MC-38 murine adenocarcinoma absent the NK-cell-mediated toxicity seen with wild-type IL-12. Thus, the structural mechanism of receptor sharing used by IL-12 family cytokines provides a protein interface blueprint for tuning this cytokine axis for therapeutics.


Asunto(s)
Interleucina-12/química , Interleucina-12/metabolismo , Células Asesinas Naturales/metabolismo , Receptores de Interleucina/química , Receptores de Interleucina/metabolismo , Linfocitos T/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Microscopía por Crioelectrón , Cristalografía por Rayos X , Epítopos/inmunología , Femenino , Células HEK293 , Humanos , Inmunidad , Interleucina-12/agonistas , Subunidad p40 de la Interleucina-12/química , Subunidad p40 de la Interleucina-12/metabolismo , Ratones Endogámicos C57BL , Modelos Moleculares , Neoplasias/inmunología , Neoplasias/patología , Estructura Cuaternaria de Proteína , Receptores de Interleucina/ultraestructura , Receptores de Interleucina-12/metabolismo , Transducción de Señal , Relación Estructura-Actividad
2.
Bioorg Med Chem Lett ; 29(12): 1497-1501, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31000154

RESUMEN

Receptor-interacting protein kinase 1 (RIPK1), a key component of the cellular necroptosis pathway, has gained recognition as an important therapeutic target. Pharmacologic inhibition or genetic inactivation of RIPK1 has shown promise in animal models of disease ranging from acute ischemic conditions, chronic inflammation, and neurodegeneration. We present here a class of RIPK1 inhibitors that is distinguished by a lack of a lipophilic aromatic group present in most literature inhibitors that typically occupies a hydrophobic back pocket of the protein active site. Despite not having this ubiquitous feature of many known RIPK1 inhibitors, we were able to obtain compounds with good potency, kinase selectivity, and pharmacokinetic properties in rats. The use of the lipophilic yet metabolically stable pentafluoroethyl group was critical to balancing the potency and properties of optimized analogs.


Asunto(s)
Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Humanos , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Relación Estructura-Actividad
3.
Proc Natl Acad Sci U S A ; 111(22): 8025-30, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24843152

RESUMEN

Janus kinases (JAKs) are receptor-associated multidomain tyrosine kinases that act downstream of many cytokines and interferons. JAK kinase activity is regulated by the adjacent pseudokinase domain via an unknown mechanism. Here, we report the 2.8-Å structure of the two-domain pseudokinase-kinase module from the JAK family member TYK2 in its autoinhibited form. We find that the pseudokinase and kinase interact near the kinase active site and that most reported mutations in cancer-associated JAK alleles cluster in or near this interface. Mutation of residues near the TYK2 interface that are analogous to those in cancer-associated JAK alleles, including the V617F and "exon 12" JAK2 mutations, results in increased kinase activity in vitro. These data indicate that JAK pseudokinases are autoinhibitory domains that hold the kinase domain inactive until receptor dimerization stimulates transition to an active state.


Asunto(s)
Janus Quinasa 1/metabolismo , Janus Quinasa 2/metabolismo , Janus Quinasa 3/metabolismo , Neoplasias/enzimología , Transducción de Señal/fisiología , TYK2 Quinasa/metabolismo , Animales , Línea Celular , Cristalografía por Rayos X , Dimerización , Activación Enzimática/fisiología , Humanos , Insectos/citología , Janus Quinasa 1/genética , Janus Quinasa 2/genética , Janus Quinasa 3/genética , Mutación , Neoplasias/genética , Estructura Terciaria de Proteína , Relación Estructura-Actividad , TYK2 Quinasa/química , TYK2 Quinasa/genética
4.
Mol Cell ; 31(5): 737-48, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18775332

RESUMEN

gp130 is a shared receptor for at least nine cytokines and can signal either as a homodimer or as a heterodimer with Leukemia Inhibitory Factor Receptor (LIF-R). Here, we biophysically and structurally characterize the full-length, transmembrane form of a quaternary cytokine receptor complex consisting of gp130, LIF-R, the cytokine Ciliary Neurotrophic Factor (CNTF), and its alpha receptor (CNTF-Ralpha). Thermodynamic analysis indicates that, unlike the cooperative assembly of the symmetric gp130/Interleukin-6/IL-6Ralpha hexameric complex, CNTF/CNTF-Ralpha heterodimerizes gp130 and LIF-R via noncooperative energetics to form an asymmetric 1:1:1:1 complex. Single particle electron microscopic analysis of the full-length gp130/LIF-R/CNTF-Ralpha/CNTF quaternary complex elucidates an asymmetric structural arrangement, in which the receptor extracellular and transmembrane segments join as a continuous, rigid unit, poised to sensitively transduce ligand engagement to the membrane-proximal intracellular signaling regions. These studies also enumerate the organizing principles for assembly of the "tall" class of gp130 family cytokine receptor complexes including LIF, IL-27, IL-12, and others.


Asunto(s)
Receptor gp130 de Citocinas/química , Complejos Multiproteicos/química , Estructura Cuaternaria de Proteína , Receptores OSM-LIF/química , Transducción de Señal/fisiología , Animales , Factor Neurotrófico Ciliar/química , Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/metabolismo , Cristalografía por Rayos X , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos/ultraestructura , Receptor de Factor Neurotrófico Ciliar/química , Receptor de Factor Neurotrófico Ciliar/genética , Receptor de Factor Neurotrófico Ciliar/metabolismo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Termodinámica
5.
J Biol Chem ; 288(37): 26926-43, 2013 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-23897821

RESUMEN

Histone deacetylases (HDACs) are critical in the control of gene expression, and dysregulation of their activity has been implicated in a broad range of diseases, including cancer, cardiovascular, and neurological diseases. HDAC inhibitors (HDACi) employing different zinc chelating functionalities such as hydroxamic acids and benzamides have shown promising results in cancer therapy. Although it has also been suggested that HDACi with increased isozyme selectivity and potency may broaden their clinical utility and minimize side effects, the translation of this idea to the clinic remains to be investigated. Moreover, a detailed understanding of how HDACi with different pharmacological properties affect biological functions in vitro and in vivo is still missing. Here, we show that a panel of benzamide-containing HDACi are slow tight-binding inhibitors with long residence times unlike the hydroxamate-containing HDACi vorinostat and trichostatin-A. Characterization of changes in H2BK5 and H4K14 acetylation following HDACi treatment in the neuroblastoma cell line SH-SY5Y revealed that the timing and magnitude of histone acetylation mirrored both the association and dissociation kinetic rates of the inhibitors. In contrast, cell viability and microarray gene expression analysis indicated that cell death induction and changes in transcriptional regulation do not correlate with the dissociation kinetic rates of the HDACi. Therefore, our study suggests that determining how the selective and kinetic inhibition properties of HDACi affect cell function will help to evaluate their therapeutic utility.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/química , Histonas/química , Acetilación , Benzamidas/química , Unión Competitiva , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/química , Concentración 50 Inhibidora , Cinética , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Piridinas/química , Transcripción Genética , Vorinostat
6.
Bioorg Med Chem Lett ; 23(21): 5923-30, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24042009

RESUMEN

A highly ligand efficient, novel 8-oxo-pyridopyrimidine containing inhibitor of Jak1 and Jak2 isoforms with a pyridone moiety as the hinge-binding motif was discovered. Structure-based design strategies were applied to significantly improve enzyme potency and the polarity of the molecule was adjusted to gain cellular activity. The crystal structures of two representative inhibitors bound to Jak1 were obtained to enable SAR exploration.


Asunto(s)
Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Humanos , Janus Quinasa 1/química , Janus Quinasa 1/metabolismo , Janus Quinasa 2/química , Janus Quinasa 2/metabolismo , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad
7.
Bioorg Med Chem Lett ; 23(12): 3592-8, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23642482

RESUMEN

The identification of a novel fused triazolo-pyrrolopyridine scaffold, optimized derivatives of which display nanomolar inhibition of Janus kinase 1, is described. Prototypical example 3 demonstrated lower cell potency shift, better permeability in cells and higher oral exposure in rat than the corresponding, previously reported, imidazo-pyrrolopyridine analogue 2. Examples 6, 7 and 18 were subsequently identified from an optimization campaign and demonstrated modest selectivity over JAK2, moderate to good oral bioavailability in rat with overall pharmacokinetic profiles comparable to that reported for an approved pan-JAK inhibitor (tofacitinib).


Asunto(s)
Janus Quinasa 1/antagonistas & inhibidores , Piridinas/farmacología , Animales , Cristalografía por Rayos X , Janus Quinasa 1/química , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/química , Cinética , Modelos Moleculares , Piridinas/química , Pirroles/química , Pirroles/farmacología , Ratas
8.
Bioorg Med Chem Lett ; 22(24): 7627-33, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23107482

RESUMEN

Herein we describe our successful efforts in obtaining C-2 substituted imidazo-pyrrolopyridines with improved JAK1 selectivity relative to JAK2 by targeting an amino acid residue that differs between the two isoforms (JAK1: E966; JAK2: D939). Efforts to improve cellular potency by reducing the polarity of the inhibitors are also detailed. The X-ray crystal structure of a representative inhibitor in complex with the JAK1 enzyme is also disclosed.


Asunto(s)
Descubrimiento de Drogas , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirroles/farmacología , Animales , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Imidazoles/química , Janus Quinasa 1/metabolismo , Janus Quinasa 2/metabolismo , Masculino , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Piridinas/administración & dosificación , Piridinas/química , Pirroles/administración & dosificación , Pirroles/química , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
9.
Science ; 376(6589): 163-169, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35271300

RESUMEN

Cytokines signal through cell surface receptor dimers to initiate activation of intracellular Janus kinases (JAKs). We report the 3.6-angstrom-resolution cryo-electron microscopy structure of full-length JAK1 complexed with a cytokine receptor intracellular domain Box1 and Box2 regions captured as an activated homodimer bearing the valine→phenylalanine (VF) mutation prevalent in myeloproliferative neoplasms. The seven domains of JAK1 form an extended structural unit, the dimerization of which is mediated by close-packing of the pseudokinase (PK) domains from the monomeric subunits. The oncogenic VF mutation lies within the core of the JAK1 PK interdimer interface, enhancing packing complementarity to facilitate ligand-independent activation. The carboxy-terminal tyrosine kinase domains are poised for transactivation and to phosphorylate the receptor STAT (signal transducer and activator of transcription)-recruiting motifs projecting from the overhanging FERM (four-point-one, ezrin, radixin, moesin)-SH2 (Src homology 2)-domains. Mapping of constitutively active JAK mutants supports a two-step allosteric activation mechanism and reveals opportunities for selective therapeutic targeting of oncogenic JAK signaling.


Asunto(s)
Janus Quinasa 1 , Receptores de Citocinas , Dominios Homologos src , Regulación Alostérica , Microscopía por Crioelectrón , Activación Enzimática , Humanos , Janus Quinasa 1/química , Janus Quinasa 1/metabolismo , Mutación , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/genética , Fosforilación , Multimerización de Proteína , Receptores de Citocinas/química , Factores de Transcripción STAT/metabolismo
10.
Nat Chem Biol ; 5(7): 469-78, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19465933

RESUMEN

MARTX toxins modulate the virulence of a number of Gram-negative Vibrio species. This family of toxins is defined by the presence of a cysteine protease domain (CPD), which proteolytically activates the Vibrio cholerae MARTX toxin. Although recent structural studies of the CPD have uncovered a new allosteric activation mechanism, the mechanism of CPD substrate recognition or toxin processing is unknown. Here we show that interdomain cleavage of MARTXVc enhances effector domain function. We also identify the first small-molecule inhibitors of this protease domain and present the 2.35-A structure of the CPD bound to one of these inhibitors. This structure, coupled with biochemical and mutational studies of the toxin, reveals the molecular basis of CPD substrate specificity and underscores the evolutionary relationship between the CPD and the clan CD caspase proteases. These studies are likely to prove valuable for devising new antitoxin strategies for a number of bacterial pathogens.


Asunto(s)
Toxina del Cólera/química , Toxina del Cólera/metabolismo , Proteasas de Cisteína/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Vibrio cholerae/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Western Blotting , Dominio Catalítico , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Secundaria de Proteína , Alineación de Secuencia , Espectroscopía Infrarroja por Transformada de Fourier , Especificidad por Sustrato , Vibrio cholerae/efectos de los fármacos , Vibrio cholerae/enzimología
11.
Sci Transl Med ; 13(625): eabg7565, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34936383

RESUMEN

Chimeric antigen receptor (CAR) T cells induce durable responses in patients with refractory hematological tumors. However, low CAR T cell activity, poor engraftment, or short in-patient persistence can lead to tumor progression or relapse. Furthermore, excessive CAR T cell expansion and activation can result in life-threatening cytokine release syndrome (CRS). Thus, in-patient control of the CAR T cell population is essential. Interleukin-2 (IL-2) is a critical cytokine for T cell proliferation and effector function, but its clinical use is limited by immune-mediated toxicity. Here, we report on an orthogonal IL-2 receptor and ligand system that enables specific in vivo control of CAR T cell expansion and activation, wherein an orthogonal human IL-2 (STK-009) selectively pairs with an orthogonal human IL-2Rß (hoRb) expressed on CAR T cells. STK-009 expands hoRb-expressing CAR T cells in the presence and absence of tumor antigen and maintains the presence of stem cell memory T cells (TSCM) and effector T cells. In preclinical models of human CAR-refractory lymphoma, STK-009 treatment resulted in systemic and intratumoral expansion and activation of hoRb-expressing anti­CD19-CD28ζ CAR T cells (SYNCAR). The orthogonal IL-2 receptor/ligand system delivers complete responses in large subcutaneous lymphomas, even with substantially reduced CAR T cell doses, by selectively expanding and activating CAR T cells in vivo. STK-009 withdrawal allowed normal CAR T cell contraction, thereby limiting CRS induced by tumor antigen­specific T cell activation. These data suggest that the orthogonal IL-2 receptor/ligand system provides the in vivo control necessary to maximize efficacy of CAR T therapies.


Asunto(s)
Interleucina-2 , Linfoma , Antígenos CD19 , Humanos , Inmunoterapia Adoptiva , Linfoma/terapia , Recurrencia Local de Neoplasia/terapia , Receptores de Antígenos de Linfocitos T , Linfocitos T
12.
ACS Med Chem Lett ; 11(3): 327-333, 2020 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-32184965

RESUMEN

IRAK4 kinase activity transduces signaling from multiple IL-1Rs and TLRs to regulate cytokines and chemokines implicated in inflammatory diseases. As such, there is high interest in identifying selective IRAK4 inhibitors for the treatment of these disorders. We previously reported the discovery of potent and selective dihydrobenzofuran inhibitors of IRAK4. Subsequent studies, however, showed inconsistent inhibition in disease-relevant pharmacodynamic models. Herein, we describe application of a human whole blood assay to the discovery of a series of benzolactam IRAK4 inhibitors. We identified potent molecule 19 that achieves robust in vivo inhibition of cytokines relevant to human disease.

13.
Mol Cell Biol ; 26(2): 425-37, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16382135

RESUMEN

Fanconi anemia (FA) is a multigene cancer susceptibility disorder characterized by cellular hypersensitivity to DNA interstrand cross-linking agents such as mitomycin C (MMC). FA proteins are suspected to function at the interface between cell cycle checkpoints, DNA repair, and DNA replication. Using replicating extracts from Xenopus eggs, we developed cell-free assays for FA proteins (xFA). Recruitment of the xFA core complex and xFANCD2 to chromatin is strictly dependent on replication initiation, even in the presence of MMC indicating specific recruitment to DNA lesions encountered by the replication machinery. The increase in xFA chromatin binding following treatment with MMC is part of a caffeine-sensitive S-phase checkpoint that is controlled by xATR. Recruitment of xFANCD2, but not xFANCA, is dependent on the xATR-xATR-interacting protein (xATRIP) complex. Immunodepletion of either xFANCA or xFANCD2 from egg extracts results in accumulation of chromosomal DNA breaks during replicative synthesis. Our results suggest coordinated chromatin recruitment of xFA proteins in response to replication-associated DNA lesions and indicate that xFA proteins function to prevent the accumulation of DNA breaks that arise during unperturbed replication.


Asunto(s)
Proteínas Portadoras/metabolismo , Daño del ADN/fisiología , Replicación del ADN , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Proteínas de Xenopus/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Cafeína/farmacología , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Reactivos de Enlaces Cruzados/farmacología , Reparación del ADN/fisiología , Femenino , Técnicas In Vitro , Mitomicina/farmacología , Datos de Secuencia Molecular , Oocitos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S/efectos de los fármacos , Fase S/fisiología , Homología de Secuencia de Aminoácido , Xenopus laevis
14.
Mol Biol Cell ; 17(4): 1559-69, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16436514

RESUMEN

The DNA damage checkpoint pathways sense and respond to DNA damage to ensure genomic stability. The ATR kinase is a central regulator of one such pathway and phosphorylates a number of proteins that have roles in cell cycle progression and DNA repair. Using the Xenopus egg extract system, we have investigated regulation of the Rad1/Hus1/Rad9 complex. We show here that phosphorylation of Rad1 and Hus1 occurs in an ATR- and TopBP1-dependent manner on T5 of Rad1 and S219 and T223 of Hus1. Mutation of these sites has no effect on the phosphorylation of Chk1 by ATR. Interestingly, phosphorylation of Rad1 is independent of Claspin and the Rad9 carboxy terminus, both of which are required for Chk1 phosphorylation. These data suggest that an active ATR signaling complex exists in the absence of the carboxy terminus of Rad9 and that this carboxy-terminal domain may be a specific requirement for Chk1 phosphorylation and not necessary for all ATR-mediated signaling events. Thus, Rad1 phosphorylation provides an alternate and early readout for the study of ATR activation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Proteínas Cromosómicas no Histona/genética , Activación Enzimática , Datos de Secuencia Molecular , Fosforilación , Proteínas Quinasas/metabolismo , Xenopus , Proteínas de Xenopus/genética
15.
J Med Chem ; 62(13): 6223-6240, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31082230

RESUMEN

A series of pyrazolopyrimidine inhibitors of IRAK4 were developed from a high-throughput screen (HTS). Modification of an HTS hit led to a series of bicyclic heterocycles with improved potency and kinase selectivity but lacking sufficient solubility to progress in vivo. Structure-based drug design, informed by cocrystal structures with the protein and small-molecule crystal structures, yielded a series of dihydrobenzofurans. This semisaturated bicycle provided superior druglike properties while maintaining excellent potency and selectivity. Improved physicochemical properties allowed for progression into in vivo experiments, where lead molecules exhibited low clearance and showed target-based inhibition of IRAK4 signaling in an inflammation-mediated PK/PD mouse model.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Aminoquinolinas/síntesis química , Aminoquinolinas/metabolismo , Aminoquinolinas/farmacología , Animales , Benzofuranos/síntesis química , Benzofuranos/metabolismo , Benzofuranos/farmacología , Dominio Catalítico , Femenino , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Ratones Endogámicos C57BL , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Pirazoles/síntesis química , Pirazoles/metabolismo , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
16.
Elife ; 72018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30044226

RESUMEN

Cytokines and interferons initiate intracellular signaling via receptor dimerization and activation of Janus kinases (JAKs). How JAKs structurally respond to changes in receptor conformation induced by ligand binding is not known. Here, we present two crystal structures of the human JAK2 FERM and SH2 domains bound to Leptin receptor (LEPR) and Erythropoietin receptor (EPOR), which identify a novel dimeric conformation for JAK2. This 2:2 JAK2/receptor dimer, observed in both structures, identifies a previously uncharacterized receptor interaction essential to dimer formation that is mediated by a membrane-proximal peptide motif called the 'switch' region. Mutation of the receptor switch region disrupts STAT phosphorylation but does not affect JAK2 binding, indicating that receptor-mediated formation of the JAK2 FERM dimer is required for kinase activation. These data uncover the structural and molecular basis for how a cytokine-bound active receptor dimer brings together two JAK2 molecules to stimulate JAK2 kinase activity.


Asunto(s)
Janus Quinasa 2/química , Fragmentos de Péptidos/química , Conformación Proteica , Receptores de Eritropoyetina/química , Receptores de Leptina/química , Cristalografía por Rayos X , Dimerización , Dominios FERM/genética , Humanos , Janus Quinasa 2/genética , Mutación , Fragmentos de Péptidos/genética , Fosforilación/genética , Unión Proteica/genética , Receptores de Eritropoyetina/genética , Receptores de Leptina/genética , Factores de Transcripción STAT/química , Factores de Transcripción STAT/genética , Transducción de Señal/genética , Dominios Homologos src/genética
17.
MAbs ; 10(7): 979-991, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30102105

RESUMEN

Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are a growing health threat worldwide. Efforts to identify novel antibodies that target S. aureus cell surface antigens are a promising direction in the development of antibiotics that can halt MRSA infection. We biochemically and structurally characterized three patient-derived MRSA-targeting antibodies that bind to wall teichoic acid (WTA), which is a polyanionic surface glycopolymer. In S. aureus, WTA exists in both α- and ß-forms, based on the stereochemistry of attachment of a N-acetylglucosamine residue to the repeating phosphoribitol sugar unit. We identified a panel of antibodies cloned from human patients that specifically recognize the α or ß form of WTA, and can bind with high affinity to pathogenic wild-type strains of S. aureus bacteria. To investigate how the ß-WTA specific antibodies interact with their target epitope, we determined the X-ray crystal structures of the three ß-WTA specific antibodies, 4462, 4497, and 6078 (Protein Data Bank IDs 6DWI, 6DWA, and 6DW2, respectively), bound to a synthetic WTA epitope. These structures reveal that all three of these antibodies, while utilizing distinct antibody complementarity-determining region sequences and conformations to interact with ß-WTA, fulfill two recognition principles: binding to the ß-GlcNAc pyranose core and triangulation of WTA phosphate residues with polar contacts. These studies reveal the molecular basis for targeting a unique S. aureus cell surface epitope and highlight the power of human patient-based antibody discovery techniques for finding novel pathogen-targeting therapeutics.


Asunto(s)
Antibacterianos/química , Anticuerpos Antibacterianos/química , Complejo Antígeno-Anticuerpo/metabolismo , Pared Celular/química , Regiones Determinantes de Complementariedad/química , Staphylococcus aureus Resistente a Meticilina/fisiología , Infecciones Estafilocócicas/inmunología , Ácidos Teicoicos/química , Antibacterianos/sangre , Anticuerpos Antibacterianos/sangre , Complejo Antígeno-Anticuerpo/genética , Pared Celular/metabolismo , Cristalografía por Rayos X/métodos , Humanos , Inmunidad Humoral , Unión Proteica , Conformación Proteica , Infecciones Estafilocócicas/terapia , Relación Estructura-Actividad , Ácidos Teicoicos/metabolismo , Transgenes/genética
18.
Artículo en Inglés | MEDLINE | ID: mdl-28458652

RESUMEN

The Janus kinases (JAKs) are non-receptor tyrosine kinases essential for signaling in response to cytokines and interferons and thereby control many essential functions in growth, development, and immune regulation. JAKs are unique among tyrosine kinases for their constitutive yet non-covalent association with class I and II cytokine receptors, which upon cytokine binding bring together two JAKs to create an active signaling complex. JAK association with cytokine receptors is facilitated by N-terminal FERM and SH2 domains, both of which are classical mediators of peptide interactions. Together, the JAK FERM and SH2 domains mediate a bipartite interaction with two distinct receptor peptide motifs, the proline-rich "Box1" and hydrophobic "Box2," which are present in the intracellular domain of cytokine receptors. While the general sidechain chemistry of Box1 and Box2 peptides is conserved between receptors, they share very weak primary sequence homology, making it impossible to posit why certain JAKs preferentially interact with and signal through specific subsets of cytokine receptors. Here, we review the structure and function of the JAK FERM and SH2 domains in light of several recent studies that reveal their atomic structure and elucidate interaction mechanisms with both the Box1 and Box2 receptor motifs. These crystal structures demonstrate how evolution has repurposed the JAK FERM and SH2 domains into a receptor-binding module that facilitates interactions with multiple receptors possessing diverse primary sequences.

19.
Neuropharmacology ; 121: 204-218, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28457974

RESUMEN

Ionotropic glutamate receptors (iGluRs) mediate fast excitatory neurotransmission and are key nervous system drug targets. While diverse pharmacological tools have yielded insight into iGluR extracellular domain function, less is known about molecular mechanisms underlying the ion conduction gating process within the transmembrane domain (TMD). We have discovered a novel NMDAR positive allosteric modulator (PAM), GNE-9278, with a unique binding site on the extracellular surface of the TMD. Mutation of a single residue near the Lurcher motif on GluN1 M3 can convert GNE-9278 modulation from positive to negative, and replacing three AMPAR pre-M1 residues with corresponding NMDAR residues can confer GNE-9278 sensitivity to AMPARs. Modulation by GNE-9278 is state-dependent and significantly alters extracellular domain pharmacology. The unique properties and structural determinants of GNE-9278 reveal new modulatory potential of the iGluR TMD.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Doxiciclina/farmacología , Estimulación Eléctrica , Fármacos actuantes sobre Aminoácidos Excitadores/química , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Ácido Glutámico/farmacología , Glicina/metabolismo , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Técnicas de Placa-Clamp , Dominios Proteicos/efectos de los fármacos , Dominios Proteicos/genética , Pirimidinonas/química , Pirimidinonas/farmacología , Receptores de N-Metil-D-Aspartato/genética , Sulfonamidas/química , Sulfonamidas/farmacología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Transfección
20.
ACS Med Chem Lett ; 8(1): 84-89, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-28105280

RESUMEN

The N-methyl-d-aspartate receptor (NMDAR) is an ionotropic glutamate receptor, gated by the endogenous coagonists glutamate and glycine, permeable to Ca2+ and Na+. NMDAR dysfunction is associated with numerous neurological and psychiatric disorders, including schizophrenia, depression, and Alzheimer's disease. Recently, we have disclosed GNE-0723 (1), a GluN2A subunit-selective and brain-penetrant positive allosteric modulator (PAM) of NMDARs. This work highlights the discovery of a related pyridopyrimidinone core with distinct structure-activity relationships, despite the structural similarity to GNE-0723. GNE-5729 (13), a pyridopyrimidinone-based NMDAR PAM, was identified with both an improved pharmacokinetic profile and increased selectivity against AMPARs. We also include X-ray structure analysis and modeling to propose hypotheses for the activity and selectivity differences.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA