Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Nat Commun ; 14(1): 7216, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37940670

RESUMEN

Single cell spatial interrogation of the immune-structural interactions in COVID -19 lungs is challenging, mainly because of the marked cellular infiltrate and architecturally distorted microstructure. To address this, we develop a suite of mathematical tools to search for statistically significant co-locations amongst immune and structural cells identified using 37-plex imaging mass cytometry. This unbiased method reveals a cellular map interleaved with an inflammatory network of immature neutrophils, cytotoxic CD8 T cells, megakaryocytes and monocytes co-located with regenerating alveolar progenitors and endothelium. Of note, a highly active cluster of immature neutrophils and CD8 T cells, is found spatially linked with alveolar progenitor cells, and temporally with the diffuse alveolar damage stage. These findings offer further insights into how immune cells interact in the lungs of severe COVID-19 disease. We provide our pipeline [Spatial Omics Oxford Pipeline (SpOOx)] and visual-analytical tool, Multi-Dimensional Viewer (MDV) software, as a resource for spatial analysis.


Asunto(s)
COVID-19 , Neutrófilos , Humanos , Linfocitos T CD8-positivos , Pulmón , Linfocitos T Citotóxicos
2.
Reprod Fertil ; 3(3): 198-206, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36001349

RESUMEN

Abstract: Mitochondrial quality is implicated as a contributor to declining fertility with aging. We investigated mitochondrial transcripts in oocytes and their associated cumulus cells from mice of different ages using RNA-seq. Mice aged 3 weeks, 9 weeks, and 1 year were superovulated, and 48 h later, oocyte cumulus complexes were collected by follicle puncture. We did not detect any major differences that could be attributed to aging. However, mitochondrial RNA transcripts which deviated from the consensus sequence were found at a higher frequency in cumulus cells than in their corresponding oocyte. Previous investigations have shown that variation in the sequence of mtRNA transcripts is substantial, and at least some of this can be accounted for by post-transcriptional modifications which impact base calling during sequencing. Our data would be consistent with either less post-transcriptional modification in mitochondrial RNA from oocytes than cumulus cells or with lower mtDNA mutational load. Lay summary: Women become less fertile as they age. Shortage of energy contributes to this, caused by a decline in the quality of mitochondria (the powerhouses of the cell) in the egg. Genes are the blueprint for the cell. They are made of DNA which is copied into an RNA message, or instructions, for making proteins. We counted differences in the RNA message of developing eggs and the cells that support them during development (cumulus cells). We compared the number of these differences in mice of different ages. These age groups represent mice had not reached puberty, those of prime reproductive age, and old mothers. We did not find any differences linked to the age of the mice. However, we did find differences between the egg and the cumulus cells. In most cases, there were lower levels of mutations in eggs than there were in cumulus cells.


Asunto(s)
Oocitos , Folículo Ovárico , Femenino , Animales , Ratones , ARN Mitocondrial/genética , ARN Mitocondrial/metabolismo , Oocitos/metabolismo , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , ARN/genética , ARN/metabolismo
3.
Nat Commun ; 13(1): 3485, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35710802

RESUMEN

The chromatin remodeller ATRX interacts with the histone chaperone DAXX to deposit the histone variant H3.3 at sites of nucleosome turnover. ATRX is known to bind repetitive, heterochromatic regions of the genome including telomeres, ribosomal DNA and pericentric repeats, many of which are putative G-quadruplex forming sequences (PQS). At these sites ATRX plays an ancillary role in a wide range of nuclear processes facilitating replication, chromatin modification and transcription. Here, using an improved protocol for chromatin immunoprecipitation, we show that ATRX also binds active regulatory elements in euchromatin. Mutations in ATRX lead to perturbation of gene expression associated with a reduction in chromatin accessibility, histone modification, transcription factor binding and deposition of H3.3 at the sequences to which it normally binds. In erythroid cells where downregulation of α-globin expression is a hallmark of ATR-X syndrome, perturbation of chromatin accessibility and gene expression occurs in only a subset of cells. The stochastic nature of this process suggests that ATRX acts as a general facilitator of cell specific transcriptional and epigenetic programmes, both in heterochromatin and euchromatin.


Asunto(s)
Cromatina , Heterocromatina , ADN Helicasas/genética , ADN Helicasas/metabolismo , Eucromatina/genética , Heterocromatina/genética , Histonas/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína Nuclear Ligada al Cromosoma X/metabolismo , Talasemia alfa
4.
Sci Immunol ; 6(59)2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-34021026

RESUMEN

Cutaneous group 2 innate lymphoid cells (ILC2) are spatially and epigenetically poised to respond to barrier compromise and associated immunological threats. ILC2, lacking rearranged antigen-specific receptors, are primarily activated by damage-associated cytokines and respond with type 2 cytokine production. To investigate ILC2 potential for direct sensing of skin pathogens and allergens, we performed RNA sequencing of ILC2 derived from in vivo challenged human skin or blood. We detected expression of NOD2 and TLR2 by skin and blood ILC2. Stimulation of ILC2 with TLR2 agonist alone not only induced interleukin-5 (IL-5) and IL-13 expression but also elicited IL-6 expression in combination with Staphylococcus aureus muramyl dipeptide (MDP). Heat-killed skin-resident bacteria provoked an IL-6 profile in ILC2 in vitro that was notably impaired in ILC2 derived from patients with nucleotide-binding oligomerization domain-containing protein 2 (NOD2) mutations. In addition, we show that NOD2 signaling can stimulate autophagy in ILC2, which was also impaired in patients with NOD2 mutations. Here, we have identified a role for ILC2 NOD2 signaling in the differential regulation of ILC2-derived IL-6 and have reported a previously unrecognized pathway of direct ILC2 bacterial sensing.


Asunto(s)
Citocinas/inmunología , Linfocitos/inmunología , Proteína Adaptadora de Señalización NOD2/inmunología , Infecciones Estafilocócicas/inmunología , Adulto , Alérgenos/inmunología , Antígenos Dermatofagoides/inmunología , Humanos , Inmunidad Innata , Mutación , Proteína Adaptadora de Señalización NOD2/genética , Piel/inmunología , Piel/microbiología , Staphylococcus aureus , Receptor Toll-Like 2/inmunología
5.
Front Immunol ; 12: 623430, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746960

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is the most severe form of chronic lung fibrosis. Circulating monocytes have been implicated in immune pathology in IPF but their phenotype is unknown. In this work, we determined the immune phenotype of monocytes in IPF using multi-colour flow cytometry, RNA sequencing and corresponding serum factors, and mapped the main findings to amount of lung fibrosis and single cell transcriptomic landscape of myeloid cells in IPF lungs. We show that monocytes from IPF patients displayed increased expression of CD64 (FcγR1) which correlated with amount of lung fibrosis, and an amplified type I IFN response ex vivo. These were accompanied by markedly raised CSF-1 levels, IL-6, and CCL-2 in serum of IPF patients. Interrogation of single cell transcriptomic data from human IPF lungs revealed increased proportion of CD64hi monocytes and "transitional macrophages" with higher expression of CCL-2 and type I IFN genes. Our study shows that monocytes in IPF patients are phenotypically distinct from age-matched controls, with a primed type I IFN pathway that may contribute to driving chronic inflammation and fibrosis. These findings strengthen the potential role of monocytes in the pathogenesis of IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática/inmunología , Interferón Tipo I/metabolismo , Pulmón/inmunología , Monocitos/inmunología , Estudios de Casos y Controles , Células Cultivadas , Quimiocina CCL2/sangre , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Inmunofenotipificación , Interferón Tipo I/genética , Interleucina-6/sangre , Pulmón/metabolismo , Pulmón/patología , Factor Estimulante de Colonias de Macrófagos/sangre , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/metabolismo , Fenotipo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Análisis de la Célula Individual
6.
Nat Commun ; 12(1): 223, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431820

RESUMEN

Enhancers are DNA sequences that enable complex temporal and tissue-specific regulation of genes in higher eukaryotes. Although it is not entirely clear how enhancer-promoter interactions can increase gene expression, this proximity has been observed in multiple systems at multiple loci and is thought to be essential for the maintenance of gene expression. Bromodomain and Extra-Terminal domain (BET) and Mediator proteins have been shown capable of forming phase condensates and are thought to be essential for super-enhancer function. Here, we show that targeting of cells with inhibitors of BET proteins or pharmacological degradation of BET protein Bromodomain-containing protein 4 (BRD4) has a strong impact on transcription but very little impact on enhancer-promoter interactions. Dissolving phase condensates reduces BRD4 and Mediator binding at enhancers and can also strongly affect gene transcription, without disrupting enhancer-promoter interactions. These results suggest that activation of transcription and maintenance of enhancer-promoter interactions are separable events. Our findings further indicate that enhancer-promoter interactions are not dependent on high levels of BRD4 and Mediator, and are likely maintained by a complex set of factors including additional activator complexes and, at some sites, CTCF and cohesin.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Transcripción Genética , Factor de Unión a CCCTC/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Glicoles/farmacología , Histonas/metabolismo , Humanos , Leucemia/genética , Leucemia/patología , Modelos Genéticos , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética , Transcripción Genética/efectos de los fármacos , Cohesinas
7.
Nat Med ; 26(9): 1480-1490, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32747828

RESUMEN

Colonic antigen-experienced lymphocytes such as tissue-resident memory CD8+ T cells can respond rapidly to repeated antigen exposure. However, their cellular phenotypes and the mechanisms by which they drive immune regulation and inflammation remain unclear. Here we compiled an unbiased atlas of human colonic CD8+ T cells in health and ulcerative colitis (UC) using single-cell transcriptomics with T-cell receptor repertoire analysis and mass cytometry. We reveal extensive heterogeneity in CD8+ T-cell composition, including expanded effector and post-effector terminally differentiated CD8+ T cells. While UC-associated CD8+ effector T cells can trigger tissue destruction and produce tumor necrosis factor (TNF)-α, post-effector cells acquire innate signatures to adopt regulatory functions that may mitigate excessive inflammation. Thus, we identify colonic CD8+ T-cell phenotypes in health and UC, define their clonal relationships and characterize terminally differentiated dysfunctional UC CD8+ T cells expressing IL-26, which attenuate acute colitis in a humanized IL-26 transgenic mouse model.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Colitis Ulcerosa/patología , Interleucinas/metabolismo , Mucosa Intestinal/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Colon/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transcriptoma/genética
8.
Nat Commun ; 10(1): 4533, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31586071

RESUMEN

Multiple myeloma is an incurable, bone marrow-dwelling malignancy that disrupts bone homeostasis causing skeletal damage and pain. Mechanisms underlying myeloma-induced bone destruction are poorly understood and current therapies do not restore lost bone mass. Using transcriptomic profiling of isolated bone lining cell subtypes from a murine myeloma model, we find that bone morphogenetic protein (BMP) signalling is upregulated in stromal progenitor cells. BMP signalling has not previously been reported to be dysregulated in myeloma bone disease. Inhibition of BMP signalling in vivo using either a small molecule BMP receptor antagonist or a solubilized BMPR1a-FC receptor ligand trap prevents trabecular and cortical bone volume loss caused by myeloma, without increasing tumour burden. BMP inhibition directly reduces osteoclastogenesis, increases osteoblasts and bone formation, and suppresses bone marrow sclerostin levels. In summary we describe a novel role for the BMP pathway in myeloma-induced bone disease that can be therapeutically targeted.


Asunto(s)
Enfermedades Óseas/tratamiento farmacológico , Proteínas Morfogenéticas Óseas/metabolismo , Mieloma Múltiple/complicaciones , Pirazoles/farmacología , Pirimidinas/farmacología , Células Madre/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Densidad Ósea/efectos de los fármacos , Enfermedades Óseas/etiología , Enfermedades Óseas/patología , Médula Ósea/patología , Receptores de Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Fémur/citología , Fémur/efectos de los fármacos , Fémur/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos , Mieloma Múltiple/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , RNA-Seq , Transducción de Señal/efectos de los fármacos , Células Madre/patología , Tibia/citología , Tibia/efectos de los fármacos , Tibia/patología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Dev Cell ; 51(2): 255-276.e7, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31639368

RESUMEN

Precise control of developmental processes is encoded in the genome in the form of gene regulatory networks (GRNs). Such multi-factorial systems are difficult to decode in vertebrates owing to their complex gene hierarchies and dynamic molecular interactions. Here we present a genome-wide in vivo reconstruction of the GRN underlying development of the multipotent neural crest (NC) embryonic cell population. By coupling NC-specific epigenomic and transcriptional profiling at population and single-cell levels with genome/epigenome engineering in vivo, we identify multiple regulatory layers governing NC ontogeny, including NC-specific enhancers and super-enhancers, novel trans-factors, and cis-signatures allowing reverse engineering of the NC-GRN at unprecedented resolution. Furthermore, identification and dissection of divergent upstream combinatorial regulatory codes has afforded new insights into opposing gene circuits that define canonical and neural NC fates early during NC ontogeny. Our integrated approach, allowing dissection of cell-type-specific regulatory circuits in vivo, has broad implications for GRN discovery and investigation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Cresta Neural/embriología , Activación Transcripcional/genética , Animales , Heterogeneidad Genética , Vertebrados/genética
10.
Nat Commun ; 10(1): 2803, 2019 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-31243293

RESUMEN

Enhancer elements are a key regulatory feature of many important genes. Several general features including the presence of specific histone modifications are used to demarcate potentially active enhancers. Here we reveal that putative enhancers marked with H3 lysine 79 (H3K79) di or trimethylation (me2/3) (which we name H3K79me2/3 enhancer elements or KEEs) can be found in multiple cell types. Mixed lineage leukemia gene (MLL) rearrangements (MLL-r) such as MLL-AF4 are a major cause of incurable acute lymphoblastic leukemias (ALL). Using the DOT1L inhibitor EPZ-5676 in MLL-AF4 leukemia cells, we show that H3K79me2/3 is required for maintaining chromatin accessibility, histone acetylation and transcription factor binding specifically at KEEs but not non-KEE enhancers. We go on to show that H3K79me2/3 is essential for maintaining enhancer-promoter interactions at a subset of KEEs. Together, these data implicate H3K79me2/3 as having a functional role at a subset of active enhancers in MLL-AF4 leukemia cells.


Asunto(s)
Elementos de Facilitación Genéticos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Metiltransferasas/metabolismo , Bencimidazoles/farmacología , Línea Celular Tumoral , Estudio de Asociación del Genoma Completo , N-Metiltransferasa de Histona-Lisina , Histonas/genética , Humanos , Metilación , Metiltransferasas/genética
11.
Haematologica ; 104(11): 2215-2224, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30975913

RESUMEN

Somatic mutations in acute myeloid leukemia are acquired sequentially and hierarchically. First, pre-leukemic mutations, such as t(8;21) that encodes AML1-ETO, are acquired within the hematopoietic stem cell (HSC) compartment, while signaling pathway mutations, including KRAS activating mutations, are late events acquired during transformation of leukemic progenitor cells and are rarely detectable in HSC. This raises the possibility that signaling pathway mutations are detrimental to clonal expansion of pre-leukemic HSC. To address this hypothesis, we used conditional genetics to introduce Aml1-ETO and K-RasG12D into murine HSC, either individually or in combination. In the absence of activated Ras, Aml1-ETO-expressing HSC conferred a competitive advantage. However, activated K-Ras had a marked detrimental effect on Aml1-ETO-expressing HSC, leading to loss of both phenotypic and functional HSC. Cell cycle analysis revealed a loss of quiescence in HSC co-expressing Aml1-ETO and K-RasG12D, accompanied by an enrichment in E2F and Myc target gene expression and depletion of HSC self-renewal-associated gene expression. These findings provide a mechanistic basis for the observed absence of KRAS signaling mutations in the pre-malignant HSC compartment.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Mutación , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína 1 Compañera de Translocación de RUNX1/genética , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Animales , Proliferación Celular/genética , Expresión Génica , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Transgénicos , Modelos Animales , Modelos Biológicos , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo
12.
Stem Cell Res Ther ; 9(1): 351, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30567594

RESUMEN

BACKGROUND: Human bone marrow-derived mesenchymal stem/stromal cells (hBM MSCs) have multiple functions, critical for skeletal formation and function. Their functional heterogeneity, however, represents a major challenge for their isolation and in developing potency and release assays to predict their functionality prior to transplantation. Additionally, potency, biomarker profiles and defining mechanisms of action in a particular clinical setting are increasing requirements of Regulatory Agencies for release of hBM MSCs as Advanced Therapy Medicinal Products for cellular therapies. Since the healing of bone fractures depends on the coupling of new blood vessel formation with osteogenesis, we hypothesised that a correlation between the osteogenic and vascular supportive potential of individual hBM MSC-derived CFU-F (colony forming unit-fibroblastoid) clones might exist. METHODS: We tested this by assessing the lineage (i.e. adipogenic (A), osteogenic (O) and/or chondrogenic (C)) potential of individual hBM MSC-derived CFU-F clones and determining if their osteogenic (O) potential correlated with their vascular supportive profile in vitro using lineage differentiation assays, endothelial-hBM MSC vascular co-culture assays and transcriptomic (RNAseq) analyses. RESULTS: Our results demonstrate that the majority of CFU-F (95%) possessed tri-lineage, bi-lineage or uni-lineage osteogenic capacity, with 64% of the CFU-F exhibiting tri-lineage AOC potential. We found a correlation between the osteogenic and vascular tubule supportive activity of CFU-F clones, with the strength of this association being donor dependent. RNAseq of individual clones defined gene fingerprints relevant to this correlation. CONCLUSIONS: This study identified a donor-dependent correlation between osteogenic and vascular supportive potential of hBM MSCs and important gene signatures that support these functions that are relevant to their bone regenerative properties.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Osteogénesis/genética , Adulto , Proliferación Celular , Células Cultivadas , Humanos , Adulto Joven
13.
Nat Commun ; 9(1): 5375, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30560907

RESUMEN

During development, it is unclear if lineage-fated cells derive from multilineage-primed progenitors and whether active mechanisms operate to restrict cell fate. Here we investigate how mesoderm specifies into blood-fated cells. We document temporally restricted co-expression of blood (Scl/Tal1), cardiac (Mesp1) and paraxial (Tbx6) lineage-affiliated transcription factors in single cells, at the onset of blood specification, supporting the existence of common progenitors. At the same time-restricted stage, absence of SCL results in expansion of cardiac/paraxial cell populations and increased cardiac/paraxial gene expression, suggesting active suppression of alternative fates. Indeed, SCL normally activates expression of co-repressor ETO2 and Polycomb-PRC1 subunits (RYBP, PCGF5) and maintains levels of Polycomb-associated histone marks (H2AK119ub/H3K27me3). Genome-wide analyses reveal ETO2 and RYBP co-occupy most SCL target genes, including cardiac/paraxial loci. Reduction of Eto2 or Rybp expression mimics Scl-null cardiac phenotype. Therefore, SCL-mediated transcriptional repression prevents mis-specification of blood-fated cells, establishing active repression as central to fate determination processes.


Asunto(s)
Linaje de la Célula/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular , Separación Celular/métodos , Embrión de Mamíferos , Citometría de Flujo/métodos , Código de Histonas/fisiología , Mesodermo/citología , Mesodermo/fisiología , Ratones , Células Madre Embrionarias de Ratones , Proteínas Nucleares/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/genética , Factores de Transcripción/genética
14.
Dev Cell ; 47(5): 608-628.e6, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30513303

RESUMEN

The neural crest (NC) is a transient embryonic stem cell-like population characterized by its multipotency and broad developmental potential. Here, we perform NC-specific transcriptional and epigenomic profiling of foxd3-mutant cells in vivo to define the gene regulatory circuits controlling NC specification. Together with global binding analysis obtained by foxd3 biotin-ChIP and single cell profiles of foxd3-expressing premigratory NC, our analysis shows that, during early steps of NC formation, foxd3 acts globally as a pioneer factor to prime the onset of genes regulating NC specification and migration by re-arranging the chromatin landscape, opening cis-regulatory elements and reshuffling nucleosomes. Strikingly, foxd3 then gradually switches from an activator to its well-described role as a transcriptional repressor and potentially uses differential partners for each role. Taken together, these results demonstrate that foxd3 acts bimodally in the neural crest as a switch from "permissive" to "repressive" nucleosome and chromatin organization to maintain multipotency and define cell fates.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/genética , Cresta Neural/embriología , Pez Cebra , Proteínas de Pez Cebra/genética
15.
Blood ; 132(12): 1225-1240, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-29930011

RESUMEN

SF3B1, SRSF2, and U2AF1 are the most frequently mutated splicing factor genes in the myelodysplastic syndromes (MDS). We have performed a comprehensive and systematic analysis to determine the effect of these commonly mutated splicing factors on pre-mRNA splicing in the bone marrow stem/progenitor cells and in the erythroid and myeloid precursors in splicing factor mutant MDS. Using RNA-seq, we determined the aberrantly spliced genes and dysregulated pathways in CD34+ cells of 84 patients with MDS. Splicing factor mutations result in different alterations in splicing and largely affect different genes, but these converge in common dysregulated pathways and cellular processes, focused on RNA splicing, protein synthesis, and mitochondrial dysfunction, suggesting common mechanisms of action in MDS. Many of these dysregulated pathways and cellular processes can be linked to the known disease pathophysiology associated with splicing factor mutations in MDS, whereas several others have not been previously associated with MDS, such as sirtuin signaling. We identified aberrantly spliced events associated with clinical variables, and isoforms that independently predict survival in MDS and implicate dysregulation of focal adhesion and extracellular exosomes as drivers of poor survival. Aberrantly spliced genes and dysregulated pathways were identified in the MDS-affected lineages in splicing factor mutant MDS. Functional studies demonstrated that knockdown of the mitosis regulators SEPT2 and AKAP8, aberrantly spliced target genes of SF3B1 and SRSF2 mutations, respectively, led to impaired erythroid cell growth and differentiation. This study illuminates the effect of the common spliceosome mutations on the MDS phenotype and provides novel insights into disease pathophysiology.


Asunto(s)
Mutación , Síndromes Mielodisplásicos/genética , Factores de Empalme de ARN/genética , Empalme del ARN , Empalmosomas/genética , Estudios de Cohortes , Reparación del ADN , Regulación de la Expresión Génica , Humanos , Síndromes Mielodisplásicos/epidemiología , Fosfoproteínas/genética , Factores de Empalme Serina-Arginina/genética , Factor de Empalme U2AF/genética , Análisis de Supervivencia
16.
Blood ; 131(15): 1712-1719, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29339402

RESUMEN

Although an essential role for canonical Notch signaling in generation of hematopoietic stem cells in the embryo and in thymic T-cell development is well established, its role in adult bone marrow (BM) myelopoiesis remains unclear. Some studies, analyzing myeloid progenitors in adult mice with inhibited Notch signaling, implicated distinct roles of canonical Notch signaling in regulation of progenitors for the megakaryocyte, erythroid, and granulocyte-macrophage cell lineages. However, these studies might also have targeted other pathways. Therefore, we specifically deleted, in adult BM, the transcription factor recombination signal-binding protein J κ (Rbpj), through which canonical signaling from all Notch receptors converges. Notably, detailed progenitor staging established that canonical Notch signaling is fully dispensable for all investigated stages of megakaryocyte, erythroid, and myeloid progenitors in steady state unperturbed hematopoiesis, after competitive BM transplantation, and in stress-induced erythropoiesis. Moreover, expression of key regulators of these hematopoietic lineages and Notch target genes were unaffected by Rbpj deficiency in BM progenitor cells.


Asunto(s)
Médula Ósea/metabolismo , Eritropoyesis , Mielopoyesis , Receptores Notch/metabolismo , Transducción de Señal , Estrés Fisiológico , Animales , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones , Ratones Transgénicos , Receptores Notch/genética
17.
Nat Immunol ; 19(1): 85-97, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29167569

RESUMEN

The hierarchy of human hemopoietic progenitor cells that produce lymphoid and granulocytic-monocytic (myeloid) lineages is unclear. Multiple progenitor populations produce lymphoid and myeloid cells, but they remain incompletely characterized. Here we demonstrated that lympho-myeloid progenitor populations in cord blood - lymphoid-primed multi-potential progenitors (LMPPs), granulocyte-macrophage progenitors (GMPs) and multi-lymphoid progenitors (MLPs) - were functionally and transcriptionally distinct and heterogeneous at the clonal level, with progenitors of many different functional potentials present. Although most progenitors had the potential to develop into only one mature cell type ('uni-lineage potential'), bi- and rarer multi-lineage progenitors were present among LMPPs, GMPs and MLPs. Those findings, coupled with single-cell expression analyses, suggest that a continuum of progenitors execute lymphoid and myeloid differentiation, rather than only uni-lineage progenitors' being present downstream of stem cells.


Asunto(s)
Diferenciación Celular/genética , Perfilación de la Expresión Génica/métodos , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Mieloides/metabolismo , Análisis de la Célula Individual/métodos , Animales , Linaje de la Célula/genética , Separación Celular/métodos , Células Cultivadas , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Ratones , Trasplante Heterólogo
18.
Sci Immunol ; 2(18)2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29273672

RESUMEN

Group 2 innate lymphoid cells (ILC2) are effectors of barrier immunity, with roles in infection, wound healing, and allergy. A proportion of ILC2 express MHCII (major histocompatibility complex II) and are capable of presenting peptide antigens to T cells and amplifying the subsequent adaptive immune response. Recent studies have highlighted the importance of CD1a-reactive T cells in allergy and infection, activated by the presentation of endogenous neolipid antigens and bacterial components. Using a human skin challenge model, we unexpectedly show that human skin-derived ILC2 can express CD1a and are capable of presenting endogenous antigens to T cells. CD1a expression is up-regulated by TSLP (thymic stromal lymphopoietin) at levels observed in the skin of patients with atopic dermatitis, and the response is dependent on PLA2G4A. Furthermore, this pathway is used to sense Staphylococcus aureus by promoting Toll-like receptor-dependent CD1a-reactive T cell responses to endogenous ligands. These findings define a previously unrecognized role for ILC2 in lipid surveillance and identify shared pathways of CD1a- and PLA2G4A-dependent ILC2 inflammation amenable to therapeutic intervention.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos CD1/genética , Hipersensibilidad , Inmunidad Innata , Linfocitos/inmunología , Adulto , Antígenos CD1/inmunología , Biopsia , Citocinas/genética , Citocinas/inmunología , Dermatitis Atópica/inmunología , Femenino , Fosfolipasas A2 Grupo IV/genética , Fosfolipasas A2 Grupo IV/inmunología , Experimentación Humana , Humanos , Inflamación/inmunología , Lípidos/inmunología , Masculino , Transducción de Señal/inmunología , Piel/citología , Piel/inmunología , Piel/patología , Staphylococcus aureus/inmunología , Linfocitos T/inmunología , Receptores Toll-Like/inmunología , Linfopoyetina del Estroma Tímico
20.
Nat Commun ; 8(1): 403, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28864822

RESUMEN

Hepcidin regulates systemic iron homeostasis. Suppression of hepcidin expression occurs physiologically in iron deficiency and increased erythropoiesis but is pathologic in thalassemia and hemochromatosis. Here we show that epigenetic events govern hepcidin expression. Erythropoiesis and iron deficiency suppress hepcidin via erythroferrone-dependent and -independent mechanisms, respectively, in vivo, but both involve reversible loss of H3K9ac and H3K4me3 at the hepcidin locus. In vitro, pan-histone deacetylase inhibition elevates hepcidin expression, and in vivo maintains H3K9ac at hepcidin-associated chromatin and abrogates hepcidin suppression by erythropoietin, iron deficiency, thalassemia, and hemochromatosis. Histone deacetylase 3 and its cofactor NCOR1 regulate hepcidin; histone deacetylase 3 binds chromatin at the hepcidin locus, and histone deacetylase 3 knockdown counteracts hepcidin suppression induced either by erythroferrone or by inhibiting bone morphogenetic protein signaling. In iron deficient mice, the histone deacetylase 3 inhibitor RGFP966 increases hepcidin, and RNA sequencing confirms hepcidin is one of the genes most differentially regulated by this drug in vivo. We conclude that suppression of hepcidin expression involves epigenetic regulation by histone deacetylase 3.Hepcidin controls systemic iron levels by inhibiting intestinal iron absorption and iron recycling. Here, Pasricha et al. demonstrate that the hepcidin-chromatin locus displays HDAC3-mediated reversible epigenetic modifications during both erythropoiesis and iron deficiency.


Asunto(s)
Regulación de la Expresión Génica , Hepcidinas/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Acetilación , Secuencias de Aminoácidos , Animales , Epigénesis Genética , Eritropoyetina/genética , Eritropoyetina/metabolismo , Hepcidinas/metabolismo , Histona Desacetilasas/genética , Histonas/química , Humanos , Deficiencias de Hierro , Masculino , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...