Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Chem Biol ; 19(2): 230-238, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36302899

RESUMEN

Small-molecule tools have enabled mechanistic investigations and therapeutic targeting of the protein kinase-like (PKL) superfamily. However, such tools are still lacking for many PKL members, including the highly conserved and disease-related UbiB family. Here, we sought to develop and characterize an inhibitor for the archetypal UbiB member COQ8, whose function is essential for coenzyme Q (CoQ) biosynthesis. Guided by crystallography, activity assays and cellular CoQ measurements, we repurposed the 4-anilinoquinoline scaffold to selectively inhibit human COQ8A in cells. Our chemical tool promises to lend mechanistic insights into the activities of these widespread and understudied proteins and to offer potential therapeutic strategies for human diseases connected to their dysfunction.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Humanos , Saccharomyces cerevisiae/metabolismo , Ubiquinona/farmacología , Ubiquinona/química , Proteínas de Saccharomyces cerevisiae/metabolismo
2.
Nat Chem Biol ; 18(6): 596-604, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35314814

RESUMEN

Current small-molecule inhibitors of KRAS(G12C) bind irreversibly in the switch-II pocket (SII-P), exploiting the strong nucleophilicity of the acquired cysteine as well as the preponderance of the GDP-bound form of this mutant. Nevertheless, many oncogenic KRAS mutants lack these two features, and it remains unknown whether targeting the SII-P is a practical therapeutic approach for KRAS mutants beyond G12C. Here we use NMR spectroscopy and a cellular KRAS engagement assay to address this question by examining a collection of SII-P ligands from the literature and from our own laboratory. We show that the SII-Ps of many KRAS hotspot (G12, G13, Q61) mutants are accessible using noncovalent ligands, and that this accessibility is not necessarily coupled to the GDP state of KRAS. The results we describe here emphasize the SII-P as a privileged drug-binding site on KRAS and unveil new therapeutic opportunities in RAS-driven cancer.


Asunto(s)
Mieloma Múltiple , Proteínas Proto-Oncogénicas p21(ras) , Humanos , Ligandos , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética
3.
J Immunol ; 200(5): 1937-1950, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351998

RESUMEN

Fc γ receptors (FcγR) are involved in multiple aspects of immune cell regulation, are central to the success of mAb therapeutics, and underpin the pathology of several autoimmune diseases. However, reliable assays capable of accurately measuring FcγR interactions with their physiological ligands, IgG immune complexes (IC), are limited. A method to study and detect IC interactions with FcγRs was therefore developed. This method, designed to model the signaling pathway of the inhibitory FcγRIIB (CD32B), used NanoLuc Binary Interaction Technology to measure recruitment of the Src homology 2 domain-containing inositol phosphatase 1 to the ITIM of this receptor. Such recruitment required prior cross-linking of an ITAM-containing activatory receptor, and evoked luciferase activity in discrete clusters at the cell surface, recapitulating the known biology of CD32B signaling. The assay detected varying forms of experimental IC, including heat-aggregated IgG, rituximab-anti-idiotype complexes, and anti-trinitrophenol-trinitrophenol complexes in a sensitive manner (≤1 µg/ml), and discriminated between complexes of varying size and isotype. Proof-of-concept for the detection of circulating ICs in autoimmune disease was provided, as responses to sera from patients with systemic lupus erythematosus and rheumatoid arthritis were detected in small pilot studies. Finally, the method was translated to a stable cell line system. In conclusion, a rapid and robust method for the detection of IC was developed, which has numerous potential applications including the monitoring of IC in autoimmune diseases and the study of underlying FcγR biology.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/inmunología , Receptores de IgG/inmunología , Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/inmunología , Línea Celular , Células HEK293 , Humanos , Inmunoglobulina G/inmunología , Lupus Eritematoso Sistémico/inmunología , Fosfoproteínas/inmunología , Rituximab/inmunología , Transducción de Señal/inmunología , Dominios Homologos src/inmunología
4.
Anal Biochem ; 489: 1-8, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26278171

RESUMEN

Ligand-mediated endocytosis is a key autoregulatory mechanism governing the duration and intensity of signals emanating from cell surface receptors. Due to the mechanistic complexity of endocytosis and its emerging relevance in disease, simple methods capable of tracking this dynamic process in cells have become increasingly desirable. We have developed a bioluminescent reporter technology for real-time analysis of ligand-mediated receptor endocytosis using genetic fusions of NanoLuc luciferase with various G-protein-coupled receptors (GPCRs). This method is compatible with standard microplate formats, which should decrease work flows for high-throughput screens. This article also describes the application of this technology to endocytosis of epidermal growth factor receptor (EGFR), demonstrating potential applicability of the method beyond GPCRs.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Endocitosis , Ensayos Analíticos de Alto Rendimiento/métodos , Luciferasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Membrana Celular/metabolismo , Descubrimiento de Drogas/métodos , Endocitosis/efectos de los fármacos , Colorantes Fluorescentes/química , Genes Reporteros/efectos de los fármacos , Células HEK293 , Humanos , Interleucina-6/química , Interleucina-6/genética , Interleucina-6/metabolismo , Cinética , Ligandos , Luciferasas/química , Luciferasas/genética , Microscopía Confocal , Microscopía Fluorescente , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Señales de Clasificación de Proteína/efectos de los fármacos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
5.
Mol Ther ; 21(8): 1602-10, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23712039

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons in the brain and spinal cord. We have recently shown that human mesenchymal stem cells (hMSCs) modified to release glial cell line-derived neurotrophic factor (GDNF) decrease disease progression in a rat model of ALS when delivered to skeletal muscle. In the current study, we determined whether or not this effect could be enhanced by delivering GDNF in concert with other trophic factors. hMSC engineered to secrete GDNF (hMSC-GDNF), vascular endothelial growth factor (hMSC-VEGF), insulin-like growth factor-I (hMSC-IGF-I), or brain-derived neurotrophic factor (hMSC-BDNF), were prepared and transplanted bilaterally into three muscle groups. hMSC-GDNF and hMSC-VEGF prolonged survival and slowed the loss of motor function, but hMSC-IGF-I and hMSC-BDNF did not have any effect. We then tested the efficacy of a combined ex vivo delivery of GDNF and VEGF in extending survival and protecting neuromuscular junctions (NMJs) and motor neurons. Interestingly, the combined delivery of these neurotrophic factors showed a strong synergistic effect. These studies further support ex vivo gene therapy approaches for ALS that target skeletal muscle.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Longevidad/genética , Factor A de Crecimiento Endotelial Vascular/genética , Esclerosis Amiotrófica Lateral/mortalidad , Animales , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Neuronas Motoras/metabolismo , Músculo Esquelético/metabolismo , Unión Neuromuscular/metabolismo , Ratas
6.
Cell Chem Biol ; 30(11): 1354-1365.e6, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37643616

RESUMEN

RAF dimer inhibitors offer therapeutic potential in RAF- and RAS-driven cancers. The utility of such drugs is predicated on their capacity to occupy both RAF protomers in the RAS-RAF signaling complex. Here we describe a method to conditionally quantify drug-target occupancy at selected RAF protomers within an active RAS-RAF complex in cells. RAF target engagement can be measured in the presence or absence of any mutant KRAS allele, enabling the high-affinity state of RAF dimer inhibitors to be quantified in the cellular milieu. The intracellular protomer selectivity of clinical-stage type II RAF inhibitors revealed that ARAF protomer engagement, but not engagement of BRAF or CRAF, is commensurate with inhibition of MAPK signaling in various mutant RAS cell lines. Our results support a fundamental role for ARAF in mutant RAS signaling and reveal poor ARAF protomer vulnerability for a cohort of RAF inhibitors undergoing clinical evaluation.


Asunto(s)
Proteínas Proto-Oncogénicas B-raf , Transducción de Señal , Humanos , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Línea Celular Tumoral , Inhibidores de Proteínas Quinasas/farmacología , Mutación , Sistema de Señalización de MAP Quinasas
7.
J Med Chem ; 64(8): 4810-4840, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33830764

RESUMEN

Histone deacetylase 6 (HDAC6) is a promising therapeutic target for the treatment of neurodegenerative disorders. SW-100 (1a), a phenylhydroxamate-based HDAC6 inhibitor (HDAC6i) bearing a tetrahydroquinoline (THQ) capping group, is a highly potent and selective HDAC6i that was shown to be effective in mouse models of Fragile X syndrome and Charcot-Marie-Tooth disease type 2A (CMT2A). In this study, we report the discovery of a new THQ-capped HDAC6i, termed SW-101 (1s), that possesses excellent HDAC6 potency and selectivity, together with markedly improved metabolic stability and druglike properties compared to SW-100 (1a). X-ray crystallography data reveal the molecular basis of HDAC6 inhibition by SW-101 (1s). Importantly, we demonstrate that SW-101 (1s) treatment elevates the impaired level of acetylated α-tubulin in the distal sciatic nerve, counteracts progressive motor dysfunction, and ameliorates neuropathic symptoms in a CMT2A mouse model bearing mutant MFN2. Taken together, these results bode well for the further development of SW-101 (1s) as a disease-modifying HDAC6i.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/tratamiento farmacológico , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/uso terapéutico , Quinolinas/química , Acetilación , Animales , Benzamidas/química , Benzamidas/metabolismo , Sitios de Unión , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Semivida , Histona Desacetilasa 6/metabolismo , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Fenotipo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Quinolinas/metabolismo , Quinolinas/uso terapéutico , Relación Estructura-Actividad , Tubulina (Proteína)/metabolismo
8.
ACS Chem Biol ; 16(2): 293-309, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33539064

RESUMEN

Macrocyclic peptides open new opportunities to target intracellular protein-protein interactions (PPIs) that are often considered nondruggable by traditional small molecules. However, engineering sufficient membrane permeability into these molecules is a central challenge for identifying clinical candidates. Currently, there is a lack of high-throughput assays to assess peptide permeability, which limits our capacity to engineer this property into macrocyclic peptides for advancement through drug discovery pipelines. Accordingly, we developed a high throughput and target-agnostic cell permeability assay that measures the relative cumulative cytosolic exposure of a peptide in a concentration-dependent manner. The assay was named NanoClick as it combines in-cell Click chemistry with an intracellular NanoBRET signal. We validated the approach using known cell penetrating peptides and further demonstrated a correlation to cellular activity using a p53/MDM2 model system. With minimal change to the peptide sequence, NanoClick enables the ability to measure uptake of molecules that enter the cell via different mechanisms such as endocytosis, membrane translocation, or passive permeability. Overall, the NanoClick assay can serve as a screening tool to uncover predictive design rules to guide structure-activity-permeability relationships in the optimization of functionally active molecules.


Asunto(s)
Bioensayo/métodos , Péptidos de Penetración Celular/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Péptidos Cíclicos/metabolismo , Alquinos/química , Secuencia de Aminoácidos , Azidas/química , Permeabilidad de la Membrana Celular , Péptidos de Penetración Celular/química , Química Clic , Células HeLa , Humanos , Hidrolasas/química , Péptidos Cíclicos/química , Transporte de Proteínas
9.
Nat Commun ; 11(1): 2743, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488087

RESUMEN

Concerted multidisciplinary efforts have led to the development of Cyclin-Dependent Kinase inhibitors (CDKi's) as small molecule drugs and chemical probes of intracellular CDK function. However, conflicting data has been reported on the inhibitory potency of CDKi's and a systematic characterization of affinity and selectivity against intracellular CDKs is lacking. We have developed a panel of cell-permeable energy transfer probes to quantify target occupancy for all 21 human CDKs in live cells, and present a comprehensive evaluation of intracellular isozyme potency and selectivity for a collection of 46 clinically-advanced CDKi's and tool molecules. We observed unexpected intracellular activity profiles for a number of CDKi's, offering avenues for repurposing of highly potent molecules as probes for previously unreported targets. Overall, we provide a broadly applicable method for evaluating the selectivity of CDK inhibitors in living cells, and present a refined set of tool molecules to study CDK function.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Proteína Quinasa CDC2 , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Quinasa 9 Dependiente de la Ciclina , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Fosforilación , Relación Estructura-Actividad
10.
J Med Chem ; 62(18): 8557-8577, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31414801

RESUMEN

Isoxazole is a five-membered heterocycle that is widely used in drug discovery endeavors. Here, we report the design, synthesis, and structural and biological characterization of SS-208, a novel HDAC6-selective inhibitor containing the isoxazole-3-hydroxamate moiety as a zinc-binding group as well as a hydrophobic linker. A crystal structure of the Danio rerio HDAC6/SS-208 complex reveals a bidentate coordination of the active-site zinc ion that differs from the preferred monodentate coordination observed for HDAC6 complexes with phenylhydroxamate-based inhibitors. While SS-208 has minimal effects on the viability of murine SM1 melanoma cells in vitro, it significantly reduced in vivo tumor growth in a murine SM1 syngeneic melanoma mouse model. These findings suggest that the antitumor activity of SS-208 is mainly mediated by immune-related antitumor activity as evidenced by the increased infiltration of CD8+ and NK+ T cells and the enhanced ratio of M1 and M2 macrophages in the tumor microenvironment.


Asunto(s)
Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Isoxazoles/farmacología , Melanoma/tratamiento farmacológico , Animales , Linfocitos T CD8-positivos/citología , Dominio Catalítico , Línea Celular Tumoral , Descubrimiento de Drogas , Inhibidores de Histona Desacetilasas/química , Humanos , Ácidos Hidroxámicos/química , Isoxazoles/química , Macrófagos/citología , Ratones , Microsomas/química , Células T Asesinas Naturales/citología , Trasplante Isogénico , Pez Cebra , Zinc/química
11.
ACS Chem Neurosci ; 10(3): 1679-1695, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30511829

RESUMEN

Disease-modifying therapies are needed for Fragile X Syndrome (FXS), as at present there are no effective treatments or cures. Herein, we report on a tetrahydroquinoline-based selective histone deacetylase 6 (HDAC6) inhibitor SW-100, its pharmacological and ADMET properties, and its ability to improve upon memory performance in a mouse model of FXS, Fmr1-/- mice. This small molecule demonstrates good brain penetrance, low-nanomolar potency for the inhibition of HDAC6 (IC50 = 2.3 nM), with at least a thousand-fold selectivity over all other class I, II, and IV HDAC isoforms. Moreover, through its inhibition of the α-tubulin deacetylase domain of HDAC6 (CD2), in cells SW-100 upregulates α-tubulin acetylation with no effect on histone acetylation and selectively restores the impaired acetylated α-tubulin levels in the hippocampus of Fmr1-/- mice. Lastly, SW-100 ameliorates several memory and learning impairments in Fmr1-/- mice, thus modeling the intellectual deficiencies associated with FXS, and hence providing a strong rationale for pursuing HDAC6-based therapies for the treatment of this rare disease.


Asunto(s)
Benzamidas/farmacología , Síndrome del Cromosoma X Frágil/fisiopatología , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Quinolinas/farmacología , Animales , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Síndrome del Cromosoma X Frágil/enzimología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Procesamiento Proteico-Postraduccional/efectos de los fármacos
12.
BMC Cell Biol ; 9: 17, 2008 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-18384686

RESUMEN

BACKGROUND: The ability to specifically label proteins within living cells can provide information about their dynamics and function. To study a membrane protein, we fused a multi-functional reporter protein, HaloTag, to the extracellular domain of a truncated integrin. RESULTS: Using the HaloTag technology, we could study the localization, trafficking and processing of an integrin-HaloTag fusion, which we showed had cellular dynamics consistent with native integrins. By labeling live cells with different fluorescent impermeable and permeable ligands, we showed spatial separation of plasma membrane and internal pools of the integrin-HaloTag fusion, and followed these protein pools over time to study bi-directional trafficking. In addition to combining the HaloTag reporter protein with different fluorophores, we also employed an affinity tag to achieve cell capture. CONCLUSION: The HaloTag technology was used successfully to study expression, trafficking, spatial separation and real-time translocation of an integrin-HaloTag fusion, thereby demonstrating that this technology can be a powerful tool to investigate membrane protein biology in live cells.


Asunto(s)
Bioensayo/métodos , Colorantes Fluorescentes/metabolismo , Genes Reporteros/genética , Proteómica/métodos , Proteínas Recombinantes de Fusión/metabolismo , Coloración y Etiquetado/métodos , Animales , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Citoplasma/metabolismo , Humanos , Integrinas/metabolismo , Proteínas Luminiscentes/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína/fisiología , Transporte de Proteínas/fisiología , Proteínas Recombinantes de Fusión/genética
13.
Cell Chem Biol ; 25(2): 206-214.e11, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29174542

RESUMEN

For kinase inhibitors, intracellular target selectivity is fundamental to pharmacological mechanism. Although a number of acellular techniques have been developed to measure kinase binding or enzymatic inhibition, such approaches can fail to accurately predict engagement in cells. Here we report the application of an energy transfer technique that enabled the first broad-spectrum, equilibrium-based approach to quantitatively profile target occupancy and compound affinity in live cells. Using this method, we performed a selectivity profiling for clinically relevant kinase inhibitors against 178 full-length kinases, and a mechanistic interrogation of the potency offsets observed between cellular and biochemical analysis. For the multikinase inhibitor crizotinib, our approach accurately predicted cellular potency and revealed improved target selectivity compared with biochemical measurements. Due to cellular ATP, a number of putative crizotinib targets are unexpectedly disengaged in live cells at a clinically relevant drug dose.


Asunto(s)
Adenosina Trifosfato/metabolismo , Fosfotransferasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Transferencia de Energía , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Espectrometría de Masas , Estructura Molecular , Fosfotransferasas/metabolismo , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
14.
ACS Chem Biol ; 12(4): 1028-1037, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28195704

RESUMEN

Novel engineered NanoLuc (Nluc) luciferase being smaller, brighter, and superior to traditional firefly (Fluc) or Renilla (Rluc) provides a great opportunity for the development of numerous biological, biomedical, clinical, and food and environmental safety applications. This new platform created an urgent need for Nluc inhibitors that could allow selective bioluminescent suppression and multiplexing compatibility with existing luminescence or fluorescence assays. Starting from thienopyrrole carboxylate 1, a hit from a 42 000 PubChem compound library with a low micromolar IC50 against Nluc, we derivatized four different structural fragments to discover a family of potent, single digit nanomolar, cell permeable inhibitors. Further elaboration revealed a channel that allowed access to the external Nluc surface, resulting in a series of highly potent cell impermeable Nluc inhibitors with negatively charged groups likely extending to the protein surface. The permeability was evaluated by comparing EC50 shifts calculated from both live and lysed cells expressing Nluc cytosolically. Luminescence imaging further confirmed that cell permeable compounds inhibit both intracellular and extracellular Nluc, whereas less permeable compounds differentially inhibit extracellular Nluc and Nluc on the cell surface. The compounds displayed little to no toxicity to cells and high luciferase specificity, showing no activity against firefly luciferase or even the closely related NanoBit system. Looking forward, the structural motifs used to gain access to the Nluc surface can also be appended with other functional groups, and therefore interesting opportunities for developing assays based on relief-of-inhibition can be envisioned.


Asunto(s)
Permeabilidad de la Membrana Celular , Inhibidores Enzimáticos/farmacología , Luciferasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Concentración 50 Inhibidora , Relación Estructura-Actividad
15.
J Immunol Methods ; 431: 11-21, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26851520

RESUMEN

Receptor-mediated antibody internalization is a key mechanism underlying several anti-cancer antibody therapeutics. Delivering highly toxic drugs to cancer cells, as in the case of antibody drug conjugates (ADCs), efficient removal of surface receptors from cancer cells and changing the pharmacokinetics profile of the antibody drugs are some of key ways that internalization impacts the therapeutic efficacy of the antibodies. Over the years, several techniques have been used to study antibody internalization including radiolabels, fluorescent microscopy, flow cytometry and cellular toxicity assays. While these methods allow analysis of internalization, they have limitations including a multistep process and limited throughput and are generally endpoint assays. Here, we present a new homogeneous method that enables time and concentration dependent measurements of antibody internalization. The method uses a new hydrophilic and bright pH sensor dye (pHAb dye), which is not fluorescent at neutral pH but becomes highly fluorescent at acidic pH. For receptor mediated antibody internalization studies, antibodies against receptors are conjugated with the pHAb dye and incubated with the cells expressing the receptors. Upon binding to the receptor, the dyes conjugated to the antibody are not fluorescent because of the neutral pH of the media, but upon internalization and trafficking into endosomal and lysosomal vesicles the pH drops and dyes become fluorescent. The enabling attributes of the pHAb dyes are the hydrophilic nature to minimize antibody aggregation and bright fluorescence at acidic pH which allows development of simple plate based assays using a fluorescent reader. Using two different therapeutic antibodies--Trastuzumab (anti-HER2) and Cetuximab (anti-EGFR)--we show labeling with pHAb dye using amine and thiol chemistries and impact of chemistry and dye to antibody ration on internalization. We finally present two new approaches using the pHAb dye, which will be beneficial for screening a large number of antibody samples during early monoclonal development phase.


Asunto(s)
Anticuerpos/análisis , Colorantes Fluorescentes/química , Piperazinas/química , Rodaminas/química , Anticuerpos/inmunología , Línea Celular Tumoral , Cetuximab/química , Cetuximab/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Concentración de Iones de Hidrógeno , Estructura Molecular , Trastuzumab/química , Trastuzumab/inmunología
16.
Nat Commun ; 6: 10091, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26631872

RESUMEN

The therapeutic action of drugs is predicated on their physical engagement with cellular targets. Here we describe a broadly applicable method using bioluminescence resonance energy transfer (BRET) to reveal the binding characteristics of a drug with selected targets within intact cells. Cell-permeable fluorescent tracers are used in a competitive binding format to quantify drug engagement with the target proteins fused to Nanoluc luciferase. The approach enabled us to profile isozyme-specific engagement and binding kinetics for a panel of histone deacetylase (HDAC) inhibitors. Our analysis was directed particularly to the clinically approved prodrug FK228 (Istodax/Romidepsin) because of its unique and largely unexplained mechanism of sustained intracellular action. Analysis of the binding kinetics by BRET revealed remarkably long intracellular residence times for FK228 at HDAC1, explaining the protracted intracellular behaviour of this prodrug. Our results demonstrate a novel application of BRET for assessing target engagement within the complex milieu of the intracellular environment.


Asunto(s)
Células/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia/métodos , Inhibidores de Histona Desacetilasas/química , Preparaciones Farmacéuticas/química , Proliferación Celular , Células/química , Células/citología , Células HeLa , Histona Desacetilasa 1/química , Histona Desacetilasa 1/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Luciferasas/química , Luciferasas/genética , Luciferasas/metabolismo , Luminiscencia
17.
J Neurosci Methods ; 228: 67-78, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24675049

RESUMEN

BACKGROUND: Stem cell therapies appear promising for treating certain neurodegenerative disorders and molecular imaging methods that track these cells in vivo could answer some key questions regarding their survival and migration. Bioluminescence imaging (BLI), which relies on luciferase expression in these cells, has been used for this purpose due to its high sensitivity. NEW METHOD: In this study, we employ BLI to track luciferase-expressing human neural progenitor cells (hNPC(Luc2)) in the rat striatum long-term. RESULTS: We show that hNPC(Luc2) are detectable in the rat striatum. Furthermore, we demonstrate that using this tracking method, surviving grafts can be detected in vivo for up to 12 weeks, while those that were rejected do not produce bioluminescence signal. We also demonstrate the ability to discern hNPC(Luc2) contralateral migration. COMPARISON WITH EXISTING METHODS: Some of the advantages of BLI compared to other imaging methods used to track progenitor/stem cells include its sensitivity and specificity, low background signal and ability to distinguish surviving grafts from rejected ones over the long term while the blood-brain barrier remains intact. CONCLUSIONS: These new findings may be useful in future preclinical applications developing cell-based treatments for neurodegenerative disorders.


Asunto(s)
Cuerpo Estriado/citología , Mediciones Luminiscentes , Células-Madre Neurales/fisiología , Neuroimagen/métodos , Análisis de Varianza , Animales , Bromodesoxiuridina/metabolismo , Recuento de Células , Línea Celular Transformada , Movimiento Celular , Cuerpo Estriado/cirugía , Ciclosporina/farmacología , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/cirugía , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Trasplante de Células Madre , Factores de Tiempo , Transfección
18.
Curr Chem Genomics ; 6: 55-71, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23248739

RESUMEN

Our fundamental understanding of proteins and their biological significance has been enhanced by genetic fusion tags, as they provide a convenient method for introducing unique properties to proteins so that they can be examinedin isolation. Commonly used tags satisfy many of the requirements for applications relating to the detection and isolation of proteins from complex samples. However, their utility at low concentration becomes compromised if the binding affinity for a detection or capture reagent is not adequate to produce a stable interaction. Here, we describe HaloTag® (HT7), a genetic fusion tag based on a modified haloalkane dehalogenase designed and engineered to overcome the limitation of affinity tags by forming a high affinity, covalent attachment to a binding ligand. HT7 and its ligand have additional desirable features. The tag is relatively small, monomeric, and structurally compatible with fusion partners, while the ligand is specific, chemically simple, and amenable to modular synthetic design. Taken together, the design features and molecular evolution of HT7 have resulted in a superior alternative to common tags for the overexpression, detection, and isolation of target proteins.

19.
ACS Chem Biol ; 3(6): 373-82, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18533659

RESUMEN

We have designed a modular protein tagging system that allows different functionalities to be linked onto a single genetic fusion, either in solution, in living cells, or in chemically fixed cells. The protein tag (HaloTag) is a modified haloalkane dehalogenase designed to covalently bind to synthetic ligands (HaloTag ligands). The synthetic ligands comprise a chloroalkane linker attached to a variety of useful molecules, such as fluorescent dyes, affinity handles, or solid surfaces. Covalent bond formation between the protein tag and the chloroalkane linker is highly specific, occurs rapidly under physiological conditions, and is essentially irreversible. We demonstrate the utility of this system for cellular imaging and protein immobilization by analyzing multiple molecular processes associated with NF-kappaB-mediated cellular physiology, including imaging of subcellular protein translocation and capture of protein--protein and protein--DNA complexes.


Asunto(s)
Técnicas Biosensibles/métodos , Células/citología , Colorantes Fluorescentes/química , Mediciones Luminiscentes/métodos , Proteínas Luminiscentes/química , Coloración y Etiquetado , Animales , Sitios de Unión , Células/metabolismo , ADN/análisis , ADN/química , ADN/metabolismo , Enzimas Inmovilizadas , Humanos , Hidrocarburos Clorados/química , FN-kappa B/análisis , FN-kappa B/metabolismo , Proteínas/análisis , Proteínas/química , Proteínas/metabolismo , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA