Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 34(6): 837-850, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38977309

RESUMEN

Studies on human parathyroids are generally limited to hyperfunctioning glands owing to the difficulty in obtaining normal human tissue. We therefore obtained non-human primate (NHP) parathyroids to provide a suitable alternative for sequencing that would bear a close semblance to human organs. Single-cell RNA expression analysis of parathyroids from four healthy adult M. mulatta reveals a continuous trajectory of epithelial cell states. Pseudotime analysis based on transcriptomic signatures suggests a progression from GCM2 hi progenitors to mature parathyroid hormone (PTH)-expressing epithelial cells with increasing core mitochondrial transcript abundance along pseudotime. We sequenced, as a comparator, four histologically characterized hyperfunctioning human parathyroids with varying oxyphil and chief cell abundance and leveraged advanced computational techniques to highlight similarities and differences from non-human primate parathyroid expression dynamics. Predicted cell-cell communication analysis reveals abundant endothelial cell interactions in the parathyroid cell microenvironment in both human and NHP parathyroid glands. We show abundant RARRES2 transcripts in both human adenoma and normal primate parathyroid cells and use coimmunostaining to reveal high levels of RARRES2 protein (also known as chemerin) in PTH-expressing cells, which could indicate that RARRES2 plays an unrecognized role in parathyroid endocrine function. The data obtained are the first single-cell RNA transcriptome to characterize nondiseased parathyroid cell signatures and to show a transcriptomic progression of cell states within normal parathyroid glands, which can be used to better understand parathyroid cell biology.


Asunto(s)
Macaca mulatta , Glándulas Paratiroides , Análisis de la Célula Individual , Análisis de la Célula Individual/métodos , Humanos , Glándulas Paratiroides/metabolismo , Animales , Transcriptoma , Quimiocinas/metabolismo , Quimiocinas/genética , Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/genética , Comunicación Celular , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Transcripción Genética
2.
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
3.
J Immunother ; 47(2): 49-53, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37991241

RESUMEN

Adoptive transfer of ex vivo expanded tumor-infiltrating lymphocytes (TILs) have produced long-term response in metastatic cancers. TILs have traditionally been expanded from surgically resected specimens. Ultrasound-guided core needle biopsy (CNB) is an alternative method that avoids the morbidity of surgery and have added benefits which may include patients not amenable to surgery as well as the potential to produce TILs from multiple lesions in the same patient. We assessed the ability to produce and expand TILs from primary triple-negative breast cancer tumors from CNB (n=7) and demonstrate comparable expansion, phenotype and cytokine secretion after phorbol myristate acetate-ionomycin stimulation to TILs expanded from surgery (n=6). T cell Receptor clonality and diversity were also comparable between the two cohorts throughout the TIL culture. CNB is a safe and feasible method to obtain tumor tissue for TIL generation in patients with triple-negative breast cancer.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias de la Mama Triple Negativas , Humanos , Biopsia con Aguja Gruesa , Neoplasias de la Mama Triple Negativas/terapia , Linfocitos Infiltrantes de Tumor/patología , Fenotipo
4.
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
5.
Exp Hematol ; 124: 36-44.e3, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37271449

RESUMEN

Colony-forming unit (CFU) assays are a powerful tool in hematopoietic research because they allow researchers to functionally test the lineage potential of individual stem and progenitor cells. Assaying for lineage potential is important for determining and validating the identity of progenitor populations isolated by methods such as fluorescence-activated cell sorting (FACS). However, current methods for CFU assays are limited in their ability to robustly assay multipotent progenitors with the ability to differentiate down the myeloid, erythroid, and megakaryocytic lineages because of the lack of specific growth factors necessary for certain lineage outputs. In addition, manual counting of colony types is subjective resulting in user to user variability in assessments of cell types based on colony and cell morphologies. We demonstrate that the addition of granulocyte colony-stimulating factor (G-CSF), macrophage (M)-CSF, and granulocyte-macrophage (GM)-CSF into a collagen-based MegaCult medium containing IL-3, IL-6, SCF, EPO, and TPO allows for the differentiation of common myeloid progenitors into expected proportions of colonies containing granulocytic (G), monocytic (M), erythroid (E), and megakaryocytic (Mk) cells. Additionally, we demonstrate an objective method using in situ immunofluorescence (IF) with anti-CD66b, anti-CD14, anti-CD235a, and anti-CD41 to detect G, M, E, and Mk cells, respectively. IF stained colonies can be analyzed individually at a microscope or using high-throughput microscopy. Thus, our improvements to the culture conditions and method for assay readout increase the accuracy, reproducibility, and throughput of the myeloid CFU assay.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Interleucina-3 , Humanos , Reproducibilidad de los Resultados , Células Madre Hematopoyéticas , Ensayo de Unidades Formadoras de Colonias , Células Cultivadas
6.
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
7.
Cell Rep ; 41(11): 111797, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36516754

RESUMEN

Persistent neutrophil-dominated lung inflammation contributes to lung damage in cystic fibrosis (CF). However, the mechanisms that drive persistent lung neutrophilia and tissue deterioration in CF are not well characterized. Starting from the observation that, in patients with CF, c-c motif chemokine receptor 2 (CCR2)+ monocytes/macrophages are abundant in the lungs, we investigate the interplay between monocytes/macrophages and neutrophils in perpetuating lung tissue damage in CF. Here we show that CCR2+ monocytes in murine CF lungs drive pathogenic transforming growth factor ß (TGF-ß) signaling and sustain a pro-inflammatory environment by facilitating neutrophil recruitment. Targeting CCR2 to lower the numbers of monocytes in CF lungs ameliorates neutrophil inflammation and pathogenic TGF-ß signaling and prevents lung tissue damage. This study identifies CCR2+ monocytes as a neglected contributor to the pathogenesis of CF lung disease and as a therapeutic target for patients with CF, for whom lung hyperinflammation and tissue damage remain an issue despite recent advances in CF transmembrane conductance regulator (CFTR)-specific therapeutic agents.


Asunto(s)
Fibrosis Quística , Neumonía , Humanos , Ratones , Animales , Fibrosis Quística/patología , Monocitos/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Neumonía/patología , Pulmón/patología , Inflamación/patología , Receptores de Quimiocina/metabolismo , Macrófagos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
9.
Sci Rep ; 12(1): 16218, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36171423

RESUMEN

Single-cell assays have enriched our understanding of hematopoiesis and, more generally, stem and progenitor cell biology. However, these single-end-point approaches provide only a static snapshot of the state of a cell. To observe and measure dynamic changes that may instruct cell fate, we developed an approach for examining hematopoietic progenitor fate specification using long-term (> 7-day) single-cell time-lapse imaging for up to 13 generations with in situ fluorescence staining of primary human hematopoietic progenitors followed by algorithm-assisted lineage tracing. We analyzed progenitor cell dynamics, including the division rate, velocity, viability, and probability of lineage commitment at the single-cell level over time. We applied a Markov probabilistic model to predict progenitor division outcome over each generation in culture. We demonstrated the utility of this methodological pipeline by evaluating the effects of the cytokines thrombopoietin and erythropoietin on the dynamics of self-renewal and lineage specification in primary human bipotent megakaryocytic-erythroid progenitors (MEPs). Our data support the hypothesis that thrombopoietin and erythropoietin support the viability and self-renewal of MEPs, but do not affect fate specification. Thus, single-cell tracking of time-lapse imaged colony-forming unit assays provides a robust method for assessing the dynamics of progenitor self-renewal and lineage commitment.


Asunto(s)
Eritropoyetina , Trombopoyetina , Diferenciación Celular , Linaje de la Célula , Eritropoyetina/farmacología , Humanos , Megacariocitos , Trombopoyetina/farmacología
11.
Exp Mol Med ; 54(5): 639-652, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35581352

RESUMEN

Overwhelming neutrophilic inflammation is a leading cause of lung damage in many pulmonary diseases, including cystic fibrosis (CF). The heme oxygenase-1 (HO-1)/carbon monoxide (CO) pathway mediates the resolution of inflammation and is defective in CF-affected macrophages (MΦs). Here, we provide evidence that systemic administration of PP-007, a CO releasing/O2 transfer agent, induces the expression of HO-1 in a myeloid differentiation factor 88 (MyD88) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)-dependent manner. It also rescues the reduced HO-1 levels in CF-affected cells induced in response to lipopolysaccharides (LPS) or Pseudomonas aeruginosa (PA). Treatment of CF and muco-obstructive lung disease mouse models with a single clinically relevant dose of PP-007 leads to effective resolution of lung neutrophilia and to decreased levels of proinflammatory cytokines in response to LPS. Using HO-1 conditional knockout mice, we show that the beneficial effect of PP-007 is due to the priming of circulating monocytes trafficking to the lungs in response to infection to express high levels of HO-1. Finally, we show that PP-007 does not compromise the clearance of PA in the setting of chronic airway infection. Overall, we reveal the mechanism of action of PP-007 responsible for the immunomodulatory function observed in clinical trials for a wide range of diseases and demonstrate the potential use of PP-007 in controlling neutrophilic pulmonary inflammation by promoting the expression of HO-1 in monocytes/macrophages.


Asunto(s)
Fibrosis Quística , Neumonía , Animales , Fibrosis Quística/complicaciones , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Hemo-Oxigenasa 1 , Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Pulmón/patología , Ratones , Monocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neumonía/patología
12.
Cells ; 10(7)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206516

RESUMEN

BACKGROUND: Alveolar type 2 (AT2) cells and bronchioalveolar stem cells (BASC) perform critical regenerative functions in response to lung damage. Published data show that nonhematopoietic, bone marrow-derived "very small embryonic-like stem cells" (VSELs) can differentiate in vivo into surfactant protein C (SPC)-producing AT2 cells in the lung. Here, we test directly whether VSEL-derived BASC and AT2 cells function to produce differentiated progeny. METHODS: using a reporter mouse in which the H2B-GFP fusion protein is driven from the murine SPC promoter, we tested whether bone marrow-derived VSELs or non-VSEL/nonhematopoietic stem cells (non-VSEL/non-HSCs) can differentiate into AT2 and BASC cells that function as progenitor cells. Immediately following bleomycin administration, WT recipient mice underwent intravenous administration of VSELs or non-VSEL/non-HSCs from SPC H2B-GFP mice. GFP+ AT2 and BASC were isolated and tested for progenitor activity using in vitro organoid assays. RESULTS: after 21 days in vivo, we observed differentiation of VSELs but not non-VSEL/non-HSCs into phenotypic AT2 and BASC consistent with previous data in irradiated recipients. Subsequent in vitro organoid assays revealed that VSEL-derived AT2 and BASC maintained physiological potential for differentiation and self-renewal. CONCLUSION: these findings prove that VSELs produce functional BASC and AT2 cells, and this may open new avenues using VSELs to develop effective cell therapy approaches for patients with lung injury.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Embrionarias/trasplante , Células Epiteliales/citología , Lesión Pulmonar/patología , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/citología , Ratones Endogámicos C57BL , Organoides/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
13.
Cell Rep ; 36(4): 109421, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34320342

RESUMEN

Mitogen-activated protein kinases (MAPKs) are inactivated by dual-specificity phosphatases (DUSPs), the activities of which are tightly regulated during cell differentiation. Using knockdown screening and single-cell transcriptional analysis, we demonstrate that DUSP4 is the phosphatase that specifically inactivates p38 kinase to promote megakaryocyte (Mk) differentiation. Mechanistically, PRMT1-mediated methylation of DUSP4 triggers its ubiquitinylation by an E3 ligase HUWE1. Interestingly, the mechanistic axis of the DUSP4 degradation and p38 activation is also associated with a transcriptional signature of immune activation in Mk cells. In the context of thrombocytopenia observed in myelodysplastic syndrome (MDS), we demonstrate that high levels of p38 MAPK and PRMT1 are associated with low platelet counts and adverse prognosis, while pharmacological inhibition of p38 MAPK or PRMT1 stimulates megakaryopoiesis. These findings provide mechanistic insights into the role of the PRMT1-DUSP4-p38 axis on Mk differentiation and present a strategy for treatment of thrombocytopenia associated with MDS.


Asunto(s)
Diferenciación Celular , Fosfatasas de Especificidad Dual , Megacariocitos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos , Adulto , Animales , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Arginina/metabolismo , Línea Celular , Fosfatasas de Especificidad Dual/metabolismo , Estabilidad de Enzimas , Células HEK293 , Sistema de Señalización de MAP Quinasas , Megacariocitos/citología , Megacariocitos/enzimología , Metilación , Ratones Endogámicos C57BL , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Síndromes Mielodisplásicos/enzimología , Síndromes Mielodisplásicos/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Poliubiquitina/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteolisis , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Ubiquitinación
14.
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
15.
Science ; 371(6533): 1019-1025, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674488

RESUMEN

In vivo models that recapitulate human erythropoiesis with persistence of circulating red blood cells (RBCs) have remained elusive. We report an immunodeficient murine model in which combined human liver and cytokine humanization confer enhanced human erythropoiesis and RBC survival in the circulation. We deleted the fumarylacetoacetate hydrolase (Fah) gene in MISTRG mice expressing several human cytokines in place of their murine counterparts. Liver humanization by intrasplenic injection of human hepatocytes (huHep) eliminated murine complement C3 and reduced murine Kupffer cell density. Engraftment of human sickle cell disease (SCD)-derived hematopoietic stem cells in huHepMISTRGFah -/- mice resulted in vaso-occlusion that replicated acute SCD pathology. Combined liver-cytokine-humanized mice will facilitate the study of diseases afflicting RBCs, including bone marrow failure, hemoglobinopathies, and malaria, and also preclinical testing of therapies.


Asunto(s)
Anemia de Células Falciformes/sangre , Circulación Sanguínea , Modelos Animales de Enfermedad , Eritrocitos/citología , Eritropoyesis/fisiología , Ratones , Animales , Citocinas/metabolismo , Eritropoyesis/genética , Femenino , Eliminación de Gen , Células Madre Hematopoyéticas/citología , Humanos , Hidrolasas/genética , Hígado/fisiología , Ratones Mutantes , Persona de Mediana Edad
16.
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
17.
Nat Rev Cardiol ; 18(3): 194-209, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33214651

RESUMEN

The core pathology of coronavirus disease 2019 (COVID-19) is infection of airway cells by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that results in excessive inflammation and respiratory disease, with cytokine storm and acute respiratory distress syndrome implicated in the most severe cases. Thrombotic complications are a major cause of morbidity and mortality in patients with COVID-19. Patients with pre-existing cardiovascular disease and/or traditional cardiovascular risk factors, including obesity, diabetes mellitus, hypertension and advanced age, are at the highest risk of death from COVID-19. In this Review, we summarize new lines of evidence that point to both platelet and endothelial dysfunction as essential components of COVID-19 pathology and describe the mechanisms that might account for the contribution of cardiovascular risk factors to the most severe outcomes in COVID-19. We highlight the distinct contributions of coagulopathy, thrombocytopathy and endotheliopathy to the pathogenesis of COVID-19 and discuss potential therapeutic strategies in the management of patients with COVD-19. Harnessing the expertise of the biomedical and clinical communities is imperative to expand the available therapeutics beyond anticoagulants and to target both thrombocytopathy and endotheliopathy. Only with such collaborative efforts can we better prepare for further waves and for future coronavirus-related pandemics.


Asunto(s)
Trastornos de la Coagulación Sanguínea/sangre , Trastornos de las Plaquetas Sanguíneas/sangre , COVID-19/sangre , Endotelio Vascular/fisiopatología , Inflamación/sangre , Trombosis/sangre , Administración por Inhalación , Anticoagulantes/uso terapéutico , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/fisiopatología , Trastornos de las Plaquetas Sanguíneas/tratamiento farmacológico , Trastornos de las Plaquetas Sanguíneas/etiología , Trastornos de las Plaquetas Sanguíneas/fisiopatología , COVID-19/complicaciones , COVID-19/fisiopatología , Factores Relajantes Endotelio-Dependientes/uso terapéutico , Epoprostenol/uso terapéutico , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Iloprost/uso terapéutico , Inflamación/etiología , Inflamación/fisiopatología , Óxido Nítrico/uso terapéutico , Inhibidores de Agregación Plaquetaria/uso terapéutico , SARS-CoV-2 , Síndrome de Respuesta Inflamatoria Sistémica/sangre , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Trombosis/etiología , Trombosis/inmunología , Microangiopatías Trombóticas/sangre , Microangiopatías Trombóticas/tratamiento farmacológico , Microangiopatías Trombóticas/etiología , Microangiopatías Trombóticas/fisiopatología , Enfermedades Vasculares/sangre , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/etiología , Enfermedades Vasculares/fisiopatología , Vasodilatadores/uso terapéutico , Tromboembolia Venosa/sangre , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/etiología , Tromboembolia Venosa/fisiopatología , Tratamiento Farmacológico de COVID-19
18.
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
19.
Health Secur ; 18(5): 392-402, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33107763

RESUMEN

During an influenza pandemic, healthcare facilities are likely to be filled to capacity, leading to delays in seeing a provider and obtaining treatment. Flu on Call is a collaborative effort between the US Centers for Disease Control and Prevention and partners to develop a toll-free telephone helpline to reduce the burden on healthcare facilities and improve access to antivirals for people who are ill during an influenza pandemic. This study tested the feasibility of Flu on Call during a 1-day simulation using a severe pandemic scenario. Trained volunteer actors placed calls to the helpline using prepared scripts that were precoded for an expected outcome ("disposition") of the call. Scripts represented callers who were ill, those calling for someone else who was ill, and callers who were only seeking information. Information specialists and medical professionals managed the calls. Results demonstrated that Flu on Call may effectively assist callers during a pandemic, increase access to antiviral prescriptions, and direct patients to the appropriate level of care. Overall, 84% of calls exactly matched the expected call disposition; few calls (2%) were undermanaged (eg, the caller was ill but not transferred to a medical professional or received advice from the medical professional that was less intensive than what was warranted). Callers indicated a high level of satisfaction (83% reported their needs were met). Because of the high volume of calls that may be received during a severe pandemic, the Flu on Call platform should evolve to include additional triage channels (eg, through internet, chat, and/or text access).


Asunto(s)
Gripe Humana/epidemiología , Pandemias , Teléfono , Centers for Disease Control and Prevention, U.S. , Humanos , Triaje/métodos , Estados Unidos
20.
Endocrinology ; 161(10)2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32810225

RESUMEN

Differentiation of pluripotent stem cells into functional parathyroid-like cells would accelerate development of important therapeutic options for subjects with parathyroid-related disorders, from the design and screening of novel pharmaceutical agents to the development of durable cellular therapies. We have established a highly reproducible directed differentiation approach leading to PTH-expressing cells from human embryonic stem cells and induced pluripotent stem cells. We accomplished this through the comparison of multiple different basal media, the inclusion of the CDK inhibitor PD0332991 in both definitive endoderm and anterior foregut endoderm stages, and a 2-stage pharyngeal endoderm series. This is the first protocol to reproducibly establish PTH-expressing cells from human pluripotent stem cells and represents a first step toward the development of functional parathyroid cells with broad applicability for medicinal and scientific investigation.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias Humanas/fisiología , Glándulas Paratiroides/embriología , Hormona Paratiroidea/genética , Células Madre Pluripotentes/fisiología , Células Cultivadas , Endodermo/citología , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Organogénesis/genética , Glándulas Paratiroides/citología , Hormona Paratiroidea/metabolismo , Células Madre Pluripotentes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA