Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33500353

RESUMEN

Neuroblastoma is the most common extracranial solid tumor and accounts for ∼10% of pediatric cancer-related deaths. The exact cell of origin has yet to be elucidated, but it is generally accepted that neuroblastoma derives from the neural crest and should thus be considered an embryonal malignancy. About 50% of primary neuroblastoma tumors arise in the adrenal gland. Here, we present an atlas of the developing mouse adrenal gland at a single-cell level. Five main cell cluster groups (medulla, cortex, endothelial, stroma, and immune) make up the mouse adrenal gland during fetal development. The medulla group, which is of neural crest origin, is further divided into seven clusters. Of interest is the Schwann cell precursor ("SCP") and the "neuroblast" cluster, a highly cycling cluster that shares markers with sympathoblasts. The signature of the medullary SCP cluster differentiates neuroblastoma patients based on disease phenotype: The SCP signature score anticorrelates with ALK and MYCN expression, two indicators of poor prognosis. Furthermore, a high SCP signature score is associated with better overall survival rates. This study provides an insight into the developing adrenal gland and introduces the SCP gene signature as being of interest for further research in understanding neuroblastoma phenotype.


Asunto(s)
Glándulas Suprarrenales/patología , Neuroblastoma/patología , Células de Schwann/patología , Análisis de la Célula Individual , Médula Suprarrenal/patología , Animales , Agregación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Estadificación de Neoplasias , Células-Madre Neurales , Neuroblastoma/genética , Fenotipo
2.
Dev Dyn ; 250(11): 1568-1583, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33848015

RESUMEN

BACKGROUND: Nephron progenitor cells (NPCs) undergo a stepwise process to generate all mature nephron structures. Mesenchymal to epithelial transition (MET) is considered a multistep process of NPC differentiation to ensure progressive establishment of new nephrons. However, despite this important role, to date, no marker for NPCs undergoing MET in the nephron exists. RESULTS: Here, we identify LGR6 as a NPC marker, expressed in very early cap mesenchyme, pre-tubular aggregates, renal vesicles, and in segments of S-shaped bodies, following the trajectory of MET. By using a lineage tracing approach in embryonic explants in combination with confocal imaging and single-cell RNA sequencing, we provide evidence for the multiple fates of LGR6+ cells during embryonic nephrogenesis. Moreover, by using long-term in vivo lineage tracing, we show that postnatal LGR6+ cells are capable of generating the multiple lineages of the nephrons. CONCLUSIONS: Given the profound early mesenchymal expression and MET signature of LGR6+ cells, together with the lineage tracing of mesenchymal LGR6+ cells, we conclude that LGR6+ cells contribute to all nephrogenic segments by undergoing MET. LGR6+ cells can therefore be considered an early committed NPC population during embryonic and postnatal nephrogenesis with potential regenerative capability.


Asunto(s)
Nefronas , Células Madre , Diferenciación Celular , Mesodermo , Organogénesis/genética
3.
Development ; 145(10)2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773646

RESUMEN

Despite the essential role of the lymphatic vasculature in tissue homeostasis and disease, knowledge of the organ-specific origins of lymphatic endothelial progenitor cells remains limited. The assumption that most murine embryonic lymphatic endothelial cells (LECs) are venous derived has recently been challenged. Here, we show that the embryonic dermal blood capillary plexus constitutes an additional, local source of LECs that contributes to the formation of the dermal lymphatic vascular network. We describe a novel mechanism whereby rare PROX1-positive endothelial cells exit the capillary plexus in a Ccbe1-dependent manner to establish discrete LEC clusters. As development proceeds, these clusters expand and further contribute to the growing lymphatic system. Lineage tracing and analyses of Gata2-deficient mice confirmed that these clusters are endothelial in origin. Furthermore, ectopic expression of Vegfc in the vasculature increased the number of PROX1-positive progenitors within the capillary bed. Our work reveals a novel source of lymphatic endothelial progenitors employed during construction of the dermal lymphatic vasculature and demonstrates that the blood vasculature is likely to remain an ongoing source of LECs during organogenesis, raising the question of whether a similar mechanism operates during pathological lymphangiogenesis.


Asunto(s)
Capilares/citología , Células Endoteliales/citología , Proteínas de Homeodominio/genética , Linfangiogénesis/fisiología , Vasos Linfáticos/embriología , Células Madre/citología , Proteínas Supresoras de Tumor/genética , Animales , Proteínas de Unión al Calcio/genética , Factor de Transcripción GATA2/genética , Linfangiogénesis/genética , Vasos Linfáticos/citología , Ratones , Ratones Transgénicos , Factor C de Crecimiento Endotelial Vascular/biosíntesis , Factor C de Crecimiento Endotelial Vascular/genética
4.
Development ; 142(15): 2719-24, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26243871

RESUMEN

Endothelial-to-hematopoietic transition (EHT) occurs within a population of hemogenic endothelial cells during embryogenesis, and leads to the formation of the adult hematopoietic system. Currently, the prospective identification of specific endothelial cells that will undergo EHT, and the cellular events enabling this transition, are not known. We set out to define precisely the morphological events of EHT, and to correlate cellular morphology with the expression of the transcription factors RUNX1 and SOX17. A novel strategy was developed to allow for correlation of immunofluorescence data with the ultrastructural resolution of scanning electron microscopy. The approach can identify single endothelial cells undergoing EHT, as identified by the ratio of RUNX1 to SOX17 immunofluorescence levels, and the morphological changes associated with the transition. Furthermore, this work details a new technical resource that is widely applicable for correlative analyses of single cells in their native tissue environments.


Asunto(s)
Diferenciación Celular/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Hemangioblastos/citología , Sistema Hematopoyético/embriología , Factores de Transcripción SOXF/metabolismo , Análisis de la Célula Individual/métodos , Animales , Técnica del Anticuerpo Fluorescente , Sistema Hematopoyético/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Microscopía Electrónica de Rastreo , Microscopía Fluorescente
5.
Circ Res ; 116(10): 1660-9, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25814692

RESUMEN

RATIONALE: Collagen- and calcium-binding EGF domain-containing protein 1 (CCBE1) is essential for lymphangiogenesis in vertebrates and has been associated with Hennekam syndrome. Recently, CCBE1 has emerged as a crucial regulator of vascular endothelial growth factor-C (VEGFC) signaling. OBJECTIVE: CCBE1 is a secreted protein characterized by 2 EGF domains and 2 collagen repeats. The functional role of the different CCBE1 protein domains is completely unknown. Here, we analyzed the functional role of the different CCBE1 domains in vivo and in vitro. METHODS AND RESULTS: We analyzed the functionality of several CCBE1 deletion mutants by generating knock-in mice expressing these mutants, by analyzing their ability to enhance Vegfc signaling in vivo in zebrafish, and by testing their ability to induce VEGFC processing in vitro. We found that deleting the collagen domains of CCBE1 has a much stronger effect on CCBE1 activity than deleting the EGF domains. First, although CCBE1ΔCollagen mice fully phenocopy CCBE1 knock-out mice, CCBE1ΔEGF knock-in embryos still form rudimentary lymphatics. Second, Ccbe1ΔEGF, but not Ccbe1ΔCollagen, could partially substitute for Ccbe1 to enhance Vegfc signaling in zebrafish. Third, CCBE1ΔEGF, similarly to CCBE1, but not CCBE1ΔCollagen could activate VEGFC processing in vitro. Furthermore, a Hennekam syndrome mutation within the collagen domain has a stronger effect than a Hennekam syndrome mutation within the EGF domain. CONCLUSIONS: We propose that the collagen domains of CCBE1 are crucial for the activation of VEGFC in vitro and in vivo. The EGF domains of CCBE1 are dispensable for regulation of VEGFC processing in vitro, however, they are necessary for full lymphangiogenic activity of CCBE1 in vivo.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Vasos Linfáticos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Sitios de Unión , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Colágeno/metabolismo , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Enfermedades de los Genitales Masculinos/genética , Enfermedades de los Genitales Masculinos/metabolismo , Genotipo , Edad Gestacional , Células HEK293 , Humanos , Linfangiectasia Intestinal/genética , Linfangiectasia Intestinal/metabolismo , Vasos Linfáticos/embriología , Linfedema/genética , Linfedema/metabolismo , Ratones , Ratones Transgénicos , Mutación , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Transfección , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
6.
Development ; 137(16): 2653-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20610484

RESUMEN

The endothelial cells of the vertebrate lymphatic system assemble into complex networks, but local cues that guide the migration of this distinct set of cells are currently unknown. As a model for lymphatic patterning, we have studied the simple vascular network of the zebrafish trunk consisting of three types of lymphatic vessels that develop in close connection with the blood vasculature. We have generated transgenic lines that allow us to distinguish between arterial, venous and lymphatic endothelial cells (LECs) within a single zebrafish embryo. We found that LECs migrate exclusively along arteries in a manner that suggests that arterial endothelial cells serve as the LEC migratory substrate. In the absence of intersegmental arteries, LEC migration in the trunk is blocked. Our data therefore demonstrate a crucial role for arteries in LEC guidance.


Asunto(s)
Tipificación del Cuerpo , Células Endoteliales/citología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Arterias/citología , Movimiento Celular
7.
Circ Res ; 109(5): 486-91, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21778431

RESUMEN

RATIONALE: Collagen- and calcium-binding EGF domains 1 (CCBE1) has been associated with Hennekam syndrome, in which patients have lymphedema, lymphangiectasias, and other cardiovascular anomalies. Insight into the molecular role of CCBE1 is completely lacking, and mouse models for the disease do not exist. OBJECTIVE: CCBE1 deficient mice were generated to understand the function of CCBE1 in cardiovascular development, and CCBE1 recombinant protein was used in both in vivo and in vitro settings to gain insight into the molecular function of CCBE1. METHODS AND RESULTS: Phenotypic analysis of murine Ccbe1 mutant embryos showed a complete lack of definitive lymphatic structures, even though Prox1(+) lymphatic endothelial cells get specified within the cardinal vein. Mutant mice die prenatally. Proximity ligation assays indicate that vascular endothelial growth factor receptor 3 activation appears unaltered in mutants. Human CCBE1 protein binds to components of the extracellular matrix in vitro, and CCBE1 protein strongly enhances vascular endothelial growth factor-C-mediated lymphangiogenesis in a corneal micropocket assay. CONCLUSIONS: Our data identify CCBE1 as a factor critically required for budding and migration of Prox-1(+) lymphatic endothelial cells from the cardinal vein. CCBE1 probably exerts these effects through binding to components of the extracellular matrix. CCBE1 has little lymphangiogenic effect on its own but dramatically enhances the lymphangiogenic effect of vascular endothelial growth factor-C in vivo. Thus, our data suggest CCBE1 to be essential but not sufficient for lymphangiogenesis.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Endotelio Linfático/irrigación sanguínea , Endotelio Linfático/metabolismo , Linfangiogénesis/fisiología , Vasos Linfáticos/embriología , Vasos Linfáticos/metabolismo , Proteínas Supresoras de Tumor/fisiología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Córnea/irrigación sanguínea , Córnea/citología , Córnea/metabolismo , Endotelio Linfático/citología , Humanos , Linfangiogénesis/genética , Ratones , Ratones Noqueados , Unión Proteica/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/fisiología
8.
Circ Res ; 109(4): 382-95, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21700929

RESUMEN

RATIONALE: Neovascularization is required for embryonic development and plays a central role in diseases in adults. In atherosclerosis, the role of neovascularization remains to be elucidated. In a genome-wide microarray-screen of Flk1+ angioblasts during murine embryogenesis, the v-ets erythroblastosis virus E26 oncogene homolog 2 (Ets2) transcription factor was identified as a potential angiogenic factor. OBJECTIVES: We assessed the role of Ets2 in endothelial cells during atherosclerotic lesion progression toward plaque instability. METHODS AND RESULTS: In 91 patients treated for carotid artery disease, Ets2 levels showed modest correlations with capillary growth, thrombogenicity, and rising levels of tumor necrosis factor-α (TNFα), monocyte chemoattractant protein 1, and interleukin-6 in the atherosclerotic lesions. Experiments in ApoE(-/-) mice, using a vulnerable plaque model, showed that Ets2 expression was increased under atherogenic conditions and was augmented specifically in the vulnerable versus stable lesions. In endothelial cell cultures, Ets2 expression and activation was responsive to the atherogenic cytokine TNFα. In the murine vulnerable plaque model, overexpression of Ets2 promoted lesion growth with neovessel formation, hemorrhaging, and plaque destabilization. In contrast, Ets2 silencing, using a lentiviral shRNA construct, promoted lesion stabilization. In vitro studies showed that Ets2 was crucial for TNFα-induced expression of monocyte chemoattractant protein 1, interleukin-6, and vascular cell adhesion molecule 1 in endothelial cells. In addition, Ets2 promoted tube formation and amplified TNFα-induced loss of vascular endothelial integrity. Evaluation in a murine retina model further validated the role of Ets2 in regulating vessel inflammation and endothelial leakage. CONCLUSIONS: We provide the first evidence for the plaque-destabilizing role of Ets2 in atherosclerosis development by induction of an intraplaque proinflammatory phenotype in endothelial cells.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Células Endoteliales/metabolismo , Inflamación/metabolismo , Proteína Proto-Oncogénica c-ets-2/metabolismo , Análisis de Varianza , Animales , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/fisiopatología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/fisiopatología , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Hemorragia/metabolismo , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica , Fenotipo , Proteína Proto-Oncogénica c-ets-2/genética , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Rotura , Factores de Tiempo , Transfección , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/metabolismo
9.
Nat Biotechnol ; 39(10): 1239-1245, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34083793

RESUMEN

Despite advances in three-dimensional (3D) imaging, it remains challenging to profile all the cells within a large 3D tissue, including the morphology and organization of the many cell types present. Here, we introduce eight-color, multispectral, large-scale single-cell resolution 3D (mLSR-3D) imaging and image analysis software for the parallelized, deep learning-based segmentation of large numbers of single cells in tissues, called segmentation analysis by parallelization of 3D datasets (STAPL-3D). Applying the method to pediatric Wilms tumor, we extract molecular, spatial and morphological features of millions of cells and reconstruct the tumor's spatio-phenotypic patterning. In situ population profiling and pseudotime ordering reveals a highly disorganized spatial pattern in Wilms tumor compared to healthy fetal kidney, yet cellular profiles closely resembling human fetal kidney cells could be observed. In addition, we identify previously unreported tumor-specific populations, uniquely characterized by their spatial embedding or morphological attributes. Our results demonstrate the use of combining mLSR-3D and STAPL-3D to generate a comprehensive cellular map of human tumors.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Imagenología Tridimensional/métodos , Neoplasias/diagnóstico por imagen , Biomarcadores de Tumor/metabolismo , Aprendizaje Profundo , Colorantes Fluorescentes , Humanos , Riñón/diagnóstico por imagen , Neoplasias/metabolismo , Neoplasias/patología , Fenotipo , Programas Informáticos
10.
Transl Oncol ; 14(5): 101048, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33667892

RESUMEN

Pediatric MLL-rearranged acute myeloid leukemia (AML) has a generally unfavorable outcome, primarily due to relapse and drug resistance. To overcome these difficulties, new therapeutic agents are urgently needed. Yet, implementing novel drugs for clinical use is a time-consuming, laborious, costly and high-risk process. Therefore, we applied a drug-repositioning strategy by screening drug libraries, comprised of >4000 compounds that are mostly FDA-approved, in a high-throughput format on primary MLL-rearranged AML cells. Here we identified pyrvinium pamoate (pyrvinium) as a novel candidate drug effective against MLL-rearranged AML, eliminating all cell viability at <1000 nM. Additional screening of identified drug hits on non-leukemic bone marrow samples, resulted in a decrease in cell viability of ∼50% at 1000 nM pyrvinium, suggesting a therapeutic window for targeting leukemic cells specifically. Validation of pyrvinium on an extensive panel of AML cell lines and primary AML samples showed comparable viabilities as the drug screen data, with pyrvinium achieving IC50 values of <80 nM in these samples. Remarkably, pyrvinium also induced cell toxicity in primary MLL-AF10+ AML cells, an MLL-rearrangement associated with a poor outcome. While pyrvinium is able to inhibit the Wnt pathway in other diseases, this unlikely explains the efficacy we observed as ß-catenin was not expressed in the AML cells tested. Rather, we show that pyrvinium co-localized with the mitochondrial stain in cells, and hence may act by inhibiting mitochondrial respiration. Overall, this study shows that pyrvinium is highly effective against MLL-rearranged AML in vitro, and therefore represents a novel potential candidate for further studies in MLL-rearranged AML.

11.
ACS Nano ; 12(3): 2138-2150, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29320626

RESUMEN

Up to 99% of systemically administered nanoparticles are cleared through the liver. Within the liver, most nanoparticles are thought to be sequestered by macrophages (Kupffer cells), although significant nanoparticle interactions with other hepatic cells have also been observed. To achieve effective cell-specific targeting of drugs through nanoparticle encapsulation, improved mechanistic understanding of nanoparticle-liver interactions is required. Here, we show the caudal vein of the embryonic zebrafish ( Danio rerio) can be used as a model for assessing nanoparticle interactions with mammalian liver sinusoidal (or scavenger) endothelial cells (SECs) and macrophages. We observe that anionic nanoparticles are primarily taken up by SECs and identify an essential requirement for the scavenger receptor, stabilin-2 ( stab2) in this process. Importantly, nanoparticle-SEC interactions can be blocked by dextran sulfate, a competitive inhibitor of stab2 and other scavenger receptors. Finally, we exploit nanoparticle-SEC interactions to demonstrate targeted intracellular drug delivery resulting in the selective deletion of a single blood vessel in the zebrafish embryo. Together, we propose stab2 inhibition or targeting as a general approach for modifying nanoparticle-liver interactions of a wide range of nanomedicines.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Hepatocitos/metabolismo , Macrófagos/metabolismo , Nanopartículas/metabolismo , Receptores Depuradores/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Embrión no Mamífero/metabolismo , Embrión no Mamífero/ultraestructura , Liposomas/análisis , Liposomas/metabolismo , Ratones , Nanopartículas/análisis , Distribución Tisular , Pez Cebra/metabolismo
12.
Nat Commun ; 8(1): 128, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28743859

RESUMEN

Hematopoietic stem and progenitor cells arise from the vascular endothelium of the dorsal aorta and subsequently switch niche to the fetal liver through unknown mechanisms. Here we report that vascular endothelium-specific deletion of mouse Drosha (Drosha cKO), an enzyme essential for microRNA biogenesis, leads to anemia and death. A similar number of hematopoietic stem and progenitor cells emerge from Drosha-deficient and control vascular endothelium, but Drosha cKO-derived hematopoietic stem and progenitor cells accumulate in the dorsal aorta and fail to colonize the fetal liver. Depletion of the let-7 family of microRNAs is a primary cause of this defect, as it leads to activation of leukotriene B4 signaling and induction of the α4ß1 integrin cell adhesion complex in hematopoietic stem and progenitor cells. Inhibition of leukotriene B4 or integrin rescues maturation and migration of Drosha cKO hematopoietic stem and progenitor cells to the fetal liver, while it hampers hematopoiesis in wild-type animals. Our study uncovers a previously undefined role of innate leukotriene B4 signaling as a gatekeeper of the hematopoietic niche transition.Hematopoietic stem and progenitor cells are generated first from the vascular endothelium of the dorsal aorta and then the fetal liver but what regulates this switch is unknown. Here, the authors show that changing miRNA biogenesis and leukotriene B4 signaling in mice modulates this switch in the niche.


Asunto(s)
Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Leucotrieno B4/metabolismo , MicroARNs/genética , Nicho de Células Madre/genética , Animales , Aorta/metabolismo , Endotelio Vascular/metabolismo , Hígado/embriología , Hígado/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Transducción de Señal/genética
13.
Reprod Sci ; 23(9): 1179-92, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26944948

RESUMEN

BACKGROUND: The human placenta is a source of hematopoietic stem and progenitor cells (HSPCs). The RUNX1 transcription factor is required for the formation of functional HSPCs. The impact of preeclampsia (PE) and preterm labor (PTL, spontaneous preterm labor [sPTL] and inflammatory preterm labor [iPTL]) on HSPC localization and RUNX1 expression in the human placenta is unknown. METHODS: We compared the frequency and density of HSPC in control samples from sPTL (n = 6) versus PE (n = 6) and iPTL (n = 6). We examined RUNX1 protein and RNA expression in placentas from normal pregnancies (5-22 weeks, n = 8 total) and in placentas from the aforementioned pregnancy complications (n = 5/group). RESULTS: Hematopoietic stem and progenitor cells were rare cell types, associated predominantly with the vasculature of placental villi. The HSPC density was greater in the chorionic plate (CP) compared to the villi (P < .001) and greater in PE and iPTL samples as compared to controls within the CP (not significant) and overall (P < .05). During the fetal period, RUNX1 was expressed in the mesenchyme of the CP and villi. Inflammatory PTL samples were more likely to exhibit intraluminal RUNX1(+) cell populations (P < .001) and RUNX1(+) cell clusters attached to arterial endothelial cells. CONCLUSION: Placental HSPCs likely arise from hematopoietic niches comprised RUNX1(+) mesenchyme and vascular endothelium. Pregnancy complications that result in preterm birth differentially affect placental HSPC localization and RUNX1 expression. Our results support previous findings that inflammation positively regulates hematopoiesis. We present new evidence that hemogenic endothelium may be active at later stages of human fetal development in the context of inflammation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Trabajo de Parto Prematuro/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Recuento de Células , Femenino , Humanos , Inflamación/complicaciones , Inflamación/metabolismo , Trabajo de Parto Prematuro/etiología , Trabajo de Parto Prematuro/patología , Placenta/patología , Preeclampsia/patología , Embarazo , ARN Mensajero/metabolismo
14.
Cardiovasc Res ; 110(1): 129-39, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26822228

RESUMEN

AIMS: Impairment of the endothelial barrier leads to microvascular breakdown in cardiovascular disease and is involved in intraplaque haemorrhaging and the progression of advanced atherosclerotic lesions that are vulnerable to rupture. The exact mechanism that regulates vascular integrity requires further definition. Using a microarray screen for angiogenesis-associated genes during murine embryogenesis, we identified thrombospondin type I domain 1 (THSD1) as a new putative angiopotent factor with unknown biological function. We sought to characterize the role of THSD1 in endothelial cells during vascular development and cardiovascular disease. METHODS AND RESULTS: Functional knockdown of Thsd1 in zebrafish embryos and in a murine retina vascularization model induced severe haemorrhaging without affecting neovascular growth. In human carotid endarterectomy specimens, THSD1 expression by endothelial cells was detected in advanced atherosclerotic lesions with intraplaque haemorrhaging, but was absent in stable lesions, implying involvement of THSD1 in neovascular bleeding. In vitro, stimulation with pro-atherogenic factors (3% O2 and TNFα) decreased THSD1 expression in human endothelial cells, whereas stimulation with an anti-atherogenic factor (IL10) showed opposite effect. Therapeutic evaluation in a murine advanced atherosclerosis model showed that Thsd1 overexpression decreased plaque vulnerability by attenuating intraplaque vascular leakage, subsequently reducing macrophage accumulation and necrotic core size. Mechanistic studies in human endothelial cells demonstrated that THSD1 activates FAK-PI3K, leading to Rac1-mediated actin cytoskeleton regulation of adherens junctions and focal adhesion assembly. CONCLUSION: THSD1 is a new regulator of endothelial barrier function during vascular development and protects intraplaque microvessels against haemorrhaging in advanced atherosclerotic lesions.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Microvasos/metabolismo , Neovascularización Patológica/metabolismo , Trombospondinas/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Placa Aterosclerótica/patología , Trombospondina 1/metabolismo
15.
Nat Commun ; 6: 7739, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26204127

RESUMEN

Changes in cell fate and identity are essential for endothelial-to-haematopoietic transition (EHT), an embryonic process that generates the first adult populations of haematopoietic stem cells (HSCs) from hemogenic endothelial cells. Dissecting EHT regulation is a critical step towards the production of in vitro derived HSCs. Yet, we do not know how distinct endothelial and haematopoietic fates are parsed during the transition. Here we show that genes required for arterial identity function later to repress haematopoietic fate. Tissue-specific, temporally controlled, genetic loss of arterial genes (Sox17 and Notch1) during EHT results in increased production of haematopoietic cells due to loss of Sox17-mediated repression of haematopoietic transcription factors (Runx1 and Gata2). However, the increase in EHT can be abrogated by increased Notch signalling. These findings demonstrate that the endothelial haematopoietic fate switch is actively repressed in a population of endothelial cells, and that derepression of these programs augments haematopoietic output.


Asunto(s)
Vasos Sanguíneos/embriología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Factor de Transcripción GATA2/metabolismo , Proteínas HMGB/fisiología , Hemangioblastos/fisiología , Factores de Transcripción SOXF/fisiología , Animales , Femenino , Genes Reporteros , Hematopoyesis , Ratones , Embarazo , Receptor Notch1/metabolismo
16.
Cancer Res ; 70(12): 4891-900, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20516114

RESUMEN

Genetic mutations in the mitotic regulatory kinase BUBR1 are associated with the cancer-susceptible disorder mosaic variegated aneuploidy (MVA). In patients with biallelic mutations, a missense mutation pairs with a truncating mutation. Here, we show that cell lines derived from MVA patients with biallelic mutations have an impaired mitotic checkpoint, chromosome alignment defects, and low overall BUBR1 abundance. Ectopic expression of BUBR1 restored mitotic checkpoint activity, proving that BUBR1 dysfunction causes chromosome segregation errors in the patients. Combined analysis of patient cells and functional protein replacement shows that all MVA mutations fall in two distinct classes: those that impose specific defects in checkpoint activity or microtubule attachment and those that lower BUBR1 protein abundance. Low protein abundance is the direct result of the absence of transcripts from truncating mutants combined with high protein turnover of missense mutants. In this group of missense mutants, the amino acid change consistently occurs in or near the BUBR1 kinase domain. Our findings provide a molecular explanation for chromosomal instability in patients with biallelic genetic mutations in BUBR1.


Asunto(s)
Anomalías Múltiples/genética , Aneuploidia , Segregación Cromosómica/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Northern Blotting , Citometría de Flujo , Genes cdc/fisiología , Células HeLa , Humanos , Immunoblotting , Mosaicismo , Neoplasias/patología , Plásmidos , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/patología , Síndrome , Transfección , Células Tumorales Cultivadas
17.
Nat Genet ; 41(4): 396-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19287381

RESUMEN

Lymphatic vessels have important roles in fluid homeostasis, fat absorption, inflammation and cancer metastasis and develop in a dynamic process (called lymphangiogenesis) involving budding, migration and proliferation of lymphangioblasts. Using a genetic screen in zebrafish we identify ccbe1 (collagen and calcium-binding EGF domain-1) as indispensible for embryonic lymphangiogenesis. Ccbe1 acts at the same stage of development as Vegfc and is required for lymphangioblast budding and angiogenic sprouting from venous endothelium.


Asunto(s)
Factor de Crecimiento Epidérmico/genética , Linfangiogénesis/genética , Vasos Linfáticos/fisiología , Mutación , Sustitución de Aminoácidos , Animales , Sitios de Unión , Calcio/metabolismo , División Celular , Movimiento Celular , Embrión no Mamífero/fisiología , Desarrollo Embrionario , Humanos , Vasos Linfáticos/citología , Vasos Linfáticos/embriología , Modelos Animales , Factor C de Crecimiento Endotelial Vascular/genética , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA