Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1293883, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38455057

RESUMEN

Fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF) and systemic scleroderma (SSc), are commonly associated with high morbidity and mortality, thereby representing a significant unmet medical need. Interleukin 11 (IL11)-mediated cell activation has been identified as a central mechanism for promoting fibrosis downstream of TGFß. IL11 signaling has recently been reported to promote fibroblast-to-myofibroblast transition, thus leading to various pro-fibrotic phenotypic changes. We confirmed increased mRNA expression of IL11 and IL11Rα in fibrotic diseases by OMICs approaches and in situ hybridization. However, the vital role of IL11 as a driver for fibrosis was not recapitulated. While induction of IL11 secretion was observed downstream of TGFß signaling in human lung fibroblasts and epithelial cells, the cellular responses induced by IL11 was quantitatively and qualitatively inferior to that of TGFß at the transcriptional and translational levels. IL11 blocking antibodies inhibited IL11Rα-proximal STAT3 activation but failed to block TGFß-induced profibrotic signals. In summary, our results challenge the concept of IL11 blockade as a strategy for providing transformative treatment for fibrosis.


Asunto(s)
Interleucina-11 , Factor de Crecimiento Transformador beta , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Fibrosis , Miofibroblastos/metabolismo
2.
J Immunol ; 206(5): 1102-1113, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33495237

RESUMEN

CTLA4-Ig/abatacept dampens activation of naive T cells by blocking costimulation via CD28. It is an approved drug for rheumatoid arthritis but failed to deliver efficacy in a number of other autoimmune diseases. One explanation is that activated T cells rely less on CD28 signaling and use alternate coreceptors for effector function. ICOS is critical for activation of T-dependent humoral immune responses, which drives pathophysiology of IgG-mediated autoimmune diseases. In this study, we asked whether CD28 and ICOS play nonredundant roles for maintenance of T-dependent responses in mouse models. Using a hapten-protein immunization model, we show that during an ongoing germinal center response, combination treatment with CTLA4-Ig and ICOS ligand (ICOSL) blocking Ab completely dissolves ongoing germinal center responses, whereas single agents show only partial activity. Next, we took two approaches to engineer a therapeutic molecule that blocks both pathways. First, we engineered CTLA4-Ig to enhance binding to ICOSL while retaining affinity to CD80/CD86. Using a library approach, binding affinity of CTLA4-Ig to human ICOSL was increased significantly from undetectable to 15-42 nM; however, the affinity was still insufficient to completely block binding of ICOSL to ICOS. Second, we designed a bispecific costimulation inhibitor with high-affinity CTLA4 extracellular domains fused to anti-ICOSL Ab termed bifunctional costimulation inhibitor. With this bispecific approach, we achieved complete inhibition of CD80 and CD86 binding to CD28 as well as ICOS binding to ICOSL. Such bispecific molecules may provide greater therapeutic benefit in IgG-mediated inflammatory diseases compared with CTLA4-Ig alone.


Asunto(s)
Antígenos CD28/metabolismo , Antígeno CTLA-4/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/farmacología , Proteína Coestimuladora de Linfocitos T Inducibles/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Abatacept/farmacología , Animales , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Femenino , Centro Germinal/efectos de los fármacos , Centro Germinal/metabolismo , Inmunidad Humoral/efectos de los fármacos , Inmunoglobulina G/metabolismo , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Linfocitos T/metabolismo
3.
BMC Mol Cell Biol ; 20(1): 29, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31382872

RESUMEN

BACKGROUND: CD40 is a 48 kDa type I transmembrane protein that is constitutively expressed on hematopoietic cells such as dendritic cells, macrophages, and B cells. Engagement of CD40 by CD40L expressed on T cells results in the production of proinflammatory cytokines, induces T helper cell function, and promotes macrophage activation. The involvement of CD40 in chronic immune activation has resulted in CD40 being proposed as a therapeutic target for a range of chronic inflammatory diseases. CD40 antagonists are currently being explored for the treatment of autoimmune diseases and several anti-CD40 agonist mAbs have entered clinical development for oncological indications. RESULTS: To better understand the mode of action of anti-CD40 mAbs, we have determined the x-ray crystal structures of the ABBV-323 (anti-CD40 antagonist, ravagalimab) Fab alone, ABBV-323 Fab complexed to human CD40 and FAB516 (anti-CD40 agonist) complexed to human CD40. These three crystals structures 1) identify the conformational CD40 epitope for ABBV-323 recognition 2) illustrate conformational changes which occur in the CDRs of ABBV-323 Fab upon CD40 binding and 3) develop a structural hypothesis for an agonist/antagonist switch in the LCDR1 of this proprietary class of CD40 antibodies. CONCLUSIONS: The structure of ABBV-323 Fab demonstrates a unique method for antagonism by stabilizing the proposed functional antiparallel dimer for CD40 receptor via novel contacts to LCDR1, namely residue position R32 which is further supported by a closely related agonist antibody FAB516 which shows only monomeric recognition and no contacts with LCDR1 due to a mutation to L32 on LCDR1. These data provide a structural basis for the full antagonist activity of ABBV-323.


Asunto(s)
Anticuerpos Monoclonales/química , Complejo Antígeno-Anticuerpo/química , Antígenos CD40/agonistas , Antígenos CD40/antagonistas & inhibidores , Antígenos CD40/química , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Modelos Moleculares , Transducción de Señal , Electricidad Estática
4.
Sci Rep ; 9(1): 9089, 2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31235749

RESUMEN

IL-36 cytokines are pro-inflammatory members of the IL-1 family that are upregulated in inflammatory disorders. Specifically, IL-36γ is highly expressed in active psoriatic lesions and can drive pro-inflammatory processes in 3D human skin equivalents supporting a role for this target in skin inflammation. Small molecule antagonists of interleukins have been historically challenging to generate. Nevertheless, we performed a small molecule high-throughput screen to identify IL-36 antagonists using a novel TR-FRET binding assay. Several compounds, including 2-oxypyrimidine containing structural analogs of the marketed endothelin receptor A antagonist Ambrisentan, were identified as hits from the screen. A-552 was identified as a the most potent antagonist of human IL-36γ, but not the closely related family member IL-36α, was capable of attenuating IL-36γ induced responses in mouse and human disease models. Additionally, x-ray crystallography studies identified key amino acid residues in the binding pocket present in human IL-36γ that are absent in human IL-36α. A-552 represents a first-in-class small molecule antagonist of IL-36 signaling that could be used as a chemical tool to further investigate the role of this pathway in inflammatory skin diseases such as psoriasis.


Asunto(s)
Interleucina-1/antagonistas & inhibidores , Psoriasis/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Psoriasis/metabolismo , Psoriasis/patología , Piel/efectos de los fármacos , Piel/patología , Bibliotecas de Moléculas Pequeñas/uso terapéutico
5.
Exp Dermatol ; 28(2): 113-120, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30417427

RESUMEN

Psoriasis vulgaris (PV) results from activation of IL-23/Th17 immune pathway and is further amplified by cytokines/chemokines from skin cells. Among skin-derived pro-inflammatory cytokines, IL-36 family members are highly upregulated in PV patients and play a critical role in general pustular psoriasis. However, there is limited data showing crosstalk between the IL-23 and IL-36 pathways in PV. Herein, potential attenuation of skin inflammation in the IL-23-induced mouse model of psoriasiform dermatitis by functional inhibition of IL-36 receptor (IL-36R) was interrogated. Anti-mouse IL-36R monoclonal antibodies (mAbs) were generated and validated in vitro by inhibiting IL-36α-induced secretion of CXCL1 from NIH 3T3 cells. Antibody target engagement was demonstrated by inhibition of CXCL1 production in a novel acute model of IL-36α systemic injection in mice. In addition, anti-IL-36R mAbs inhibited tissue inflammation and inflammatory gene expression in an IL-36α ear injection model of psoriasiform dermatitis demonstrating engagement of the target in the ear skin. To elucidate the possible role of IL-36 signalling in IL-23/Th17 pathway, the ability of anti-IL-36R mAbs to inhibit skin inflammation in an IL-23 ear injection model was assessed. Inhibiting the IL-36 pathway resulted in significant attenuation of skin thickening and psoriasis-relevant gene expression. Taken together, these data suggest a role for IL-36 signalling in the IL-23/Th17 signalling axis in PV.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Dermatitis/inmunología , Inflamación/inmunología , Interleucinas/inmunología , Psoriasis/inmunología , Receptores de Interleucina/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/uso terapéutico , Quimiocina CXCL1/metabolismo , Citocinas/metabolismo , Dermatitis/terapia , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Inflamación/metabolismo , Interleucina-1/inmunología , Interleucina-23/farmacología , Ligandos , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Psoriasis/terapia , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina/inmunología , Receptores de Interleucina-1/inmunología , Transducción de Señal , Piel/metabolismo , Piel/patología , Células Th17/citología
6.
J Biol Chem ; 293(2): 403-411, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29180446

RESUMEN

IL-36 cytokines signal through the IL-36 receptor (IL-36R) and a shared subunit, IL-1RAcP (IL-1 receptor accessory protein). The activation mechanism for the IL-36 pathway is proposed to be similar to that of IL-1 in that an IL-36R agonist (IL-36α, IL-36ß, or IL-36γ) forms a binary complex with IL-36R, which then recruits IL-1RAcP. Recent studies have shown that IL-36R interacts with IL-1RAcP even in the absence of an agonist. To elucidate the IL-36 activation mechanism, we considered all possible binding events for IL-36 ligands/receptors and examined these events in direct binding assays. Our results indicated that the agonists bind the IL-36R extracellular domain with micromolar affinity but do not detectably bind IL-1RAcP. Using surface plasmon resonance (SPR), we found that IL-1RAcP also does not bind IL-36R when no agonist is present. In the presence of IL-36α, however, IL-1RAcP bound IL-36R strongly. These results suggested that the main pathway to the IL-36R·IL-36α·IL-1RAcP ternary complex is through the IL-36R·IL-36α binary complex, which recruits IL-1RAcP. We could not measure the binding affinity of IL-36R to IL-1RAcP directly, so we engineered a fragment crystallizable-linked construct to induce IL-36R·IL-1RAcP heterodimerization and predicted the binding affinity during a complete thermodynamic cycle to be 74 µm The SPR analysis also indicated that the IL-36R antagonist IL-36Ra binds IL-36R with higher affinity and a much slower off rate than the IL-36R agonists, shedding light on IL-36 pathway inhibition. Our results reveal the landscape of IL-36 ligand and receptor interactions, improving our understanding of IL-36 pathway activation and inhibition.


Asunto(s)
Quimiocina CXCL1/metabolismo , Interleucina-1/metabolismo , Receptores de Interleucina/metabolismo , Línea Celular Tumoral , Células HEK293 , Humanos , Proteína Accesoria del Receptor de Interleucina-1/metabolismo , Cinética , Unión Proteica , Resonancia por Plasmón de Superficie
7.
J Am Chem Soc ; 139(46): 16822-16829, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29068676

RESUMEN

Ligand-binding assays are the linchpin of drug discovery and medicinal chemistry. Cell-surface receptors and their ligands have traditionally been characterized by radioligand-binding assays, which have low temporal and spatial resolution and entail safety risks. Here, we report a powerful alternative (GlycoFRET), where terbium-labeled fluorescent reporters are irreversibly attached to receptors by metabolic glycan engineering. For the first time, we show time-resolved fluorescence resonance energy transfer between receptor glycans and fluorescently labeled ligands. We describe GlycoFRET for a GPI-anchored receptor, a G-protein-coupled receptor, and a heterodimeric cytokine receptor in living cells with excellent sensitivity and high signal-to-background ratios. In contrast to previously described methods, GlycoFRET does not require genetic engineering or antibodies to label receptors. Given that all cell-surface receptors are glycosylated, we expect that GlycoFRET can be generalized with applications in chemical biology and biotechnology, such as target engagement, receptor pharmacology, and high-throughput screening.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Ligadas a GPI/metabolismo , Polisacáridos/metabolismo , Receptores de Superficie Celular/metabolismo , Supervivencia Celular , Receptores de Folato Anclados a GPI/metabolismo , Humanos , Ligandos , Receptores Histamínicos H3/metabolismo , Receptores de Interleucina/metabolismo , Terbio
8.
MAbs ; 9(2): 269-284, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27929753

RESUMEN

Exudative age-related macular degeneration (AMD) is the most common cause of moderate and severe vision loss in developed countries. Intraocular injections of vascular endothelial growth factor (VEGF or VEGF-A)-neutralizing proteins provide substantial benefit, but frequent, long-term injections are needed. In addition, many patients experience initial visual gains that are ultimately lost due to subretinal fibrosis. Preclinical studies and early phase clinical trials suggest that combined suppression of VEGF and platelet-derived growth factor-BB (PDGF-BB) provides better outcomes than suppression of VEGF alone, due to more frequent regression of neovascularization (NV) and suppression of subretinal fibrosis. We generated a dual variable domain immunoglobulin molecule, ABBV642 that specifically and potently binds and neutralizes VEGF and PDGF-BB. ABBV642 has been optimized for treatment of exudative AMD based on the following design characteristics: 1) high affinity binding to all VEGF-A isoforms and both soluble and extracellular matrix (ECM)-associated PDGF-BB; 2) potential for extended residence time in the vitreous cavity to decrease the frequency of intraocular injections; 3) rapid clearance from systemic circulation compared with molecules with wild type Fc region for normal FcRn binding, which may reduce the risk of systemic complications; and 4) low risk of potential effector function. The bispecificity of ABBV642 allows for a single injection of a single therapeutic agent, and thus a more streamlined development and regulatory path compared with combination products. In a mouse model of exudative AMD, ABBV642 was observed to be more effective than aflibercept. ABBV642 has potential to improve efficacy with reduced injection frequency in patients with exudative AMD, thereby reducing the enormous disease burden for patients and society.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Biespecíficos/farmacología , Degeneración Macular/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-sis/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Becaplermina , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Ingeniería de Proteínas , Conejos
9.
BMC Struct Biol ; 12: 22, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22995073

RESUMEN

BACKGROUND: Structure-based drug design (SBDD) can accelerate inhibitor lead design and optimization, and efficient methods including protein purification, characterization, crystallization, and high-resolution diffraction are all needed for rapid, iterative structure determination. Janus kinases are important targets that are amenable to structure-based drug design. Here we present the first mouse Tyk2 crystal structures, which are complexed to 3-aminoindazole compounds. RESULTS: A comprehensive construct design effort included N- and C-terminal variations, kinase-inactive mutations, and multiple species orthologs. High-throughput cloning and expression methods were coupled with an abbreviated purification protocol to optimize protein solubility and stability. In total, 50 Tyk2 constructs were generated. Many displayed poor expression, inadequate solubility, or incomplete affinity tag processing. One kinase-inactive murine Tyk2 construct, complexed with an ATP-competitive 3-aminoindazole inhibitor, provided crystals that diffracted to 2.5-2.6 Å resolution. This structure revealed initial "hot-spot" regions for SBDD, and provided a robust platform for ligand soaking experiments. Compared to previously reported human Tyk2 inhibitor crystal structures (Chrencik et al. (2010) J Mol Biol 400:413), our structures revealed a key difference in the glycine-rich loop conformation that is induced by the inhibitor. Ligand binding also conferred resistance to proteolytic degradation by thermolysin. As crystals could not be obtained with the unliganded enzyme, this enhanced stability is likely important for successful crystallization and inhibitor soaking methods. CONCLUSIONS: Practical criteria for construct performance and prioritization, the optimization of purification protocols to enhance protein yields and stability, and use of high-throughput construct exploration enable structure determination methods early in the drug discovery process. Additionally, specific ligands stabilize Tyk2 protein and may thereby enable crystallization.


Asunto(s)
Diseño de Fármacos , Indazoles/química , Indazoles/farmacología , TYK2 Quinasa/antagonistas & inhibidores , TYK2 Quinasa/química , Secuencia de Aminoácidos , Animales , Cristalización , Cristalografía por Rayos X , Estabilidad de Enzimas/efectos de los fármacos , Humanos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Ratones , Datos de Secuencia Molecular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Estructura Secundaria de Proteína , Proteolisis/efectos de los fármacos , Relación Estructura-Actividad , TYK2 Quinasa/aislamiento & purificación
10.
Bioorg Med Chem Lett ; 20(1): 330-3, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19919896

RESUMEN

MK2 is a Ser/Thr kinase of significant interest as an anti-inflammatory drug discovery target. Here we describe the development of in vitro tools for the identification and characterization of MK2 inhibitors, including validation of inhibitor interactions with the crystallography construct and determination of the unique binding mode of 2,4-diaminopyrimidine inhibitors in the MK2 active site. Use of these tools in the optimization of a potent and selective inhibitor lead series is described in the accompanying Letter.


Asunto(s)
Antiinflamatorios/química , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/química , Adenosina Trifosfato/química , Antiinflamatorios/síntesis química , Antiinflamatorios/farmacología , Sitios de Unión , Unión Competitiva , Simulación por Computador , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/síntesis química , Pirimidinas/farmacología , Relación Estructura-Actividad
11.
BMC Struct Biol ; 9: 16, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19296855

RESUMEN

BACKGROUND: Structure-based drug design (SBDD) can provide valuable guidance to drug discovery programs. Robust construct design and expression, protein purification and characterization, protein crystallization, and high-resolution diffraction are all needed for rapid, iterative inhibitor design. We describe here robust methods to support SBDD on an oral anti-cytokine drug target, human MAPKAP kinase 2 (MK2). Our goal was to obtain useful diffraction data with a large number of chemically diverse lead compounds. Although MK2 structures and structural methods have been reported previously, reproducibility was low and improved methods were needed. RESULTS: Our construct design strategy had four tactics: N- and C-terminal variations; entropy-reducing surface mutations; activation loop deletions; and pseudoactivation mutations. Generic, high-throughput methods for cloning and expression were coupled with automated liquid dispensing for the rapid testing of crystallization conditions with minimal sample requirements. Initial results led to development of a novel, customized robotic crystallization screen that yielded MK2/inhibitor complex crystals under many conditions in seven crystal forms. In all, 44 MK2 constructs were generated, ~500 crystals were tested for diffraction, and ~30 structures were determined, delivering high-impact structural data to support our MK2 drug design effort. CONCLUSION: Key lessons included setting reasonable criteria for construct performance and prioritization, a willingness to design and use customized crystallization screens, and, crucially, initiation of high-throughput construct exploration very early in the drug discovery process.


Asunto(s)
Diseño de Fármacos , Péptidos y Proteínas de Señalización Intracelular/química , Mutagénesis Sitio-Dirigida , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Sustitución de Aminoácidos , Simulación por Computador , Cristalización , Cristalografía por Rayos X , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Isoenzimas/biosíntesis , Isoenzimas/química , Isoenzimas/aislamiento & purificación , Conformación Proteica , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética
12.
J Immunol Methods ; 334(1-2): 134-41, 2008 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-18355834

RESUMEN

Interleukin 13 is a key cytokine that mediates airway hyper-responsiveness and mucus over-production, and several anti-IL-13 therapeutic antibodies are currently in clinical development for the treatment of asthma. Conventional cell-based bioassays for evaluating neutralization potencies of IL-13 antagonists are semi-quantitative or with a low sensitivity. Here, we report the development of a highly sensitive bioassay to assess the potency of IL-13 antagonists using human lung epithelial A-549 cells that produce thymus and activation-regulated chemokine (TARC) in response to IL-13 stimulation. The A-549 cells were responsive to both wild type and a variant form of recombinant human IL-13 in a concentration-dependent manner, with a 16 to 24 h exposure time found to be within the linear portion of the bioassay response range. The Effective Concentration at 50% of the maximal response (ED50) of IL-13 determined for the assay was 1-5 ng/mL. With this level of IL-13, an anti-IL-13 antibody B-B13 yielded an approximate median Inhibitory Concentration (IC50) value of 0.2 nM. Bioassay optimization was performed to achieve best assay condition and sensitivity. Additionally, IL-13 antagonist potency against natural human IL-13 was also investigated in the A-549 cell bioassay.


Asunto(s)
Bioensayo/métodos , Quimiocina CCL17/metabolismo , Interleucina-13/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Quimiocina CCL17/inmunología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Interleucina-13/inmunología , Interleucina-13/metabolismo , Subunidad alfa1 del Receptor de Interleucina-13/inmunología , Subunidad alfa1 del Receptor de Interleucina-13/metabolismo , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Interleucina-2/inmunología , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Sensibilidad y Especificidad , Factor de Necrosis Tumoral alfa/metabolismo
13.
Protein Expr Purif ; 46(2): 299-308, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16256367

RESUMEN

A truncated form of human procaspase-9 missing the first 111 amino acids, and a variety of mutants derived therefrom, have been expressed in Escherichia coli inclusion bodies. Upon refolding to active enzymes, Delta(1-111) procaspase-9 and mutants were recovered at purity greater than 95% and with a final yield of 20-35 mg/L cell culture. Our active procaspase-9 retains its pro-segment, while undergoing major auto processing at Asp315 and a minor (20%) cleavage at Glu306. This unusual cleavage at a Glu-X bond also took place in the D315E mutant, and we describe herein the inhibitor Z-VAE-fmk that shows enhanced inactivation of procaspase-9 over caspases-3. The bond at Asp330, not processed by procaspase-9, is cleaved by caspase-3 and the resulting procaspase-9 variant, missing the 316-330 bridge, is six times as active as the non-mutated Delta(1-111) proenzyme. A deletion mutant lacking residues 316-330 underwent auto activation by cleavage at Asp315-Ala331 bond. Moreover, substitution of Glu306 by an Asp residue in this mutant led to rapid removal of the peptide spanning Ser307 to Asp330, and resulted in an enzyme that was 7.6 times as active as the non-mutated Delta(1-111) procaspase-9. Finally, replacing both Asp315 and Glu306 with Ala generated a procaspase-9 mutant incapable of auto processing. This single chain procaspase-9 was fully as active as the non-mutated Delta(1-111) enzyme processed at Asp315 or Glu306. Our demonstration that unprocessed procaspase-9 mutants are active as proteases with caspase-type specificity suggests that the role of procaspase-9 in cascade activation of executioner caspases might, in some circumstances, be carried out alone and without association of the apoptosome.


Asunto(s)
Caspasas/biosíntesis , Caspasas/química , Expresión Génica , Pliegue de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Errores Innatos del Metabolismo de los Aminoácidos , Caspasa 9 , Caspasas/genética , Activación Enzimática/genética , Escherichia coli , Humanos , Cuerpos de Inclusión/química , Cuerpos de Inclusión/genética , Proteínas Recombinantes/genética , Eliminación de Secuencia
14.
J Biol Chem ; 279(38): 40227-36, 2004 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-15252021

RESUMEN

Both germinal (gACE) and somatic (sACE) isozymes of angiotensin-converting enzyme (ACE) are type I ectoproteins whose enzymatically active ectodomains are cleaved and shed by a membrane-bound protease. Here, we report a role of protein tyrosine phosphorylation in regulating this process. Strong enhancements of ACE cleavage secretion was observed upon enhancing protein Tyr phosphorylation by treating gACE- or sACE-expressing cells with pervanadate, an inhibitor of protein Tyr phosphatases. Secreted gACE, cell-bound mature gACE and its precursors were all Tyr-phosphorylated, as was the endoplasmic reticulum protein, immunoglobulin heavy chain-binding protein, that co-immunoprecipitated with ACE. The enhancement of cleavage secretion by pervanadate did not require the presence of the cytoplasmic domain of ACE, and it was not accomplished by enhancing the rate of intracellular processing of the protein. The observed enhancement of cleavage secretion of ACE in pervanadate-treated cells was specifically blocked by an inhibitor of the p38 mitogen-activated protein (MAP) kinase but not by inhibitors of many other Ser/Thr and Tyr protein kinases, including a specific inhibitor of protein kinase C that, however, could block the enhancement of cleavage secretion elicited by phorbol ester. These results indicate that ACE Tyr phosphorylation, probably in the endoplasmic reticulum, enhances the rate of its cleavage secretion at the plasma membrane using a regulatory pathway that may involve p38 MAP kinase.


Asunto(s)
Peptidil-Dipeptidasa A/metabolismo , Animales , Carcinógenos/farmacología , Línea Celular , Retículo Endoplásmico/enzimología , Chaperón BiP del Retículo Endoplásmico , Inhibidores Enzimáticos/farmacología , Proteínas de Choque Térmico/metabolismo , Riñón/citología , Espectrometría de Masas , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Chaperonas Moleculares/metabolismo , Peptidil-Dipeptidasa A/química , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína , Acetato de Tetradecanoilforbol/farmacología , Tirosina/metabolismo , Vanadatos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...