Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 213(1): 29-39, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38767437

RESUMO

High-dose (HD) IL-2 was the first immuno-oncology agent approved for treating advanced renal cell carcinoma and metastatic melanoma, but its use was limited because of substantial toxicities. Multiple next-generation IL-2 agents are being developed to improve tolerability. However, a knowledge gap still exists for the genomic markers that define the target pharmacology for HD IL-2 itself. In this retrospective observational study, we collected PBMC samples from 23 patients with metastatic renal cell carcinoma who were treated with HD IL-2 between 2009 and 2015. We previously reported the results of flow cytometry analyses. In this study, we report the results of our RNA-sequencing immunogenomic survey, which was performed on bulk PBMC samples from immediately before (day 1), during (day 3), and after treatment (day 5) in cycle 1 and/or cycle 2 of the first course of HD IL-2. As part of a detailed analysis of immunogenomic response to HD IL-2 treatment, we analyzed the changes in individual genes and immune gene signatures. By day 3, most lymphoid cell types had transiently decreased, whereas myeloid transcripts increased. Although most genes and/or signatures generally returned to pretreatment expression levels by day 5, certain ones representative of B cell, NK cell, and T cell proliferation and effector functions continued to increase, along with B cell (but not T cell) oligoclonal expansion. Regulatory T cells progressively expanded during and after treatment. They showed strong negative correlation with myeloid effector cells. This detailed RNA-sequencing immunogenomic survey of IL-2 pharmacology complements results of prior flow cytometry analyses. These data provide valuable pharmacological context for assessing PBMC gene expression data from patients dosed with IL-2-related compounds that are currently in development.


Assuntos
Carcinoma de Células Renais , Imunoterapia , Interleucina-2 , Neoplasias Renais , Humanos , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/genética , Interleucina-2/administração & dosagem , Interleucina-2/genética , Neoplasias Renais/imunologia , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/patologia , Masculino , Pessoa de Meia-Idade , Feminino , Imunoterapia/métodos , Idoso , Estudos Retrospectivos , Adulto , Leucócitos Mononucleares/imunologia , Metástase Neoplásica
2.
Cancer Res Commun ; 2(8): 894-903, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-36923304

RESUMO

Recombinant human high-dose IL2 (HD-IL2; aldesleukin) was one of the first approved immune-oncology agents based upon clinical activity in renal cell carcinoma (RCC) and metastatic melanoma but use was limited due to severe toxicity. Next-generation IL2 agents designed to improve tolerability are in development, increasing the need for future identification of genomic markers of clinical benefit and/or clinical response. In this retrospective study, we report clinical and tumor molecular profiling from patients with metastatic RCC (mRCC) treated with HD-IL2 and compare findings with patients with RCC treated with anti-PD-1 therapy. Genomic characteristics common and unique to IL2 and/or anti-PD-1 therapy response are presented, with insight into rational combination strategies for these agents. Residual pretreatment formalin-fixed paraffin embedded tumor samples from n = 36 patients with HD-IL2 mRCC underwent RNA-sequencing and corresponding clinical data were collected. A de novo 40-gene nearest centroid IL2 treatment response classifier and individual gene and/or immune marker signature differences were correlated to clinical response and placed into context with a separate dataset of n = 35 patients with anti-PD-1 mRCC. Immune signatures and genes, comprising suppressor and effector cells, were increased in patients with HD-IL2 clinical benefit. The 40-gene response classifier was also highly enriched for immune genes. While several effector immune signatures and genes were common between IL2 and anti-PD-1 treated patients, multiple inflammatory and/or immunosuppressive genes, previously reported to predict poor response to anti-PD-L1 immunotherapy, were only increased in IL2-responsive tumors. These findings suggest that common and distinct immune-related response markers for IL2 and anti-PD-1 therapy may help guide their use, either alone or in combination. Significance: Next-generation IL2 agents, designed for improved tolerability over traditional HD-IL2 (aldesleukin), are in clinical development. Retrospective molecular tumor profiling of patients treated with HD-IL2 or anti-PD-1 therapy provides insights into genomic characteristics of therapy response. This study revealed common and distinct immune-related predictive response markers for IL2 and anti-PD-1 therapy which may play a role in therapy guidance, and rational combination strategies for these agents.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Carcinoma de Células Renais/tratamento farmacológico , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Interleucina-2/genética , Neoplasias Renais/tratamento farmacológico , Estudos Retrospectivos
3.
Nat Commun ; 12(1): 4785, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34373459

RESUMO

The implementation of applied engineering principles to create synthetic biological systems promises to revolutionize medicine, but application of fundamentally redesigned organisms has thus far not impacted practical drug development. Here we utilize an engineered microbial organism with a six-letter semi-synthetic DNA code to generate a library of site-specific, click chemistry compatible amino acid substitutions in the human cytokine IL-2. Targeted covalent modification of IL-2 variants with PEG polymers and screening identifies compounds with distinct IL-2 receptor specificities and improved pharmacological properties. One variant, termed THOR-707, selectively engages the IL-2 receptor beta/gamma complex without engagement of the IL-2 receptor alpha. In mice, administration of THOR-707 results in large-scale activation and amplification of CD8+ T cells and NK cells, without Treg expansion characteristic of IL-2. In syngeneic B16-F10 tumor-bearing mice, THOR-707 enhances drug accumulation in the tumor tissue, stimulates tumor-infiltrating CD8+ T and NK cells, and leads to a dose-dependent reduction of tumor growth. These results support further characterization of the immune modulatory, anti-tumor properties of THOR-707 and represent a fundamental advance in the application of synthetic biology to medicine, leveraging engineered semi-synthetic organisms as cellular factories to facilitate discovery and production of differentiated classes of chemically modified biologics.


Assuntos
Antineoplásicos/uso terapêutico , Interleucina-2/química , Interleucina-2/metabolismo , Interleucina-2/farmacologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Descoberta de Drogas , Engenharia Genética , Humanos , Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2 , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Linfócitos/efeitos dos fármacos , Camundongos , Biologia Sintética
4.
Circ Res ; 125(7): 699-706, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31426700

RESUMO

RATIONALE: Gq signaling in cardiac myocytes is classically considered toxic. Targeting Gq directly to test this is problematic, because cardiac myocytes have many Gq-coupled receptors. OBJECTIVE: Test whether Gq coupling is required for the cardioprotective effects of an alpha-1A-AR (adrenergic receptor) agonist. METHODS AND RESULTS: In recombinant cells, a mouse alpha-1A-AR with a 6-residue substitution in the third intracellular loop does not couple to Gq signaling. Here we studied a knockin mouse with this alpha-1A-AR mutation. Heart alpha-1A receptor levels and antagonist affinity in the knockin were identical to wild-type. In wild-type cardiac myocytes, the selective alpha-1A agonist A61603-stimulated phosphoinositide-phospholipase C and myocyte contraction. In myocytes with the alpha-1A knockin, both A61603 effects were absent, indicating that Gq coupling was absent. Surprisingly, A61603 activation of cardioprotective ERK (extracellular signal-regulated kinase) was markedly impaired in the KI mutant myocytes, and A61603 did not protect mutant myocytes from doxorubicin toxicity in vitro. Similarly, mice with the α1A KI mutation had increased mortality after transverse aortic constriction, and A61603 did not rescue cardiac function in mice with the Gq coupling-defective alpha-1A receptor. CONCLUSIONS: Gq coupling is required for cardioprotection by an alpha-1A-AR agonist. Gq signaling can be adaptive.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Cardiotônicos/farmacologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Imidazóis/farmacologia , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Tetra-Hidronaftalenos/farmacologia , Substituição de Aminoácidos , Animais , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Contração Miocárdica , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Fosfoinositídeo Fosfolipase C/metabolismo , Domínios Proteicos , Receptores Adrenérgicos alfa 1/química , Receptores Adrenérgicos alfa 1/genética , Transdução de Sinais
5.
J Med Chem ; 60(8): 3511-3517, 2017 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-28300404

RESUMO

A prevalent observation in high-throughput screening and drug discovery programs is the inhibition of protein function by small-molecule compound aggregation. Here, we present the X-ray structural description of aggregation-based inhibition of a protein-protein interaction involving tumor necrosis factor α (TNFα). An ordered conglomerate of an aggregating small-molecule inhibitor (JNJ525) induces a quaternary structure switch of TNFα that inhibits the protein-protein interaction between TNFα and TNFα receptors. SPD-304 may employ a similar mechanism of inhibition.


Assuntos
Fator de Necrose Tumoral alfa/antagonistas & inibidores , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Cristalografia por Raios X , Humanos , Estrutura Molecular , Ligação Proteica , Espectroscopia de Prótons por Ressonância Magnética , Fator de Necrose Tumoral alfa/química
6.
Sci Rep ; 6: 35598, 2016 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-27982031

RESUMO

Dysregulated activity of A Disintegrin And Metalloproteinase 17 (ADAM17)/TNFα Converting Enzyme (TACE) is associated with inflammatory disorders and cancer progression by releasing regulatory membrane-tethered proteins like TNFα, IL6R and EGFR ligands. Although specific inhibition of TACE is thought to be a viable strategy for inflammatory disorders and for malignancies treatment, the generation of effective inhibitors in vivo has been proven to be challenging. Here we report on the development of a protein inhibitor that leverages the endogenous modulator of TACE. We have generated a stable form of the auto-inhibitory TACE prodomain (TPD), which specifically inhibits in vitro and cell-surface TACE, but not the related ADAM10, and effectively modulated TNFα secretion in cells. TPD significantly attenuated TACE-mediated disease models of sepsis, rheumatoid arthritis (RA) and inflammatory bowel disease (IBD), and reduced TNFα in synovial fluids from RA patients. Our results demonstrate that intervening with endogenous ADAM sheddase modulatory mechanisms holds potential as a general strategy for the design of ADAM inhibitors.


Assuntos
Proteína ADAM17/química , Artrite/tratamento farmacológico , Colite/tratamento farmacológico , Inibidores Enzimáticos/administração & dosagem , Choque Séptico/tratamento farmacológico , Proteína ADAM10/metabolismo , Proteína ADAM17/antagonistas & inibidores , Animais , Artrite/induzido quimicamente , Artrite/metabolismo , Células Cultivadas , Colite/induzido quimicamente , Colite/metabolismo , Colágeno/efeitos adversos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Lipopolissacarídeos/efeitos adversos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Domínios Proteicos , Choque Séptico/induzido quimicamente , Choque Séptico/metabolismo , Ácido Trinitrobenzenossulfônico/efeitos adversos
7.
J Biomol Screen ; 21(2): 127-35, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26442913

RESUMO

Leukotrienes (LTs) and related species are proinflammatory lipid mediators derived from arachidonic acid (AA) that have pathological roles in autoimmune and inflammatory conditions, cardiovascular diseases, and cancer. 5-Lipoxygenase activating protein (FLAP) plays a critical accessory role in the conversion of AA to LTA4, and its subsequent conversion to LTC4 by LTC4 synthase. Pharmacological inhibition of FLAP results in a loss of LT production by preventing the biosynthesis of both LTB4 and LTC4, making it an attractive target for the treatment of inflammatory diseases in which LTs likely play a role. Small-molecule (SM) drugs often exhibit polypharmacology through various pathways, which may explain the differential therapeutic efficacies of compounds sharing structural similarity. We have profiled a series of SM FLAP modulators for their selectivity across enzymes of AA cascade in human whole blood (HWB), using a recently developed LC/MS (liquid chromatography-mass spectrometry)-based high-throughput lipidomics platform that monitors 122 eicosanoids in multiplex. Highly efficient data acquisition coupled with fast and accurate data analysis allowed facile compound profiling from ex vivo study samples. This platform allowed us to quantitatively map the effects of those SMs on the entire AA cascade, demonstrating its potential to discriminate structurally related compounds.


Assuntos
Proteínas Ativadoras de 5-Lipoxigenase/química , Bibliotecas de Moléculas Pequenas/química , Eicosanoides/química , Glutationa Transferase/química , Humanos , Leucotrienos/química , Polifarmacologia
8.
J Biomol Screen ; 20(7): 876-86, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25948491

RESUMO

An EPIC label-free phenotypic platform was developed to explore B cell receptor (BCR) and CD40R-mediated B cell activation. The phenotypic assay measured the association of RL non-Hodgkin's lymphoma B cells expressing lymphocyte function-associated antigen 1 (LFA-1) to intercellular adhesion molecule 1 (ICAM-1)-coated EPIC plates. Anti-IgM (immunoglobulin M) mediated BCR activation elicited a response that was blocked by LFA-1/ICAM-1 specific inhibitors and a panel of Bruton's tyrosine kinase (BTK) inhibitors. LFA-1/ICAM-1 association was further increased on coapplication of anti-IgM and mega CD40L when compared to individual application of either. Anti-IgM, mega CD40L, or the combination of both displayed distinct kinetic profiles that were inhibited by treatment with a BTK inhibitor. We also established a FLIPR-based assay to measure B cell activation in Ramos Burkitt's lymphoma B cells and an RL cell line. Anti-IgM-mediated BCR activation elicited a robust calcium response that was inhibited by a panel of BTK inhibitors. Conversely, CD40R activation did not elicit a calcium response in the FLIPR assay. Compared to the FLIPR, the EPIC assay has the propensity to identify inhibitors of both BCR and CD40R-mediated B cell activation and may provide more pharmacological depth or novel mechanisms of action for inhibition of B cell activation.


Assuntos
Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Descoberta de Drogas/métodos , Imunossupressores/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Fenótipo , Anticorpos Anti-Idiotípicos/farmacologia , Linfócitos B/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Humanos , Concentração Inibidora 50 , Molécula 1 de Adesão Intercelular/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo
9.
J Lipid Res ; 56(8): 1492-500, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26022804

RESUMO

It is widely accepted that small-molecule drugs, despite their selectivity at primary targets, exert pharmacological effects (and safety liabilities) through a multiplicity of pathways. As such, it has proved extremely difficult to experimentally assess polypharmacology in an agnostic fashion. Profiling of metabolites produced as part of physiological responses to pharmacological stimuli provides a unique opportunity to explore drug pharmacology. A total of 122 eicosanoid lipids in human whole blood were monitored from 10 different donors upon stimulation with several inducers of immunological responses and treatment with modulators of prostaglandin (PG) and leukotriene biosynthesis, including clinical and investigational molecules. Such analysis revealed differentiation between drugs nominally targeting different eicosanoid biosynthetic enzymes, or even those designed to target the same enzyme. Profiled agents, some of them marketed products, affect eicosanoid biosynthesis in ways that cannot be predicted from information on their intended targets. As an example, we used this platform to discriminate drugs based on their ability to silence PG biosynthesis in response to bacterial lipopolysaccharide, resulting in differential pharmacological activity in an in vivo model of endotoxemia. Some of the observed effects are subject to variability among individuals, indicating a potential application of this methodology to the patient stratification, based on their responses to benchmark drugs and experimental compounds read on the eicosanome via a simple blood test.


Assuntos
Eicosanoides/sangue , Metabolômica , Fenótipo , Polifarmacologia , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Medicina de Precisão
10.
J Pharmacol Exp Ther ; 350(2): 455-68, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24917542

RESUMO

Both preclinical evidence and clinical evidence suggest that α7 nicotinic acetylcholine receptor activation (α7nAChR) improves cognitive function, the decline of which is associated with conditions such as Alzheimer's disease and schizophrenia. Moreover, allosteric modulation of α7nAChR is an emerging therapeutic strategy in an attempt to avoid the rapid desensitization properties associated with the α7nAChR after orthosteric activation. We used a calcium assay to screen for positive allosteric modulators (PAMs) of α7nAChR and report on the pharmacologic characterization of the novel compound RO5126946 (5-chloro-N-[(1S,3R)-2,2-dimethyl-3-(4-sulfamoyl-phenyl)-cyclopropyl]-2-methoxy-benzamide), which allosterically modulates α7nAChR activity. RO5126946 increased acetylcholine-evoked peak current and delayed current decay but did not affect the recovery of α7nAChRs from desensitization. In addition, RO5126946's effects were absent when nicotine-evoked currents were completely blocked by coapplication of the α7nAChR-selective antagonist methyl-lycaconitine. RO5126946 enhanced α7nAChR synaptic transmission and positively modulated GABAergic responses. The absence of RO5126946 effects at human α4ß2nAChR and 5-hydroxytryptamine 3 receptors, among others, indicated selectivity for α7nAChRs. In vivo, RO5126946 is orally bioavailable and brain-penetrant and improves associative learning in a scopolamine-induced deficit model of fear conditioning in rats. In addition, procognitive effects of RO5126946 were investigated in the presence of nicotine to address potential pharmacologic interactions on behavior. RO5126946 potentiated nicotine's effects on fear memory when both compounds were administered at subthreshold doses and did not interfere with procognitive effects observed when both compounds were administered at effective doses. Overall, RO5126946 is a novel α7nAChR PAM with cognitive-enhancing properties.


Assuntos
Benzamidas/farmacologia , Sulfonamidas/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/efeitos dos fármacos , Regulação Alostérica , Animais , Células Cultivadas , Cognição/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Humanos , Aprendizagem/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Nicotina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/fisiologia , Receptores de Glutamato/fisiologia
11.
Br J Pharmacol ; 160(6): 1387-98, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20590629

RESUMO

BACKGROUND AND PURPOSE: Purinoceptors containing the P2X3 subunit (P2X3 homotrimeric and P2X2/3 heterotrimeric) are members of the P2X family of ion channels gated by ATP and may participate in primary afferent sensitization in a variety of pain-related diseases. The current work describes the in vitro pharmacological characteristics of AF-353, a novel, orally bioavailable, highly potent and selective P2X3/P2X2/3 receptor antagonist. EXPERIMENTAL APPROACH: The antagonistic potencies (pIC(50)) of AF-353 for rat and human P2X3 and human P2X2/3 receptors were determined using methods of radioligand binding, intracellular calcium flux and whole cell voltage-clamp electrophysiology. KEY RESULTS: The pIC(50) estimates for these receptors ranged from 7.3 to 8.5, while concentrations 300-fold higher had little or no effect on other P2X channels or on an assortment of receptors, enzymes and transporter proteins. In contrast to A-317491 and TNP-ATP, competition binding and intracellular calcium flux experiments suggested that AF-353 inhibits activation by ATP in a non-competitive fashion. Favourable pharmacokinetic parameters were observed in rat, with good oral bioavailability (%F = 32.9), reasonable half-life (t(1/2) = 1.63 h) and plasma-free fraction (98.2% protein bound). CONCLUSIONS AND IMPLICATIONS: The combination of a favourable pharmacokinetic profile with the antagonist potency and selectivity for P2X3 and P2X2/3 receptors suggests that AF-353 is an excellent in vivo tool compound for study of these channels in animal models and demonstrates the feasibility of identifying and optimizing molecules into potential clinical candidates, and, ultimately, into a novel class of therapeutics for the treatment of pain-related disorders.


Assuntos
Trifosfato de Adenosina/metabolismo , Éteres Fenílicos/farmacologia , Antagonistas do Receptor Purinérgico P2 , Pirimidinas/farmacologia , Administração Oral , Animais , Disponibilidade Biológica , Células CHO , Cálcio/metabolismo , Cricetinae , Cricetulus , Meia-Vida , Humanos , Concentração Inibidora 50 , Masculino , Técnicas de Patch-Clamp , Éteres Fenílicos/administração & dosagem , Éteres Fenílicos/farmacocinética , Pirimidinas/administração & dosagem , Pirimidinas/farmacocinética , Ratos , Ratos Wistar , Receptores Purinérgicos P2X2 , Receptores Purinérgicos P2X3
12.
J Neurosci ; 28(46): 12052-61, 2008 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19005070

RESUMO

Tumor necrosis factor-alpha (TNFalpha) is a proinflammatory cytokine that is elevated in Alzheimer's disease (AD) brains. Because TNFalpha is released from cell membranes by the TNFalpha-converting enzyme (TACE), inhibition of TACE has the potential to mitigate TNFalpha effects in AD brain. TACE also cleaves amyloid precursor protein (APP) and generates sAPPalpha, precluding the formation of potentially harmful amyloid beta (Abeta) peptides by beta-site APP cleaving enzymes (BACE). Hence, the anti-inflammatory benefits of TACE inhibition might be offset by an increase in Abeta. We have examined the effects of the highly selective TACE inhibitor, BMS-561392, on APP processing in vitro and in vivo. In Chinese hamster ovary cells expressing APP, BMS-561392 significantly reduced secretion of sAPPalpha without a corresponding increase in Abeta production. Conversely, a BACE inhibitor decreased sAPPbeta and Abeta peptides with no change in the secretion of sAPPalpha. These data indicate an absence of TACE and BACE competition for the APP substrate. Despite this, we observed competition for APP when TACE activity was enhanced via phorbol ester treatment or if APP was modified such that it was retained within the trans-Golgi network (TGN). These results suggest that BACE and TACE share a common TGN localization, but under normal conditions do not compete for APP. To confirm this finding in vivo, BMS-561392 was infused into the brains of Tg2576 and wild-type mice. Although decreased brain sAPPalpha levels were observed, steady-state Abeta levels were not significantly changed. Accordingly, it is possible that TACE inhibitors could reduce TNFalpha levels without increasing Abeta levels within the AD brain.


Assuntos
Proteínas ADAM/metabolismo , Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Placa Amiloide/metabolismo , Proteínas ADAM/antagonistas & inibidores , Proteína ADAM17 , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Células CHO , Cricetinae , Cricetulus , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ésteres de Forbol/farmacologia , Placa Amiloide/patologia , Quinolinas/farmacologia , Rede trans-Golgi/efeitos dos fármacos , Rede trans-Golgi/metabolismo
13.
Biochemistry ; 47(37): 9911-9, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18707126

RESUMO

The TNFalpha converting enzyme (TACE) is a zinc metalloproteinase that mediates shedding of multiple cell surface proteins. Regulation of TACE enzymatic activity is ultimately mediated via proteolytic removal of its inhibitory prodomain. Sequence determinants for TACE prodomain inhibition of the catalytic domain are yet to be identified. Surprisingly, although TACE and ADAM 10 (closest homologue) share only 23% sequence identity at their prodomains, the latter in isolation inhibits TACE with the same potency as TACE own prodomain. In contrast, the prodomain of ADAM 9 inhibited TACE only weakly. Detailed analysis of ADAM prodomains revealed two short regions for which TACE and ADAM 10 depart dramatically from all other family members. We prepared TACE prodomain variants containing full or partial switches to ADAM 9 residues at those two regions and examined their functional properties. Variants containing ADAM 9 substitutions including amino acid residues 72-82 and 126-137 were fully inactive for TACE inhibition. A third variant comprising residues 114-125 was active but at lower potency relative to wild type. All inactive variants appeared to be correctly folded. Finally, the amino acid residue Phe72 and the motif Asp-Asp-Val-Ile137 were identified within those regions as key determinants for TACE prodomain inhibitory function. We conclude that TACE and ADAM 10 prodomains are functionally equivalent in a way that separates them from the rest of the ADAM family.


Assuntos
Proteínas ADAM/química , Proteínas ADAM/metabolismo , Estrutura Terciária de Proteína , Proteínas ADAM/genética , Proteína ADAM17 , Sequência de Aminoácidos , Domínio Catalítico , Dicroísmo Circular , Dados de Sequência Molecular , Dobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
15.
Proc Natl Acad Sci U S A ; 104(12): 4931-6, 2007 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-17360351

RESUMO

Despite their key roles in many normal and pathological processes, the molecular details by which zinc-dependent proteases hydrolyze their physiological substrates remain elusive. Advanced theoretical analyses have suggested reaction models for which there is limited and controversial experimental evidence. Here we report the structure, chemistry and lifetime of transient metal-protein reaction intermediates evolving during the substrate turnover reaction of a metalloproteinase, the tumor necrosis factor-alpha converting enzyme (TACE). TACE controls multiple signal transduction pathways through the proteolytic release of the extracellular domain of a host of membrane-bound factors and receptors. Using stopped-flow x-ray spectroscopy methods together with transient kinetic analyses, we demonstrate that TACE's catalytic zinc ion undergoes dynamic charge transitions before substrate binding to the metal ion. This indicates previously undescribed communication pathways taking place between distal protein sites and the enzyme catalytic core. The observed charge transitions are synchronized with distinct phases in the reaction kinetics and changes in metal coordination chemistry mediated by the binding of the peptide substrate to the catalytic metal ion and product release. Here we report key local charge transitions critical for proteolysis as well as long sought evidence for the proposed reaction model of peptide hydrolysis. This study provides a general approach for gaining critical insights into the molecular basis of substrate recognition and turnover by zinc metalloproteinases that may be used for drug design.


Assuntos
Proteínas ADAM/metabolismo , Aminoácidos/metabolismo , Domínio Catalítico , Proteína ADAM17 , Sítios de Ligação , Catálise , Desenho de Fármacos , Humanos , Hidrólise , Cinética , Ligantes , Modelos Moleculares , Dinâmica não Linear , Peptídeo Hidrolases/metabolismo , Peptídeos/metabolismo , Especificidade por Substrato , Zinco/metabolismo
16.
Proc Natl Acad Sci U S A ; 104(5): 1621-6, 2007 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-17244710

RESUMO

Semaphorin 4D (sema4D; CD100) is an integral membrane protein and the ligand for two receptors, CD72 and plexin-B1. Soluble sema4D has been shown to evoke angiogenic responses from endothelial cells and impair monocyte migration, but the origin of soluble sema4D, particularly at sites of vascular injury, has been unclear. Here we show that platelets express sema4D and both of its receptors and provide evidence that these molecules promote thrombus formation. We also show that the surface expression of sema4D and CD72 increases during platelet activation, followed by the gradual shedding of the sema4D extracellular domain. Shedding is blocked by metalloprotease inhibitors and abolished in mouse platelets that lack the metalloprotease ADAM17 (TACE). Mice that lack sema4D exhibit delayed arterial occlusion after vascular injury in vivo, and their platelets show impaired collagen responses in vitro. In resting platelets, as in B lymphocytes, CD72 is associated with the protein tyrosine phosphatase SHP-1. Platelet activation causes dissociation of the complex, as does the addition of soluble sema4D. These findings suggest a dual role for sema4D in vascular responses to injury. As thrombus formation begins, platelet-associated sema4D can bind to its receptors on nearby platelets, promoting thrombus formation. As thrombus formation continues, sema4D is shed from the platelet surface and becomes available to interact with receptors on endothelial cells and monocytes, as well as continuing to interact with platelets.


Assuntos
Antígenos CD/química , Plaquetas/metabolismo , Vasos Sanguíneos/lesões , Regulação da Expressão Gênica , Semaforinas/química , Proteínas ADAM/química , Proteína ADAM17 , Animais , Vasos Sanguíneos/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Ativação Plaquetária , Estrutura Terciária de Proteína , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Trombose/metabolismo
17.
Cell Biochem Biophys ; 44(3): 342-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16679521

RESUMO

The tumor necrosis factor-alpha-converting enzyme (TACE) is a member of the disintegrin family of metalloproteinases (ADAMs) that plays a central role in the regulated shedding of a host of cell surface proteins. TACE is biosynthesized as a precursor protein with latent proteolytic activity (zymogen). TACE's zymogen inhibition is mediated by its Pro domain, a 197-amino acid region that serves this function as well as aiding in the secretion of this enzyme through the secretory pathway. We have discovered that a conserved "cysteine switch" consensus motif within TACE's Pro domain is, contrary to expectations, not required for maintenance of the inactive precursor state or for the secretion of this metalloproteinase in its functional form. The only role for this motif seems to be in decreasing TACE's susceptibility to proteolytic degradation during its biogenesis and maturation within the secretory pathway. Interestingly, the Pro domain of TACE seems to carry both its inhibitory and secretory functions through the same mechanism: it seems to prevent the Catalytic domain from accessing its native, functional state, resembling the function of true molecular chaperones. Recent evidence suggests that TACE may also be switched out of the active conformation even by small, drug-like molecules such as the synthetic compound SB-3CT. These findings point at the possibility of developing, in the near future, a new generation of antiinflammatory, noncompetitive TACE inhibitors that would exert negative allosteric modulation over the activity of this key enzyme, mediating several inflammatory diseases and certain cancers.


Assuntos
Proteínas ADAM/metabolismo , Cisteína/metabolismo , Precursores Enzimáticos/metabolismo , Genes de Troca/fisiologia , Proteínas ADAM/biossíntese , Proteínas ADAM/química , Proteínas ADAM/genética , Proteína ADAM17 , Domínio Catalítico , Sequência Conservada , Ativação Enzimática , Precursores Enzimáticos/fisiologia , Regulação da Expressão Gênica/fisiologia , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/química , Metaloendopeptidases/metabolismo , Chaperonas Moleculares/química , Mutação/genética , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes , Fator de Necrose Tumoral alfa/metabolismo
18.
J Biol Chem ; 280(50): 41155-64, 2005 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-16215261

RESUMO

The receptor activator of NF-kappaB (RANK) belongs to the neuregulin/tumor necrosis factor (TNF) receptor superfamily and is activated by RANK ligand (RANK-L), a homotrimeric, TNF-like cytokine. RANK is present on the surface of osteoclast cell precursors, where its interaction with RANK-L induces their terminal differentiation into osteoclasts, thus increasing bone breakdown. The secreted, soluble receptor osteoprotegerin (OPG) interrupts this activation by binding directly to RANK-L. Therefore, osteoclast maturation (and bone homeostasis) is regulated in vivo by OPG levels of expression. We have studied the assembly state and affinity of OPG for RANK-L by sedimentation analyses and surface plasmon resonance (Biacore). Full-length, homodimeric OPG binds to RANK-L with a KD of 10 nM. OPG is also a member of the TNF receptor superfamily and contains four disulfide-rich ligand-binding domains, yet lacks a transmembrane region separating the ligand-binding region from the two death domains, as observed for other receptor family members. We showed that dimerization of OPG results from noncovalent interactions mediated by the death domains and to a lesser extent by a C-terminal heparin-binding region. In contrast, a C-terminal intermolecular disulfide bond does not contribute to the formation or stability of OPG dimers. A truncate of osteoprotegerin, containing the ligand-binding domains but lacking the dimerization domains, bound RANK-L with a KD of approximately 3 microM, indicating that monomer oligomerization for the OPG provides an increase of 3 orders of magnitude in the affinity for RANK-L. Therefore, OPG dimer formation is required for the mechanism of inhibition of the RANK-L/RANK receptor interaction.


Assuntos
Proteínas de Transporte/metabolismo , Glicoproteínas/química , Glicoproteínas de Membrana/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Receptores do Fator de Necrose Tumoral/química , Animais , Afinidade de Anticorpos , Bioensaio , Diferenciação Celular , Membrana Celular/metabolismo , Dicroísmo Circular , Citocinas/metabolismo , Dimerização , Dissulfetos/química , Escherichia coli/metabolismo , Vetores Genéticos , Glicoproteínas/metabolismo , Heparina/química , Humanos , Insetos , Cinética , Ligantes , Modelos Químicos , Mutação , Osteoclastos/metabolismo , Osteoprotegerina , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/química , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Ressonância de Plasmônio de Superfície , Fatores de Tempo , Vírus/genética
19.
Biochem J ; 387(Pt 3): 797-805, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15581423

RESUMO

The prodomain of TACE [TNFalpha (tumour necrosis factor alpha)-converting enzyme] is essential for the secretion of the functional enzyme. Previously, we showed that a TACE truncate was not secreted in the absence of the prodomain and that it was subjected to intracellular degradation. In the present study, we show that full-length TACE was also degraded when expressed without the prodomain. We demonstrate that the prodomain can rescue TACE's secretion in trans, suggesting an intramolecular chaperone function. We addressed the question whether a cysteine switch consensus motif is needed for the secretion of active TACE. The cysteine switch mutants [C184A (Cys184-->Ala)] of TACE resembled the wild-type functionally and in their sensitivity to inhibitors. Interestingly, TACE zymogen forms expressed in the context of the C184A mutation were susceptible to intracellular degradation, suggesting that the prodomain-bound TACE zymogen may be more accessible to intracellular proteinases when compared with mature TACE. Two independent findings confirmed that the catalytic domain of TACE is in a more open state when bound to its prodomain: (i) core tryptophan residues were exposed to the solvent in the procatalytic domain complex and (ii) LysC rapidly proteolysed the procatalytic domain complex but not mature TACE. Therefore the prodomain of TACE is a specific intramolecular chaperone that aids in the secretion of this enzyme, while keeping the catalytic domain in a relatively open conformation. The cysteine switch of TACE is not essential for the secretion of the functional enzyme, but may prevent intracellular degradation of the TACE zymogen.


Assuntos
Proteínas ADAM/biossíntese , Proteínas ADAM/química , Regulação da Expressão Gênica/fisiologia , Chaperonas Moleculares/química , Proteínas ADAM/genética , Proteína ADAM17 , Sequência de Aminoácidos , Cisteína/química , Ativação Enzimática , Mutação , Estrutura Terciária de Proteína
20.
J Biol Chem ; 279(30): 31638-45, 2004 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-15100227

RESUMO

Tumor necrosis factor-alpha-converting enzyme (TACE) is a disintegrin metalloproteinase that processes tumor necrosis factor and a host of other ectodomains. TACE is biosynthesized as a zymogen, and activation requires the removal of an inhibitory pro domain. Little is known about how the pro domain exerts inhibition for this class of enzymes. To study the inhibitory properties of the pro domain of TACE, we have expressed it in isolation from the rest of the protease. Here we show that the TACE pro domain (TACE Pro) is a stably folded protein that is able to inhibit this enzyme. TACE Pro inhibited the catalytic domain of TACE with an IC(50) of 70 nm. In contrast, this inhibitory potency decreased over 30-fold against a TACE form containing the catalytic plus disintegrin/cysteine-rich domains (IC(50) greater that 2 microm). The disintegrin/cysteine-rich region in isolation also decreases the interaction of TACE Pro with the catalytic domain. Surprisingly, we found that the cysteine switch motif located in TACE Pro was not essential for inhibition of the enzymatic activity of TACE; the pro domain variant C184A showed the same inhibitory potency against both TACE forms as wild type TACE Pro. X-ray absorption spectroscopy experiments indicate that binding of TACE Pro to the catalytic domain does include ligation of the catalytic zinc ion via the sulfur atom of its conserved Cys(184) residue. Moreover, the binding of TACE Pro to the catalytic zinc ion partially oxidizes the catalytic zinc ion of the enzyme. Despite this, the nature of the interaction between the pro and catalytic domains of TACE is not consistent with a simple competitive model of inhibition based on cysteine switch ligation of the zinc ion within the active site of TACE.


Assuntos
Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/química , Fator de Necrose Tumoral alfa/metabolismo , Proteínas ADAM , Proteína ADAM17 , Sequência de Aminoácidos , Animais , Domínio Catalítico/genética , Cisteína/química , Técnicas In Vitro , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Mutagênese Sítio-Dirigida , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA