Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Neurooncol Adv ; 4(1): vdac079, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733514

RESUMEN

Background: Diffuse midline gliomas (DMG) are highly malignant incurable pediatric brain tumors. A lack of effective treatment options highlights the need to investigate novel therapeutic strategies. This includes the use of immunotherapy, which has shown promise in other hard-to-treat tumors. To facilitate preclinical immunotherapeutic research, immunocompetent mouse models that accurately reflect the unique genetic, anatomical, and histological features of DMG patients are warranted. Methods: We established cell cultures from primary DMG mouse models (C57BL/6) that were generated by brainstem targeted intra-uterine electroporation (IUE). We subsequently created allograft DMG mouse models by orthotopically implanting these tumor cells into syngeneic mice. Immunohistochemistry and -fluorescence, mass cytometry, and cell-viability assays were then used to verify that these murine tumors recapitulated human DMG. Results: We generated three genetically distinct allograft models representing histone 3 wildtype (H3WT) and K27M-mutant DMG (H3.3K27M and H3.1K27M). These allograft models recapitulated the histopathologic phenotype of their human counterparts, including their diffuse infiltrative growth and expression of DMG-associated antigens. These murine pontine tumors also exhibited an immune microenvironment similar to human DMG, characterized by considerable myeloid cell infiltration and a paucity of T-lymphocytes and NK cells. Finally, we show that these murine DMG cells display similar sensitivity to histone deacetylase (HDAC) inhibition as patient-derived DMG cells. Conclusions: We created and validated an accessible method to generate immunocompetent allograft models reflecting different subtypes of DMG. These models adequately recapitulated the histopathology, immune microenvironment, and therapeutic response of human DMG, providing useful tools for future preclinical studies.

2.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35210365

RESUMEN

The antigen specificity and long serum half-life of monoclonal antibodies have made them a critical part of modern therapeutics. These properties have been coopted in a number of synthetic formats, such as antibody-drug conjugates, bispecific antibodies, or Fc-fusion proteins to generate novel biologic drug modalities. Historically, these new therapies have been generated by covalently linking multiple molecular moieties through chemical or genetic methods. This irreversible fusion of different components means that the function of the molecule is static, as determined by the structure. Here, we report the development of a technology for switchable assembly of functional antibody complexes using chemically induced dimerization domains. This approach enables control of the antibody's intended function in vivo by modulating the dose of a small molecule. We demonstrate this switchable assembly across three therapeutically relevant functionalities in vivo, including localization of a radionuclide-conjugated antibody to an antigen-positive tumor, extension of a cytokine's half-life, and activation of bispecific, T cell-engaging antibodies.


Asunto(s)
Anticuerpos/metabolismo , Inmunoconjugados/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Especificidad de Anticuerpos , Humanos
3.
J Immunother Cancer ; 9(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34083417

RESUMEN

BACKGROUND: Glioblastoma (GBM) is refractory to immune checkpoint inhibitor (ICI) therapy. We sought to determine to what extent this immune evasion is due to intrinsic properties of the tumor cells versus the specialized immune context of the brain, and if it can be reversed. METHODS: We used CyTOF mass cytometry to compare the tumor immune microenvironments (TIME) of human tumors that are generally ICI-refractory (GBM and sarcoma) or ICI-responsive (renal cell carcinoma), as well as mouse models of GBM that are ICI-responsive (GL261) or ICI-refractory (SB28). We further compared SB28 tumors grown intracerebrally versus subcutaneously to determine how tumor site affects TIME and responsiveness to dual CTLA-4/PD-1 blockade. Informed by these data, we explored rational immunotherapeutic combinations. RESULTS: ICI-sensitivity in human and mouse tumors was associated with increased T cells and dendritic cells (DCs), and fewer myeloid cells, in particular PD-L1+ tumor-associated macrophages. The SB28 mouse model of GBM responded to ICI when grown subcutaneously but not intracerebrally, providing a system to explore mechanisms underlying ICI resistance in GBM. The response to ICI in the subcutaneous SB28 model required CD4 T cells and NK cells, but not CD8 T cells. Recombinant FLT3L expanded DCs, improved antigen-specific T cell priming, and prolonged survival of mice with intracerebral SB28 tumors, but at the cost of increased Tregs. Targeting PD-L1 also prolonged survival, especially when combined with stereotactic radiation. CONCLUSIONS: Our data suggest that a major obstacle for effective immunotherapy of GBM is poor antigen presentation in the brain, rather than intrinsic immunosuppressive properties of GBM tumor cells. Deep immune profiling identified DCs and PD-L1+ tumor-associated macrophages as promising targetable cell populations, which was confirmed using therapeutic interventions in vivo.


Asunto(s)
Neoplasias Encefálicas/terapia , Antígeno CTLA-4/metabolismo , Glioblastoma/terapia , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Proteínas de la Membrana/administración & dosificación , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Neoplasias Encefálicas/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Línea Celular Tumoral , Glioblastoma/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Proteínas de la Membrana/farmacología , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T Reguladores/metabolismo , Escape del Tumor/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Cancer Ther ; 18(9): 1565-1576, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31270152

RESUMEN

Amplification of the epidermal growth factor receptor gene (EGFR) represents one of the most commonly observed genetic lesions in glioblastoma (GBM); however, therapies targeting this signaling pathway have failed clinically. Here, using human tumors, primary patient-derived xenografts (PDX), and a murine model for GBM, we demonstrate that EGFR inhibition leads to increased invasion of tumor cells. Further, EGFR inhibitor-treated GBM demonstrates altered oxidative stress, with increased lipid peroxidation, and generation of toxic lipid peroxidation products. A tumor cell subpopulation with elevated aldehyde dehydrogenase (ALDH) levels was determined to comprise a significant proportion of the invasive cells observed in EGFR inhibitor-treated GBM. Our analysis of the ALDH1A1 protein in newly diagnosed GBM revealed detectable ALDH1A1 expression in 69% (35/51) of the cases, but in relatively low percentages of tumor cells. Analysis of paired human GBM before and after EGFR inhibitor therapy showed an increase in ALDH1A1 expression in EGFR-amplified tumors (P < 0.05, n = 13 tumor pairs), and in murine GBM ALDH1A1-high clones were more resistant to EGFR inhibition than ALDH1A1-low clones. Our data identify ALDH levels as a biomarker of GBM cells with high invasive potential, altered oxidative stress, and resistance to EGFR inhibition, and reveal a therapeutic target whose inhibition should limit GBM invasion.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Familia de Aldehído Deshidrogenasa 1/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dasatinib/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/farmacología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Estrés Oxidativo/efectos de los fármacos , Retinal-Deshidrogenasa/metabolismo
5.
Methods Mol Biol ; 1989: 55-81, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31077099

RESUMEN

The advent of mass cytometry (CyTOF®) has permitted simultaneous detection of more than 40 antibody parameters at the single-cell level, although a limited number of metal-labeled antibodies are commercially available. Here we present optimized and scalable protocols for conjugation of lanthanide as well as bismuth ions to immunoglobulin (Ig) using a maleimide-functionalized chelating polymer and for characterization of the conjugate. The maleimide functional group is reactive with cysteine sulfhydryl groups generated through partial reduction of the Ig Fc region. Incubation of Ig with polymer pre-loaded with lanthanide ions produces metal-labeled Ig without disrupting antigen specificity. Antibody recovery rates can be determined by UV spectrophotometry and frequently exceeds 60%. Each custom-conjugated antibody is validated using positive and negative cellular control populations and is titrated for optimal staining at concentrations ranging from 0.1 to 10 µg/mL. The preparation of metal-labeled antibodies can be completed in 4.5 h, and titration requires an additional 3-5 h.


Asunto(s)
Anticuerpos/química , Células/citología , Citometría de Flujo/métodos , Inmunofenotipificación/métodos , Isótopos/análisis , Espectrometría de Masas/métodos , Análisis de la Célula Individual/métodos , Quelantes/química , Humanos , Inmunoglobulinas/inmunología , Metales/química , Polímeros/química , Compuestos de Sulfhidrilo/química
6.
Blood ; 133(10): 1119-1129, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30591526

RESUMEN

Follicular lymphoma (FL) is a low-grade B-cell malignancy that transforms into a highly aggressive and lethal disease at a rate of 2% per year. Perfect isolation of the malignant B-cell population from a surgical biopsy is a significant challenge, masking important FL biology, such as immune checkpoint coexpression patterns. To resolve the underlying transcriptional networks of follicular B-cell lymphomas, we analyzed the transcriptomes of 34 188 cells derived from 6 primary FL tumors. For each tumor, we identified normal immune subpopulations and malignant B cells, based on gene expression. We used multicolor flow cytometry analysis of the same tumors to confirm our assignments of cellular lineages and validate our predictions of expressed proteins. Comparison of gene expression between matched malignant and normal B cells from the same patient revealed tumor-specific features. Malignant B cells exhibited restricted immunoglobulin (Ig) light chain expression (either Igκ or Igλ), as well the expected upregulation of the BCL2 gene, but also downregulation of the FCER2, CD52, and major histocompatibility complex class II genes. By analyzing thousands of individual cells per patient tumor, we identified the mosaic of malignant B-cell subclones that coexist within a FL and examined the characteristics of tumor-infiltrating T cells. We identified genes coexpressed with immune checkpoint molecules, such as CEBPA and B2M in regulatory T (Treg) cells, providing a better understanding of the gene networks involved in immune regulation. In summary, parallel measurement of single-cell expression in thousands of tumor cells and tumor-infiltrating lymphocytes can be used to obtain a systems-level view of the tumor microenvironment and identify new avenues for therapeutic development.


Asunto(s)
Linfoma de Células B/genética , Linfoma Folicular/genética , Linfocitos T Reguladores/citología , Biopsia , Proteínas Potenciadoras de Unión a CCAAT/genética , Linfocitos T CD4-Positivos/citología , Antígeno CD52/genética , Linaje de la Célula , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/citología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Sistema Inmunológico , Inmunoglobulina G , Lectinas Tipo C/genética , Leucocitos Mononucleares/citología , Linfoma de Células B/sangre , Linfoma Folicular/sangre , Tonsila Palatina/metabolismo , Receptores de IgE/genética , Análisis de Secuencia de ARN , Transcriptoma , Microambiente Tumoral , Microglobulina beta-2/genética
7.
Sci Rep ; 8(1): 10770, 2018 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018331

RESUMEN

Single-cell barcoding enables the combined processing and acquisition of multiple individual samples as one. This maximizes assay efficiency and eliminates technical variability in both sample preparation and analysis. Remaining challenges are the barcoding of live, unprocessed cells to increase downstream assay performance combined with the flexibility of the approach towards a broad range of cell types. To that end, we developed a novel antibody-based platform that allows the robust barcoding of live human cells for mass cytometry (CyTOF). By targeting both the MHC class I complex (beta-2-microglobulin) and a broadly expressed sodium-potassium ATPase-subunit (CD298) with platinum-conjugated antibodies, human immune cells, stem cells as well as tumor cells could be multiplexed in the same single-cell assay. In addition, we present a novel palladium-based covalent viability reagent compatible with this barcoding strategy. Altogether, this platform enables mass cytometry-based, live-cell barcoding across a multitude of human sample types and provides a scheme for multiplexed barcoding of human single-cell assays in general.


Asunto(s)
Análisis de la Célula Individual/métodos , Línea Celular , Células Cultivadas , Cisplatino , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Células Jurkat
8.
Bioinformatics ; 34(23): 4131-4133, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29850785

RESUMEN

Motivation: High-parameter single-cell technologies can reveal novel cell populations of interest, but studying or validating these populations using lower-parameter methods remains challenging. Results: Here, we present GateFinder, an algorithm that enriches high-dimensional cell types with simple, stepwise polygon gates requiring only two markers at a time. A series of case studies of complex cell types illustrates how simplified enrichment strategies can enable more efficient assays, reveal novel biomarkers and clarify underlying biology. Availability and implementation: The GateFinder algorithm is implemented as a free and open-source package for BioConductor: https://nalab.stanford.edu/gatefinder. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Biomarcadores/análisis , Citometría de Flujo , Programas Informáticos
9.
Nat Med ; 24(4): 474-483, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29505032

RESUMEN

Insight into the cancer cell populations that are responsible for relapsed disease is needed to improve outcomes. Here we report a single-cell-based study of B cell precursor acute lymphoblastic leukemia at diagnosis that reveals hidden developmentally dependent cell signaling states that are uniquely associated with relapse. By using mass cytometry we simultaneously quantified 35 proteins involved in B cell development in 60 primary diagnostic samples. Each leukemia cell was then matched to its nearest healthy B cell population by a developmental classifier that operated at the single-cell level. Machine learning identified six features of expanded leukemic populations that were sufficient to predict patient relapse at diagnosis. These features implicated the pro-BII subpopulation of B cells with activated mTOR signaling, and the pre-BI subpopulation of B cells with activated and unresponsive pre-B cell receptor signaling, to be associated with relapse. This model, termed 'developmentally dependent predictor of relapse' (DDPR), significantly improves currently established risk stratification methods. DDPR features exist at diagnosis and persist at relapse. By leveraging a data-driven approach, we demonstrate the predictive value of single-cell 'omics' for patient stratification in a translational setting and provide a framework for its application to human cancer.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras B/clasificación , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Análisis de la Célula Individual , Adulto , Linfocitos B/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Recurrencia , Medición de Riesgo , Transducción de Señal , Análisis de Supervivencia , Serina-Treonina Quinasas TOR/metabolismo , Adulto Joven
10.
Cancer Cell ; 31(3): 424-435, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-28292440

RESUMEN

Although signaling from phosphatidylinositol 3-kinase (PI3K) and AKT to mechanistic target of rapamycin (mTOR) is prominently dysregulated in high-grade glial brain tumors, blockade of PI3K or AKT minimally affects downstream mTOR activity in glioma. Allosteric mTOR inhibitors, such as rapamycin, incompletely block mTORC1 compared with mTOR kinase inhibitors (TORKi). Here, we compared RapaLink-1, a TORKi linked to rapamycin, with earlier-generation mTOR inhibitors. Compared with rapamycin and Rapalink-1, TORKi showed poor durability. RapaLink-1 associated with FKBP12, an abundant mTOR-interacting protein, enabling accumulation of RapaLink-1. RapaLink-1 showed better efficacy than rapamycin or TORKi, potently blocking cancer-derived, activating mutants of mTOR. Our study re-establishes mTOR as a central target in glioma and traces the failure of existing drugs to incomplete/nondurable inhibition of mTORC1.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Complejos Multiproteicos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Femenino , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos BALB C , Sirolimus/uso terapéutico , Proteína 1A de Unión a Tacrolimus/fisiología
11.
Neuro Oncol ; 19(2): 153-161, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27282398

RESUMEN

High-grade gliomas in children are different from those that arise in adults. Recent collaborative molecular analyses of these rare cancers have revealed previously unappreciated connections among chromatin regulation, developmental signaling, and tumorigenesis. As we begin to unravel the unique developmental origins and distinct biological drivers of this heterogeneous group of tumors, clinical trials need to keep pace. It is important to avoid therapeutic strategies developed purely using data obtained from studies on adult glioblastoma. This approach has resulted in repetitive trials and ineffective treatments being applied to these children, with limited improvement in clinical outcome. The authors of this perspective, comprising biology and clinical expertise in the disease, recently convened to discuss the most effective ways to translate the emerging molecular insights into patient benefit. This article reviews our current understanding of pediatric high-grade glioma and suggests approaches for innovative clinical management.


Asunto(s)
Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Glioma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Niño , Glioma/genética , Glioma/metabolismo , Humanos , Clasificación del Tumor , Pronóstico
12.
Cell ; 162(1): 184-97, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26095251

RESUMEN

Acute myeloid leukemia (AML) manifests as phenotypically and functionally diverse cells, often within the same patient. Intratumor phenotypic and functional heterogeneity have been linked primarily by physical sorting experiments, which assume that functionally distinct subpopulations can be prospectively isolated by surface phenotypes. This assumption has proven problematic, and we therefore developed a data-driven approach. Using mass cytometry, we profiled surface and intracellular signaling proteins simultaneously in millions of healthy and leukemic cells. We developed PhenoGraph, which algorithmically defines phenotypes in high-dimensional single-cell data. PhenoGraph revealed that the surface phenotypes of leukemic blasts do not necessarily reflect their intracellular state. Using hematopoietic progenitors, we defined a signaling-based measure of cellular phenotype, which led to isolation of a gene expression signature that was predictive of survival in independent cohorts. This study presents new methods for large-scale analysis of single-cell heterogeneity and demonstrates their utility, yielding insights into AML pathophysiology.


Asunto(s)
Biología Computacional/métodos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/fisiopatología , Análisis de la Célula Individual/métodos , Médula Ósea/patología , Niño , Estudios de Cohortes , Heterogeneidad Genética , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Transcriptoma
13.
Cancer Cell ; 26(3): 414-427, 2014 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-25175806

RESUMEN

MYC proteins are major drivers of cancer yet are considered undruggable because their DNA binding domains are composed of two extended alpha helices with no apparent surfaces for small-molecule binding. Proteolytic degradation of MYCN protein is regulated in part by a kinase-independent function of Aurora A. We describe a class of inhibitors that disrupts the native conformation of Aurora A and drives the degradation of MYCN protein across MYCN-driven cancers. Comparison of cocrystal structures with structure-activity relationships across multiple inhibitors and chemotypes, coupled with mechanistic studies and biochemical assays, delineates an Aurora A conformation-specific effect on proteolytic degradation of MYCN, rather than simple nanomolar-level inhibition of Aurora A kinase activity.


Asunto(s)
Antineoplásicos/farmacología , Aurora Quinasa A/química , Neuroblastoma/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Compuestos de Fenilurea/farmacología , Pirimidinas/farmacología , Regulación Alostérica , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Área Bajo la Curva , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Modelos Moleculares , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/patología , Proteínas Nucleares/química , Proteínas Oncogénicas/química , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacocinética , Fosforilación , Procesamiento Proteico-Postraduccional , Estructura Secundaria de Proteína , Proteolisis , Pirimidinas/química , Pirimidinas/farmacocinética , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Relación Estructura-Actividad , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell ; 157(3): 714-25, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24766814

RESUMEN

Tissue regeneration is an orchestrated progression of cells from an immature state to a mature one, conventionally represented as distinctive cell subsets. A continuum of transitional cell states exists between these discrete stages. We combine the depth of single-cell mass cytometry and an algorithm developed to leverage this continuum by aligning single cells of a given lineage onto a unified trajectory that accurately predicts the developmental path de novo. Applied to human B cell lymphopoiesis, the algorithm (termed Wanderlust) constructed trajectories spanning from hematopoietic stem cells through to naive B cells. This trajectory revealed nascent fractions of B cell progenitors and aligned them with developmentally cued regulatory signaling including IL-7/STAT5 and cellular events such as immunoglobulin rearrangement, highlighting checkpoints across which regulatory signals are rewired paralleling changes in cellular state. This study provides a comprehensive analysis of human B lymphopoiesis, laying a foundation to apply this approach to other tissues and "corrupted" developmental processes including cancer.


Asunto(s)
Algoritmos , Linfocitos B/citología , Linfopoyesis , Humanos , Interleucina-7/metabolismo , Células Precursoras de Linfocitos B/citología , Factor de Transcripción STAT5/metabolismo , Recombinación V(D)J
15.
Nat Biotechnol ; 31(6): 545-52, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23685480

RESUMEN

New high-dimensional, single-cell technologies offer unprecedented resolution in the analysis of heterogeneous tissues. However, because these technologies can measure dozens of parameters simultaneously in individual cells, data interpretation can be challenging. Here we present viSNE, a tool that allows one to map high-dimensional cytometry data onto two dimensions, yet conserve the high-dimensional structure of the data. viSNE plots individual cells in a visual similar to a scatter plot, while using all pairwise distances in high dimension to determine each cell's location in the plot. We integrated mass cytometry with viSNE to map healthy and cancerous bone marrow samples. Healthy bone marrow automatically maps into a consistent shape, whereas leukemia samples map into malformed shapes that are distinct from healthy bone marrow and from each other. We also use viSNE and mass cytometry to compare leukemia diagnosis and relapse samples, and to identify a rare leukemia population reminiscent of minimal residual disease. viSNE can be applied to any multi-dimensional single-cell technology.


Asunto(s)
Neoplasias de la Médula Ósea/patología , Citometría de Imagen , Inmunofenotipificación , Leucemia/patología , Análisis de la Célula Individual/métodos , Biomarcadores de Tumor/metabolismo , Neoplasias de la Médula Ósea/diagnóstico , Linaje de la Célula , Humanos , Leucemia/diagnóstico , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/patología , Recurrencia
16.
Cytometry A ; 83(5): 483-94, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23512433

RESUMEN

Mass cytometry uses atomic mass spectrometry combined with isotopically pure reporter elements to currently measure as many as 40 parameters per single cell. As with any quantitative technology, there is a fundamental need for quality assurance and normalization protocols. In the case of mass cytometry, the signal variation over time due to changes in instrument performance combined with intervals between scheduled maintenance must be accounted for and then normalized. Here, samples were mixed with polystyrene beads embedded with metal lanthanides, allowing monitoring of mass cytometry instrument performance over multiple days of data acquisition. The protocol described here includes simultaneous measurements of beads and cells on the mass cytometer, subsequent extraction of the bead-based signature, and the application of an algorithm enabling correction of both short- and long-term signal fluctuations. The variation in the intensity of the beads that remains after normalization may also be used to determine data quality. Application of the algorithm to a one-month longitudinal analysis of a human peripheral blood sample reduced the range of median signal fluctuation from 4.9-fold to 1.3-fold.


Asunto(s)
Citometría de Flujo/métodos , Elementos de la Serie de los Lantanoides , Leucocitos Mononucleares/citología , Espectrometría de Masas/métodos , Microesferas , Poliestirenos , Algoritmos , Citometría de Flujo/instrumentación , Humanos , Espectrometría de Masas/instrumentación , Ensayo de Materiales/métodos , Control de Calidad , Valores de Referencia , Programas Informáticos
17.
Nat Biotechnol ; 30(9): 858-67, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22902532

RESUMEN

Mass cytometry facilitates high-dimensional, quantitative analysis of the effects of bioactive molecules on human samples at single-cell resolution, but instruments process only one sample at a time. Here we describe mass-tag cellular barcoding (MCB), which increases mass cytometry throughput by using n metal ion tags to multiplex up to 2n samples. We used seven tags to multiplex an entire 96-well plate, and applied MCB to characterize human peripheral blood mononuclear cell (PBMC) signaling dynamics and cell-to-cell communication, signaling variability between PBMCs from eight human donors, and the effects of 27 inhibitors on this system. For each inhibitor, we measured 14 phosphorylation sites in 14 PBMC types at 96 conditions, resulting in 18,816 quantified phosphorylation levels from each multiplexed sample. This high-dimensional, systems-level inquiry allowed analysis across cell-type and signaling space, reclassified inhibitors and revealed off-target effects. High-content, high-throughput screening with MCB should be useful for drug discovery, preclinical testing and mechanistic investigation of human disease.


Asunto(s)
Citometría de Flujo/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Leucocitos Mononucleares/citología , Biología de Sistemas/métodos , Quelantes , Compuestos Heterocíclicos con 1 Anillo , Humanos , Células K562 , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Fosforilación , Análisis de Componente Principal , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal
18.
Bioinformatics ; 28(18): 2400-1, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22782546

RESUMEN

MOTIVATION: Recent advances in flow cytometry enable simultaneous single-cell measurement of 30+ surface and intracellular proteins. CytoSPADE is a high-performance implementation of an interface for the Spanning-tree Progression Analysis of Density-normalized Events algorithm for tree-based analysis and visualization of this high-dimensional cytometry data. AVAILABILITY: Source code and binaries are freely available at http://cytospade.org and via Bioconductor version 2.10 onwards for Linux, OSX and Windows. CytoSPADE is implemented in R, C++ and Java. CONTACT: michael.linderman@mssm.edu SUPPLEMENTARY INFORMATION: Additional documentation available at http://cytospade.org.


Asunto(s)
Algoritmos , Citometría de Flujo/métodos , Programas Informáticos , Gráficos por Computador
19.
Cytometry A ; 81(6): 467-75, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22577098

RESUMEN

In fluorescence-based flow cytometry, cellular viability is determined with membrane-impermeable fluorescent reagents that specifically enter and label plasma membrane-compromised nonviable cells. A recent technological advance in flow cytometry uses antibodies conjugated to elemental metal isotopes, rather than to fluorophores, to allow signal detection by atomic mass spectrometry. Unhampered by the limitations of overlapping emission fluorescence, mass cytometry increases the number of parameters that can be measured in single cells. However, mass cytometry is unable to take advantage of current fluorescent viability dyes. An alternative methodology was therefore developed here in which the platinum-containing chemotherapy drug cisplatin was used to resolve live and dead cells by mass cytometry. In a 1-min incubation step, cisplatin preferentially labeled nonviable cells from both adherent and suspension cultures, resulting in a platinum signal quantifiable by mass cytometry. This protocol was compatible with established sample processing steps for intracellular cytometry. Furthermore, the live/dead ratios were comparable between mass- and fluorescence-based cytometry. Importantly, although cisplatin is a known DNA-damaging agent, a 1-min "pulse" of cisplatin did not induce observable DNA damage or apoptotic responses even within 6-h post-exposure. Cisplatin can therefore be used as a viability reagent for a wide range of mass cytometry protocols.


Asunto(s)
Cisplatino/química , Citometría de Flujo/métodos , Espectrometría de Masas/métodos , Platino (Metal)/química , Coloración y Etiquetado/métodos , Adhesión Celular , Permeabilidad de la Membrana Celular , Supervivencia Celular , ADN/análisis , ADN/química , Femenino , Colorantes Fluorescentes , Humanos , Análisis de la Célula Individual , Células Tumorales Cultivadas
20.
Nat Biotechnol ; 29(10): 886-91, 2011 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-21964415

RESUMEN

The ability to analyze multiple single-cell parameters is critical for understanding cellular heterogeneity. Despite recent advances in measurement technology, methods for analyzing high-dimensional single-cell data are often subjective, labor intensive and require prior knowledge of the biological system. To objectively uncover cellular heterogeneity from single-cell measurements, we present a versatile computational approach, spanning-tree progression analysis of density-normalized events (SPADE). We applied SPADE to flow cytometry data of mouse bone marrow and to mass cytometry data of human bone marrow. In both cases, SPADE organized cells in a hierarchy of related phenotypes that partially recapitulated well-described patterns of hematopoiesis. We demonstrate that SPADE is robust to measurement noise and to the choice of cellular markers. SPADE facilitates the analysis of cellular heterogeneity, the identification of cell types and comparison of functional markers in response to perturbations.


Asunto(s)
Algoritmos , Células de la Médula Ósea/citología , Citometría de Flujo/métodos , Adulto , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Simulación por Computador , Bases de Datos como Asunto , Hematopoyesis/efectos de los fármacos , Humanos , Ratones , Coloración y Etiquetado , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA