Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Leukemia ; 35(2): 573-584, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32457357

RESUMO

CD38-targeted antibody, daratumumab, is approved for the treatment of multiple myeloma (MM). Phase 1/2 studies GEN501/SIRIUS revealed a novel immunomodulatory mechanism of action (MOA) of daratumumab that enhanced the immune response, reducing natural killer (NK) cells without affecting efficacy or safety. We further evaluated daratumumab's effects on immune cells in whole blood samples of relapsed/refractory MM patients from both treatment arms of the phase 3 POLLUX study (lenalidomide/dexamethasone [Rd] or daratumumab plus Rd [D-Rd]) at baseline (D-Rd, 40; Rd, 45) and after 2 months on treatment (D-Rd, 31; Rd, 33) using cytometry by time-of-flight. We confirmed previous reports of NK cell reduction with D-Rd. Persisting NK cells were phenotypically distinct, with increased expression of HLA-DR, CD69, CD127, and CD27. The proportion of T cells increased preferentially in deep responders to D-Rd, with a higher proportion of CD8+ versus CD4+ T cells. The expansion of CD8+ T cells correlated with clonality, indicating generation of adaptive immune response with D-Rd. D-Rd resulted in a higher proportion of effector memory T cells versus Rd. D-Rd reduced immunosuppressive CD38+ regulatory T cells. This study confirms daratumumab's immunomodulatory MOA in combination with immunomodulatory drugs and provides further insight into immune cell changes and activation status following daratumumab-based therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores/análise , Células Matadoras Naturais/imunologia , Mieloma Múltiplo/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T/imunologia , Anticorpos Monoclonais/administração & dosagem , Dexametasona/administração & dosagem , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Lenalidomida/administração & dosagem , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos
2.
Sci Rep ; 10(1): 13262, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32764586

RESUMO

Phenomic profiles are high-dimensional sets of readouts that can comprehensively capture the biological impact of chemical and genetic perturbations in cellular assay systems. Phenomic profiling of compound libraries can be used for compound target identification or mechanism of action (MoA) prediction and other applications in drug discovery. To devise an economical set of phenomic profiling assays, we assembled a library of 1,008 approved drugs and well-characterized tool compounds manually annotated to 218 unique MoAs, and we profiled each compound at four concentrations in live-cell, high-content imaging screens against a panel of 15 reporter cell lines, which expressed a diverse set of fluorescent organelle and pathway markers in three distinct cell lineages. For 41 of 83 testable MoAs, phenomic profiles accurately ranked the reference compounds (AUC-ROC ≥ 0.9). MoAs could be better resolved by screening compounds at multiple concentrations than by including replicates at a single concentration. Screening additional cell lineages and fluorescent markers increased the number of distinguishable MoAs but this effect quickly plateaued. There remains a substantial number of MoAs that were hard to distinguish from others under the current study's conditions. We discuss ways to close this gap, which will inform the design of future phenomic profiling efforts.


Assuntos
Produtos Biológicos/farmacologia , Proteínas Luminescentes/genética , Fenômica/métodos , Bibliotecas de Moléculas Pequenas/farmacologia , Células A549 , Linhagem Celular , Descoberta de Drogas , Regulação da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Proteínas Luminescentes/metabolismo
3.
Cytometry A ; 95(3): 279-289, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30536810

RESUMO

Daratumumab is a CD38-targeted human monoclonal antibody with direct anti-myeloma cell mechanisms of action. Flow cytometry in relapsed and/or refractory multiple myeloma (RRMM) patients treated with daratumumab revealed cytotoxic T-cell expansion and reduction of immune-suppressive populations, suggesting immune modulation as an additional mechanism of action. Here, we performed an in-depth analysis of the effects of daratumumab on immune-cell subpopulations using high-dimensional mass cytometry. Whole-blood and bone-marrow baseline and on-treatment samples from RRMM patients who participated in daratumumab monotherapy studies (SIRIUS and GEN501) were evaluated with high-throughput immunophenotyping. In daratumumab-treated patients, the intensity of CD38 marker expression decreased on many immune cells in SIRIUS whole-blood samples. Natural killer (NK) cells were depleted with daratumumab, with remaining NK cells showing increased CD69 and CD127, decreased CD45RA, and trends for increased CD25, CD27, and CD137 and decreased granzyme B. Immune-suppressive population depletion paralleled previous findings, and a newly observed reduction in CD38+ basophils was seen in patients who received monotherapy. After 2 months of daratumumab, the T-cell population in whole-blood samples from responders shifted to a CD8 prevalence with higher granzyme B positivity (P = 0.017), suggesting increased killing capacity and supporting monotherapy-induced CD8+ T-cell activation. High-throughput cytometry immune profiling confirms and builds upon previous flow cytometry data, including comparable CD38 marker intensity on plasma cells, NK cells, monocytes, and B/T cells. Interestingly, a shift toward cytolytic granzyme B+ T cells was also observed and supports adaptive responses in patients that may contribute to depth of response. © 2018 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/imunologia , Antígenos de Diferenciação de Linfócitos T/metabolismo , Basófilos/citologia , Basófilos/efeitos dos fármacos , Basófilos/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Citometria de Fluxo , Granzimas/metabolismo , Humanos , Imunofenotipagem , Células Matadoras Naturais/citologia , Mieloma Múltiplo/sangue , Mieloma Múltiplo/metabolismo , Recidiva
4.
Cell Chem Biol ; 25(5): 611-618.e3, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29503208

RESUMO

In both academia and the pharmaceutical industry, large-scale assays for drug discovery are expensive and often impractical, particularly for the increasingly important physiologically relevant model systems that require primary cells, organoids, whole organisms, or expensive or rare reagents. We hypothesized that data from a single high-throughput imaging assay can be repurposed to predict the biological activity of compounds in other assays, even those targeting alternate pathways or biological processes. Indeed, quantitative information extracted from a three-channel microscopy-based screen for glucocorticoid receptor translocation was able to predict assay-specific biological activity in two ongoing drug discovery projects. In these projects, repurposing increased hit rates by 50- to 250-fold over that of the initial project assays while increasing the chemical structure diversity of the hits. Our results suggest that data from high-content screens are a rich source of information that can be used to predict and replace customized biological assays.


Assuntos
Reposicionamento de Medicamentos/métodos , Processamento de Imagem Assistida por Computador/métodos , Aprendizado de Máquina , Redes Neurais de Computação , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Ensaios de Triagem em Larga Escala/métodos , Humanos , Neoplasias/tratamento farmacológico
5.
Genome Med ; 9(1): 80, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28854983

RESUMO

The identification of functional non-coding mutations is a key challenge in the field of genomics. Here we introduce µ-cisTarget to filter, annotate and prioritize cis-regulatory mutations based on their putative effect on the underlying "personal" gene regulatory network. We validated µ-cisTarget by re-analyzing the TAL1 and LMO1 enhancer mutations in T-ALL, and the TERT promoter mutation in melanoma. Next, we re-sequenced the full genomes of ten cancer cell lines and used matched transcriptome data and motif discovery to identify master regulators with de novo binding sites that result in the up-regulation of nearby oncogenic drivers. µ-cisTarget is available from http://mucistarget.aertslab.org .


Assuntos
Análise Mutacional de DNA/métodos , Redes Reguladoras de Genes , Genes Neoplásicos , Mutação , Neoplasias/genética , Sequências Reguladoras de Ácido Nucleico , Algoritmos , Sítios de Ligação , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Genômica/métodos , Humanos , Masculino , Neoplasias/metabolismo , Medicina de Precisão/métodos , Fatores de Transcrição/metabolismo
6.
J Cheminform ; 9: 17, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28316655

RESUMO

Chemogenomics data generally refers to the activity data of chemical compounds on an array of protein targets and represents an important source of information for building in silico target prediction models. The increasing volume of chemogenomics data offers exciting opportunities to build models based on Big Data. Preparing a high quality data set is a vital step in realizing this goal and this work aims to compile such a comprehensive chemogenomics dataset. This dataset comprises over 70 million SAR data points from publicly available databases (PubChem and ChEMBL) including structure, target information and activity annotations. Our aspiration is to create a useful chemogenomics resource reflecting industry-scale data not only for building predictive models of in silico polypharmacology and off-target effects but also for the validation of cheminformatics approaches in general.

7.
Nucleic Acids Res ; 38(18): 6135-47, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20484370

RESUMO

Lens epithelium-derived growth factor/p75 (LEDGF/p75) is a transcriptional coactivator involved in stress response, autoimmune disease, cancer and HIV replication. A fusion between the nuclear pore protein NUP98 and LEDGF/p75 has been found in human acute and chronic myeloid leukemia and association of LEDGF/p75 with mixed-lineage leukemia (MLL)/menin is critical for leukemic transformation. During lentiviral replication, LEDGF/p75 tethers the pre-integration complex to the host chromatin resulting in a bias of integration into active transcription units (TUs). The consensus function of LEDGF/p75 is tethering of cargos to chromatin. In this regard, we determined the LEDGF/p75 chromatin binding profile. To this purpose, we used DamID technology and focused on the highly annotated ENCODE (Encyclopedia of DNA Elements) regions. LEDGF/p75 primarily binds downstream of the transcription start site of active TUs in agreement with the enrichment of HIV-1 integration sites at these locations. We show that LEDGF/p75 binding is not restricted to stress response elements in the genome, and correlation analysis with more than 200 genomic features revealed an association with active chromatin markers, such as H3 and H4 acetylation, H3K4 monomethylation and RNA polymerase II binding. Interestingly, some associations did not correlate with HIV-1 integration indicating that not all LEDGF/p75 complexes on the chromosome are amenable to HIV-1 integration.


Assuntos
Cromatina/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Transcrição Gênica , Sítios de Ligação , Linhagem Celular , Cromatina/metabolismo , DNA/química , HIV-1/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas Recombinantes de Fusão/metabolismo , Sítio de Iniciação de Transcrição , Integração Viral
8.
Mol Cell Biol ; 29(3): 758-70, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19029251

RESUMO

Cellular senescence is an irreversible proliferation arrest, tumor suppression process and likely contributor to tissue aging. Senescence is often characterized by domains of facultative heterochromatin, called senescence-associated heterochromatin foci (SAHF), which repress expression of proliferation-promoting genes. Given its likely contribution to tumor suppression and tissue aging, it is essential to identify all components of the SAHF assembly pathway. Formation of SAHF in human cells is driven by a complex of histone chaperones, namely, HIRA and ASF1a. In yeast, the complex orthologous to HIRA/ASF1a contains two additional proteins, Hpc2p and Hir3p. Using a sophisticated approach to search for remote orthologs conserved in multiple species through evolution, we identified the HIRA-associated proteins, UBN1 and UBN2, as candidate human orthologs of Hpc2p. We show that the Hpc2-related domain of UBN1, UBN2, and Hpc2p is an evolutionarily conserved HIRA/Hir-binding domain, which directly interacts with the N-terminal WD repeats of HIRA/Hir. UBN1 binds to proliferation-promoting genes that are repressed by SAHF and associates with histone methyltransferase activity that methylates lysine 9 of histone H3, a site that is methylated in SAHF. UBN1 is indispensable for formation of SAHF. We conclude that UBN1 is an ortholog of yeast Hpc2p and a novel regulator of senescence.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Senescência Celular , Montagem e Desmontagem da Cromatina , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Homologia de Sequência de Aminoácidos , Transativadores/química , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Sequência Conservada , Ciclina A/genética , Ciclina A2 , Evolução Molecular , Heterocromatina/metabolismo , Chaperonas de Histonas , Humanos , Corpos de Inclusão/metabolismo , Metiltransferases/metabolismo , Chaperonas Moleculares , Dados de Sequência Molecular , Proteínas Nucleares/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Transporte Proteico , Sequências Repetitivas de Aminoácidos , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/química
9.
J Biol Chem ; 280(48): 40003-11, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16216881

RESUMO

Maternal embryonic leucine zipper kinase (MELK) is a protein Ser/Thr kinase that has been implicated in stem cell renewal, cell cycle progression, and pre-mRNA splicing, but its substrates and regulation are not yet known. We show here that MELK has a rather broad substrate specificity and does not appear to require a specific sequence surrounding its (auto)phosphorylation sites. We have mapped no less than 16 autophosphorylation sites including serines, threonines, and a tyrosine residue and show that the phosphorylation of Thr167 and Ser171 is required for the activation of MELK. The expression of MELK activity also requires reducing agents such as dithiothreitol or reduced glutathione. Furthermore, we show that MELK is a Ca2+-binding protein and is inhibited by physiological Ca2+ concentrations. The smallest MELK fragment that was still catalytically active comprises the N-terminal catalytic domain and the flanking ubiquitin-associated domain. A C-terminal fragment of MELK functions as an autoinhibitory domain. Our data show that the activity of MELK is regulated in a complex manner and offer new perspectives for the further elucidation of its biological function.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteínas Serina-Treonina Quinases/fisiologia , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Soluções Tampão , Células COS , Cálcio/química , Cálcio/metabolismo , Ciclo Celular , Linhagem Celular , Chlorocebus aethiops , Ditiotreitol/química , Ativação Enzimática , Regulação da Expressão Gênica , Glutationa/química , Humanos , Íons , Cinética , Ligantes , Espectrometria de Massas , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mapeamento de Peptídeos , Peptídeos/química , Fosforilação , Análise Serial de Proteínas , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Serina/química , Células-Tronco/citologia , Especificidade por Substrato , Treonina/química , Tirosina/química
10.
J Virol ; 78(13): 6758-65, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15194750

RESUMO

Two types of geminate structures were purified from African cassava mosaic geminivirus (ACMV)-infected Nicotiana benthamiana plants and analyzed by electron cryomicroscopy and image reconstruction. After cesium sulfate density gradient centrifugation, they were separated into lighter top (T) and heavier bottom (B) components. T particles comigrated with host proteins, whereas B particles were concentrated in a cesium density typical for complete virions. Both particles were composed of two incomplete icosahedra of 11 capsomers each, but T particles were slightly larger (diameter, 22.5 nm) and less dense in the interior than B particles (diameter, 21.5 nm). T particles were frequently associated with small globules of approximately 14 nm diameter of unknown origin. The overall structure of ACMV, a begomovirus transmitted by whiteflies, was similar to that of Maize streak virus (MSV), a mastrevirus transmitted by leafhoppers, although the vertices of the icosahedra were less pronounced. Models of ACMV coat proteins based on Satellite tobacco necrosis virus support the exposure of parts of the molecule essential for transmission specificity by whiteflies and provide possible structural explanations for the smaller protrusion of the ACMV capsid relative to MSV. The differences of ACMV and MSV virion shapes are discussed with reference to their different animal vectors.


Assuntos
Geminiviridae/química , Sequência de Aminoácidos , Microscopia Crioeletrônica , Geminiviridae/genética , Geminiviridae/isolamento & purificação , Processamento de Imagem Assistida por Computador , Manihot/virologia , Modelos Moleculares , Dados de Sequência Molecular , Doenças das Plantas/virologia , Nicotiana/virologia , Vírion/química , Vírion/ultraestrutura
11.
Biochem J ; 371(Pt 2): 321-30, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12533192

RESUMO

The ubiquitous nucleotide pyrophosphatases/phosphodiesterases NPP1-3 consist of a short intracellular N-terminal domain, a single transmembrane domain and a large extracellular part, comprising two somatomedin-B-like domains, a catalytic domain and a poorly defined C-terminal domain. We show here that the C-terminal domain of NPP1-3 is structurally related to a family of DNA/RNA non-specific endonucleases. However, none of the residues that are essential for catalysis by the endonucleases are conserved in NPP1-NPP3, suggesting that the nuclease-like domain of NPP1-3 does not represent a second catalytic domain. Truncation analysis revealed that the nuclease-like domain of NPP1 is required for protein stability, for the targeting of NPP1 to the plasma membrane and for the expression of catalytic activity. We also demonstrate that 16 conserved cysteines in the somatomedin-B-like domains of NPP1, in concert with two flanking cysteines, mediate the dimerization of NPP1. The K173Q polymorphism of NPP1, which maps to the second somatomedin-B-like domain and has been associated with the aetiology of insulin resistance, did not affect the dimerization or catalytic activity of NPP1, and did not endow NPP1 with an affinity for the insulin receptor. Our data suggest that the non-catalytic ectodomains contribute to the subunit structure, stability and function of NPP1-3.


Assuntos
Diester Fosfórico Hidrolases/química , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/química , Pirofosfatases/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Catálise , Chlorocebus aethiops , Dimerização , Endonucleases/química , Células HeLa , Humanos , Insulina/fisiologia , Cinética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Frações Subcelulares/enzimologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA