Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
MAbs ; 12(1): 1795492, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32744170

RESUMO

Given the increasing use of combination therapy with multiple monoclonal antibodies (mAbs), there is a clinical need for multiplexing assays. For the frequently co-administered anti-human epidermal growth factor receptor 2 (HER2) mAbs trastuzumab and pertuzumab, we developed a high-throughput and robust hybrid ligand-binding liquid chromatography-mass spectrometry (LC-MS)/MS quantitative assay. Nanomolar concentrations of trastuzumab and pertuzumab were determined in 10 µL serum samples after extraction by affinity purification through protein A beads, followed by on-bead reduction, alkylation, and trypsin digestion. After electrospray ionization, quantification was obtained by multiple reaction monitoring LC-MS/MS using SILuMab as an internal standard. The method was validated according to the current guidelines from the US Food and Drug Administration and the European Medicines Agency. Assay linearity was established in the ranges 0.250-250 µg/mL for trastuzumab and 0.500-500 µg/mL for pertuzumab. The method was accurate and selective for the simultaneous determination of trastuzumab and pertuzumab in clinical samples, thereby overcoming the limitation of ligand binding assays that cannot quantify mAbs targeting the same receptor. Furthermore, this method requires a small blood volume, which reduces blood collection time and stress for patients. The assay robustness was verified in a clinical trial where trastuzumab and pertuzumab concentrations were determined in 670 serum samples. As we used commercially available reagents and standards, the described generic bioanalytical strategy can easily be adapted to multiplex quantifications of other mAb combinations in non-clinical and clinical samples.


Assuntos
Anticorpos Monoclonais Humanizados , Espectrometria de Massas em Tandem , Trastuzumab , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacocinética , Feminino , Humanos , Masculino , Trastuzumab/administração & dosagem , Trastuzumab/farmacocinética
2.
PLoS One ; 10(5): e0128585, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26016997

RESUMO

Leukemic stem cells (LSCs) reside within bone marrow niches that maintain their relatively quiescent state and convey resistance to conventional treatment. Many of the microenvironmental signals converge on RAC GTPases. Although it has become clear that RAC proteins fulfill important roles in the hematopoietic compartment, little has been revealed about the downstream effectors and molecular mechanisms. We observed that in BCR-ABL-transduced human hematopoietic stem/progenitor cells (HSPCs) depletion of RAC2 but not RAC1 induced a marked and immediate decrease in proliferation, progenitor frequency, cobblestone formation and replating capacity, indicative for reduced self-renewal. Cell cycle analyses showed reduced cell cycle activity in RAC2-depleted BCR-ABL leukemic cobblestones coinciding with an increased apoptosis. Moreover, a decrease in mitochondrial membrane potential was observed upon RAC2 downregulation, paralleled by severe mitochondrial ultrastructural malformations as determined by automated electron microscopy. Proteome analysis revealed that RAC2 specifically interacted with a set of mitochondrial proteins including mitochondrial transport proteins SAM50 and Metaxin 1, and interactions were confirmed in independent co-immunoprecipitation studies. Downregulation of SAM50 also impaired the proliferation and replating capacity of BCR-ABL-expressing cells, again associated with a decreased mitochondrial membrane potential. Taken together, these data suggest an important role for RAC2 in maintaining mitochondrial integrity.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Mitocôndrias/genética , Doenças Mitocondriais/genética , Células-Tronco/metabolismo , Proteínas rac de Ligação ao GTP/genética , Apoptose/genética , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Ciclo Celular/genética , Linhagem Celular , Proliferação de Células/genética , Regulação para Baixo/genética , Proteínas de Fusão bcr-abl/genética , Células HEK293 , Células-Tronco Hematopoéticas/patologia , Humanos , Imunoprecipitação/métodos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Potencial da Membrana Mitocondrial/genética , Proteínas de Membrana/genética , Mitocôndrias/patologia , Doenças Mitocondriais/patologia , Proteínas de Transporte da Membrana Mitocondrial , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Proteínas Mitocondriais/genética , Proteínas/genética , Células-Tronco/patologia , Proteína RAC2 de Ligação ao GTP
3.
Blood ; 121(13): 2452-61, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23349393

RESUMO

The Polycomb group (PcG) protein BMI1 is a key factor in regulating hematopoietic stem cell (HSC) and leukemic stem cell self-renewal and functions in the context of the Polycomb repressive complex 1 (PRC1). In humans, each of the 5 subunits of PRC1 has paralog family members of which many reside in PRC1 complexes, likely in a mutually exclusive manner, pointing toward a previously unanticipated complexity of Polycomb-mediated silencing. We used an RNA interference screening approach to test the functionality of these paralogs in human hematopoiesis. Our data demonstrate a lack of redundancy between various paralog family members, suggestive of functional diversification between PcG proteins. By using an in vivo biotinylation tagging approach followed by liquid chromatography-tandem mass spectrometry to identify PcG interaction partners, we confirmed the existence of multiple specific PRC1 complexes. We find that CBX2 is a nonredundant CBX paralog vital for HSC and progenitor function that directly regulates the expression of the cyclin-dependent kinase inhibitor p21, independently of BMI1 that dominantly controls expression of the INK4A/ARF locus. Taken together, our data show that different PRC1 paralog family members have nonredundant and locus-specific gene regulatory activities that are essential for human hematopoiesis.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Inativação Gênica , Loci Gênicos/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Feminino , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica/fisiologia , Hematopoese/genética , Células-Tronco Hematopoéticas/fisiologia , Humanos , Recém-Nascido , Família Multigênica/genética , Família Multigênica/fisiologia , Gravidez , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Homologia de Sequência , Especificidade por Substrato/genética
4.
Mol Cell Proteomics ; 12(3): 626-37, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23233446

RESUMO

Interactions between hematopoietic stem cells and their niche are mediated by proteins within the plasma membrane (PM) and changes in these interactions might alter hematopoietic stem cell fate and ultimately result in acute myeloid leukemia (AML). Here, using nano-LC/MS/MS, we set out to analyze the PM profile of two leukemia patient samples. We identified 867 and 610 unique CD34(+) PM (-associated) proteins in these AML samples respectively, including previously described proteins such as CD47, CD44, CD135, CD96, and ITGA5, but also novel ones like CD82, CD97, CD99, PTH2R, ESAM, MET, and ITGA6. Further validation by flow cytometry and functional studies indicated that long-term self-renewing leukemic stem cells reside within the CD34(+)/ITGA6(+) fraction, at least in a subset of AML cases. Furthermore, we combined proteomics with transcriptomics approaches using a large panel of AML CD34(+) (n = 60) and normal bone marrow CD34(+) (n = 40) samples. Thus, we identified eight subgroups of AML patients based on their specific PM expression profile. GSEA analysis revealed that these eight subgroups are enriched for specific cellular processes.


Assuntos
Perfilação da Expressão Gênica/métodos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteômica/métodos , Doença Aguda , Antígenos CD34/genética , Antígenos CD34/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Membrana Celular/metabolismo , Cromatografia Líquida , Citometria de Fluxo , Regulação Leucêmica da Expressão Gênica , Humanos , Integrina alfa6/genética , Integrina alfa6/metabolismo , Nanotecnologia/métodos , Análise de Componente Principal , Proteoma/análise , Espectrometria de Massas em Tandem
5.
Mol Biosyst ; 8(3): 709-19, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22258412

RESUMO

In the past decade among the main developments in the field of bionanotechnology is the application of proteins in devices. Research focuses on the modification of enzyme systems by means of chemical and physical tools in order to achieve full control of their function and to employ them for specific tasks. Membrane protein channels are intriguing biological devices as they allow the recognition and passage of a variety of macromolecules through an otherwise impermeable lipid bilayer. Hence, membrane proteins can be used as sensory devices for detection or as molecular nanovalves to allow for the controlled release of molecules. Here, we discuss the structure and function of three different channel proteins that mediate the membrane passage of macromolecules using different mechanisms. These systems are described in a comparative manner and an overview is provided of the technological advances in employing these proteins in external (or human) controllable devices.


Assuntos
Proteínas de Membrana/química , Humanos , Canais Iônicos/química , Canais Iônicos/metabolismo , Proteínas de Membrana/metabolismo , Modelos Biológicos , Conformação Proteica
6.
Proc Natl Acad Sci U S A ; 108(19): 7775-80, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21518907

RESUMO

Protein translocation in Escherichia coli is mediated by the translocase that in its minimal form consists of the protein-conducting channel SecYEG, and the motor protein, SecA. SecYEG forms a narrow pore in the membrane that allows passage of unfolded proteins only. Molecular dynamics simulations suggest that the maximal width of the central pore of SecYEG is limited to . To access the functional size of the SecYEG pore, the precursor of outer membrane protein A was modified with rigid spherical tetraarylmethane derivatives of different diameters at a unique cysteine residue. SecYEG allowed the unrestricted passage of the precursor of outer membrane protein A conjugates carrying tetraarylmethanes with diameters up to , whereas a sized molecule blocked the translocation pore. Translocation of the protein-organic molecule hybrids was strictly proton motive force-dependent and occurred at a single pore. With an average diameter of an unfolded polypeptide chain of , the pore accommodates structures of at least , which is vastly larger than the predicted maximal width of a single pore by molecular dynamics simulations.


Assuntos
Proteínas de Escherichia coli/química , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Simulação de Dinâmica Molecular , Estrutura Molecular , Conformação Proteica , Precursores de Proteínas/química , Precursores de Proteínas/metabolismo , Transporte Proteico , Força Próton-Motriz , Canais de Translocação SEC , Proteínas SecA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...