Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 5(1): 1231, 2022 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-36371461

RESUMO

Cell-cell communication and physical interactions play a vital role in cancer initiation, homeostasis, progression, and immune response. Here, we report a system that combines live capture of different cell types, co-incubation, time-lapse imaging, and gene expression profiling of doublets using a microfluidic integrated fluidic circuit that enables measurement of physical distances between cells and the associated transcriptional profiles due to cell-cell interactions. We track the temporal variations in natural killer-triple-negative breast cancer cell distances and compare them with terminal cellular transcriptome profiles. The results show the time-bound activities of regulatory modules and allude to the existence of transcriptional memory. Our experimental and bioinformatic approaches serve as a proof of concept for interrogating live-cell interactions at doublet resolution. Together, our findings highlight the use of our approach across different cancers and cell types.


Assuntos
Transcriptoma , Neoplasias de Mama Triplo Negativas , Humanos , Microfluídica , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica
2.
Cell Rep ; 25(8): 2083-2093.e4, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30463007

RESUMO

Megakaryocytic-erythroid progenitors (MEPs) give rise to the cells that produce red blood cells and platelets. Although the mechanisms underlying megakaryocytic (MK) and erythroid (E) maturation have been described, those controlling their specification from MEPs are unknown. Single-cell RNA sequencing of primary human MEPs, common myeloid progenitors (CMPs), megakaryocyte progenitors, and E progenitors revealed a distinct transitional MEP signature. Inferred regulatory transcription factors (TFs) were associated with differential expression of cell cycle regulators. Genetic manipulation of selected TFs validated their role in lineage specification and demonstrated coincident modulation of the cell cycle. Genetic and pharmacologic modulation demonstrated that cell cycle activation is sufficient to promote E versus MK specification. These findings, obtained from healthy human cells, lay a foundation to study the mechanisms underlying benign and malignant disease states of the megakaryocytic and E lineages.


Assuntos
Ciclo Celular , Linhagem da Célula , Células Progenitoras de Megacariócitos e Eritrócitos/citologia , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Células HEK293 , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo
3.
Leukemia ; 32(7): 1670, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29891936

RESUMO

At the time of publication the funding information was omitted from the article - this has now been corrected in both the HTML and the PDF.

4.
Leukemia ; 32(7): 1575-1586, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29467489

RESUMO

The presence, within the human bone marrow, of cells with both endothelial and hemogenic potential has been controversial. Herein, we identify, within the human fetal bone marrow, prior to establishment of hematopoiesis, a unique APLNR+, Stro-1+ cell population, co-expressing markers of early mesodermal precursors and/or hemogenic endothelium. In adult marrow, cells expressing similar markers are also found, but at very low frequency. These adult-derived cells can be extensively culture expanded in vitro without loss of potential, they preserve a biased hemogenic transcriptional profile, and, upon in vitro induction with OCT4, assume a hematopoietic phenotype. In vivo, these cells, upon transplantation into a fetal microenvironment, contribute to the vasculature, and generate hematopoietic cells that provide multilineage repopulation upon serial transplantation. The identification of this human somatic cell population provides novel insights into human ontogenetic hematovascular potential, which could lead to a better understanding of, and new target therapies for, malignant and nonmalignant hematologic disorders.

5.
Blood ; 128(7): 923-33, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27268089

RESUMO

Bipotent megakaryocyte/erythroid progenitors (MEPs) give rise to progeny limited to the megakaryocyte (Mk) and erythroid (E) lineages. We developed a novel dual-detection functional in vitro colony-forming unit (CFU) assay for single cells that differentiates down both the Mk and E lineages (CFU-Mk/E), which allowed development and validation of a novel purification strategy for the identification and quantitation of primary functional human MEPs from granulocyte colony-stimulating factor-mobilized peripheral blood and bone marrow. Applying this assay to fluorescence-activated cell sorter-sorted cell populations, we found that the Lin(-)CD34(+)CD38(mid)CD45RA(-)FLT3(-)MPL(+)CD36(-)CD41(-) population is much more highly enriched for bipotent MEPs than any previously reported subpopulations. We also developed purification strategies for primary human lineage-committed Mk and E progenitors identified as CFU-Mk and burst forming unit-E. Comparative expression analyses in MEP, MkP, and ErP populations revealed differential expression of MYB We tested whether alterations in MYB concentration affect the Mk-E fate decision at the single cell level in MEPs and found that short hairpin RNA-mediated MYB knockdown promoted commitment of MEPs to the Mk lineage, further defining its role in MEP lineage fate. There are numerous applications for these novel enrichment strategies, including facilitating mechanistic studies of MEP lineage commitment, improving approaches for in vitro expansion of Mk and E cells, and developing improved therapies for benign and malignant hematologic disease.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Antígenos CD34/metabolismo , Células Progenitoras de Megacariócitos e Eritrócitos/citologia , Adulto , Linhagem da Célula , Separação Celular , Ensaio de Unidades Formadoras de Colônias , Células Eritroides/citologia , Células Eritroides/metabolismo , Humanos , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Megacariócitos/citologia , Fenótipo , Proteínas Proto-Oncogênicas c-myb/metabolismo , Receptores de Trombopoetina/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo
6.
J Cell Physiol ; 228(5): 1010-6, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23042590

RESUMO

Besides the liver, it has been difficult to identify which organ(s) and/or cellular component(s) contribute significantly to the production of human FVIII:c (FVIII). Thus far, only endothelial cells have been shown to constitute a robust extrahepatic source of FVIII, possibly explaining both the diverse presence of FVIII mRNA in the body, and the observed increase in FVIII levels during liver failure. Here, we investigate whether human mesenchymal stem cells (MSC), ubiquitously present in different organs, could also contribute to FVIII production. MSC isolated from human lung, liver, brain, and bone marrow expressed FVIII message as determined by quantitative-RT-PCR. Using an antibody specific for FVIII, confocal microscopy, and umbilical cord-derived endothelial cells (HUVEC) as a negative control, we demonstrated that, in MSC, FVIII protein was not stored in granules; rather, it localized to the perinuclear region. Furthermore, functional FVIII was detected in MSC supernatants and cell lysates by aPTT and chromogenic assays. These results demonstrate that MSC can contribute at low levels to the functional FVIII pool, and advance the understanding of the physiology of FVIII production and secretion.


Assuntos
Fator VIII/biossíntese , Fator VIII/metabolismo , Células-Tronco Mesenquimais , Diferenciação Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Células-Tronco Mesenquimais/metabolismo , Microscopia Confocal , RNA Mensageiro/metabolismo , Vesículas Secretórias/metabolismo , Distribuição Tecidual
7.
Exp Hematol ; 39(12): 1124-1135.e4, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21906573

RESUMO

We recently re-established a line of sheep that accurately mimics the clinical symptoms and genetics of severe hemophilia A (HA). Here, we tested a novel, nonablative transplantation therapy in two pediatric HA animals. Paternal mesenchymal stem cells (MSC) were transduced with a porcine FVIII-encoding lentivector and transplanted via the intraperitoneal route without preconditioning. At the time of transplantation, these animals had received multiple human FVIII treatments for various spontaneous bleeds and had developed debilitating hemarthroses, which produced severe defects in posture and gait. Transplantation of transduced MSC resolved all existent hemarthroses, and spontaneous bleeds ceased. Damaged joints recovered fully; the animals regained normal posture and gait and resumed normal activity. Despite achieving factor-independence, a sharp rise in pre-existent Bethesda titers occurred following transplantation, decreasing the effectiveness and duration of therapy. Postmortem examination revealed widespread engraftment, with MSC present within the lung, liver, intestine, and thymus, but particularly within joints affected at the time of transplantation, suggesting MSC homed to sites of ongoing injury/inflammation to release FVIII, explaining the dramatic improvement in hemarthrotic joints. In summary, this novel, nonablative MSC transplantation was straightforward, safe, and converted life-threatening, debilitating HA to a moderate phenotype in a large animal model.


Assuntos
Modelos Animais de Doenças , Fator VIII/genética , Hemofilia A/cirurgia , Transplante de Células-Tronco Mesenquimais , Ovinos/genética , Animais , Linhagem da Célula , Movimento Celular , Fator VIII/imunologia , Feminino , Vetores Genéticos/genética , Sobrevivência de Enxerto , Hemartrose/etiologia , Hemartrose/patologia , Hemofilia A/complicações , Hemofilia A/tratamento farmacológico , Hemorragia/etiologia , Humanos , Injeções Intraperitoneais , Isoanticorpos/biossíntese , Isoanticorpos/imunologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/virologia , Fenótipo , Proteínas Recombinantes/uso terapêutico , Indução de Remissão , Ovinos/sangue , Sus scrofa/genética , Distribuição Tecidual
8.
Exp Hematol ; 36(12): 1739-49, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19007686

RESUMO

OBJECTIVE: We and many others have long used sheep as a predictive model system in which to explore stem cell transplantation. Unfortunately, while numerous markers are available to identify and isolate human hematopoietic stem cells (HSC), no reagents exist that allow HSC/progenitors from sheep to be identified or purified, greatly impeding the application of this well-established large animal model to the study of autologous or allogeneic HSC transplantation. The current studies were undertaken to create a monoclonal antibody to sheep CD34 that would enable isolation and study of sheep HSC/progenitors. MATERIALS AND METHODS: A partial cDNA to the extracellular domain of the sheep CD34 antigen was polymerase chain reaction cloned, characterized, and used to genetically immunize mice and create hybridomas. RESULTS: The resultant monoclonal antibody to sheep CD34 allows flow cytometric detection of sheep HSC/progenitors present within bone marrow, cord blood, and mobilized peripheral blood. Moreover, this antibody can be used to enrich for HSC/progenitors with enhanced in vitro colony-forming potential, and also identifies endothelial cells in situ within paraffin-embedded tissue sections, similarly to antibodies to human CD34. CONCLUSIONS: The availability of this monoclonal antibody recognizing the stem cell antigen CD34 in sheep will greatly facilitate the study of autologous and allogeneic HSC transplantation using this clinically relevant large animal model.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD34/imunologia , Células-Tronco Hematopoéticas/imunologia , Ovinos/imunologia , Animais , Especificidade de Anticorpos/imunologia , Citometria de Fluxo , Humanos , Camundongos , Modelos Biológicos , Transplante de Células-Tronco , Transplante Autólogo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA