Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(11): e0277937, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36409750

RESUMO

The importance of human cell-based in vitro tools to drug development that are robust, accurate, and predictive cannot be understated. There has been significant effort in recent years to develop such platforms, with increased interest in 3D models that can recapitulate key aspects of biology that 2D models might not be able to deliver. We describe the development of a 3D human cell-based in vitro assay for the investigation of nephrotoxicity, using RPTEC-TERT1 cells. These RPTEC-TERT1 proximal tubule organoids 'tubuloids' demonstrate marked differences in physiologically relevant morphology compared to 2D monolayer cells, increased sensitivity to nephrotoxins observable via secreted protein, and with a higher degree of similarity to native human kidney tissue. Finally, tubuloids incubated with nephrotoxins demonstrate altered Na+/K+-ATPase signal intensity, a potential avenue for a high-throughput, translatable nephrotoxicity assay.


Assuntos
Túbulos Renais Proximais , Organoides , Humanos , Linhagem Celular , Túbulos Renais Proximais/metabolismo , Túbulos Renais , Rim
3.
Commun Biol ; 4(1): 56, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420321

RESUMO

Overcoming tumor-mediated immunosuppression and enhancing cytotoxic T-cell activity within the tumor microenvironment are two central goals of immuno-oncology (IO) drug discovery initiatives. However, exploratory assays involving immune components are often plagued by low-throughput and poor clinical relevance. Here we present an innovative ultra-high-content assay platform for interrogating T-cell-mediated killing of 3D multicellular tumor spheroids. Employing this assay platform in a chemical genomics screen of 1800 annotated compounds enabled identification of small molecule perturbagens capable of enhancing cytotoxic CD8+ T-cell activity in an antigen-dependent manner. Specifically, cyclin-dependent kinase (CDK) and bromodomain (BRD) protein inhibitors were shown to significantly augment anti-tumor T-cell function by increasing cytolytic granule and type II interferon secretion in T-cells in addition to upregulating major histocompatibility complex (MHC) expression and antigen presentation in tumor cells. The described biotechnology screening platform yields multi-parametric, clinically-relevant data and can be employed kinetically for the discovery of first-in-class IO therapeutic agents.


Assuntos
Antígenos de Neoplasias/imunologia , Bioensaio/métodos , Descoberta de Drogas/métodos , Neoplasias/imunologia , Linfócitos T/fisiologia , Apresentação de Antígeno , Biomimética , Técnicas de Cocultura , Esferoides Celulares , Células Tumorais Cultivadas
4.
BMC Bioinformatics ; 21(1): 280, 2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32615917

RESUMO

BACKGROUND: Image-based high throughput (HT) screening provides a rich source of information on dynamic cellular response to external perturbations. The large quantity of data generated necessitates computer-aided quality control (QC) methodologies to flag imaging and staining artifacts. Existing image- or patch-level QC methods require separate thresholds to be simultaneously tuned for each image quality metric used, and also struggle to distinguish between artifacts and valid cellular phenotypes. As a result, extensive time and effort must be spent on per-assay QC feature thresholding, and valid images and phenotypes may be discarded while image- and cell-level artifacts go undetected. RESULTS: We present a novel cell-level QC workflow built on machine learning approaches for classifying artifacts from HT image data. First, a phenotype sampler based on unlabeled clustering collects a comprehensive subset of cellular phenotypes, requiring only the inspection of a handful of images per phenotype for validity. A set of one-class support vector machines are then trained on each biologically valid image phenotype, and used to classify individual objects in each image as valid cells or artifacts. We apply this workflow to two real-world large-scale HT image datasets and observe that the ratio of artifact to total object area (ARcell) provides a single robust assessment of image quality regardless of the underlying causes of quality issues. Gating on this single intuitive metric, partially contaminated images can be salvaged and highly contaminated images can be excluded before image-level phenotype summary, enabling a more reliable characterization of cellular response dynamics. CONCLUSIONS: Our cell-level QC workflow enables identification of artificial cells created not only by staining or imaging artifacts but also by the limitations of image segmentation algorithms. The single readout ARcell that summaries the ratio of artifacts contained in each image can be used to reliably rank images by quality and more accurately determine QC cutoff thresholds. Machine learning-based cellular phenotype clustering and sampling reduces the amount of manual work required for training example collection. Our QC workflow automatically handles assay-specific phenotypic variations and generalizes to different HT image assays.


Assuntos
Células/metabolismo , Processamento de Imagem Assistida por Computador , Fluxo de Trabalho , Algoritmos , Animais , Artefatos , Linhagem Celular , Humanos , Aprendizado de Máquina , Fenótipo , Controle de Qualidade , Máquina de Vetores de Suporte
5.
Future Med Chem ; 7(4): 513-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25875876

RESUMO

Increasingly, organotypic cellular platforms are being recognized as useful tools in drug discovery. This review offers an industry-centric perspective on the benefits of emerging complex cell models over conventional 2D systems, as well as the challenges and opportunities for incorporating these multidimensional platforms into high-density formats. We particularly highlight the need for novel chemical sensors to noninvasively quantitate 3D structures in real time, and we contend that the use of more focused chemical and genomics libraries will enable screening of complex cell models derived from primary and induced pluripotent stem cells. Finally, we offer outlooks on several emerging technologies that show great potential for future integration of complex cell systems into contemporary drug screening.


Assuntos
Células/ultraestrutura , Modelos Biológicos , Animais , Desenho de Fármacos , Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Humanos , Células-Tronco Pluripotentes Induzidas , Fenótipo
6.
Artigo em Inglês | MEDLINE | ID: mdl-24753373

RESUMO

Time-dependent extracellular manipulations of human pluripotent stem cells can yield as much as 90% pure populations of cardiomyocytes. While the extracellular control of differentiation generally entails dynamic regulation of well-known pathways such as Wnt, BMP, and Nodal signaling, the underlying genetic networks are far more complex and are poorly understood. Notably, the identification of these networks holds promise for understanding heart disease and regeneration. The availability of genome-wide experimentation, such as RNA and DNA sequencing, as well as high throughput surveying with small molecule and small interfering RNA libraries, now enables us to map the genetic interactions underlying cardiac differentiation on a global scale. Initial studies demonstrate the complexity of the genetic regulation of cardiac differentiation, exposing unanticipated novel mechanisms. However, the large datasets generated tend to be overwhelming and systematic approaches are needed to process the vast amount of data to improve our mechanistic understanding of the complex biology. Systems biology methods spur high hopes for parsing vast amounts of data into genetic interaction models that can be verified experimentally and ultimately yield functional networks that expose the genetic connections underlying biological processes.


Assuntos
Diferenciação Celular , Coração/fisiologia , Células-Tronco Pluripotentes/citologia , Proteômica/métodos , Animais , Regulação da Expressão Gênica , Genoma , Genômica/métodos , Humanos , Camundongos , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , RNA Interferente Pequeno/metabolismo , Análise de Sequência de DNA , Análise de Sequência de RNA , Biologia de Sistemas
7.
Methods Mol Biol ; 1029: 163-89, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23756949

RESUMO

Despite advances in understanding pluripotency through traditional cell biology and gene expression profiling, the signaling networks responsible for maintenance of pluripotency and lineage-specific differentiation are poorly defined. To aid in an improved understanding of these networks at the systems level, we present procedures for the combined analysis of the total proteome and total phosphoproteome (termed (phospho)proteome) from human embryonic stem cells (hESCs), human induced pluripotent stem cells (hiPSCs), and their differentiated derivatives. Because there has been considerable heterogeneity in the literature on the culture of pluripotent cells, we first briefly describe our feeder-free cell culture protocol. The focus, however, is on procedures necessary to generate large-scale (phospho)proteomic data from the cells. Human cells are described here, but the (phospho)proteomic procedures are broadly applicable. Detailed procedures are given for lysis of the cells, protein sample preparation and digestion, multidimensional liquid chromatography, analysis by tandem mass spectrometry, and database searches for peptide/protein identification (ID). We summarize additional data analysis procedures, the subject of ongoing efforts.


Assuntos
Fosfoproteínas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteômica/métodos , Alquilação , Sulfato de Amônio , Animais , Fracionamento Celular , Células Cultivadas , Precipitação Química , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Cromatografia por Troca Iônica , Cromatografia de Fase Reversa , Bases de Dados de Proteínas , Humanos , Espectrometria de Massas , Camundongos , Fosfopeptídeos/isolamento & purificação , Estatística como Assunto , Titânio
8.
J Cardiovasc Transl Res ; 4(3): 340-50, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21424858

RESUMO

Heart failure is one of the major causes of death in the Western world because cardiac muscle loss is largely irreversible and can lead to a relentless decline in cardiac function. Novel therapies are needed since the only therapy to effectively replace lost myocytes today is transplantation of the entire heart. The advent of embryonic and induced pluripotent stem cell (ESC/iPSC) technologies offers the unprecedented possibility of devising cell replacement therapies for numerous degenerative disorders. Not only are ESCs and iPSCs a plausible source of cardiomyocytes in vitro for transplantation, they are also useful tools to elucidate the biology of stem cells that reside in the adult heart and define signaling molecules that might enhance the limited regenerative capability of the adult human heart. Here, we review the extracellular factors that control stem cell cardiomyogenesis and describe new approaches that combine embryology with stem cell biology to discover drug-like small molecules that stimulate cardiogenesis and potentially contribute to the development of pharmaceutical strategies for heart muscle regeneration.


Assuntos
Fármacos Cardiovasculares/uso terapêutico , Células-Tronco Embrionárias/efeitos dos fármacos , Insuficiência Cardíaca/tratamento farmacológico , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Animais , Fármacos Cardiovasculares/química , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Desenho de Fármacos , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/cirurgia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/transplante , Transdução de Sinais/efeitos dos fármacos , Transplante de Células-Tronco , Resultado do Tratamento
9.
J Neurosci ; 30(27): 9027-37, 2010 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-20610737

RESUMO

Phosphodiesterase 10A (PDE10A) is a striatum-enriched, dual-specific cyclic nucleotide phosphodiesterase that has gained considerable attention as a potential therapeutic target for psychiatric disorders such as schizophrenia. As such, a PDE10A-selective inhibitor compound, MP-10, has recently entered clinical testing. Since little is known about the cellular regulation of PDE10A, we sought to elucidate the mechanisms that govern its subcellular localization in striatal medium spiny neurons. Previous reports suggest that PDE10A is primarily membrane bound and is transported throughout medium spiny neuron axons and dendrites. Moreover, it has been shown in PC12 cells that the localization of the major splice form, PDE10A2, may be regulated by protein kinase A phosphorylation at threonine 16 (Thr-16). Using an antibody that specifically recognizes phosphorylated Thr-16 (pThr-16) of PDE10A2, we provide evidence that phosphorylation at Thr-16 is critical for the regulation of PDE10A subcellular localization in vivo. Furthermore, we demonstrate in primary mouse striatal neuron cultures that PDE10A membrane association and transport throughout dendritic processes requires palmitoylation of cysteine 11 (Cys-11) of PDE10A2, likely by the palmitoyl acyltransferases DHHC-7 and -19. Finally, we show that Thr-16 phosphorylation regulates PDE10A trafficking and localization by preventing palmitoylation of Cys-11 rather than by interfering with palmitate-lipid interactions. These data support a model whereby PDE10A trafficking and localization can be regulated in response to local fluctuations in cAMP levels. Given this, we propose that excessive striatal dopamine release, as occurs in schizophrenia, might exert differential effects on the regulation of PDE10A localization in the two striatal output pathways.


Assuntos
Lipoilação/fisiologia , Neurônios/fisiologia , Diester Fosfórico Hidrolases/metabolismo , Análise de Variância , Animais , Células Cultivadas , Corpo Estriado/citologia , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Cisteína/genética , Cisteína/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Embrião de Mamíferos , Humanos , Hipoglicemiantes/farmacologia , Imunoprecipitação/métodos , Lipoilação/efeitos dos fármacos , Lipoilação/genética , Camundongos , Microscopia Confocal/métodos , Mutagênese Sítio-Dirigida/métodos , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Oligopeptídeos/farmacologia , Palmitatos/farmacologia , Diester Fosfórico Hidrolases/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fosforilação/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Ratos , Treonina/metabolismo , Transfecção/métodos
10.
Cell Signal ; 18(6): 795-806, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16126371

RESUMO

Shc adapter proteins are thought to regulate cellular proliferation, differentiation and apoptosis by activating the SOS-Grb2-RAS-MAPK signaling cascade. Using the small hairpin RNA (shRNA) technique, we found that decreasing ShcA mRNA reduced the proliferative ability of HEK293 mammalian culture cells. We then recapitulated phosphorylation-dependent Shc-Grb2 complex formation in Saccharomyces cerevisiae. Immunoprecipitation followed by Western analysis demonstrated that activated TrkB, composed of the intracellular domain of TrkB fused to glutathione S-transferase (GST-TrkB(ICD)), promoted the association of ShcC and Grb2 in yeast. The Ras-recruitment system (RRS), in which a myristoylated (Myr)-bait and son of sevenless (hSOS)-prey are brought together to complement the defective Ras-cAMP pathway in a thermosensitive cdc25H mutant yeast strain, was used to validate a phenotypic assay. Yeast cells transformed with both Myr-ShcC and hSOS-Grb2 (referred to as scheme 1) or Myr-Grb2 and hSOS-ShcC (scheme 2) did not grow at non-permissive temperature; the additional transformation of GST-TrkB(ICD) enabled growth. GST-TrkB(ICD) also enabled growth with hSOS-Grb2 and either Myr-ShcA or Myr-SHP2. Mutational analysis of TrkB showed that its kinase activity was essential for complementation, while its docking site for Shc proteins was not. Mutational analysis of ShcC showed that the PTB and SH2 domains were not essential for complementation but phosphorylation at Y304 in the CH1 domain was. Phosphorylation at Y304 could not be substituted by an acidic amino acid. The RRS provides a genetic system to probe Shc proteins and potentially identify member specific protein partners and pharmacological reagents.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proliferação de Células , Proteína Adaptadora GRB2/metabolismo , Neuropeptídeos/metabolismo , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Divisão Celular/fisiologia , Linhagem Celular , Células Cultivadas , Proteínas Fúngicas/metabolismo , Proteína Adaptadora GRB2/genética , Técnicas de Transferência de Genes , Teste de Complementação Genética , Humanos , Neuropeptídeos/genética , Fosforilação , Ligação Proteica , Receptor trkB/metabolismo , Saccharomyces cerevisiae/citologia , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais/genética , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Proteína 3 de Transformação que Contém Domínio 2 de Homologia de Src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA