Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(52): e2304900120, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38109529

RESUMEN

Diacylglycerol lipase-beta (DAGLß) serves as a principal 2-arachidonoylglycerol (2-AG) biosynthetic enzyme regulating endocannabinoid and eicosanoid metabolism in immune cells including macrophages and dendritic cells. Genetic or pharmacological inactivation of DAGLß ameliorates inflammation and hyper-nociception in preclinical models of pathogenic pain. These beneficial effects have been assigned principally to reductions in downstream proinflammatory lipid signaling, leaving alternative mechanisms of regulation largely underexplored. Here, we apply quantitative chemical- and phospho-proteomics to find that disruption of DAGLß in primary macrophages leads to LKB1-AMPK signaling activation, resulting in reprogramming of the phosphoproteome and bioenergetics. Notably, AMPK inhibition reversed the antinociceptive effects of DAGLß blockade, thereby directly supporting DAGLß-AMPK crosstalk in vivo. Our findings uncover signaling between endocannabinoid biosynthetic enzymes and ancient energy-sensing kinases to mediate cell biological and pain responses.


Asunto(s)
Endocannabinoides , Glicéridos , Humanos , Endocannabinoides/metabolismo , Glicéridos/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Lipoproteína Lipasa/metabolismo , Ácidos Araquidónicos/metabolismo , Dolor
2.
Chembiochem ; 25(16): e202400382, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-38819848

RESUMEN

Imidazole-1-sulfonyl and -sulfonate (imidazylate) are widely used in synthetic chemistry as nucleofuges for diazotransfer, nucleophilic substitution, and cross-coupling reactions. The utility of these reagents for protein bioconjugation, in contrast, have not been comprehensively explored and important considering the prevalence of imidazoles in biomolecules and drugs. Here, we synthesized a series of alkyne-modified sulfonyl- and sulfonate-imidazole probes to investigate the utility of this electrophile for protein binding. Alkylation of the distal nitrogen activated the nucleofuge capability of the imidazole to produce sulfonyl-imidazolium electrophiles that were highly reactive but unstable for biological applications. In contrast, arylsulfonyl imidazoles functioned as a tempered electrophile for assessing ligandability of select tyrosine and lysine sites in cell proteomes and when mated to a recognition element could produce targeted covalent inhibitors with reduced off-target activity. In summary, imidazole nucleofuges show balanced stability and tunability to produce sulfone-based electrophiles that bind functional tyrosine and lysine sites in the proteome.


Asunto(s)
Imidazoles , Tirosina , Imidazoles/química , Imidazoles/síntesis química , Humanos , Tirosina/química , Estructura Molecular , Alquilación
3.
J Am Chem Soc ; 145(20): 11056-11066, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37159397

RESUMEN

Stress granules (SGs) and processing-bodies (PBs, P-bodies) are ubiquitous and widely studied ribonucleoprotein (RNP) granules involved in cellular stress response, viral infection, and the tumor microenvironment. While proteomic and transcriptomic investigations of SGs and PBs have provided insights into molecular composition, chemical tools to probe and modulate RNP granules remain lacking. Herein, we combine an immunofluorescence (IF)-based phenotypic screen with chemoproteomics to identify sulfonyl-triazoles (SuTEx) capable of preventing or inducing SG and PB formation through liganding of tyrosine (Tyr) and lysine (Lys) sites in stressed cells. Liganded sites were enriched for RNA-binding and protein-protein interaction (PPI) domains, including several sites found in RNP granule-forming proteins. Among these, we functionally validate G3BP1 Y40, located in the NTF2 dimerization domain, as a ligandable site that can disrupt arsenite-induced SG formation in cells. In summary, we present a chemical strategy for the systematic discovery of condensate-modulating covalent small molecules.


Asunto(s)
Gránulos Citoplasmáticos , ADN Helicasas , ADN Helicasas/química , ADN Helicasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Gránulos Citoplasmáticos/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteómica , ARN Helicasas/química
4.
J Biol Chem ; 296: 100070, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33187980

RESUMEN

Lipids in complex, protein-enriched films at air/liquid interfaces reduce surface tension. In the absence of this benefit, the light refracting and immunoprotective tear film on eyes would collapse. Premature collapse, coupled with chronic inflammation compromising visual acuity, is a hallmark of dry eye disease affecting 7 to 10% of individuals worldwide. Although collapse seems independent of mutation (unlike newborn lung alveoli), selective proteome and possible lipidome changes have been noted. These include elevated tissue transglutaminase and consequent inactivation through C-terminal cross-linking of the tear mitogen lacritin, leading to significant loss of lacritin monomer. Lacritin monomer restores homeostasis via autophagy and mitochondrial fusion and promotes basal tearing. Here, we discover that lacritin monomer C-terminal processing, inclusive of cysteine, serine, and metalloproteinase activity, generates cationic amphipathic α-helical proteoforms. Such proteoforms (using synthetic peptide surrogates) act like alveolar surfactant proteins to rapidly bind and stabilize the tear lipid layer. Immunodepletion of C- but not N-terminal proteoforms nor intact lacritin, from normal human tears promotes loss of stability akin to human dry eye tears. Stability of these and dry eye tears is rescuable with C- but not N-terminal proteoforms. Repeated topical application in rabbits reveals a proteoform turnover time of 7 to 33 h with gradual loss from human tear lipid that retains bioactivity without further processing. Thus, the processed C-terminus of lacritin that is deficient or absent in dry eye tears appears to play a key role in preventing tear film collapse and as a natural slow release mechanism that restores epithelial homeostasis.


Asunto(s)
Síndromes de Ojo Seco/fisiopatología , Proteínas del Ojo/metabolismo , Glicoproteínas/fisiología , Isoformas de Proteínas/fisiología , Lágrimas/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Glándulas Tarsales/fisiología , Conejos
5.
Nat Chem Biol ; 16(2): 170-178, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932721

RESUMEN

C1 domains are lipid-binding modules that regulate membrane activation of kinases, nucleotide exchange factors and other C1-containing proteins to trigger signal transduction. Despite annotation of typical C1 domains as diacylglycerol (DAG) and phorbol ester sensors, the function of atypical counterparts remains ill-defined. Here, we assign a key role for atypical C1 domains in mediating DAG fatty acyl specificity of diacylglycerol kinases (DGKs) in live cells. Activity-based proteomics mapped C1 probe binding as a principal differentiator of type 1 DGK active sites that combined with global metabolomics revealed a role for C1s in lipid substrate recognition. Protein engineering by C1 domain swapping demonstrated that exchange of typical and atypical C1s is functionally tolerated and can directly program DAG fatty acyl specificity of type 1 DGKs. Collectively, we describe a protein engineering strategy for studying metabolic specificity of lipid kinases to assign a role for atypical C1 domains in cell metabolism.


Asunto(s)
Diacilglicerol Quinasa/química , Diacilglicerol Quinasa/metabolismo , Ingeniería de Proteínas/métodos , Animales , Dominio Catalítico , Cromatografía Liquida , Diacilglicerol Quinasa/genética , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Humanos , Metabolómica/métodos , Sondas Moleculares/química , Ácidos Fosfatidicos/metabolismo , Dominios Proteicos , Proteómica/métodos , Ratas , Especificidad por Sustrato , Espectrometría de Masas en Tándem
6.
Nat Chem Biol ; 16(2): 150-159, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31768034

RESUMEN

Covalent probes serve as valuable tools for global investigation of protein function and ligand binding capacity. Despite efforts to expand coverage of residues available for chemical proteomics (e.g., cysteine and lysine), a large fraction of the proteome remains inaccessible with current activity-based probes. Here, we introduce sulfur-triazole exchange (SuTEx) chemistry as a tunable platform for developing covalent probes with broad applications for chemical proteomics. We show modifications to the triazole leaving group can furnish sulfonyl probes with ~5-fold enhanced chemoselectivity for tyrosines over other nucleophilic amino acids to investigate more than 10,000 tyrosine sites in lysates and live cells. We discover that tyrosines with enhanced nucleophilicity are enriched in enzymatic, protein-protein interaction and nucleotide recognition domains. We apply SuTEx as a chemical phosphoproteomics strategy to monitor activation of phosphotyrosine sites. Collectively, we describe SuTEx as a biocompatible chemistry for chemical biology investigations of the human proteome.


Asunto(s)
Sondas Moleculares/química , Proteómica/métodos , Azufre/química , Triazoles/química , Tirosina/análisis , Tirosina/química , Células A549 , Sitios de Unión , Flúor/química , Gutatión-S-Transferasa pi/genética , Gutatión-S-Transferasa pi/metabolismo , Células HEK293 , Humanos , Sondas Moleculares/síntesis química , Fosforilación , Fosfotirosina/química , Fosfotirosina/metabolismo , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Ácidos Sulfínicos/química , Tirosina/metabolismo
7.
Anal Chem ; 93(35): 11946-11955, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34431655

RESUMEN

Chemical proteomics is widely used for the global investigation of protein activity and binding of small molecule ligands. Covalent probe binding and inhibition are assessed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) to gain molecular information on targeted proteins and probe-modified sites. The identification of amino acid sites modified by large complex probes, however, is particularly challenging because of the increased size, hydrophobicity, and charge state of peptides derived from modified proteins. These studies are important for direct evaluation of proteome-wide selectivity of inhibitor scaffolds used to develop targeted covalent inhibitors. Here, we disclose reverse-phase chromatography and MS dissociation conditions tailored for binding site identification using a clickable covalent kinase inhibitor containing a sulfonyl-triazole reactive group (KY-26). We applied this LC-MS/MS strategy to identify tyrosine and lysine sites modified by KY-26 in functional sites of kinases and other ATP-/NAD-binding proteins (>65 in total) in live cells. Our studies revealed key bioanalytical conditions to guide future chemical proteomic workflows for direct target site identification of complex irreversible probes and inhibitors.


Asunto(s)
Proteómica , Espectrometría de Masas en Tándem , Cromatografía Liquida , Proteoma , Triazoles
8.
Chembiochem ; 22(12): 2134-2139, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33861519

RESUMEN

Sulfonyl-triazoles have emerged as a new reactive group for covalent modification of tyrosine sites on proteins through sulfur-triazole exchange (SuTEx) chemistry. The extent to which this sulfur electrophile can be tuned for developing ligands with cellular activity remains largely underexplored. Here, we performed fragment-based ligand discovery in live cells to identify SuTEx compounds capable of liganding tyrosine sites on diverse protein targets. We verified our quantitative chemical proteomic findings by demonstrating concentration-dependent activity of SuTEx ligands, but not inactive counterparts, against recombinant protein targets directly in live cells. Our structure-activity relationship studies identified the SuTEx ligand HHS-0701 as a cell-active inhibitor capable of blocking prostaglandin reductase 2 (PTGR2) biochemical activity.


Asunto(s)
15-Oxoprostaglandina 13-Reductasa/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Azufre/farmacología , Triazoles/farmacología , 15-Oxoprostaglandina 13-Reductasa/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Ligandos , Estructura Molecular , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Azufre/química , Compuestos de Azufre , Triazoles/química
9.
J Am Chem Soc ; 142(18): 8270-8280, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32329615

RESUMEN

Tuning reactivity of sulfur electrophiles is key for advancing click chemistry and chemical probe discovery. To date, activation of the sulfur electrophile for protein modification has been ascribed principally to stabilization of a fluoride leaving group (LG) in covalent reactions of sulfonyl fluorides and arylfluorosulfates. We recently introduced sulfur-triazole exchange (SuTEx) chemistry to demonstrate the triazole as an effective LG for activating nucleophilic substitution reactions on tyrosine sites of proteins. Here, we probed tunability of SuTEx for fragment-based ligand discovery by modifying the adduct group (AG) and LG with functional groups of differing electron-donating and -withdrawing properties. We discovered the sulfur electrophile is highly sensitive to the position of modification (AG versus LG), which enabled both coarse and fine adjustments in solution and proteome activity. We applied these reactivity principles to identify a large fraction of tyrosine sites (∼30%) on proteins (∼44%) that can be liganded across >1500 probe-modified sites quantified by chemical proteomics. Our proteomic studies identified noncatalytic tyrosine and phosphotyrosine sites that can be liganded by SuTEx fragments with site specificity in lysates and live cells to disrupt protein function. Collectively, we describe SuTEx as a versatile covalent chemistry with broad applications for chemical proteomics and protein ligand discovery.


Asunto(s)
Proteínas/química , Azufre/química , Triazoles/química , Tirosina/química , Células HEK293 , Humanos , Ligandos , Estructura Molecular , Proteómica , Relación Estructura-Actividad
10.
Anal Chem ; 92(2): 2005-2010, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31869197

RESUMEN

Spatially targeted optical microproteomics (STOMP) is a method to study region-specific protein complexity in primary cells and tissue samples. STOMP uses a confocal microscope to visualize structures of interest and to tag the proteins within those structures by a photodriven cross-linking reaction so that they can be affinity purified and identified by mass spectrometry (eLife 2015, 4, e09579). However, the use of a custom photo-cross-linker and the requirement for extensive user intervention during sample tagging have posed barriers to the utilization of STOMP. To address these limitations, we built automated STOMP (autoSTOMP) which uses a customizable code in SikuliX to coordinate image capture and cross-linking functions in Zeiss Zen Black with image processing in FIJI. To increase protocol accessibility, we implemented a commercially available biotin-benzophenone photo-cross-linking and purification protocol. Here we demonstrate that autoSTOMP can efficiently label, purify, and identify proteins belonging to 1-2 µm structures in primary human foreskin fibroblasts or mouse bone marrow-derived dendritic cells infected with the protozoan parasite Toxoplasma gondii (Tg). AutoSTOMP can easily be adapted to address a range of research questions using Zeiss Zen Black microscopy systems and LC-MS protocols that are standard in many research cores.


Asunto(s)
Automatización , Proteínas/análisis , Proteómica , Animales , Células Dendríticas/química , Fibroblastos/química , Humanos , Ratones , Estructura Molecular , Imagen Óptica , Conformación Proteica
11.
Curr Top Microbiol Immunol ; 420: 175-210, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30128827

RESUMEN

As a major sentinel of adaptive immunity, T cells seek and destroy diseased cells using antigen recognition to achieve molecular specificity. Strategies to block checkpoint inhibition of T cell activity and thus reawaken the patient's antitumor immune responses are rapidly becoming standard of care for treatment of diverse cancers. Adoptive transfer of patient T cells genetically engineered with tumor-targeting capabilities is redefining the field of personalized medicines. The diverse opportunities for exploiting T cell biology in the clinic have prompted new efforts to expand the scope of targets amenable to immuno-oncology. Given the complex spatiotemporal regulation of T cell function and fate, new technologies capable of global molecular profiling in vivo are needed to guide selection of appropriate T cell targets and subsets. In this chapter, we describe the use of activity-based protein profiling (ABPP) to illuminate different aspects of T cell metabolism and signaling as fertile starting points for investigation. We highlight the merits of ABPP methods to enable target, inhibitor, and biochemical pathway discovery of T cells in the burgeoning field of immuno-oncology.


Asunto(s)
Proteoma/análisis , Proteoma/metabolismo , Proteómica/métodos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Inmunidad Adaptativa/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Proteoma/química , Transducción de Señal/inmunología
12.
J Pharmacol Exp Ther ; 371(3): 652-662, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31601683

RESUMEN

Oncogenic protein tyrosine phosphatases (PTPs) are overexpressed in numerous human cancers but they have been challenging pharmacological targets. The emblematic oncogenic PTP4A tyrosine phosphatase family regulates many fundamental malignant processes. 7-Imino-2-phenylthieno[3,2-c]pyridine-4,6(5H,7H)-dione (JMS-053) is a novel, potent, and selective PTP4A inhibitor but its mechanism of action has not been fully elucidated, nor has the chemotype been fully investigated. Because tyrosine phosphatases are notoriously susceptible to oxidation, we interrogated JMS-053 and three newly synthesized analogs with specific attention on the role of oxidation. JMS-053 and its three analogs were potent in vitro PTP4A3 inhibitors, but 7-imino-5-methyl-2-phenylthieno[3,2-c]pyridine-4,6(5H,7H)-dione (NRT-870-59) appeared unique among the thienopyridinediones with respect to its inhibitory specificity for PTP4A3 versus both a PTP4A3 A111S mutant and an oncogenic dual specificity tyrosine phosphatase, CDC25B. Like JMS-053, NRT-870-59 was a reversible PTP4A3 inhibitor. All of the thienopyridinediones retained cytotoxicity against human ovarian and breast cancer cells grown as pathologically relevant three-dimensional spheroids. Inhibition of cancer cell colony formation by NRT-870-59, like JMS-053, required PTP4A3 expression. JMS-053 failed to generate significant detectable reactive oxygen species in vitro or in cancer cells. Mass spectrometry results indicated no disulfide bond formation or oxidation of the catalytic Cys104 after in vitro incubation of PTP4A3 with JMS-053 or NRT-870-59. Gene expression profiling of cancer cells exposed to JMS-053 phenocopied many of the changes seen with the loss of PTP4A3 and did not indicate oxidative stress. These data demonstrate that PTP4A phosphatases can be selectively targeted with small molecules that lack prominent reactive oxygen species generation and encourage further studies of this chemotype. SIGNIFICANCE STATEMENT: Protein tyrosine phosphatases are emerging as important contributors to human cancers. We report on a new class of reversible protein phosphatase small molecule inhibitors that are cytotoxic to human ovarian and breast cancer cells, do not generate significant reactive oxygen species in vitro and in cells, and could be valuable lead molecules for future studies of PTP4A phosphatases.


Asunto(s)
Antineoplásicos/farmacología , Iminas/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Piridinas/farmacología , Piridonas/farmacología , Línea Celular Tumoral , Humanos , Mutación , Proteínas de Neoplasias/genética , Proteínas Tirosina Fosfatasas/genética , Especies Reactivas de Oxígeno/metabolismo
13.
Nat Chem Biol ; 18(7): 686-687, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35710618
14.
Proc Natl Acad Sci U S A ; 113(4): 1086-91, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26755579

RESUMEN

Chronic nicotine exposure (CNE) alters synaptic transmission in the ventral tegmental area (VTA) in a manner that enhances dopaminergic signaling and promotes nicotine use. The present experiments identify a correlation between enhanced production of the endogenous cannabinoid 2-arachidonoylglycerol (2-AG) and diminished release of the inhibitory neurotransmitter GABA in the VTA following CNE. To study the functional role of on-demand 2-AG signaling in GABAergic synapses, we used 1,2,3-triazole urea compounds to selectively inhibit 2-AG biosynthesis by diacylglycerol lipase (DAGL). The potency and selectivity of these inhibitors were established in rats in vitro (rat brain proteome), ex vivo (brain slices), and in vivo (intracerebroventricular administration) using activity-based protein profiling and targeted metabolomics analyses. Inhibition of DAGL (2-AG biosynthesis) rescues nicotine-induced VTA GABA signaling following CNE. Conversely, enhancement of 2-AG signaling in naïve rats by inhibiting 2-AG degradation recapitulates the loss of nicotine-induced GABA signaling evident following CNE. DAGL inhibition reduces nicotine self-administration without disrupting operant responding for a nondrug reinforcer or motor activity. Collectively, these findings provide a detailed characterization of selective inhibitors of rat brain DAGL and demonstrate that excessive 2-AG signaling contributes to a loss of inhibitory GABAergic constraint of VTA excitability following CNE.


Asunto(s)
Lipoproteína Lipasa/antagonistas & inhibidores , Nicotina/farmacología , Área Tegmental Ventral/efectos de los fármacos , Animales , Ácidos Araquidónicos/análisis , Ácidos Araquidónicos/antagonistas & inhibidores , Ácidos Araquidónicos/fisiología , Endocannabinoides/análisis , Endocannabinoides/antagonistas & inhibidores , Endocannabinoides/fisiología , Glicéridos/análisis , Glicéridos/antagonistas & inhibidores , Glicéridos/fisiología , Masculino , Ratas , Ratas Wistar , Autoadministración , Área Tegmental Ventral/fisiología , Ácido gamma-Aminobutírico/metabolismo
15.
Biochemistry ; 57(2): 231-236, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29155586

RESUMEN

Diacylglycerol kinases (DGKs) regulate lipid metabolism and cell signaling through ATP-dependent phosphorylation of diacylglycerol to biosynthesize phosphatidic acid. Selective chemical probes for studying DGKs are currently lacking and are needed to annotate isoform-specific functions of these elusive lipid kinases. Previously, we explored fragment-based approaches to discover a core fragment of DGK-α (DGKα) inhibitors responsible for selective binding to the DGKα active site. Here, we utilize quantitative chemical proteomics to deconstruct widely used DGKα inhibitors to identify structural regions mediating off-target activity. We tested the activity of a fragment (RLM001) derived from a nucleotide-like region found in the DGKα inhibitors R59022 and ritanserin and discovered that RLM001 mimics ATP in its ability to broadly compete at ATP-binding sites of DGKα as well as >60 native ATP-binding proteins (kinases and ATPases) detected in cell proteomes. Equipotent inhibition of activity-based probe labeling by RLM001 supports a contiguous ligand-binding site composed of C1, DAGKc, and DAGKa domains in the DGKα active site. Given the lack of available crystal structures of DGKs, our studies highlight the utility of chemical proteomics in revealing active-site features of lipid kinases to enable development of inhibitors with enhanced selectivity against the human proteome.


Asunto(s)
Diacilglicerol Quinasa/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Proteómica/métodos , Ritanserina/análogos & derivados , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Diacilglicerol Quinasa/metabolismo , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Estructura Molecular , Proteínas Recombinantes/metabolismo , Ritanserina/química , Ritanserina/metabolismo , Ritanserina/farmacología , Relación Estructura-Actividad
16.
Mol Pharmacol ; 94(5): 1246-1255, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30158316

RESUMEN

Ritanserin was tested in the clinic as a serotonin receptor inverse agonist but recently emerged as a novel kinase inhibitor with potential applications in cancer. Here, we discovered that ritanserin induced apoptotic cell death of non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) cells via a serotonin-independent mechanism. We used quantitative chemical proteomics to reveal a ritanserin-dependent kinase network that includes key mediators of lipid [diacylglycerol kinase α, phosphatidylinositol 4-kinase ß] and protein [feline encephalitis virus-related kinase, rapidly accelerated fibrosarcoma (RAF)] signaling, metabolism [eukaryotic elongation factor 2 kinase, eukaryotic translation initiation factor 2-α kinase 4], and DNA damage response [tousled-like kinase 2] to broadly kill lung tumor cell types. Whereas ritanserin exhibited polypharmacology in NSCLC proteomes, this compound showed unexpected specificity for c-RAF in the SCLC subtype, with negligible activity against other kinases mediating mitogen-activated protein kinase signaling. Here we show that ritanserin blocks c-RAF but not B-RAF activation of established oncogenic signaling pathways in live cells, providing evidence in support of c-RAF as a key target mediating its anticancer activity. Given the role of c-RAF activation in RAS-mutated cancers resistant to clinical B-RAF inhibitors, our findings may have implications in overcoming resistance mechanisms associated with c-RAF biology. The unique target landscape combined with acceptable safety profiles in humans provides new opportunities for repositioning ritanserin in cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Pequeñas/metabolismo , Carcinoma de Células Pequeñas/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteómica , Ritanserina/farmacología , Secuencia de Aminoácidos , Línea Celular Tumoral , Reposicionamiento de Medicamentos , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos , Serotonina/metabolismo
17.
J Pharmacol Exp Ther ; 366(1): 169-183, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29540562

RESUMEN

Although paclitaxel effectively treats various cancers, its debilitating peripheral neuropathic pain side effects often persist long after treatment has ended. Therefore, a compelling need exists for the identification of novel pharmacologic strategies to mitigate this condition. As inhibitors of monoacylglycerol lipase (MAGL), the primary hydrolytic enzyme of the endogenous cannabinoid, 2-arachidonyolglycerol, produces antinociceptive effects in numerous rodent models of pain, we investigated whether inhibitors of this enzyme (i.e., JZL184 and MJN110) would reverse paclitaxel-induced mechanical allodynia in mice. These drugs dose dependently reversed allodynia with respective ED50 values (95% confidence limit) of 8.4 (5.2-13.6) and 1.8 (1.0-3.3) mg/kg. Complementary genetic and pharmacologic approaches revealed that the antiallodynic effects of each drug require both cannabinoid receptors, CB1 and CB2 MJN110 reduced paclitaxel-mediated increased expression of monocyte chemoattractant protein-1 (MCP-1, CCL2) and phospho-p38 MAPK in dorsal root ganglia as well as MCP-1 in spinal dorsal horn. Whereas the antinociceptive effects of high dose JZL184 (40 mg/kg) underwent tolerance following 6 days of repeated dosing, repeated administration of a threshold dose (i.e., 4 mg/kg) completely reversed paclitaxel-induced allodynia. In addition, we found that the administration of MJN110 to control mice lacked intrinsic rewarding effects in the conditioned place preference (CPP) paradigm. However, it produced a CPP in paclitaxel-treated animals, suggesting a reduced paclitaxel-induced aversive state. Importantly, JZL184 did not alter the antiproliferative and apoptotic effects of paclitaxel in A549 and H460 non-small cell lung cancer cells. Taken together, these data indicate that MAGL inhibitors reverse paclitaxel-induced neuropathic pain without interfering with chemotherapeutic efficacy.


Asunto(s)
Antineoplásicos/efectos adversos , Inhibidores Enzimáticos/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Monoacilglicerol Lipasas/antagonistas & inhibidores , Nocicepción/efectos de los fármacos , Paclitaxel/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Benzodioxoles/farmacología , Benzodioxoles/uso terapéutico , Biomarcadores/metabolismo , Carbamatos/farmacología , Carbamatos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/uso terapéutico , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Inflamación/metabolismo , Masculino , Ratones , Fosfoproteínas/metabolismo , Piperidinas/farmacología , Piperidinas/uso terapéutico , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Succinimidas/farmacología , Succinimidas/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Mol Pharm ; 15(3): 721-728, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-28901776

RESUMEN

Diacylglycerol lipase-beta (DAGLß) hydrolyzes arachidonic acid (AA)-containing diacylglycerols to produce bioactive lipids including endocannabinoids and AA-derived eicosanoids involved in regulation of inflammatory signaling. Previously, we demonstrated that DAGLß inactivation using the triazole urea inhibitor KT109 blocked macrophage inflammatory signaling and reversed allodynic responses of mice in inflammatory and neuropathic pain models. Here, we tested whether we could exploit the phagocytic capacity of macrophages to localize delivery of DAGLß inhibitors to these cells in vivo using liposome encapsulated KT109. We used DAGLß-tailored activity-based probes and chemical proteomic methods to measure potency and selectivity of liposomal KT109 in macrophages and tissues from treated mice. Surprisingly, delivery of ∼5 µg of liposomal KT109 was sufficient to achieve ∼80% inactivation of DAGLß in macrophages with no apparent activity in other tissues in vivo. Our macrophage-targeted delivery resulted in a >100-fold enhancement in antinociceptive potency compared with free compound in a mouse inflammatory pain model. Our studies describe a novel anti-inflammatory strategy that is achieved by targeted in vivo delivery of DAGLß inhibitors to macrophages.


Asunto(s)
Antiinflamatorios/farmacología , Inhibidores Enzimáticos/farmacología , Lipoproteína Lipasa/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Dolor/tratamiento farmacológico , Fagocitosis/efectos de los fármacos , Triazoles/farmacología , Urea/análogos & derivados , Urea/farmacología , Animales , Antiinflamatorios/uso terapéutico , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Humanos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Inflamación/complicaciones , Inflamación/inmunología , Lipopolisacáridos/inmunología , Lipoproteína Lipasa/metabolismo , Liposomas , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Dolor/inmunología , Fagocitosis/inmunología , Resultado del Tratamiento , Triazoles/uso terapéutico , Urea/uso terapéutico
19.
Biochem Biophys Res Commun ; 490(4): 1226-1231, 2017 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-28684316

RESUMEN

The serine hydrolase inhibitors pyrrophenone and KT195 inhibit cell death induced by A23187 and H2O2 by blocking the release of calcium from the endoplasmic reticulum and mitochondrial calcium uptake. The effect of pyrrophenone and KT195 on these processes is not due to inhibition of their known targets, cytosolic phospholipase A2 and α/ß-hydrolase domain-containing (ABHD) 6, respectively, but represent off-target effects. To identify targets of KT195, fibroblasts were treated with KT195-alkyne to covalently label protein targets followed by click chemistry with biotin azide, enrichment on streptavidin beads and tryptic peptide analysis by mass spectrometry. Although several serine hydrolases were identified, α/ß-hydrolase domain-containing 2 (ABHD2) was the only target in which both KT195 and pyrrophenone competed for binding to KT195-alkyne. ABHD2 is a serine hydrolase with a predicted transmembrane domain consistent with its pull-down from the membrane proteome. Subcellular fractionation showed localization of ABHD2 to the endoplasmic reticulum but not to mitochondria or mitochondrial-associated membranes. Knockdown of ABHD2 with shRNA attenuated calcium release from the endoplasmic reticulum, mitochondrial calcium uptake and cell death in fibroblasts stimulated with A23187. The results describe a novel mechanism for regulating calcium transfer from the endoplasmic reticulum to mitochondria that involves the serine hydrolase ABHD2.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Hidrolasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Fosfolipasas A2 Grupo IV/antagonistas & inhibidores , Fosfolipasas A2 Grupo IV/deficiencia , Fosfolipasas A2 Grupo IV/metabolismo , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/farmacología , Hidrolasas/antagonistas & inhibidores , Ratones , Ratones Noqueados , Inhibidores de Fosfolipasa A2/farmacología , Pirrolidinas/farmacología , Relación Estructura-Actividad
20.
Proc Natl Acad Sci U S A ; 111(11): 4338-43, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591635

RESUMEN

Cell surface glycans form a critical interface with the biological milieu, informing diverse processes from the inflammatory cascade to cellular migration. Assembly of discrete carbohydrate structures requires the coordinated activity of a repertoire of proteins, including glycosyltransferases and glycosidases. Little is known about the regulatory networks controlling this complex biosynthetic process. Recent work points to a role for microRNA (miRNA) in the regulation of specific glycan biosynthetic enzymes. Herein we take a unique systems-based approach to identify connections between miRNA and the glycome. By using our glycomic analysis platform, lectin microarrays, we identify glycosylation signatures in the NCI-60 cell panel that point to the glycome as a direct output of genomic information flow. Integrating our glycomic dataset with miRNA data, we map miRNA regulators onto genes in glycan biosynthetic pathways (glycogenes) that generate the observed glycan structures. We validate three of these predicted miRNA/glycogene regulatory networks: high mannose, fucose, and terminal ß-GalNAc, identifying miRNA regulation that would not have been observed by traditional bioinformatic methods. Overall, our work reveals critical nodes in the global glycosylation network accessible to miRNA regulation, providing a bridge between miRNA-mediated control of cell phenotype and the glycome.


Asunto(s)
Vías Biosintéticas/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Redes Reguladoras de Genes/genética , MicroARNs/metabolismo , Polisacáridos/biosíntesis , Western Blotting , Línea Celular , Regulación Enzimológica de la Expresión Génica/genética , Glicómica/métodos , Glicosilación/efectos de los fármacos , Humanos , Luciferasas , MicroARNs/farmacología , Análisis por Micromatrices , Microscopía Fluorescente , Reacción en Cadena en Tiempo Real de la Polimerasa , Biología de Sistemas/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA