Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
RSC Med Chem ; 15(5): 1539-1546, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38784453

RESUMO

Dysregulation of the networking of RNA-binding proteins (RBPs) and RNAs drives many human diseases, including cancers, and the targeting of RNA-protein interactions (RPIs) has emerged as an exciting area of RNA-targeted drug discovery. Accordingly, methods that enable the discovery of cell-active small molecule modulators of RPIs are needed to propel this emerging field forward. Herein, we describe the application of live-cell assay technology, RNA interaction with protein-mediated complementation assay (RiPCA), for high-throughput screening to identify small molecule inhibitors of the pre-let-7d-Lin28A RPI. Utilizing a combination of RNA-biased small molecules and virtual screening hits, we discovered an RNA-binding small molecule that can disrupt the pre-let-7-Lin28 interaction demonstrating the potential of RiPCA for advancing RPI-targeted drug discovery.

2.
J Med Chem ; 66(15): 10734-10745, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37471629

RESUMO

Eukaryotic translation initiation factor 4E (eIF4E) is an RNA-binding protein that binds to the m7GpppX-cap at the 5' terminus of coding mRNAs to initiate cap-dependent translation. While all cells require cap-dependent translation, cancer cells become addicted to enhanced translational capacity, driving the production of oncogenic proteins involved in proliferation, evasion of apoptosis, metastasis, and angiogenesis, among other cancerous phenotypes. eIF4E is the rate-limiting translation factor, and its activation has been shown to drive cancer initiation, progression, metastasis, and drug resistance. These findings have established eIF4E as a translational oncogene and promising, albeit challenging, anti-cancer therapeutic target. Although significant effort has been put forth toward inhibiting eIF4E, the design of cell-permeable, cap-competitive inhibitors remains a challenge. Herein, we describe our work toward solving this long-standing challenge. By employing an acyclic nucleoside phosphonate prodrug strategy, we report the synthesis of cell-permeable inhibitors of eIF4E binding to capped mRNA to inhibit cap-dependent translation.


Assuntos
Fator de Iniciação 4E em Eucariotos , Neoplasias , Fator de Iniciação 4E em Eucariotos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ligação Proteica , Biossíntese de Proteínas , Neoplasias/tratamento farmacológico
3.
bioRxiv ; 2023 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-37292917

RESUMO

Eukaryotic translation initiation factor 4E (eIF4E) is an RNA-binding protein that binds to the m 7 GpppX-cap at the 5' terminus of coding mRNAs to initiate cap-dependent translation. While all cells require cap-dependent translation, cancer cells become addicted to enhanced translational capacity, driving the production of oncogenic proteins involved in proliferation, evasion of apoptosis, metastasis, and angiogenesis among other cancerous phenotypes. eIF4E is the rate-limiting translation factor and its activation has been shown to drive cancer initiation, progression, metastasis, and drug resistance. These findings have established eIF4E as a translational oncogene and promising, albeit challenging, anti-cancer therapeutic target. Although significant effort has been put forth towards inhibiting eIF4E, the design of cell-permeable, cap-competitive inhibitors remains a challenge. Herein, we describe our work towards solving this long-standing challenge. By employing an acyclic nucleoside phosphonate prodrug strategy, we report the synthesis of cell-permeable inhibitors of eIF4E binding to capped mRNA to inhibit cap-dependent translation.

4.
Biochemistry ; 62(11): 1725-1734, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37130292

RESUMO

Dicer is an RNase III enzyme that is responsible for the maturation of small RNAs such as microRNAs. As Dicer's cleavage products play key roles in promoting cellular homeostasis through the fine-tuning of gene expression, dysregulation of Dicer activity can lead to several human diseases, including cancers. Mutations in Dicer have been found to induce tumorigenesis and lead to the development of a rare pleiotropic tumor predisposition syndrome found in children and young adults called DICER1 syndrome. These patients harbor germline and somatic mutations in Dicer that lead to defective microRNA processing and activity. While most mutations occur within Dicer's catalytic RNase III domains, alterations within the Platform-PAZ (Piwi-Argonaute-Zwille) domain also cause loss of microRNA production. Using a combination of in vitro biochemical and cellular studies, we characterized the effect of disease-relevant Platform-PAZ-associated mutations on the processing of a well-studied oncogenic microRNA, pre-microRNA-21. We then compared these results to those of a representative from another Dicer substrate class, the small nucleolar RNA, snord37. From this analysis, we provide evidence that mutations within the Platform-PAZ domain result in differential impacts on RNA binding and processing, adding new insights into the complexities of Dicer processing of small RNA substrates.


Assuntos
MicroRNAs , RNA Nucleolar Pequeno , Criança , Humanos , RNA Nucleolar Pequeno/genética , Ribonuclease III/química , MicroRNAs/química , Mutação , RNA Helicases DEAD-box/genética
5.
ACS Med Chem Lett ; 12(6): 878-886, 2021 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-34141065

RESUMO

MicroRNAs (miRNAs) are a family of small noncoding RNAs that regulate gene expression. Due to their important activity in the fine-tuning of protein translation, abnormal expression of miRNAs has been linked to many human diseases, making the targeting of miRNAs attractive as a novel therapeutic strategy. Accordingly, researchers have been heavily engaged in the discovery of small molecule modulators of miRNAs. With an interest in the identification of new chemical space for targeting miRNAs, we developed a high-throughput screening (HTS) technology, catalytic enzyme-linked click chemistry assay (cat-ELCCA), aimed at the discovery of small molecule ligands for pre-miR-21, a miRNA that is frequently overexpressed in human cancers. From our HTS campaign, we found that natural products, a source of many impactful human medicines, may be a promising source of potential pre-miR-21-selective maturation inhibitors. Herein we describe our first efforts in natural product inhibitor discovery leading to the identification of a depsipeptide class of natural products as RNA-binding inhibitors of Dicer-mediated miRNA processing.

6.
Eur J Med Chem ; 205: 112655, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32739551

RESUMO

Eukaryotic translation initiation factor 4E (eIF4E) has emerged as a promising cancer therapeutic target due to its role in the initiation of cap-dependent translation, a process that is accelerated during tumorigenesis. To regulate the initiation of cap-dependent translation, eIF4E participates in protein-protein interactions (PPI) with binding partners, 4E-BP1 and eIF4G, which act as an inhibitor and stimulator of translation, respectively. As both of these proteins interact with eIF4E by utilizing a short, α-helical stretch of amino acids, our laboratory has been working to develop helical mimetics of these proteins, in particular 4E-BP1, to inhibit eIF4E PPIs. Herein, we describe our continued efforts in this area and report the development and characterization of a cell-penetrant lactam stapled peptide for targeting cellular eIF4E.


Assuntos
Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/farmacologia , Fator de Iniciação 4E em Eucariotos/metabolismo , Lactamas/química , Linhagem Celular Tumoral , Humanos , Terapia de Alvo Molecular , Ligação Proteica/efeitos dos fármacos , Biossíntese de Proteínas
7.
FEBS Lett ; 594(8): 1307-1318, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31853978

RESUMO

Phosphorylation of translational repressor eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) controls the initiation of cap-dependent translation, a type of protein synthesis that is frequently upregulated in human diseases such as cancer. Because of its critical cellular function, it is not surprising that multiple kinases can post-translationally modify 4E-BP1 to drive aberrant cap-dependent translation. We recently reported a site-selective chemoproteomic method for uncovering kinase-substrate interactions, and using this approach, we discovered the cyclin-dependent kinase (CDK)4 as a new 4E-BP1 kinase. Herein, we describe our extension of this work and reveal the role of CDK4 in modulating 4E-BP1 activity in the transition from mitosis to G1, thereby demonstrating a novel role for this kinase in cell cycle regulation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Mitose/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Ciclo Celular/genética , Fase G1/genética , Células HeLa , Humanos , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Biossíntese de Proteínas , Piridinas/farmacologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
8.
J Biol Chem ; 294(46): 17188-17196, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31619517

RESUMO

MicroRNAs (miRNAs or miRs) are small, noncoding RNAs that are implicated in the regulation of most biological processes. Global miRNA biogenesis is altered in many cancers, and RNA-binding proteins play a role in miRNA biogenesis, presenting a promising avenue for targeting miRNA dysregulation in diseases. miR-34a exhibits tumor-suppressive activities by targeting cell cycle regulators CDK4/6 and anti-apoptotic factor BCL-2, among other regulatory pathways such as Wnt, TGF-ß, and Notch signaling. Many cancers exhibit down-regulation or loss of miR-34a, and synthetic miR-34a supplementation has been shown to inhibit tumor growth in vivo However, the post-transcriptional mechanisms that cause miR-34a loss in cancer are not entirely understood. Here, using a proteomics-mediated approach in non-small-cell lung cancer (NSCLC) cells, we identified squamous cell carcinoma antigen recognized by T-cells 3 (SART3) as a putative pre-miR-34a-binding protein. SART3 is a spliceosome recycling factor and nuclear RNA-binding protein with no previously reported role in miRNA regulation. We found that SART3 binds pre-miR-34a with higher specificity than pre-let-7d (used as a negative control) and elucidated a new functional role for SART3 in NSCLC cells. SART3 overexpression increased miR-34a levels, down-regulated the miR-34a target genes CDK4/6, and caused a cell cycle arrest in the G1 phase. In vitro binding experiments revealed that the RNA-recognition motifs within the SART3 sequence are responsible for selective pre-miR-34a binding. Our results provide evidence for a significant role of SART3 in miR-34a biogenesis and cell cycle progression in NSCLC cells.


Assuntos
Antígenos de Neoplasias/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Proteínas de Neoplasias/genética , Proteínas de Ligação a RNA/genética , Apoptose/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Ligação Proteica/genética , Proteômica/métodos , Spliceossomos/genética
9.
Org Biomol Chem ; 17(26): 6414-6419, 2019 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-31215581

RESUMO

Hydrocarbon stapled (HCS) peptides are a class of cross-linked α-helix mimetics. The technology relies on the use of α,α'-disubstituted alkenyl amino acids, which fully contrain the helical region to typically yield peptides with enhanced structural ordering and biological activity. Recently, monosubstituted alkenyl amino acids were disclosed for peptide stapling; however, the impact that this tether has on HCS peptide structure and activity has not yet been fully explored. By applying this HCS to the disordered peptide eIF4E-binding protein 1 (4E-BP1), we discovered that this type of tethering has a dramatic effect on olefin geometry and activity of the resultant stapled peptides, where the putative trans isomer was found to exhibit enhanced in vitro and cellular inhibitory activity against eIF4E protein-protein interactions. We further demonstrated that the metathesis catalyst used for ring-closing metathesis can influence monosubstituted HCS peptide activity, presumably through alteration of the cis/trans olefin ratio. This study represents one of the first in-depth analyses of olefin isomers of a stapled peptide and highlights an additional feature for medicinal chemistry optimization of this class of peptide-based probes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Alcenos/química , Proteínas de Ciclo Celular/química , Peptídeos/química , Humanos , Modelos Moleculares , Peptídeos/síntese química , Especificidade por Substrato
10.
Cell Chem Biol ; 26(7): 980-990.e8, 2019 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-31056462

RESUMO

Recent estimates of the human proteome suggest there are ∼20,000 protein-coding genes, the protein products of which contain >145,000 phosphosites. Unfortunately, in-depth examination of the human phosphoproteome has outpaced the ability to annotate the kinases that mediate these post-translational modifications. To obtain actionable information about phosphorylation-driven signaling cascades, it is essential to identify the kinases responsible for phosphorylating sites that differ across disease states. To fill in these gaps we have developed an unbiased, chemoproteomic approach for identifying high-confidence kinase-substrate interactions with phosphosite specificity. Using this assay, we uncovered the role of cyclin-dependent kinase 4 (CDK4), a clinically validated kinase important for cell-cycle progression, in regulating cap-dependent translation via phosphorylation of the tumor suppressor 4E-BP1. The discovery of this signaling axis sheds light on the mechanisms by which CDK4/6 inhibitors control cell proliferation and constitutes a successful example of kinase discovery using an activity-based, kinase-directed probe.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Ensaios de Triagem em Larga Escala/instrumentação , Ensaios de Triagem em Larga Escala/métodos , Proteínas Adaptadoras de Transdução de Sinal/genética , Adolescente , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Quinase 4 Dependente de Ciclina/genética , Feminino , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Pessoa de Meia-Idade , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteômica/métodos , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
11.
J Med Chem ; 62(10): 4967-4978, 2019 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-31033289

RESUMO

Protein disorder plays a crucial role in signal transduction and is key for many cellular processes including transcription, translation, and cell cycle. Within the intrinsically disordered protein interactome, the α-helix is commonly used for binding, which is induced via a disorder-to-order transition. Because the targeting of protein-protein interactions (PPIs) remains an important challenge in medicinal chemistry, efforts have been made to mimic this secondary structure for rational inhibitor design through the use of stapled peptides. Cap-dependent mRNA translation is regulated by two disordered proteins, 4E-BP1 and eIF4G, that inhibit or stimulate the activity of the m7G cap-binding translation initiation factor, eIF4E, respectively. Both use an α-helical motif for eIF4E binding, warranting the investigation of stapled peptide mimics for manipulating eIF4E PPIs. Herein, we describe our efforts toward this goal, resulting in the synthesis of a cell-active stapled peptide for further development in manipulating aberrant cap-dependent translation in human diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas de Ciclo Celular/química , Desenho de Fármacos , Fator de Iniciação 4E em Eucariotos/química , Fator de Iniciação Eucariótico 4G/química , Fragmentos de Peptídeos/síntese química , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fator de Iniciação 4E em Eucariotos/antagonistas & inibidores , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação Eucariótico 4G/antagonistas & inibidores , Fator de Iniciação Eucariótico 4G/genética , Humanos , Concentração Inibidora 50 , Cinética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Plasmídeos , Ligação Proteica
12.
Eur J Med Chem ; 166: 339-350, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30735900

RESUMO

Eukaryotic translation initiation factor 4E (eIF4E) is a key player in the initiation of cap-dependent translation through recognition of the m7GpppX cap at the 5' terminus of coding mRNAs. As eIF4E overexpression has been observed in a number of human diseases, most notably cancer, targeting this oncogenic translation initiation factor has emerged as a promising strategy for the development of novel anti-cancer therapeutics. Toward this end, in the present study, we have rationally designed a series of Bn7GxP-based PROTACs for the targeted degradation of eIF4E. Herein we describe our synthetic efforts, in addition to biochemical and cellular characterization of these compounds.


Assuntos
Desenho de Fármacos , Fator de Iniciação 4E em Eucariotos/metabolismo , Guanosina/análogos & derivados , Proteólise/efeitos dos fármacos , Linhagem Celular Tumoral , Técnicas de Química Sintética , Fator de Iniciação 4E em Eucariotos/química , Guanosina/síntese química , Guanosina/química , Guanosina/farmacologia , Humanos , Modelos Moleculares , Conformação Proteica
13.
Chembiochem ; 20(1): 40-45, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30137694

RESUMO

Proteins containing intrinsic disorder often form secondary structure upon interaction with a binding partner. Modulating such structures presents an approach for manipulating the resultant functional outcomes. Translational repressor protein 4E-BP1 is an example of an intrinsically disordered protein that forms an α-helix upon binding to its protein ligand, eIF4E. Current biophysical methods for analyzing binding-induced structural changes are low-throughput, require large amounts of sample, or are extremely sensitive to signal interference by the ligand itself. Herein, we describe the discovery and development of a conditionally fluorescent 4E-BP1 peptide that reports structural changes of its helix in high-throughput format. This reporter peptide is based on conditional quenching of fluorescein by thioamides. In this case, fluorescence signal increases as the peptide becomes more ordered. Conversely, destabilization of the α-helix results in decreased fluorescence signal. The low concentration and low volume of peptide required make this approach amenable for high-throughput screening to discover ligands that alter peptide secondary structure.


Assuntos
Proteínas de Transporte/metabolismo , Corantes Fluorescentes/química , Peptídeos/metabolismo , Tioamidas/química , Sequência de Aminoácidos , Proteínas de Transporte/síntese química , Proteínas de Transporte/química , Fator de Iniciação 4E em Eucariotos/metabolismo , Fluoresceína-5-Isotiocianato/química , Humanos , Peptídeos/síntese química , Peptídeos/química , Conformação Proteica em alfa-Hélice , Dobramento de Proteína
15.
Proc Natl Acad Sci U S A ; 111(34): 12556-61, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25114221

RESUMO

In a fluorescence polarization screen for the MYC-MAX interaction, we have identified a novel small-molecule inhibitor of MYC, KJ-Pyr-9, from a Kröhnke pyridine library. The Kd of KJ-Pyr-9 for MYC in vitro is 6.5 ± 1.0 nM, as determined by backscattering interferometry; KJ-Pyr-9 also interferes with MYC-MAX complex formation in the cell, as shown in a protein fragment complementation assay. KJ-Pyr-9 specifically inhibits MYC-induced oncogenic transformation in cell culture; it has no or only weak effects on the oncogenic activity of several unrelated oncoproteins. KJ-Pyr-9 preferentially interferes with the proliferation of MYC-overexpressing human and avian cells and specifically reduces the MYC-driven transcriptional signature. In vivo, KJ-Pyr-9 effectively blocks the growth of a xenotransplant of MYC-amplified human cancer cells.


Assuntos
Antineoplásicos/farmacologia , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Piridinas/farmacologia , Pirimidinas/farmacologia , Animais , Antineoplásicos/química , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/antagonistas & inibidores , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/química , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Células Cultivadas , Embrião de Galinha , Avaliação Pré-Clínica de Medicamentos , Feminino , Polarização de Fluorescência , Genes myc , Humanos , Interferometria , Camundongos , Camundongos Nus , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/química , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/química , Piridinas/química , Pirimidinas/química , Ensaios Antitumorais Modelo de Xenoenxerto
16.
ACS Chem Biol ; 8(6): 1117-20, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23517377

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) preferentially induces apoptosis in cancer cells over normal cells; however, tumor cells may develop TRAIL resistance. Here, we demonstrate that this resistance can be overcome in the presence of bacterial acylhomoserine lactones (AHLs) or AHL-producing bacteria through the combined effect of TRAIL-induced apoptosis and AHL-mediated inhibition of inflammation regulated by NF-κB signaling. This discovery unveils a previously unrecognized symbiotic link between bacteria and host immunosurveillance.


Assuntos
Acil-Butirolactonas/imunologia , Citocinas/imunologia , Neoplasias/imunologia , Neoplasias/microbiologia , Pseudomonas aeruginosa/imunologia , Acil-Butirolactonas/química , Apoptose , Linhagem Celular Tumoral , Humanos , NF-kappa B/imunologia , Pseudomonas aeruginosa/química , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/imunologia
18.
J Med Chem ; 54(11): 3963-72, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21534605

RESUMO

Onchocerciasis, or river blindness, is a neglected tropical disease that affects more than 37 million people worldwide, primarily in Africa and Central and South America. We have disclosed evidence that the larval-stage-specific chitinase, OvCHT1, may be a potential biological target for affecting nematode development. On the basis of screening efforts, closantel, a known anthelmintic drug, was discovered as a potent and highly specific OvCHT1 inhibitor. Originally, closantel's anthelmintic mode of action was believed to rely solely on its role as a proton ionophore; thus, the impact of each of its biological activities on O. volvulus L3 molting was investigated. Structure-activity relationship studies on an active closantel fragment are detailed, and remarkably, by use of a simple salicylanilide scaffold, compounds acting only as protonophores or chitinase inhibitors were identified. From these data, unexpected synergistic protonophore and chitinase inhibition activities have also been found to be critical for molting in O. volvulus L3 larvae.


Assuntos
Quitinases/antagonistas & inibidores , Filaricidas/síntese química , Filaricidas/farmacologia , Onchocerca volvulus/efeitos dos fármacos , Salicilanilidas/síntese química , Salicilanilidas/farmacologia , Animais , Quitinases/química , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Células Epiteliais , Filaricidas/química , Células HEK293 , Haplorrinos , Humanos , Ionóforos/farmacologia , Larva/efeitos dos fármacos , Estrutura Molecular , Muda/efeitos dos fármacos , Onchocerca volvulus/enzimologia , Onchocerca volvulus/crescimento & desenvolvimento , Oncocercose/tratamento farmacológico , Oncocercose/parasitologia , Oncocercose Ocular/tratamento farmacológico , Oncocercose Ocular/parasitologia , Prótons , Salicilanilidas/química , Relação Estrutura-Atividade
19.
Br J Haematol ; 153(6): 741-52, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21517817

RESUMO

We present previously unknown evidence that the immunoglobulin heavy chain binding protein BIP/HSPA5, also known as glucose regulated protein (GRP)78, serving as a pivotal component of the pro-survival axis of the unfolded protein response (UPR) signalling network, is abundantly expressed in relapsed B-lineage acute lymphoblastic leukaemia (ALL) and contributes to chemotherapy resistance of leukaemic B-cell precursors. The resistance of B-lineage ALL cells to the standard anti-leukaemic drug vincristine was overcome by the HSPA5 inhibitor epigallocatechin gallate, which inhibits the anti-apoptotic function of HSPA5 by targeting its ATP-binding domain. Notably, chemotherapy-resistant B-lineage ALL cells underwent apoptosis within 48 h of exposure to a doxorubicin-conjugated cell-penetrating cyclic anti-HSPA5 peptide targeting surface-expressed HSPA5 molecules on leukaemia cells. The identification of the HSPA5 as a chemoresistance biomarker and molecular target for B-lineage ALL may lead to new anti-leukaemic treatment strategies that are much needed.


Assuntos
Proteínas de Choque Térmico/antagonistas & inibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Adolescente , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/fisiologia , Catequina/análogos & derivados , Catequina/farmacologia , Criança , Pré-Escolar , Doxorrubicina/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Chaperona BiP do Retículo Endoplasmático , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiologia , Humanos , Lactente , Terapia de Alvo Molecular/métodos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Recidiva , Transdução de Sinais/genética , Células Tumorais Cultivadas , Resposta a Proteínas não Dobradas/genética
20.
J Biol Chem ; 286(19): 17351-8, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21454495

RESUMO

In the postantibiotic era, available treatment options for severe bacterial infections caused by methicillin-resistant Staphylococcus aureus have become limited. Therefore, new and innovative approaches are needed to combat such life-threatening infections. Virulence factor expression in S. aureus is regulated in a cell density-dependent manner using "quorum sensing," which involves generation and secretion of autoinducing peptides (AIPs) into the surrounding environment to activate a bacterial sensor kinase at a particular threshold concentration. Mouse monoclonal antibody AP4-24H11 was shown previously to blunt quorum sensing-mediated changes in gene expression in vitro and protect mice from a lethal dose of S. aureus by sequestering the AIP signal. We have elucidated the crystal structure of the AP4-24H11 Fab in complex with AIP-4 at 2.5 Å resolution to determine its mechanism of ligand recognition. A key Glu(H95) provides much of the binding specificity through formation of hydrogen bonds with each of the four amide nitrogens in the AIP-4 macrocyclic ring. Importantly, these structural data give clues as to the interactions between the cognate staphylococcal AIP receptors AgrC and the AIPs, as AP4-24H11·AIP-4 binding recapitulates features that have been proposed for AgrC-AIP recognition. Additionally, these structural insights may enable the engineering of AIP cross-reactive antibodies or quorum quenching vaccines for use in active or passive immunotherapy for prevention or treatment of S. aureus infections.


Assuntos
Ligantes , Percepção de Quorum/genética , Staphylococcus aureus/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X/métodos , Regulação Bacteriana da Expressão Gênica , Fragmentos de Imunoglobulinas/química , Imunoglobulina G/química , Proteínas Luminescentes/metabolismo , Modelos Moleculares , Peptídeos/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Percepção de Quorum/imunologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA