Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Annu Rev Biochem ; 84: 765-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26034893

RESUMEN

Hydrogen peroxide (H2O2) is a prime member of the reactive oxygen species (ROS) family of molecules produced during normal cell function and in response to various stimuli, but if left unchecked, it can inflict oxidative damage on all types of biological macromolecules and lead to cell death. In this context, a major source of H2O2 for redox signaling purposes is the NADPH oxidase (Nox) family of enzymes, which were classically studied for their roles in phagocytic immune response but have now been found to exist in virtually all mammalian cell types in various isoforms with distinct tissue and subcellular localizations. Downstream of this tightly regulated ROS generation, site-specific, reversible covalent modification of proteins, particularly oxidation of cysteine thiols to sulfenic acids, represents a prominent posttranslational modification akin to phosphorylation as an emerging molecular mechanism for transforming an oxidant signal into a dynamic biological response. We review two complementary types of chemical tools that enable (a) specific detection of H2O2 generated at its sources and (b) mapping of sulfenic acid posttranslational modification targets that mediate its signaling functions, which can be used to study this important chemical signal in biological systems.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , NADPH Oxidasas/metabolismo , Transducción de Señal , Animales , Humanos , Oxidación-Reducción , Ácidos Sulfénicos/metabolismo
2.
Mol Cell ; 80(6): 1013-1024.e6, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33338401

RESUMEN

Impaired DNA crosslink repair leads to Fanconi anemia (FA), characterized by a unique manifestation of bone marrow failure and pancytopenia among diseases caused by DNA damage response defects. As a germline disorder, why the hematopoietic hierarchy is specifically affected is not fully understood. We find that reprogramming transcription during hematopoietic differentiation results in an overload of genotoxic stress, which causes aborted differentiation and depletion of FA mutant progenitor cells. DNA damage onset most likely arises from formaldehyde, an obligate by-product of oxidative protein demethylation during transcription regulation. Our results demonstrate that rapid and extensive transcription reprogramming associated with hematopoietic differentiation poses a major threat to genome stability and cell viability in the absence of the FA pathway. The connection between differentiation and DNA damage accumulation reveals a novel mechanism of genome scarring and is critical to exploring therapies to counteract the aplastic anemia for the treatment of FA patients.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Reprogramación Celular/genética , Anemia de Fanconi/genética , Formaldehído/toxicidad , Daño del ADN/efectos de los fármacos , Reparación del ADN/genética , Anemia de Fanconi/sangre , Anemia de Fanconi/patología , Formaldehído/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Inestabilidad Genómica/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células K562 , Transcripción Genética
3.
Nature ; 560(7716): 70-75, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30068953

RESUMEN

Despite the widespread success of transition-metal-catalysed cross-coupling methodologies, considerable limitations still exist in reactions at sp3-hybridized carbon atoms, with most approaches relying on prefunctionalized alkylmetal or bromide coupling partners1,2. Although the use of native functional groups (for example, carboxylic acids, alkenes and alcohols) has improved the overall efficiency of such transformations by expanding the range of potential feedstocks3-5, the direct functionalization of carbon-hydrogen (C-H) bonds-the most abundant moiety in organic molecules-represents a more ideal approach to molecular construction. In recent years, an impressive range of reactions that form C(sp3)-heteroatom bonds from strong C-H bonds has been reported6,7. Additionally, valuable technologies have been developed for the formation of carbon-carbon bonds from the corresponding C(sp3)-H bonds via substrate-directed transition-metal C-H insertion8, undirected C-H insertion by captodative rhodium carbenoid complexes9, or hydrogen atom transfer from weak, hydridic C-H bonds by electrophilic open-shell species10-14. Despite these advances, a mild and general platform for the coupling of strong, neutral C(sp3)-H bonds with aryl electrophiles has not been realized. Here we describe a protocol for the direct C(sp3) arylation of a diverse set of aliphatic, C-H bond-containing organic frameworks through the combination of light-driven, polyoxometalate-facilitated hydrogen atom transfer and nickel catalysis. This dual-catalytic manifold enables the generation of carbon-centred radicals from strong, neutral C-H bonds, which thereafter act as nucleophiles in nickel-mediated cross-coupling with aryl bromides to afford C(sp3)-C(sp2) cross-coupled products. This technology enables unprecedented, single-step access to a broad array of complex, medicinally relevant molecules directly from natural products and chemical feedstocks through functionalization at sites that are unreactive under traditional methods.


Asunto(s)
Carbono/química , Enlace de Hidrógeno , Productos Biológicos/síntesis química , Productos Biológicos/química , Catálisis , Níquel/química , Compuestos de Tungsteno/química
4.
Nature ; 548(7669): 549-554, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28813411

RESUMEN

The folate-driven one-carbon (1C) cycle is a fundamental metabolic hub in cells that enables the synthesis of nucleotides and amino acids and epigenetic modifications. This cycle might also release formaldehyde, a potent protein and DNA crosslinking agent that organisms produce in substantial quantities. Here we show that supplementation with tetrahydrofolate, the essential cofactor of this cycle, and other oxidation-prone folate derivatives kills human, mouse and chicken cells that cannot detoxify formaldehyde or that lack DNA crosslink repair. Notably, formaldehyde is generated from oxidative decomposition of the folate backbone. Furthermore, we find that formaldehyde detoxification in human cells generates formate, and thereby promotes nucleotide synthesis. This supply of 1C units is sufficient to sustain the growth of cells that are unable to use serine, which is the predominant source of 1C units. These findings identify an unexpected source of formaldehyde and, more generally, indicate that the detoxification of this ubiquitous endogenous genotoxin creates a benign 1C unit that can sustain essential metabolism.


Asunto(s)
Carbono/metabolismo , Ácido Fólico/química , Ácido Fólico/metabolismo , Formaldehído/química , Formaldehído/metabolismo , Redes y Vías Metabólicas , Mutágenos/química , Mutágenos/metabolismo , Alcohol Deshidrogenasa/metabolismo , Animales , Carbono/deficiencia , Línea Celular , Pollos , Coenzimas/metabolismo , Reactivos de Enlaces Cruzados/metabolismo , Daño del ADN , Reparación del ADN , Humanos , Inactivación Metabólica , Ratones , Nucleótidos/biosíntesis , Oxidación-Reducción , Serina/química , Serina/metabolismo , Tetrahidrofolatos/metabolismo
6.
J Am Chem Soc ; 142(37): 15917-15930, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32872768

RESUMEN

Carbon monoxide (CO) is an emerging gasotransmitter and reactive carbon species with broad anti-inflammatory, cytoprotective, and neurotransmitter functions along with therapeutic potential for the treatment of cardiovascular diseases. The study of CO chemistry in biology and medicine relative to other prominent gasotransmitters such as NO and H2S remains challenging, in large part due to limitations in available tools for the direct visualization of this transient and freely diffusing small molecule in complex living systems. Here we report a ligand-directed activity-based sensing (ABS) approach to CO detection through palladium-mediated carbonylation chemistry. Specifically, the design and synthesis of a series of ABS probes with systematic alterations in the palladium-ligand environment (e.g., sp3-S, sp3-N, sp2-N) establish structure-activity relationships for palladacycles to confer selective reactivity with CO under physiological conditions. These fundamental studies led to the development of an optimized probe, termed Carbon Monoxide Probe-3 Ester Pyridine (COP-3E-Py), which enables imaging of CO release in live cell and brain settings, including monitoring of endogenous CO production that triggers presynaptic dopamine release in fly brains. This work provides a unique tool for studying CO in living systems and establishes the utility of a synthetic methods approach to activity-based sensing using principles of organometallic chemistry.


Asunto(s)
Monóxido de Carbono/análisis , Complejos de Coordinación/química , Colorantes Fluorescentes/química , Paladio/química , Complejos de Coordinación/síntesis química , Colorantes Fluorescentes/síntesis química , Células HEK293 , Humanos , Ligandos , Estructura Molecular
7.
Proc Natl Acad Sci U S A ; 112(17): 5383-8, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25848022

RESUMEN

Cyclic dinucleotides are an expanding class of signaling molecules that control many aspects of bacterial physiology. A synthase for cyclic AMP-GMP (cAG, also referenced as 3'-5', 3'-5' cGAMP) called DncV is associated with hyperinfectivity of Vibrio cholerae but has not been found in many bacteria, raising questions about the prevalence and function of cAG signaling. We have discovered that the environmental bacterium Geobacter sulfurreducens produces cAG and uses a subset of GEMM-I class riboswitches (GEMM-Ib, Genes for the Environment, Membranes, and Motility) as specific receptors for cAG. GEMM-Ib riboswitches regulate genes associated with extracellular electron transfer; thus cAG signaling may control aspects of bacterial electrophysiology. These findings expand the role of cAG beyond organisms that harbor DncV and beyond pathogenesis to microbial geochemistry, which is important to environmental remediation and microbial fuel cell development. Finally, we have developed an RNA-based fluorescent biosensor for live-cell imaging of cAG. This selective, genetically encodable biosensor will be useful to probe the biochemistry and cell biology of cAG signaling in diverse bacteria.


Asunto(s)
Fenómenos Electrofisiológicos , Geobacter/metabolismo , Nucleótidos Cíclicos/metabolismo , ARN Bacteriano/metabolismo , Riboswitch/fisiología , Sistemas de Mensajero Secundario/fisiología , Geobacter/genética , Nucleótidos Cíclicos/genética , ARN Bacteriano/genética , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
8.
J Am Chem Soc ; 139(15): 5338-5350, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28375637

RESUMEN

Formaldehyde (FA) is a reactive signaling molecule that is continuously produced through a number of central biological pathways spanning epigenetics to one-carbon metabolism. On the other hand, aberrant, elevated levels of FA are implicated in disease states ranging from asthma to neurodegenerative disorders. In this context, fluorescence-based probes for FA imaging are emerging as potentially powerful chemical tools to help disentangle the complexities of FA homeostasis and its physiological and pathological contributions. Currently available FA indicators require direct modification of the fluorophore backbone through complex synthetic considerations to enable FA detection, often limiting the generalization of designs to other fluorophore classes. To address this challenge, we now present the rational, iterative development of a general reaction-based trigger utilizing 2-aza-Cope reactivity for selective and sensitive detection of FA in living systems. Specifically, we developed a homoallylamine functionality that can undergo a subsequent self-immolative ß-elimination, creating a FA-responsive trigger that is capable of masking a phenol on a fluorophore or any other potential chemical scaffold for related imaging and/or therapeutic applications. We demonstrate the utility of this trigger by creating a series of fluorescent probes for FA with excitation and emission wavelengths that span the UV to visible spectral regions through caging of a variety of dye units. In particular, Formaldehyde Probe 573 (FAP573), based on a resorufin scaffold, is the most red-shifted and FA sensitive in this series in terms of signal-to-noise responses and enables identification of alcohol dehydrogenase 5 (ADH5) as an enzyme that regulates FA metabolism in living cells. The results provide a starting point for the broader use of 2-aza-Cope reactivity for probing and manipulating FA biology.


Asunto(s)
Compuestos Aza/química , Formaldehído/análisis , Formaldehído/química , Imagen Óptica , Supervivencia Celular , Células HEK293 , Humanos , Estructura Molecular
9.
Proc Natl Acad Sci U S A ; 111(21): 7531-6, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24821806

RESUMEN

Development of small-molecule inhibitors of protein-protein interactions is a fundamental challenge at the interface of chemistry and cancer biology. Successful methods for design of protein-protein interaction inhibitors include computational and experimental high-throughput and fragment-based screening strategies to locate small-molecule fragments that bind protein surfaces. An alternative rational design approach seeks to mimic the orientation and disposition of critical binding residues at protein interfaces. We describe the design, synthesis, biochemical, and in vivo evaluation of a small-molecule scaffold that captures the topography of α-helices. We designed mimics of a key α-helical domain at the interface of hypoxia-inducible factor 1α and p300 to develop inhibitors of hypoxia-inducible signaling. The hypoxia-inducible factor/p300 interaction regulates the transcription of key genes, whose expression contributes to angiogenesis, metastasis, and altered energy metabolism in cancer. The designed compounds target the desired protein with high affinity and in a predetermined manner, with the optimal ligand providing effective reduction of tumor burden in experimental animal models.


Asunto(s)
Biomimética/métodos , Descubrimiento de Drogas/métodos , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Secuencia de Aminoácidos , Anaerobiosis , Animales , Western Blotting , Clonación Molecular , Perfilación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Luciferasas , Ratones , Datos de Secuencia Molecular , Estructura Molecular , Mutagénesis , Piperazina , Piperazinas/química , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
10.
J Am Chem Soc ; 137(34): 10886-9, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26306005

RESUMEN

Formaldehyde (FA) is a reactive carbonyl species (RCS) produced in living systems that has been implicated in epigenetics as well as in the pathologies of various cancers, diabetes, and heart, liver, and neurodegenerative diseases. Traditional methods for biological FA detection rely on sample destruction and/or extensive processing, resulting in a loss of spatiotemporal information. To help address this technological gap, we present the design, synthesis, and biological evaluation of a fluorescent probe for live-cell FA imaging that relies on a FA-induced aza-Cope rearrangement. Formaldehyde probe-1 (FAP-1) is capable of detecting physiologically relevant concentrations of FA in aqueous buffer and in live cells with high selectivity over potentially competing biological analytes. Moreover, FAP-1 can visualize endogenous FA produced by lysine-specific demethylase 1 in a breast cancer cell model, presaging the potential utility of this chemical approach to probe RCS biology.


Asunto(s)
Compuestos Aza/química , Colorantes Fluorescentes/química , Formaldehído/análisis , Compuestos Aza/análisis , Compuestos Aza/síntesis química , Supervivencia Celular , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/síntesis química , Células HEK293 , Humanos , Células MCF-7 , Estructura Molecular
11.
J Am Chem Soc ; 136(22): 7877-88, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24972345

RESUMEN

Protein-protein interactions encompass large surface areas, but often a handful of key residues dominate the binding energy landscape. Rationally designed small molecule scaffolds that reproduce the relative positioning and disposition of important binding residues, termed "hotspot residues", have been shown to successfully inhibit specific protein complexes. Although this strategy has led to development of novel synthetic inhibitors of protein complexes, often direct mimicry of natural amino acid residues does not lead to potent inhibitors. Experimental screening of focused compound libraries is used to further optimize inhibitors but the number of possible designs that can be efficiently synthesized and experimentally tested in academic settings is limited. We have applied the principles of computational protein design to optimization of nonpeptidic helix mimics as ligands for protein complexes. We describe the development of computational tools to design helix mimetics from canonical and noncanonical residue libraries and their application to two therapeutically important protein-protein interactions: p53-MDM2 and p300-HIF1α. The overall study provides a streamlined approach for discovering potent peptidomimetic inhibitors of protein-protein interactions.


Asunto(s)
Proteínas/química , Aminoácidos/química , Biología Computacional , Diseño de Fármacos , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Modelos Moleculares , Imitación Molecular , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
12.
Chem Sci ; 15(21): 8080-8088, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38817555

RESUMEN

Formaldehyde (FA) is both a highly reactive environmental genotoxin and an endogenously produced metabolite that functions as a signaling molecule and one-carbon (1C) store to regulate 1C metabolism and epigenetics in the cell. Owing to its signal-stress duality, cells have evolved multiple clearance mechanisms to maintain FA homeostasis, acting to avoid the established genotoxicity of FA while also redirecting FA-derived carbon units into the biosynthesis of essential nucleobases and amino acids. The highly compartmentalized nature of FA exposure, production, and regulation motivates the development of chemical tools that enable monitoring of transient FA fluxes with subcellular resolution. Here we report a mitochondrial-targeted, activity-based sensing probe for ratiometric FA detection, MitoRFAP-2, and apply this reagent to monitor endogenous mitochondrial sources and sinks of this 1C unit. We establish the utility of subcellular localization by showing that MitoRFAP-2 is sensitive enough to detect changes in mitochondrial FA pools with genetic and pharmacological modulation of enzymes involved in 1C and amino acid metabolism, including the pervasive, less active genetic mutant aldehyde dehydrogenase 2*2 (ALDH2*2), where previous, non-targeted versions of FA sensors are not. Finally, we used MitoRFAP-2 to comparatively profile basal levels of FA across a panel of breast cancer cell lines, finding that FA-dependent fluorescence correlates with expression levels of enzymes involved in 1C metabolism. By showcasing the ability of MitoRFAP-2 to identify new information on mitochondrial FA homeostasis, this work provides a starting point for the design of a broader range of chemical probes for detecting physiologically important aldehydes with subcellular resolution and a useful reagent for further studies of 1C biology.

13.
ACS Med Chem Lett ; 13(1): 84-91, 2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35059127

RESUMEN

Hematopoietic progenitor kinase 1 (HPK1) is implicated as a negative regulator of T-cell receptor-induced T-cell activation. Studies using HPK1 kinase-dead knock-in animals have demonstrated the loss of HPK1 kinase activity resulted in an increase in T-cell function and tumor growth inhibition in glioma models. Herein, we describe the discovery of a series of small molecule inhibitors of HPK1. Using a structure-based drug design approach, the kinase selectivity of the molecules was significantly improved by inducing and stabilizing an unusual P-loop folded binding mode. The metabolic liabilities of the initial 7-azaindole high-throughput screening hit were mitigated by addressing a key metabolic soft spot along with physicochemical property-based optimization. The resulting spiro-azaindoline HPK1 inhibitors demonstrated improved in vitro ADME properties and the ability to induce cytokine production in primary human T-cells.

14.
Curr Opin Chem Biol ; 39: 17-23, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28527906

RESUMEN

Formaldehyde (FA) is a common environmental toxin but is also endogenously produced through a diverse array of essential biological processes, including mitochondrial one-carbon metabolism, metabolite oxidation, and nuclear epigenetic modifications. Its high electrophilicity enables reactivity with a wide variety of biological nucleophiles, which can be beneficial or detrimental to cellular function depending on the context. New methods that enable detection of FA in living systems can help disentangle the signal/stress dichotomy of this simplest reactive carbonyl species (RCS), and fluorescent probes for FA with high selectivity and sensitivity have emerged as promising chemical tools in this regard.


Asunto(s)
Colorantes Fluorescentes/química , Formaldehído/química , Formaldehído/metabolismo , Imagen Molecular/métodos , Animales , Humanos
15.
Chem Commun (Camb) ; 53(72): 10042-10045, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28837194

RESUMEN

We report here a new bioinspired copper-based strategy of superoxide sensing and the development of sensitive (>90-fold fluorescence turn-on) and selective superoxide probes for imaging variations in the endogenous superoxide level in various live mammalian cells (HEK293T, HeLa and A431).

16.
Chem Sci ; 8(5): 4073-4081, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28580121

RESUMEN

Formaldehyde (FA) is a major reactive carbonyl species (RCS) that is naturally produced in living systems through a diverse array of cellular pathways that span from epigenetic regulation to the metabolic processing of endogenous metabolites. At the same time, however, aberrant elevations in FA levels contribute to pathologies ranging from cancer and diabetes to heart, liver, and neurodegenerative diseases. Disentangling the complex interplay between FA physiology and pathology motivates the development of chemical tools that can enable the selective detection of this RCS in biological environments with spatial and temporal fidelity. We report the design, synthesis, and biological evaluation of ratiometric formaldehyde probe (RFAP) indicators for the excitation-ratiometric fluorescence imaging of formaldehyde production in living systems. RFAP-1 and RFAP-2 utilize FA-dependent aza-Cope reactivity to convert an alkylamine-functionalized coumarin platform into its aldehyde congener with a ca. 50 nm shift in the excitation wavelength. The probes exhibit visible excitation and emission profiles, and high selectivity for FA over a variety of RCS and related reactive biological analytes, including acetaldehyde, with up to a 6-fold change in the fluorescence ratio. The RFAP indicators can be used to monitor changes in FA levels in biological samples by live-cell imaging and/or flow cytometry. Moreover, RFAP-2 is capable of visualizing differences in the resting FA levels between wild-type cells and models with a gene knockout of ADH5, a major FA-metabolizing enzyme, establishing the utility of this ratiometric detection platform for identifying and probing sources of FA fluxes in biology.

17.
Chem Sci ; 7(8): 5503-5507, 2016 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-30034690

RESUMEN

Formaldehyde (FA) is a reactive carbonyl species (RCS) that plays a broad spectrum of roles in epigenetics, toxicology, and progression of diseases ranging from cancer to diabetes to neurodegeneration, motivating the development of translatable technologies for FA imaging. Here we report formaldehyde-caged-[18F]fluorodeoxyglucose-1 ([18F]FAC-FDG-1), an aza-Cope-based reactivity probe for in vivo FA imaging using positron emission tomography (PET). [18F]FAC-FDG-1 reacts selectively with FA over potentially competing analytes to generate [18F]FDG, allowing its FA-dependent uptake and retention in cell culture as well as in animal models. The relative uptake of [18F]FAC-FDG-1 was evaluated using FA-treated PC3 prostate cancer and U87-MG glioblastoma cells demonstrating a dose-dependent response to exogenously added FA. Moreover, [18F]FAC-FDG-1 is capable of FA detection in vivo using a PC3 tumor xenograft model. In addition to providing a unique tool for monitoring FA in living animals, these data establish a general approach for translatable detection of FA and other reactive biological analytes in vivo by exploiting the widely-available clinical [18F]FDG tracer as a masked aldehyde that can be caged by analyte-responsive triggers.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA