Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biotechnol Bioeng ; 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715197

RESUMEN

The human microbiota impacts a variety of diseases and responses to therapeutics. Due to a lack of robust in vitro models, detailed mechanistic explanations of host-microbiota interactions cannot often be recapitulated. We describe the design and development of a novel, versatile and modular in vitro system that enables indirect coculture of human epithelial cells with anaerobic bacteria for the characterization of host-microbe secreted metabolite interactions. This system was designed to compartmentalize anaerobes and human cells in separate chambers conducive to each organism's requisite cell growth conditions. Using perfusion, fluidic mixing, and automated sample collection, the cells continuously received fresh media, while in contact with their corresponding compartments conditioned supernatant. Supernatants from each chamber were collected in a cell-free time-resolved fashion. The system sustained low oxygen conditions in the anaerobic chamber, while also supporting the growth of a representative anaerobe (Bacteroides thetaiotaomicron) and a human colonic epithelial cell line (Caco-2) in the aerobic chamber. Caco-2 global gene expression changes in response to coculture with B. thetaiotaomicron was characterized using RNA sequencing. Extensive, targeted metabolomics analysis of over 150 central carbon metabolites was performed on the serially collected supernatants. We observed broad metabolite changes in host-microbe coculture, compared to respective mono-culture controls. These effects were dependent both on sampling time and the compartment probed (apical vs. basolateral). Coculturing resulted in the depletion of several important metabolites, including guanine, uridine 5'-monophosphate, asparagine, and thiamine. Additionally, while Caco-2 cells cultured alone predominantly affected the basolateral metabolite milieu, increased abundance of 2,3-dihydroxyisovalerate and thymine on the basolateral side, occurred when the cells were cocultured with B. thetaiotaomicron. Thus, our system can capture the dynamic, competitive and cooperative processes between host cells and gut microbes.

2.
Bioeng Transl Med ; 8(5): e10542, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37693049

RESUMEN

Cyclic peptides are poised to target historically difficult to drug intracellular protein-protein interactions, however, their general cell impermeability poses a challenge for characterizing function. Recent advances in microfluidics have enabled permeabilization of the cytoplasmic membrane by physical cell deformation (i.e., mechanoporation), resulting in intracellular delivery of impermeable macromolecules in vector- and electrophoretic-free approaches. However, the number of payloads (e.g., peptides) and/or concentrations delivered via microfluidic mechanoporation is limited by having to pre-mix cells and payloads, a manually intensive process. In this work, we show that cells are momentarily permeable (t 1/2 = 1.1-2.8 min) after microfluidic vortex shedding (µVS) and that lower molecular weight macromolecules can be cytosolically delivered upon immediate exposure after cells are processed/permeabilized. To increase the ability to screen peptides, we built a system, dispensing-microfluidic vortex shedding (DµVS), that integrates a µVS chip with inline microplate-based dispensing. To do so, we synced an electronic pressure regulator, flow sensor, on/off dispense valve, and an x-y motion platform in a software-driven feedback loop. Using this system, we were able to deliver low microliter-scale volumes of transiently mechanoporated cells to hundreds of wells on microtiter plates in just several minutes (e.g., 96-well plate filled in <2.5 min). We validated the delivery of an impermeable peptide directed at MDM2, a negative regulator of the tumor suppressor p53, using a click chemistry- and NanoBRET-based cell permeability assay in 96-well format, with robust delivery across the full plate. Furthermore, we demonstrated that DµVS could be used to identify functional, low micromolar, cellular activity of otherwise cell-inactive MDM2-binding peptides using a p53 reporter cell assay in 96- and 384-well format. Overall, DµVS can be combined with downstream cell assays to investigate intracellular target engagement in a high-throughput manner, both for improving structure-activity relationship efforts and for early proof-of-biology of non-optimized peptide (or potentially other macromolecular) tools.

3.
Cell Chem Biol ; 30(10): 1313-1322.e7, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37499664

RESUMEN

Identifying virus-host interactions on the cell surface can improve our understanding of viral entry and pathogenesis. SARS-CoV-2, the causative agent of the COVID-19 disease, uses ACE2 as a receptor to enter cells. Yet the full repertoire of cell surface proteins that contribute to viral entry is unknown. We developed a photocatalyst-based viral-host protein microenvironment mapping platform (ViraMap) to probe the molecular neighborhood of the SARS-CoV-2 spike protein on the human cell surface. Application of ViraMap to ACE2-expressing cells captured ACE2, the established co-receptor NRP1, and several novel cell surface proteins. We systematically analyzed the relevance of these candidate proteins to SARS-CoV-2 entry by knockdown and overexpression approaches in pseudovirus and authentic infection models and identified PTGFRN and EFNB1 as bona fide viral entry factors. Our results highlight additional host targets that participate in SARS-CoV-2 infection and showcase ViraMap as a powerful platform for defining viral interactions on the cell surface.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Enzima Convertidora de Angiotensina 2 , Glicoproteína de la Espiga del Coronavirus , Proteínas Virales/metabolismo , Unión Proteica
4.
J Transl Med ; 21(1): 162, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36864446

RESUMEN

Mammalian cells responding to specific perturbations of homeostasis can undergo a regulated variant of cell death that elicits adaptive immune responses. As immunogenic cell death (ICD) can only occur in a precise cellular and organismal context, it should be conceptually differentiated from instances of immunostimulation or inflammatory responses that do not mechanistically depend on cellular demise. Here, we critically discuss key conceptual and mechanistic aspects of ICD and its implications for cancer (immuno)therapy.


Asunto(s)
Muerte Celular Inmunogénica , Neoplasias , Animales , Neoplasias/terapia , Muerte Celular , Diferenciación Celular , Homeostasis , Mamíferos
5.
Nat Chem Biol ; 18(8): 850-858, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35654846

RESUMEN

The growing appreciation of immune cell-cell interactions within disease environments has led to extensive efforts to develop immunotherapies. However, characterizing complex cell-cell interfaces in high resolution remains challenging. Thus, technologies leveraging therapeutic-based modalities to profile intercellular environments offer opportunities to study cell-cell interactions with molecular-level insight. We introduce photocatalytic cell tagging (PhoTag) for interrogating cell-cell interactions using single-domain antibodies (VHHs) conjugated to photoactivatable flavin-based cofactors. Following irradiation with visible light, the flavin photocatalyst generates phenoxy radical tags for targeted labeling. Using this technology, we demonstrate selective synaptic labeling across the PD-1/PD-L1 axis in antigen-presenting cell-T cell systems. In combination with multiomics single-cell sequencing, we monitored interactions between peripheral blood mononuclear cells and Raji PD-L1 B cells, revealing differences in transient interactions with specific T cell subtypes. The utility of PhoTag in capturing cell-cell interactions will enable detailed profiling of intercellular communication across different biological systems.


Asunto(s)
Antígeno B7-H1 , Leucocitos Mononucleares , Comunicación Celular , Flavinas , Inmunoterapia
6.
Cell Rep ; 38(8): 110399, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35139367

RESUMEN

Follicular helper T (Tfh) cells promote, whereas follicular regulatory T (Tfr) cells restrain, germinal center (GC) reactions. However, the precise roles of these cells in the complex GC reaction remain poorly understood. Here, we perturb Tfh or Tfr cells after SARS-CoV-2 spike protein vaccination in mice. We find that Tfh cells promote the frequency and somatic hypermutation (SHM) of Spike-specific GC B cells and regulate clonal diversity. Tfr cells similarly control SHM and clonal diversity in the GC but do so by limiting clonal competition. In addition, deletion of Tfh or Tfr cells during primary vaccination results in changes in SHM after vaccine boosting. Aged mice, which have altered Tfh and Tfr cells, have lower GC responses, presenting a bimodal distribution of SHM. Together, these data demonstrate that GC responses to SARS-CoV-2 spike protein vaccines require a fine balance of positive and negative follicular T cell help to optimize humoral immunity.


Asunto(s)
COVID-19/prevención & control , Centro Germinal/inmunología , Glicoproteína de la Espiga del Coronavirus/administración & dosificación , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Envejecimiento , Animales , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Linfocitos B/metabolismo , COVID-19/virología , Centro Germinal/citología , Centro Germinal/metabolismo , Inmunidad Humoral , Ratones , Ratones Endogámicos C57BL , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Vacunación , Vacunas de Subunidad/inmunología
7.
RSC Chem Biol ; 2(1): 30-46, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-34458775

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the current coronavirus disease 2019 (COVID-19) pandemic that has led to a global economic disruption and collapse. With several ongoing efforts to develop vaccines and treatments for COVID-19, understanding the molecular interaction between the coronavirus, host cells, and the immune system is critical for effective therapeutic interventions. Greater insight into these mechanisms will require the contribution and combination of multiple scientific disciplines including the techniques and strategies that have been successfully deployed by chemical biology to tease apart complex biological pathways. We highlight in this review well-established strategies and methods to study coronavirus-host biophysical interactions and discuss the impact chemical biology will have on understanding these interactions at the molecular level.

8.
Chembiochem ; 21(24): 3555-3562, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-32749732

RESUMEN

Despite the growing use of visible-light photochemistry in both chemistry and biology, no general low-heat photoreactor for use across these different disciplines exists. Herein, we describe the design and use of a standardized photoreactor for visible-light-driven activation and photocatalytic chemical transformations. Using this single benchtop photoreactor, we performed photoredox reactions across multiple visible light wavelengths, a high-throughput photocatalytic cross-coupling reaction, and in vitro labeling of proteins and live cells. Given the success of this reactor in all tested applications, we envision that this multi-use photoreactor will be widely used in biology, chemical biology, and medicinal chemistry settings.


Asunto(s)
Biotina/análisis , Luz , Fotobiorreactores , Tiramina/química , Catálisis , Línea Celular Tumoral , Diseño de Equipo , Humanos , Estructura Molecular , Procesos Fotoquímicos , Tiramina/análogos & derivados , Tiramina/síntesis química
9.
Sci Rep ; 10(1): 5321, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32210258

RESUMEN

Recent reports show that colorectal tumors contain microbiota that are distinct from those that reside in a 'normal' colon environment, and that these microbiota can contribute to cancer progression. Fusobacterium nucleatum is the most commonly observed species in the colorectal tumor microenvironment and reportedly influences disease progression through numerous mechanisms. However, a detailed understanding of the role of this organism in cancer progression is limited, in part due to challenges in maintaining F. nucleatum viability under standard aerobic cell culture conditions. Herein we describe the development of a 3-dimensional (3D) tumor spheroid model that can harbor and promote the growth of anaerobic bacteria. Bacteria-tumor cell interactions and metabolic crosstalk were extensively studied by measuring the kinetics of bacterial growth, cell morphology and lysis, cancer-related gene expression, and metabolomics. We observed that viable F. nucleatum assembles biofilm-like structures in the tumor spheroid microenvironment, whereas heat-killed F. nucleatum is internalized and sequestered in the cancer cells. Lastly, we use the model to co-culture 28 Fusobacterium clinical isolates and demonstrate that the model successfully supports co-culture with diverse fusobacterial species. This bacteria-spheroid co-culture model enables mechanistic investigation of the role of anaerobic bacteria in the tumor microenvironment.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Neoplasias Colorrectales/microbiología , Esferoides Celulares/metabolismo , Bacterias Anaerobias , Línea Celular Tumoral , Técnicas de Cocultivo/métodos , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Infecciones por Fusobacterium/microbiología , Fusobacterium nucleatum/genética , Fusobacterium nucleatum/metabolismo , Fusobacterium nucleatum/patogenicidad , Humanos , Modelos Biológicos , Microambiente Tumoral/fisiología
10.
Science ; 367(6482): 1091-1097, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32139536

RESUMEN

Many disease pathologies can be understood through the elucidation of localized biomolecular networks, or microenvironments. To this end, enzymatic proximity labeling platforms are broadly applied for mapping the wider spatial relationships in subcellular architectures. However, technologies that can map microenvironments with higher precision have long been sought. Here, we describe a microenvironment-mapping platform that exploits photocatalytic carbene generation to selectively identify protein-protein interactions on cell membranes, an approach we term MicroMap (µMap). By using a photocatalyst-antibody conjugate to spatially localize carbene generation, we demonstrate selective labeling of antibody binding targets and their microenvironment protein neighbors. This technique identified the constituent proteins of the programmed-death ligand 1 (PD-L1) microenvironment in live lymphocytes and selectively labeled within an immunosynaptic junction.


Asunto(s)
Antígeno B7-H1/metabolismo , Membrana Celular/metabolismo , Microambiente Celular , Linfocitos/metabolismo , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas , Catálisis , Membrana Celular/efectos de la radiación , Transferencia de Energía , Humanos , Células Jurkat , Linfocitos/efectos de la radiación , Metano/análogos & derivados , Metano/química , Metano/efectos de la radiación , Procesos Fotoquímicos , Rayos Ultravioleta
11.
Curr Microbiol ; 76(4): 398-409, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30603964

RESUMEN

Cell wall hydrolases are enzymes that cleave bacterial cell walls by hydrolyzing specific bonds within peptidoglycan and other portions of the envelope. Two major sources of hydrolases in nature are from hosts and microbes. This study specifically investigated whether cell wall hydrolytic enzymes could be employed as exogenous reagents to augment the efficacy of antimicrobial agents against mycobacteria. Mycobacterium smegmatis cultures were treated with ten conventional antibiotics and six anti-tuberculosis drugs-alone or in combination with cell wall hydrolases. Culture turbidity, colony-forming units (CFUs), vital staining, and oxygen consumption were all monitored. The majority of antimicrobial agents tested alone only had minimal inhibitory effects on bacterial growth. However, the combination of cell wall hydrolases and most of the antimicrobial agents tested, revealed a synergistic effect that resulted in significant enhancement of bactericidal activity. Vital staining showed increased cellular damage when M. smegmatis and Mycobacterium bovis bacillus Calmette-Guérin (M. bovis BCG) were treated with both drug and lysozyme. Respiration analysis revealed stress responses when cells were treated with lysozyme and drugs individually, and an acute increase in oxygen consumption when treated with both drug and lysozyme. Similar trends were also observed for the other three enzymes (hydrolase-30, RipA-His6 and RpfE-His6) evaluated. These findings demonstrated that cell wall hydrolytic enzymes, as a group of biological agents, have the capability to improve the potency of many current antimicrobial drugs and render ineffective antibiotics effective in killing mycobacteria. This combinatorial approach may represent an important strategy to eliminate drug-resistant bacteria.


Asunto(s)
Antibacterianos/farmacología , Pared Celular/enzimología , Hidrolasas/metabolismo , Mycobacterium/efectos de los fármacos , Antituberculosos/farmacología , Recuento de Colonia Microbiana , Sinergismo Farmacológico , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium/enzimología , Mycobacterium/crecimiento & desarrollo , Mycobacterium/metabolismo , Mycobacterium bovis/efectos de los fármacos , Mycobacterium bovis/crecimiento & desarrollo , Mycobacterium bovis/metabolismo , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/crecimiento & desarrollo , Mycobacterium smegmatis/metabolismo , Oxígeno/metabolismo
12.
Sci Signal ; 11(541)2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30065029

RESUMEN

Members of the family of nuclear factor κB (NF-κB) transcription factors are critical for multiple cellular processes, including regulating innate and adaptive immune responses, cell proliferation, and cell survival. Canonical NF-κB complexes are retained in the cytoplasm by the inhibitory protein IκBα, whereas noncanonical NF-κB complexes are retained by p100. Although activation of canonical NF-κB signaling through the IκBα kinase complex is well studied, few regulators of the NF-κB-inducing kinase (NIK)-dependent processing of noncanonical p100 to p52 and the subsequent nuclear translocation of p52 have been identified. We discovered a role for cyclin-dependent kinase 12 (CDK12) in transcriptionally regulating the noncanonical NF-κB pathway. High-content phenotypic screening identified the compound 919278 as a specific inhibitor of the lymphotoxin ß receptor (LTßR), and tumor necrosis factor (TNF) receptor superfamily member 12A (FN14)-dependent nuclear translocation of p52, but not of the TNF-α receptor-mediated nuclear translocation of p65. Chemoproteomics identified CDK12 as the target of 919278. CDK12 inhibition by 919278, the CDK inhibitor THZ1, or siRNA-mediated knockdown resulted in similar global transcriptional changes and prevented the LTßR- and FN14-dependent expression of MAP3K14 (which encodes NIK) as well as NIK accumulation by reducing phosphorylation of the carboxyl-terminal domain of RNA polymerase II. By coupling a phenotypic screen with chemoproteomics, we identified a pathway for the activation of the noncanonical NF-κB pathway that could serve as a therapeutic target in autoimmunity and cancer.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/metabolismo , Regulación Neoplásica de la Expresión Génica , FN-kappa B/metabolismo , Osteosarcoma/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , Ciclinas/metabolismo , Perfilación de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Indoles/farmacología , Receptor beta de Linfotoxina/antagonistas & inhibidores , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Propionatos/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteoma , Transducción de Señal , Receptor de TWEAK/antagonistas & inhibidores , Receptor de TWEAK/genética , Receptor de TWEAK/metabolismo , Células Tumorales Cultivadas , Quinasa de Factor Nuclear kappa B
13.
ACS Chem Biol ; 12(12): 2970-2974, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29088528

RESUMEN

Biochemical screening is a major source of lead generation for novel targets. However, during the process of small molecule lead optimization, compounds with excellent biochemical activity may show poor cellular potency, making structure-activity relationships difficult to decipher. This may be due to low membrane permeability of the molecule, resulting in insufficient intracellular drug concentration. The Cell Squeeze platform increases permeability regardless of compound structure by mechanically disrupting the membrane, which can overcome permeability limitations and bridge the gap between biochemical and cellular studies. In this study, we show that poorly permeable Janus kinase (JAK) inhibitors are delivered into primary cells using Cell Squeeze, inhibiting up to 90% of the JAK pathway, while incubation of JAK inhibitors with or without electroporation had no significant effect. We believe this robust intracellular delivery approach could enable more effective lead optimization and deepen our understanding of target engagement by small molecules and functional probes.


Asunto(s)
Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/metabolismo , Dispositivos Laboratorio en un Chip , Leucocitos Mononucleares/efectos de los fármacos , Membrana Celular , Células Cultivadas , Humanos , Inhibidores de las Cinasas Janus/química , Leucocitos Mononucleares/fisiología , Estructura Molecular , Relación Estructura-Actividad
14.
ACS Chem Biol ; 12(8): 2015-2020, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28718624

RESUMEN

Irreversible enzyme inhibitors and covalent chemical biology probes often utilize the reaction of a protein cysteine residue with an appropriately positioned electrophile (e.g., acrylamide) on the ligand template. However, cysteine residues are not always available for site-specific protein labeling, and therefore new approaches are needed to expand the toolkit of appropriate electrophiles ("warheads") that target alternative amino acids. We previously described the rational targeting of tyrosine residues in the active site of a protein (the mRNA decapping scavenger enzyme, DcpS) using inhibitors armed with a sulfonyl fluoride electrophile. These inhibitors subsequently enabled the development of clickable probe technology to measure drug-target occupancy in live cells. Here we describe a fluorosulfate-containing inhibitor (aryl fluorosulfate probe (FS-p1)) with excellent chemical and metabolic stability that reacts selectively with a noncatalytic serine residue in the same active site of DcpS as confirmed by peptide mapping experiments. Our results suggest that noncatalytic serine targeting using fluorosulfate electrophilic warheads could be a suitable strategy for the development of covalent inhibitor drugs and chemical probes.


Asunto(s)
Inhibidores Enzimáticos/química , Fluoruros/química , Serina/química , Ácidos Sulfúricos/química , Animales , Dominio Catalítico , Línea Celular , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Estabilidad de Enzimas , Humanos
15.
ACS Chem Biol ; 11(12): 3442-3451, 2016 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-27791347

RESUMEN

PF-06651600, a newly discovered potent JAK3-selective inhibitor, is highly efficacious at inhibiting γc cytokine signaling, which is dependent on both JAK1 and JAK3. PF-06651600 allowed the comparison of JAK3-selective inhibition to pan-JAK or JAK1-selective inhibition, in relevant immune cells to a level that could not be achieved previously without such potency and selectivity. In vitro, PF-06651600 inhibits Th1 and Th17 cell differentiation and function, and in vivo it reduces disease pathology in rat adjuvant-induced arthritis as well as in mouse experimental autoimmune encephalomyelitis models. Importantly, by sparing JAK1 function, PF-06651600 selectively targets γc cytokine pathways while preserving JAK1-dependent anti-inflammatory signaling such as the IL-10 suppressive functions following LPS treatment in macrophages and the suppression of TNFα and IL-1ß production in IL-27-primed macrophages. Thus, JAK3-selective inhibition differentiates from pan-JAK or JAK1 inhibition in various immune cellular responses, which could potentially translate to advantageous clinical outcomes in inflammatory and autoimmune diseases.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Janus Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Animales , Artritis Experimental/inmunología , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Interleucina-10/inmunología , Interleucina-1beta/inmunología , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/metabolismo , Janus Quinasa 3/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Modelos Moleculares , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Pirroles/farmacocinética , Pirroles/farmacología , Ratas , Células TH1/citología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/citología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/inmunología
16.
ACS Comb Sci ; 18(10): 611-615, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27494431

RESUMEN

Small molecule selectivity is an essential component of candidate drug selection and target validation. New technologies are required to better understand off-target effects, with particular emphasis needed on broad protein profiling. Here, we describe the use of a tritiated chemical probe and a 9000 human protein microarray to discern the binding selectivity of an inhibitor of the mRNA decapping scavenger enzyme DcpS. An immobilized m7GTP resin was also used to assess the selectivity of a DcpS inhibitor against mRNA cap-associated proteins in whole cell extracts. These studies confirm the exquisite selectivity of diaminoquinazoline DcpS inhibitors, and highlight the utility of relatively simple protein microarray and affinity enrichment technologies in drug discovery and chemical biology.


Asunto(s)
Endorribonucleasas/análisis , Sondas Moleculares/química , Quinazolinas/química , Proteínas de Unión a Caperuzas de ARN/análisis , Catálisis , Células Cultivadas , Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/genética , Humanos , Leucocitos Mononucleares/química , Análisis por Matrices de Proteínas , ARN Mensajero/genética , Proteína 2 para la Supervivencia de la Neurona Motora/análisis , Tritio
17.
Org Biomol Chem ; 14(26): 6179-83, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27216142

RESUMEN

Proof of drug-target engagement in physiologically-relevant contexts is a key pillar of successful therapeutic target validation. We developed two orthogonal technologies, the cellular thermal shift assay (CETSA) and a covalent chemical probe reporter approach (harnessing sulfonyl fluoride tyrosine labeling and subsequent click chemistry) to measure the occupancy of the mRNA-decapping scavenger enzyme DcpS by a small molecule inhibitor in live cells. Enzyme affinity determined using isothermal dose response fingerprinting (ITDRFCETSA) and the concentration required to occupy 50% of the enzyme (OC50) using the chemical probe reporter assay were very similar. In this case, the chemical probe method worked well due to the long offset kinetics of the reversible inhibitor (determined using a fluorescent dye-tagged probe). This work suggests that CETSA could become the first choice assay to determine in-cell target engagement due to its simplicity.


Asunto(s)
Endorribonucleasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes/química , Quinazolinas/farmacología , Temperatura , Química Clic , Relación Dosis-Respuesta a Droga , Endorribonucleasas/metabolismo , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Estructura Molecular , Quinazolinas/química , Ácidos Sulfínicos/química , Tirosina/química
19.
Mol Biosyst ; 11(10): 2709-12, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25959423

RESUMEN

Despite its diverse applications, such as identification of the protein binding partners of small molecules and investigation of intracellular drug-target engagement, photoaffinity labelling (PAL) is intrinsically challenging, primarily due to the difficulty in discovering functionally active photoaffinity probes. Here we describe the creation of a chemoproteomic library to discover a novel photoaffinity probe for DcpS, an mRNA decapping enzyme that is a putative target for Spinal Muscular Atrophy. This library approach expedites the discovery of photoaffinity probes and expands the chemical biology toolbox to include RNA cap-binding proteins.


Asunto(s)
Endorribonucleasas/metabolismo , Sondas Moleculares/química , Etiquetas de Fotoafinidad/química , Sitios de Unión , Endorribonucleasas/química , Biblioteca de Genes , Humanos , Modelos Moleculares , Sondas Moleculares/metabolismo , Quinazolinas/química
20.
ACS Chem Biol ; 10(4): 1094-8, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25571984

RESUMEN

This work describes the first rational targeting of tyrosine residues in a protein binding site by small-molecule covalent probes. Specific tyrosine residues in the active site of the mRNA-decapping scavenger enzyme DcpS were modified using reactive sulfonyl fluoride covalent inhibitors. Structure-based molecular design was used to create an alkyne-tagged probe bearing the sulfonyl fluoride warhead, thus enabling the efficient capture of the protein from a complex proteome. Use of the probe in competition experiments with a diaminoquinazoline DcpS inhibitor permitted the quantification of intracellular target occupancy. As a result, diaminoquinazoline upregulators of survival motor neuron protein that are used for the treatment of spinal muscular atrophy were confirmed as inhibitors of DcpS in human primary cells. This work illustrates the utility of sulfonyl fluoride probes designed to react with specific tyrosine residues of a protein and augments the chemical biology toolkit by these probes uses in target validation and molecular pharmacology.


Asunto(s)
Endorribonucleasas/metabolismo , Inhibidores Enzimáticos/farmacología , Sondas Moleculares/química , Ácidos Sulfínicos/química , Tirosina/metabolismo , Dominio Catalítico , Células Cultivadas , Técnicas de Química Sintética , Cristalografía por Rayos X , Endorribonucleasas/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Sondas Moleculares/síntesis química , Terapia Molecular Dirigida/métodos , Relación Estructura-Actividad , Tirosina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...