Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(4): 703-715, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38514887

RESUMEN

Analysis of the human hematopoietic progenitor compartment is being transformed by single-cell multimodal approaches. Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) enables coupled surface protein and transcriptome profiling, thereby revealing genomic programs underlying progenitor states. To perform CITE-seq systematically on primary human bone marrow cells, we used titrations with 266 CITE-seq antibodies (antibody-derived tags) and machine learning to optimize a panel of 132 antibodies. Multimodal analysis resolved >80 stem, progenitor, immune, stromal and transitional cells defined by distinctive surface markers and transcriptomes. This dataset enables flow cytometry solutions for in silico-predicted cell states and identifies dozens of cell surface markers consistently detected across donors spanning race and sex. Finally, aligning annotations from this atlas, we nominate normal marrow equivalents for acute myeloid leukemia stem cell populations that differ in clinical response. This atlas serves as an advanced digital resource for hematopoietic progenitor analyses in human health and disease.


Asunto(s)
Células Madre Hematopoyéticas , Transcriptoma , Humanos , Médula Ósea , Perfilación de la Expresión Génica , Células de la Médula Ósea
2.
Cell ; 156(4): 649-62, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24486105

RESUMEN

Reprogramming somatic cells to induced pluripotency by Yamanaka factors is usually slow and inefficient and is thought to be a stochastic process. We identified a privileged somatic cell state, from which acquisition of pluripotency could occur in a nonstochastic manner. Subsets of murine hematopoietic progenitors are privileged whose progeny cells predominantly adopt the pluripotent fate with activation of endogenous Oct4 locus after four to five divisions in reprogramming conditions. Privileged cells display an ultrafast cell cycle of ∼8 hr. In fibroblasts, a subpopulation cycling at a similar ultrafast speed is observed after 6 days of factor expression and is increased by p53 knockdown. This ultrafast cycling population accounts for >99% of the bulk reprogramming activity in wild-type or p53 knockdown fibroblasts. Our data demonstrate that the stochastic nature of reprogramming can be overcome in a privileged somatic cell state and suggest that cell-cycle acceleration toward a critical threshold is an important bottleneck for reprogramming. PAPERCLIP:


Asunto(s)
Reprogramación Celular , Células Progenitoras de Granulocitos y Macrófagos/citología , Células Madre Pluripotentes Inducidas , Animales , Células de la Médula Ósea , Diferenciación Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Genes p53 , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Ratones
3.
Hum Mol Genet ; 33(2): 138-149, 2024 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-37802886

RESUMEN

Spinocerebellar ataxia type 1 is caused by an expansion of the polyglutamine tract in ATAXIN-1. Ataxin-1 is broadly expressed throughout the brain and is involved in regulating gene expression. However, it is not yet known if mutant ataxin-1 can impact the regulation of alternative splicing events. We performed RNA sequencing in mouse models of spinocerebellar ataxia type 1 and identified that mutant ataxin-1 expression abnormally leads to diverse splicing events in the mouse cerebellum of spinocerebellar ataxia type 1. We found that the diverse splicing events occurred in a predominantly cell autonomous manner. A majority of the transcripts with misregulated alternative splicing events were previously unknown, thus allowing us to identify overall new biological pathways that are distinctive to those affected by differential gene expression in spinocerebellar ataxia type 1. We also provide evidence that the splicing factor Rbfox1 mediates the effect of mutant ataxin-1 on misregulated alternative splicing and that genetic manipulation of Rbfox1 expression modifies neurodegenerative phenotypes in a Drosophila model of spinocerebellar ataxia type 1 in vivo. Together, this study provides novel molecular mechanistic insight into the pathogenesis of spinocerebellar ataxia type 1 and identifies potential therapeutic strategies for spinocerebellar ataxia type 1.


Asunto(s)
Empalme Alternativo , Ataxias Espinocerebelosas , Ratones , Animales , Ataxina-1/genética , Ataxina-1/metabolismo , Empalme Alternativo/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología , Encéfalo/metabolismo , Ataxina-3/metabolismo
4.
Immunity ; 44(2): 246-58, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26872695

RESUMEN

Exposure to a plethora of environmental challenges commonly triggers pathological type 2 cell-mediated inflammation. Here we report the pathological role of the Wnt antagonist Dickkopf-1 (Dkk-1) upon allergen challenge or non-healing parasitic infection. The increased circulating amounts of Dkk-1 polarized T cells to T helper 2 (Th2) cells, stimulating a marked simultaneous induction of the transcription factors c-Maf and Gata-3, mediated by the kinases p38 MAPK and SGK-1, resulting in Th2 cell cytokine production. Circulating Dkk-1 was primarily from platelets, and the increase of Dkk-1 resulted in formation of leukocyte-platelet aggregates (LPA) that facilitated leukocyte infiltration to the affected tissue. Functional inhibition of Dkk-1 impaired Th2 cell cytokine production and leukocyte infiltration, protecting mice from house dust mite (HDM)-induced asthma or Leishmania major infection. These results highlight that Dkk-1 from thrombocytes is an important regulator of leukocyte infiltration and polarization of immune responses in pathological type 2 cell-mediated inflammation.


Asunto(s)
Asma/inmunología , Plaquetas/inmunología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Células Th2/inmunología , Proteínas Wnt/antagonistas & inhibidores , Animales , Antígenos Dermatofagoides/inmunología , Antígenos de Protozoos/inmunología , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Pyroglyphidae , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo
5.
Nucleic Acids Res ; 49(8): e43, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33511400

RESUMEN

Characterization of the epigenetic status of individual cells remains a challenge. Current sequencing approaches have limited coverage, and it is difficult to assign an epigenetic status to the transcription state of individual gene alleles in the same cell. To address these limitations, a targeted microscopy-based epigenetic visualization assay (EVA) was developed for detection and quantification of epigenetic marks at genes of interest in single cells. The assay is based on an in situ biochemical reaction between an antibody-conjugated alkaline phosphatase bound to the epigenetic mark of interest, and a 5'-phosphorylated fluorophore-labeled DNA oligo tethered to a target gene by gene-specific oligonucleotides. When the epigenetic mark is present at the gene, phosphate group removal by the phosphatase protects the oligo from λ-exonuclease activity providing a quantitative fluorescent readout. We applied EVA to measure 5-methylcytosine (5mC) and H3K9Ac levels at different genes and the HIV-1 provirus in human cell lines. To link epigenetic marks to gene transcription, EVA was combined with RNA-FISH. Higher 5mC levels at the silenced compared to transcribed XIST gene alleles in female somatic cells validated this approach and demonstrated that EVA can be used to relate epigenetic marks to the transcription status of individual gene alleles.


Asunto(s)
5-Metilcitosina/metabolismo , Epigénesis Genética , Histonas/metabolismo , Hibridación Fluorescente in Situ/métodos , Análisis de la Célula Individual/métodos , Acetilación , Línea Celular , Metilación de ADN , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Epigenómica , Femenino , Regulación de la Expresión Génica/genética , Silenciador del Gen , VIH-1/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Provirus/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
J Biol Chem ; 296: 100543, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33722605

RESUMEN

Myocardin-related transcription factor A (MRTFA) is a coactivator of serum response factor, a transcription factor that participates in several critical cellular functions including cell growth and apoptosis. MRTFA couples transcriptional regulation to actin cytoskeleton dynamics, and the transcriptional targets of the MRTFA-serum response factor complex include genes encoding cytoskeletal proteins as well as immediate early genes. Previous work has shown that MRTFA promotes the differentiation of many cell types, including various types of muscle cells and hematopoietic cells, and MRTFA's interactions with other protein partners broaden its cellular roles. However, despite being first identified as part of the recurrent t(1;22) chromosomal translocation in acute megakaryoblastic leukemia, the mechanisms by which MRTFA functions in malignant hematopoiesis have yet to be defined. In this review, we provide an in-depth examination of the structure, regulation, and known functions of MRTFA with a focus on hematopoiesis. We conclude by identifying areas of study that merit further investigation.


Asunto(s)
Hematopoyesis/fisiología , Transactivadores/fisiología , Animales , Humanos , Unión Proteica , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Transactivadores/química , Transactivadores/genética , Transcripción Genética
7.
Mol Cancer ; 21(1): 219, 2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36514045

RESUMEN

Adoptive cell therapy (ACT) using tumor infiltrating lymphocytes (TIL) is being studied in multiple tumor types. However, little is known about clonal cell expansion in vitro and persistence of the ACT product in vivo. We performed single-cell RNA and T-Cell Receptor (TCR) sequencing on serial blood and tumor samples from a patient undergoing ACT, who did not respond. We found that clonal expansion varied during preparation of the ACT product, and only one expanded clone was preserved in the ACT product. The TCR of the preserved clone which persisted and remained activated for five months was previously reported as specific for cytomegalovirus and had upregulation of granzyme family genes and genes associated with effector functions (HLA-DQB1, LAT, HLA-DQA1, and KLRD1). Clones that contracted during TIL preparation had features of exhaustion and apoptosis. At disease progression, all previously detected clonotypes were detected. New clonotypes appearing in blood or tumor at disease progression were enriched for genes associated with cytotoxicity or stemness (FGFBP2, GNLY, GZMH, GZMK, IL7R, SELL and KLF2), and these might be harnessed for alternative cellular therapy or cytokine therapy. In-depth single-cell analyses of serial samples from additional ACT-treated patients is warranted, and viral- versus tumor-specificity should be carefully analyzed.


Asunto(s)
Melanoma , Humanos , Melanoma/genética , Linfocitos Infiltrantes de Tumor/patología , Receptores de Antígenos de Linfocitos T/genética , Análisis de la Célula Individual , Insuficiencia del Tratamiento , Progresión de la Enfermedad , Tratamiento Basado en Trasplante de Células y Tejidos , Inmunoterapia Adoptiva
9.
PLoS Biol ; 17(9): e3000421, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31513564

RESUMEN

Decidua is a transient uterine tissue shared by mammals with hemochorial placenta and is essential for pregnancy. The decidua is infiltrated by many immune cells promoting pregnancy. Adult bone marrow (BM)-derived cells (BMDCs) differentiate into rare populations of nonhematopoietic endometrial cells in the uterus. However, whether adult BMDCs become nonhematopoietic decidual cells and contribute functionally to pregnancy is unknown. Here, we show that pregnancy mobilizes mesenchymal stem cells (MSCs) to the circulation and that pregnancy induces considerable adult BMDCs recruitment to decidua, where some differentiate into nonhematopoietic prolactin-expressing decidual cells. To explore the functional importance of nonhematopoietic BMDCs to pregnancy, we used Homeobox a11 (Hoxa11)-deficient mice, having endometrial stromal-specific defects precluding decidualization and successful pregnancy. Hoxa11 expression in BM is restricted to nonhematopoietic cells. BM transplant (BMT) from wild-type (WT) to Hoxa11-/- mice results in stromal expansion, gland formation, and marked decidualization otherwise absent in Hoxa11-/- mice. Moreover, in Hoxa11+/- mice, which have increased pregnancy losses, BMT from WT donors leads to normalized uterine expression of numerous decidualization-related genes and rescue of pregnancy loss. Collectively, these findings reveal that adult BMDCs have a previously unrecognized nonhematopoietic physiologic contribution to decidual stroma, thereby playing important roles in decidualization and pregnancy.


Asunto(s)
Células de la Médula Ósea/fisiología , Decidua/citología , Implantación del Embrión , Células Madre Mesenquimatosas/fisiología , Embarazo/fisiología , Animales , Femenino , Proteínas de Homeodominio/genética , Masculino , Ratones Noqueados
10.
Curr Opin Hematol ; 28(1): 28-35, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33186151

RESUMEN

PURPOSE OF REVIEW: This review focuses on our current understanding of fate decisions in bipotent megakaryocyte-erythroid progenitors (MEPs). Although extensive research has been carried out over decades, our understanding of how MEP commit to the erythroid versus megakaryocyte fate remains unclear. RECENT FINDINGS: We discuss the isolation of primary human MEP, and focus on gene expression patterns, epigenetics, transcription factors and extrinsic factors that have been implicated in MEP fate determination. We conclude with an overview of the open debates in the field of MEP biology. SUMMARY: Understanding MEP fate is important because defects in megakaryocyte and erythrocyte development lead to disease states such as anaemia, thrombocytopenia and leukaemia. MEP also represent a model system for studying fundamental principles underlying cell fate decisions of bipotent and pluripotent progenitors, such that discoveries in MEP are broadly applicable to stem/progenitor cell biology.


Asunto(s)
Hematopoyesis , Células Progenitoras de Megacariocitos y Eritrocitos/citología , Animales , Linaje de la Célula , Células Eritroides/citología , Células Eritroides/metabolismo , Humanos , Células Progenitoras de Megacariocitos y Eritrocitos/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Transcriptoma
11.
Blood ; 134(18): 1547-1557, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31439541

RESUMEN

The mechanisms underlying thrombocytosis in patients with iron deficiency anemia remain unknown. Here, we present findings that support the hypothesis that low iron biases the commitment of megakaryocytic (Mk)-erythroid progenitors (MEPs) toward the Mk lineage in both human and mouse. In MEPs of transmembrane serine protease 6 knockout (Tmprss6-/-) mice, which exhibit iron deficiency anemia and thrombocytosis, we observed a Mk bias, decreased labile iron, and decreased proliferation relative to wild-type (WT) MEPs. Bone marrow transplantation assays suggest that systemic iron deficiency, rather than a local role for Tmprss6-/- in hematopoietic cells, contributes to the MEP lineage commitment bias observed in Tmprss6-/- mice. Nontransgenic mice with acquired iron deficiency anemia also show thrombocytosis and Mk-biased MEPs. Gene expression analysis reveals that messenger RNAs encoding genes involved in metabolic, vascular endothelial growth factor, and extracellular signal-regulated kinase (ERK) pathways are enriched in Tmprss6-/- vs WT MEPs. Corroborating our findings from the murine models of iron deficiency anemia, primary human MEPs exhibit decreased proliferation and Mk-biased commitment after knockdown of transferrin receptor 2, a putative iron sensor. Signal transduction analyses reveal that both human and murine MEP have lower levels of phospho-ERK1/2 in iron-deficient conditions compared with controls. These data are consistent with a model in which low iron in the marrow environment affects MEP metabolism, attenuates ERK signaling, slows proliferation, and biases MEPs toward Mk lineage commitment.


Asunto(s)
Anemia Ferropénica/metabolismo , Diferenciación Celular/fisiología , Células Progenitoras de Megacariocitos/metabolismo , Megacariocitos/metabolismo , Anemia Ferropénica/complicaciones , Animales , Proliferación Celular , Humanos , Hierro , Células Progenitoras de Megacariocitos/citología , Megacariocitos/citología , Ratones , Ratones Noqueados , Trombocitosis/etiología , Trombocitosis/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 39(4): 603-612, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30727757

RESUMEN

Smooth muscle cells (SMCs) are a critical component of blood vessel walls that provide structural support, regulate vascular tone, and allow for vascular remodeling. These cells also exhibit a remarkable plasticity that contributes to vascular growth and repair but also to cardiovascular pathologies, including atherosclerosis, intimal hyperplasia and restenosis, aneurysm, and transplant vasculopathy. Mouse models have been an important tool for the study of SMC functions. The development of smooth muscle-expressing Cre-driver lines has allowed for exciting discoveries, including recent advances revealing the diversity of phenotypes derived from mature SMC transdifferentiation in vivo using inducible CreER T2 lines. We review SMC-targeting Cre lines driven by the Myh11, Tagln, and Acta2 promoters, including important technical considerations associated with these models. Limitations that can complicate study of the vasculature include expression in visceral SMCs leading to confounding phenotypes, and expression in multiple nonsmooth muscle cell types, such as Acta2-Cre expression in myofibroblasts. Notably, the frequently employed Tagln/ SM22α- Cre driver expresses in the embryonic heart but can also confer expression in nonmuscular cells including perivascular adipocytes and their precursors, myeloid cells, and platelets, with important implications for interpretation of cardiovascular phenotypes. With new Cre-driver lines under development and the increasing use of fate mapping methods, we are entering an exciting new era in SMC research.


Asunto(s)
Marcación de Gen/métodos , Músculo Liso Vascular/fisiología , Regiones Promotoras Genéticas , Actinas/biosíntesis , Actinas/genética , Animales , Línea Celular , Linaje de la Célula , Transdiferenciación Celular , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/genética , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Miocitos del Músculo Liso/fisiología , Miofibroblastos/fisiología , Cadenas Pesadas de Miosina/biosíntesis , Cadenas Pesadas de Miosina/genética , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica , Fenotipo , Proteínas Recombinantes de Fusión/metabolismo
13.
Stem Cells ; 36(8): 1138-1145, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29658164

RESUMEN

Hematopoietic stem and progenitor cells maintain blood formation throughout our lifetime by undergoing long- and short-term self-renewal, respectively. As progenitor cells progress through the hematopoiesis process, their differentiation capabilities narrow, such that the precursors become committed to only one or two lineages. This Review focuses on recent advances in the identification and characterization of bipotent megakaryocytic-erythroid progenitors (MEP), the cells that can further produce two completely different functional outputs: platelets and red blood cells. The existence of MEP has sparked controversy as studies describing the requirement for this intermediate progenitor stage prior to commitment to the erythroid and megakaryocytic lineages have been potentially contradictory. Interpretation of these studies is complicated by the variety of species, cell sources, and analytical approaches used along with inherent challenges in the continuum of hematopoiesis, where hematopoietic progenitors do not stop at discrete steps on single paths as classically drawn in hematopoietic hierarchy models. With the goal of improving our understanding of human hematopoiesis, we discuss findings in both human and murine cells. Based on these data, MEP clearly represent a transitional stage of differentiation in at least one route to the generation of both megakaryocytes and erythroid cells. Stem Cells 2018;36:1138-1145.


Asunto(s)
Células Progenitoras de Megacariocitos y Eritrocitos/citología , Animales , Linaje de la Célula , Eritropoyesis , Humanos
14.
Am J Physiol Lung Cell Mol Physiol ; 314(5): L882-L892, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29345196

RESUMEN

Surfactant protein C (SPC), a key component of pulmonary surfactant, also plays a role in regulating inflammation. SPC deficiency in patients and mouse models is associated with increased inflammation and delayed repair, but the key drivers of SPC-regulated inflammation in response to injury are largely unknown. This study focuses on a new mechanism of SPC as an anti-inflammatory molecule using SPC-TK/SPC-KO (surfactant protein C-thymidine kinase/surfactant protein C knockout) mice, which represent a novel sterile injury model that mimics clinical acute respiratory distress syndrome (ARDS). SPC-TK mice express the inducible suicide gene thymidine kinase from by the SPC promoter, which targets alveolar type 2 (AT2) cells for depletion in response to ganciclovir (GCV). We compared GCV-induced injury and repair in SPC-TK mice that have normal endogenous SPC expression with SPC-TK/SPC-KO mice lacking SPC expression. In contrast to SPC-TK mice, SPC-TK/SPC-KO mice treated with GCV exhibited more severe inflammation, resulting in over 90% mortality; there was only 8% mortality of SPC-TK animals. SPC-TK/SPC-KO mice had highly elevated inflammatory cytokines and granulocyte infiltration in the bronchoalveolar lavage (BAL) fluid. Consistent with a proinflammatory phenotype, immunofluorescence revealed increased phosphorylated signal transduction and activation of transcription 3 (pSTAT3), suggesting enhanced Janus kinase (JAK)/STAT activation in inflammatory and AT2 cells of SPC-TK/SPC-KO mice. The level of suppressor of cytokine signaling 3, an anti-inflammatory mediator that decreases pSTAT3 signaling, was significantly decreased in the BAL fluid of SPC-TK/SPC-KO mice. Hyperactivation of pSTAT3 and inflammation were rescued by AZD1480, a JAK1/2 inhibitor. Our findings showing a novel role for SPC in regulating inflammation via JAK/STAT may have clinical applications.


Asunto(s)
Modelos Animales de Enfermedad , Janus Quinasa 1/metabolismo , Lesión Pulmonar/prevención & control , Péptidos/fisiología , Neumonía/prevención & control , Factor de Transcripción STAT3/metabolismo , Timidina Quinasa/fisiología , Animales , Péptidos y Proteínas de Señalización Intercelular , Janus Quinasa 1/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Neumonía/metabolismo , Neumonía/patología , Proteína C Asociada a Surfactante Pulmonar , Factor de Transcripción STAT3/genética
15.
Blood ; 127(11): 1386, 2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26989190

RESUMEN

Platelet adhesion to collagen via collagen receptors is an important part of thrombosis. In this issue of Blood, Matsuura et al identify collagen receptors as previously unrecognized targets of the extracellular enzyme lysyl oxidase (LOX), the level of which is increased in myeloproliferative neoplasms (MPNs) and other conditions associated with pathological thromboses.


Asunto(s)
Plaquetas/efectos de los fármacos , Colágeno/farmacología , Activación Plaquetaria/fisiología , Proteína-Lisina 6-Oxidasa/fisiología , Trombofilia/enzimología , Animales
16.
Blood ; 128(7): 923-33, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27268089

RESUMEN

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.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Antígenos CD34/metabolismo , Células Progenitoras de Megacariocitos y Eritrocitos/citología , Adulto , Linaje de la Célula , Separación Celular , Ensayo de Unidades Formadoras de Colonias , Células Eritroides/citología , Células Eritroides/metabolismo , Humanos , Células Progenitoras de Megacariocitos y Eritrocitos/metabolismo , Megacariocitos/citología , Fenotipo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Receptores de Trombopoyetina/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo
17.
Blood ; 126(4): 520-30, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25964668

RESUMEN

The actin cytoskeleton is important for platelet biogenesis. Tropomodulin-3 (Tmod3), the only Tmod isoform detected in platelets and megakaryocytes (MKs), caps actin filament (F-actin) pointed ends and binds tropomyosins (TMs), regulating actin polymerization and stability. To determine the function of Tmod3 in platelet biogenesis, we studied Tmod3(-/-) embryos, which are embryonic lethal by E18.5. Tmod3(-/-) embryos often show hemorrhaging at E14.5 with fewer and larger platelets, indicating impaired platelet biogenesis. MK numbers are moderately increased in Tmod3(-/-) fetal livers, with only a slight increase in the 8N population, suggesting that MK differentiation is not significantly affected. However, Tmod3(-/-) MKs fail to develop a normal demarcation membrane system (DMS), and cytoplasmic organelle distribution is abnormal. Moreover, cultured Tmod3(-/-) MKs exhibit impaired proplatelet formation with a wide range of proplatelet bud sizes, including abnormally large proplatelet buds containing incorrect numbers of von Willebrand factor-positive granules. Tmod3(-/-) MKs exhibit F-actin disturbances, and Tmod3(-/-) MKs spreading on collagen fail to polymerize F-actin into actomyosin contractile bundles. Tmod3 associates with TM4 and the F-actin cytoskeleton in wild-type MKs, and confocal microscopy reveals that Tmod3, TM4, and F-actin partially colocalize near the membrane of proplatelet buds. In contrast, the abnormally large proplatelets from Tmod3(-/-) MKs show increased F-actin and redistribution of F-actin and TM4 from the cortex to the cytoplasm, but normal microtubule coil organization. We conclude that F-actin capping by Tmod3 regulates F-actin organization in mouse fetal liver-derived MKs, thereby controlling MK cytoplasmic morphogenesis, including DMS formation and organelle distribution, as well as proplatelet formation and sizing.


Asunto(s)
Citoesqueleto de Actina/patología , Plaquetas/patología , Membrana Celular/patología , Embrión de Mamíferos/patología , Hemorragia/etiología , Megacariocitos/patología , Tropomodulina/fisiología , Citoesqueleto de Actina/metabolismo , Animales , Apoptosis , Plaquetas/metabolismo , Western Blotting , Membrana Celular/metabolismo , Proliferación Celular , Células Cultivadas , Citoplasma/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Hematopoyesis/fisiología , Hemorragia/metabolismo , Hemorragia/patología , Inmunoprecipitación , Megacariocitos/metabolismo , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Ploidias , Polimerizacion
19.
Am J Physiol Lung Cell Mol Physiol ; 310(8): L711-9, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26851259

RESUMEN

Cystic fibrosis (CF) is caused by homozygous mutations of the CF transmembrane conductance regulator (CFTR) Cl(-) channel, which result in chronic pulmonary infection and inflammation, the major cause of morbidity and mortality. Although these processes are clearly related to each other, each is likely to contribute to the pathology differently. Understanding the contribution of each of these processes to the overall pathology has been difficult, because they are usually so intimately connected. Various CF mouse models have demonstrated abnormal immune responses compared with wild-type (WT) littermates when challenged with live bacteria or bacterial products acutely. However, these studies have not investigated the consequences of persistent inflammation on lung tissue in CF mice, which may better model the lung pathology in patients. We characterized the lung pathology and immune response of Cftr(-/-) (CF) and Cftr(+/+) (WT) mice to chronic administration of Pseudomonas aeruginosa lipopolysaccharide (LPS). We show that, after long-term repeated LPS exposure, CF mice develop an abnormal and persistent immune response, which is associated with more robust structural changes in the lung than those observed in WT mice. Although CF mice and their WT littermates develop lung pathology after chronic exposure to LPS, the inflammation and damage resolve in WT mice. However, CF mice do not recover efficiently, and, as a consequence of their chronic inflammation, CF mice are more susceptible to morphological changes and lung remodeling. This study shows that chronic inflammation alone contributes significantly to aspects of CF lung pathology.


Asunto(s)
Fibrosis Quística/patología , Lipopolisacáridos/farmacología , Pulmón/patología , Neumonía/inmunología , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Quimiocina CXCL10/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/inmunología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Pulmón/inmunología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos CFTR , Ratones Noqueados , Neumonía/patología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
20.
Blood ; 133(19): 2004-2005, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-31072961
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA