Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Biol Chem ; 299(1): 102769, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36470427

RESUMEN

Programmed death-ligand 1 (PD-L1) is a key immune regulatory protein that interacts with programmed cell death protein 1 (PD-1), leading to T-cell suppression. Whilst this interaction is key in self-tolerance, cancer cells evade the immune system by overexpressing PD-L1. Inhibition of the PD-1/PD-L1 pathway with standard monoclonal antibodies has proven a highly effective cancer treatment; however, single domain antibodies (VHH) may offer numerous potential benefits. Here, we report the identification and characterization of a diverse panel of 16 novel VHHs specific to PD-L1. The panel of VHHs demonstrate affinities of 0.7 nM to 5.1 µM and were able to completely inhibit PD-1 binding to PD-L1. The binding site for each VHH on PD-L1 was determined using NMR chemical shift perturbation mapping and revealed a common binding surface encompassing the PD-1-binding site. Additionally, we solved crystal structures of two representative VHHs in complex with PD-L1, which revealed unique binding modes. Similar NMR experiments were used to identify the binding site of CD80 on PD-L1, which is another immune response regulatory element and interacts with PD-L1 localized on the same cell surface. CD80 and PD-1 were revealed to share a highly overlapping binding site on PD-L1, with the panel of VHHs identified expected to inhibit CD80 binding. Comparison of the CD80 and PD-1 binding sites on PD-L1 enabled the identification of a potential antibody binding region able to confer specificity for the inhibition of PD-1 binding only, which may offer therapeutic benefits to counteract cancer cell evasion of the immune system.


Asunto(s)
Anticuerpos , Antígeno B7-1 , Antígeno B7-H1 , Receptor de Muerte Celular Programada 1 , Humanos , Antígeno B7-1/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/metabolismo , Unión Proteica , Sitios de Unión , Cristalografía , Anticuerpos/química , Anticuerpos/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(6): 3093-3102, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-31980531

RESUMEN

The catalytic activity of the protease MALT1 is required for adaptive immune responses and regulatory T (Treg)-cell development, while dysregulated MALT1 activity can lead to lymphoma. MALT1 activation requires its monoubiquitination on lysine 644 (K644) within the Ig3 domain, localized adjacent to the protease domain. The molecular requirements for MALT1 monoubiquitination and the mechanism by which monoubiquitination activates MALT1 had remained elusive. Here, we show that the Ig3 domain interacts directly with ubiquitin and that an intact Ig3-ubiquitin interaction surface is required for the conjugation of ubiquitin to K644. Moreover, by generating constitutively active MALT1 mutants that overcome the need for monoubiquitination, we reveal an allosteric communication between the ubiquitination site K644, the Ig3-protease interaction surface, and the active site of the protease domain. Finally, we show that MALT1 mutants that alter the Ig3-ubiquitin interface impact the biological response of T cells. Thus, ubiquitin binding by the Ig3 domain promotes MALT1 activation by an allosteric mechanism that is essential for its biological function.


Asunto(s)
Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Ubiquitina , Ubiquitinación , Regulación Alostérica , Células HEK293 , Humanos , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/química , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Mutación , Unión Proteica , Dominios Proteicos , Ubiquitina/química , Ubiquitina/metabolismo , Ubiquitinación/genética , Ubiquitinación/fisiología
3.
Biochim Biophys Acta Proteins Proteom ; 1866(1): 88-96, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28578073

RESUMEN

Human aromatase is the cytochrome P450 catalysing the conversion of androgens into estrogens playing a key role in the endocrine system. Due to this role, it is likely to be a target of the so-called endocrine disrupting chemicals, a series of compounds able to interfere with the hormone system with toxic effects. If on one side the toxicity of some compounds such as bisphenol A is well known, on the other side the toxic concentrations of such compounds as well as the effect of the many other molecules that are in contact with us in everyday life still need a deep investigation. The availability of biological assays able to detect the interaction of chemicals with key molecular targets of the endocrine system represents a possible solution to identify potential endocrine disrupting chemicals. Here the so-called alkali assay previously developed in our laboratory is applied to test the effect of different compounds on the activity of human aromatase. The assay is based on the detection of the alkali product that forms upon strong alkali treatment of the NADP+ released upon enzyme turnover. Here it is applied on human aromatase and validated using anastrozole and sildenafil as known aromatase inhibitors. Out of the small library of compounds tested, resveratrol and ketoconazole resulted to inhibit aromatase activity, while bisphenol A and nicotine were found to exert an inhibitory effect at relatively high concentrations (100µM), and other molecules such as lindane and four plasticizers did not show any significant effect. These data are confirmed by quantification of the product estrone in the same reaction mixtures through ELISA. Overall, the results show that the alkali assay is suitable to screen for molecules that interfere with aromatase activity. As a consequence it can also be applied to other molecular targets of EDCs that use NAD(P)H for catalysis in a high throughput format for the fast screening of many different compounds as endocrine disrupting chemicals. This article is part of a Special Issue entitled: Cytochrome P450 biodiversity and biotechnology, edited by Erika Plettner, Gianfranco Gilardi, Luet Wong, Vlada Urlacher, Jared Goldstone.


Asunto(s)
Inhibidores de la Aromatasa/química , Aromatasa/química , Bioensayo , Disruptores Endocrinos/química , Anastrozol , Aromatasa/genética , Inhibidores de la Aromatasa/análisis , Compuestos de Bencidrilo/análisis , Compuestos de Bencidrilo/química , Disruptores Endocrinos/análisis , Ensayo de Inmunoadsorción Enzimática , Estrona/química , Expresión Génica , Humanos , Cetoconazol/análisis , Cetoconazol/química , Ligandos , NADP/química , Nicotina/análisis , Nicotina/química , Nitrilos/análisis , Nitrilos/química , Fenoles/análisis , Fenoles/química , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Resveratrol , Citrato de Sildenafil/análisis , Citrato de Sildenafil/química , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/química , Estilbenos/análisis , Estilbenos/química , Triazoles/análisis , Triazoles/química
4.
Biotechnol Appl Biochem ; 65(1): 46-53, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28926141

RESUMEN

Aromatase catalyzes the biosynthesis of estrogens from androgens. Owing to the physiological importance of this conversion of lipophilic substrates, the interaction with the lipid bilayer for this cytochrome P450 is crucial for its dynamics that must allow an easy access to substrates and inhibitors. Here, the aromatase-anastrozole interaction is studied by combining computational methods to identify possible access/egress routes with the protein inserted in the membrane and experimental tools aimed at the investigation of the effect of the inhibitor on the protein conformation. By means of molecular dynamics simulations of the protein inserted in the membrane, two channels, not detected in the starting crystal structure, are found after a 20-nSec simulation. Trypsin digestion on the recombinant protein shows that the enzyme is strongly protected by the presence of the substrate and even more by the inhibitor. DSC experiments show an increase in the melting temperature of the protein in complex with the substrate (49.3 °C) and the inhibitor (58.7 °C) compared to the ligand-free enzyme (45.9 °C), consistent with a decrease of flexibility of the protein. The inhibitor anastrozole enters the active site of the protein through a channel different from that used from the substrate and promotes a conformational change that stiffens the protein conformation and decreases the protein-protein interaction between different aromatase molecules.


Asunto(s)
Aromatasa/química , Simulación de Dinámica Molecular , Nitrilos/química , Triazoles/química , Anastrozol , Aromatasa/metabolismo , Humanos , Ligandos , Nitrilos/metabolismo , Estructura Cuaternaria de Proteína , Triazoles/metabolismo
5.
Front Immunol ; 14: 1216967, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37483614

RESUMEN

Interleukin-13 (IL-13) is a cytokine involved in T-cell immune responses and is a well validated therapeutic target for the treatment of asthma, along with other allergic and inflammatory diseases. IL-13 signals through a ternary signalling complex formed with the receptors IL-13Rα1 and IL-4Rα. This complex is assembled by IL-13 initially binding IL-13Rα1, followed by association of the binary IL-13:IL-13Rα1 complex with IL-4Rα. The receptors are shared with IL-4, but IL-4 initially binds IL-4Rα. Here we report the identification and characterisation of a diverse panel of single-domain antibodies (VHHs) that bind to IL-13 (KD 40 nM-5.5 µM) and inhibit downstream IL-13 signalling (IC50 0.2-53.8 µM). NMR mapping showed that the VHHs recognise a number of epitopes on IL-13, including previously unknown allosteric sites. Further NMR investigation of VHH204 bound to IL-13 revealed a novel allosteric mechanism of inhibition, with the antibody stabilising IL-13 in a conformation incompatible with receptor binding. This also led to the identification of a conformational equilibrium for free IL-13, providing insights into differing receptor signalling complex assembly seen for IL-13 compared to IL-4, with formation of the IL-13:IL-13Rα1 complex required to stabilise IL-13 in a conformation with high affinity for IL-4Rα. These findings highlight new opportunities for therapeutic targeting of IL-13 and we report a successful 19F fragment screen of the IL-13:VHH204 complex, including binding sites identified for several hits. To our knowledge, these 19F containing fragments represent the first small-molecules shown to bind to IL-13 and could provide starting points for a small-molecule drug discovery programme.


Asunto(s)
Interleucina-13 , Anticuerpos de Dominio Único , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Subunidad alfa1 del Receptor de Interleucina-13/metabolismo , Citocinas
6.
J Steroid Biochem Mol Biol ; 167: 23-32, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27702664

RESUMEN

The cytochrome P450 aromatase is involved in the last step of sex hormones biosynthesis by converting androgens into estrogens. The human enzyme is highly polymorphic and literature data correlate aromatase single nucleotide polymorphisms to the onset of pathologies such as breast cancer and neurodegenerative diseases. The aims of this study were i) to study the influence of the mutations R264C and R264H on the structure-function of the enzyme also upon phosphorylation by selected kinases and ii) to compare the activity of the variants to that of aromatase wild type in two different cell lines. Far-UV circular dichroism spectroscopy, thermal denaturation experiments and CO-binding assay showed that the two polymorphic variants are correctly folded. Steady-state kinetics experiments showed that rArom R264C and R264H exhibit a 1.5 and 3.4 folds lower catalytic efficiency, respectively, when compared to the wild type protein. Since R264 is part of the consensus motif of PKA and PKG1, phosphorylation experiments were performed to study the effect on aromatase function. Phosphorylation by PKA caused a decrease in activity by 36.2%, 49.3% and 27.9% in the wild type, R264C and R264H proteins respectively. Phosphorylation by PKG1 was also found to decrease the activity by 30.3%, 30.5% and 15.4% in the wild type, R264C and R264H proteins respectively. Experiments performed on the three full-length proteins expressed in human MCF-7 breast cancer cells and rat ST14A neuronal cells showed that, depending on the cell line used, the activity of the proteins is different, implicating different cellular mechanisms modulating aromatase activity. This work demonstrate that R264 polymorphism causes an intrinsic alteration of aromatase activity together with a different consensus for phosphorylation by different kinases, indicating that estrogen production can be different when such mutations are present. These findings are significant in understanding the onset and treatment of pathologies in which aromatase has been shown to be involved.


Asunto(s)
Aromatasa/metabolismo , Polimorfismo Genético , Secuencias de Aminoácidos , Animales , Aromatasa/química , Neoplasias de la Mama/metabolismo , Catálisis , Línea Celular , Dicroismo Circular , Técnicas de Cocultivo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Estrógenos/metabolismo , Femenino , Humanos , Células MCF-7 , Conformación Molecular , Mutagénesis Sitio-Dirigida , Mutación , Neuronas/metabolismo , Fosforilación , Ratas , Proteínas Recombinantes/metabolismo , Espectrofotometría Ultravioleta
7.
J Steroid Biochem Mol Biol ; 165(Pt B): 438-447, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27616271

RESUMEN

Aromatase catalyses the conversion of androgens into estrogens and is a well-known target for breast cancer therapy. As it has been suggested that its activity is affected by inhibitors of phosphodiesterase-5, this work investigates the potential interaction of sildenafil with aromatase. This is carried out both at molecular level through structural and kinetics assays applied to the purified enzyme, and at cellular level using neuronal and breast cancer cell lines. Sildenafil is found to bind to aromatase with a KD of 0.58±0.05µM acting as a partial and mixed inhibitor with a maximal inhibition of 35±2%. Hyperfine sublevel correlation spectroscopy and docking studies show that sildenafil binds to the heme iron via its 6th axial water ligand. These results also provide information on the starting molecular scaffold for the development of new generations of drugs designed to inhibit aromatase as well as phosphodiesterase-5, a new emerging target for breast cancer therapy.


Asunto(s)
Inhibidores de la Aromatasa/química , Aromatasa/metabolismo , Citrato de Sildenafil/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Catálisis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Relación Dosis-Respuesta a Droga , Espectroscopía de Resonancia por Spin del Electrón , Femenino , Hemo/química , Humanos , Concentración 50 Inhibidora , Hierro/química , Cinética , Ligandos , Células MCF-7 , Simulación del Acoplamiento Molecular , Unión Proteica , Proteínas Recombinantes/química , Espectrofotometría , Espectrofotometría Ultravioleta , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA