Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Cell Dev Biol ; 12: 1344070, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38440076

RESUMEN

The lymphatic vasculature regulates lung homeostasis through drainage of fluid and trafficking of immune cells and plays a key role in the response to lung injury in several disease states. We have previously shown that lymphatic dysfunction occurs early in the pathogenesis of chronic obstructive pulmonary disease (COPD) caused by cigarette smoke (CS) and that this is associated with increased thrombin and fibrin clots in lung lymph. However, the direct effects of CS and thrombin on lymphatic endothelial cells (LECs) in COPD are not entirely clear. Studies of the blood vasculature have shown that COPD is associated with increased thrombin after CS exposure that causes endothelial dysfunction characterized by changes in the expression of coagulation factors and leukocyte adhesion proteins. Here, we determined whether similar changes occur in LECs. We used an in vitro cell culture system and treated human lung microvascular lymphatic endothelial cells with cigarette smoke extract (CSE) and/or thrombin. We found that CSE treatment led to decreased fibrinolytic activity in LECs, which was associated with increased expression of plasminogen activator inhibitor 1 (PAI-1). LECs treated with both CSE and thrombin together had a decreased expression of tissue factor pathway inhibitor (TFPI) and increased expression of adhesion molecules. RNA sequencing of lung LECs isolated from mice exposed to CS also showed upregulation of prothrombotic and inflammatory pathways at both acute and chronic exposure time points. Analysis of publicly available single-cell RNA sequencing of LECs as well as immunohistochemical staining of lung tissue from COPD patients supported these data and showed increased expression of inflammatory markers in LECs from COPD patients compared to those from controls. These studies suggest that in parallel with blood vessels, the lymphatic endothelium undergoes inflammatory changes associated with CS exposure and increased thrombin in COPD. Further research is needed to unravel the mechanisms by which these changes affect lymphatic function and drive tissue injury in COPD.

2.
bioRxiv ; 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37961242

RESUMEN

Chronic Obstructive Pulmonary Disease (COPD) is a heterogeneous disease that is characterized by many clinical phenotypes. One such phenotype of COPD is defined by emphysema, pathogenic lung tertiary lymphoid organs (TLOs), and autoantibody production. We have previously shown that lymphatic dysfunction can cause lung TLO formation and lung injury in mice. We now sought to uncover whether underlying lymphatic dysfunction may be a driver of lung injury in cigarette smoke (CS)-induced COPD. We found that lung TLOs in mice with lymphatic dysfunction produce autoantibodies and are associated with a lymphatic endothelial cell subtype that expresses antigen presentation genes. Mice with underlying lymphatic dysfunction develop increased emphysema after CS exposure, with increased size and activation of TLOs. CS further increased autoantibody production in mice with lymphatic dysfunction. B-cell blockade prevented TLO formation and decreased lung injury after CS in mice with lymphatic dysfunction. Using tissue from human COPD patients, we also found evidence of a lymphatic gene signature that was specific to patients with emphysema and prominent TLOs compared to COPD patients without emphysema. Taken together, these data suggest that lymphatic dysfunction may underlie lung injury in a subset of COPD patients with an autoimmune emphysema phenotype.

3.
Cell Stem Cell ; 29(4): 593-609.e7, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35364013

RESUMEN

The liver vascular network is patterned by sinusoidal and hepatocyte co-zonation. How intra-liver vessels acquire their hierarchical specialized functions is unknown. We study heterogeneity of hepatic vascular cells during mouse development through functional and single-cell RNA-sequencing. The acquisition of sinusoidal endothelial cell identity is initiated during early development and completed postnatally, originating from a pool of undifferentiated vascular progenitors at E12. The peri-natal induction of the transcription factor c-Maf is a critical switch for the sinusoidal identity determination. Endothelium-restricted deletion of c-Maf disrupts liver sinusoidal development, aberrantly expands postnatal liver hematopoiesis, promotes excessive postnatal sinusoidal proliferation, and aggravates liver pro-fibrotic sensitivity to chemical insult. Enforced c-Maf overexpression in generic human endothelial cells switches on a liver sinusoidal transcriptional program that maintains hepatocyte function. c-Maf represents an inducible intra-organotypic and niche-responsive molecular determinant of hepatic sinusoidal cell identity and lays the foundation for the strategies for vasculature-driven liver repair.


Asunto(s)
Capilares , Células Endoteliales , Animales , Endotelio , Hígado/patología , Cirrosis Hepática/patología , Regeneración Hepática , Ratones , Proteínas Proto-Oncogénicas c-maf
4.
Nat Cell Biol ; 24(1): 99-111, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34961794

RESUMEN

Histone variants and the associated post-translational modifications that govern the stemness of haematopoietic stem cells (HSCs) and differentiation thereof into progenitors (HSPCs) have not been well defined. H3.3 is a replication-independent H3 histone variant in mammalian systems that is enriched at both H3K4me3- and H3K27me3-marked bivalent genes as well as H3K9me3-marked endogenous retroviral repeats. Here we show that H3.3, but not its chaperone Hira, prevents premature HSC exhaustion and differentiation into granulocyte-macrophage progenitors. H3.3-null HSPCs display reduced expression of stemness and lineage-specific genes with a predominant gain of H3K27me3 marks at their promoter regions. Concomitantly, loss of H3.3 leads to a reduction of H3K9me3 marks at endogenous retroviral repeats, opening up binding sites for the interferon regulatory factor family of transcription factors, allowing the survival of rare, persisting H3.3-null HSCs. We propose a model whereby H3.3 maintains adult HSC stemness by safeguarding the delicate interplay between H3K27me3 and H3K9me3 marks, enforcing chromatin adaptability.


Asunto(s)
Cromatina/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Histonas/metabolismo , Mielopoyesis/fisiología , Animales , Linfocitos T CD8-positivos/citología , Proteínas de Ciclo Celular , Línea Celular , Granulocitos/citología , Hematopoyesis/fisiología , Chaperonas de Histonas , Células Endoteliales de la Vena Umbilical Humana , Humanos , Macrófagos/citología , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/fisiología , Factores de Transcripción
5.
Genes Dev ; 35(21-22): 1475-1489, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34675061

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are generated de novo in the embryo from hemogenic endothelial cells (HECs) via an endothelial-to-hematopoietic transition (EHT) that requires the transcription factor RUNX1. Ectopic expression of RUNX1 alone can efficiently promote EHT and HSPC formation from embryonic endothelial cells (ECs), but less efficiently from fetal or adult ECs. Efficiency correlated with baseline accessibility of TGFß-related genes associated with endothelial-to-mesenchymal transition (EndoMT) and participation of AP-1 and SMAD2/3 to initiate further chromatin remodeling along with RUNX1 at these sites. Activation of TGFß signaling improved the efficiency with which RUNX1 specified fetal ECs as HECs. Thus, the ability of RUNX1 to promote EHT depends on its ability to recruit the TGFß signaling effectors AP-1 and SMAD2/3, which in turn is determined by the changing chromatin landscape in embryonic versus fetal ECs. This work provides insight into regulation of EndoMT and EHT that will guide reprogramming efforts for clinical applications.


Asunto(s)
Hemangioblastos , Diferenciación Celular/genética , Cromatina/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Feto , Hemangioblastos/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1
6.
Commun Biol ; 4(1): 406, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33767339

RESUMEN

Jak3 is the only non-promiscuous member of the Jak family of secondary messengers. Studies to date have focused on understanding and targeting the cell-autonomous role of Jak3 in immunity, while functional Jak3 expression outside the hematopoietic system remains largely unreported. We show that Jak3 is expressed in endothelial cells across hematopoietic and non-hematopoietic organs, with heightened expression in the bone marrow. The bone marrow niche is understood as a network of different cell types that regulate hematopoietic function. We show that the Jak3-/- bone marrow niche is deleterious for the maintenance of long-term repopulating hematopoietic stem cells (LT-HSCs) and that JAK3-overexpressing endothelial cells have increased potential to expand LT-HSCs in vitro. This work may serve to identify a novel function for a highly specific tyrosine kinase in the bone marrow vascular niche and to further characterize the LT-HSC function of sinusoidal endothelium.


Asunto(s)
Células Endoteliales/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Janus Quinasa 3/genética , Animales , Femenino , Técnicas de Sustitución del Gen , Janus Quinasa 3/metabolismo , Masculino , Ratones
7.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33542154

RESUMEN

Cells derived from pluripotent sources in vitro must resemble those found in vivo as closely as possible at both transcriptional and functional levels in order to be a useful tool for studying diseases and developing therapeutics. Recently, differentiation of human pluripotent stem cells (hPSCs) into brain microvascular endothelial cells (ECs) with blood-brain barrier (BBB)-like properties has been reported. These cells have since been used as a robust in vitro BBB model for drug delivery and mechanistic understanding of neurological diseases. However, the precise cellular identity of these induced brain microvascular endothelial cells (iBMECs) has not been well described. Employing a comprehensive transcriptomic metaanalysis of previously published hPSC-derived cells validated by physiological assays, we demonstrate that iBMECs lack functional attributes of ECs since they are deficient in vascular lineage genes while expressing clusters of genes related to the neuroectodermal epithelial lineage (Epi-iBMEC). Overexpression of key endothelial ETS transcription factors (ETV2, ERG, and FLI1) reprograms Epi-iBMECs into authentic endothelial cells that are congruent with bona fide endothelium at both transcriptomic as well as some functional levels. This approach could eventually be used to develop a robust human BBB model in vitro that resembles the human brain EC in vivo for functional studies and drug discovery.


Asunto(s)
Endotelio Vascular/citología , Células Madre Pluripotentes/citología , Factores de Transcripción/genética , Animales , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Encéfalo/citología , Diferenciación Celular , Línea Celular , Reprogramación Celular/fisiología , Endotelio Vascular/fisiología , Expresión Génica , Humanos , Ratones Endogámicos , Células Madre Pluripotentes/fisiología , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
8.
Nat Commun ; 10(1): 5705, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31836710

RESUMEN

Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders.


Asunto(s)
Capilares/crecimiento & desarrollo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glomérulos Renales/irrigación sanguínea , Animales , Capilares/citología , Capilares/metabolismo , Células Cultivadas , Embrión de Mamíferos , Endotelio Vascular/citología , Endotelio Vascular/crecimiento & desarrollo , Factor de Transcripción GATA5/genética , Factor de Transcripción GATA5/metabolismo , Perfilación de la Expresión Génica , Humanos , Glomérulos Renales/crecimiento & desarrollo , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B/metabolismo , Cultivo Primario de Células , RNA-Seq , Análisis de la Célula Individual , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
9.
FEBS Lett ; 593(23): 3253-3265, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31725897

RESUMEN

Haematopoietic stem cells (HSCs) are the only adult stem cells with a demonstrated clinical use, even though a tractable method to maintain and expand human HSCs in vitro has not yet been found. Owing to the introduction of transplantation strategies for the treatment of haematological malignancies and, more recently, the promise of gene therapy, the need to improve the generation, manipulation and scalability of autologous or allogeneic HSCs has risen steeply over the past decade. In that context, reprogramming strategies based on the expression of exogenous transcription factors have emerged as a means to produce functional HSCs in vitro. These approaches largely stem from the assumption that key master transcription factors direct the expression of downstream target genes thereby triggering haematopoiesis. Both somatic and pluripotent cells have been used to this end, yielding variable results in terms of haematopoietic phenotype and functionality. Here, we present an overview of the haematopoietic reprogramming methods reported to date, provide the appropriate historical context and offer some critical insight about where the field stands at present.


Asunto(s)
Reprogramación Celular/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Humanos , Factores de Transcripción/genética
10.
J Transl Med ; 17(1): 194, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31182109

RESUMEN

BACKGROUND: One main challenge in ovarian cancer rests on the presence of a relapse and an important metastatic disease, despite extensive surgical debulking and chemotherapy. The difficulty in containing metastatic cancer is partly due to the heterotypic interaction of tumor and its microenvironment. In this context, evidence suggests that endothelial cells (EC) play an important role in ovarian tumor growth and chemoresistance. Here, we studied the role of tumor endothelium on ovarian cancer cells (OCCs). METHODS: We evaluated the effect of activated endothelial cells on ovarian cancer cell proliferation and resistance to chemotherapy and investigated the survival pathways activated by endothelial co-culture. RESULTS: The co-culture between OCCs and E4+ECs, induced an increase of OCCs proliferation both in vitro and in vivo. This co-culture induced an increase of Notch receptors expression on OCC surface and an increase of Jagged 1 expression on E4+ECs surface and activation of survival pathways leading to chemoresistance by E4+ECs. CONCLUSION: The targeting of aberrant NOTCH signaling could constitute a strategy to disrupt the pro-tumoral endothelial niche.


Asunto(s)
Carcinoma Epitelial de Ovario/patología , Proliferación Celular , Endotelio/patología , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptores Notch/metabolismo , Animales , Carcinoma Epitelial de Ovario/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Resistencia a Antineoplásicos , Endotelio/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Neoplasias Ováricas/metabolismo , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
11.
Nat Protoc ; 13(12): 2758-2780, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30429596

RESUMEN

The ability to generate hematopoietic stem cells (HSCs) in vitro would have an immeasurable impact on many areas of clinical practice, including trauma, cancer, and congenital disease. In this protocol, we describe a stepwise approach that converts adult murine endothelial cells (ECs) to HSCs, termed 'reprogrammed ECs into hematopoietic stem and progenitor cells' (rEC-HSPCs). The conversion, which is achieved without cells transitioning through a pluripotent state, comprises three phases: induction, specification, and expansion. Adult ECs are first isolated from Runx1-IRES-GFP; Rosa26-rtTa mice and maintained in culture under EC growth factor stimulation and Tgfß inhibition. In the first (induction) phase of conversion (days 0-8), four transcription factors (TFs)-FosB, Gfi1, Runx1, and Spi1 (FGRS)-are expressed transiently, which results in endogenous Runx1 expression. During the second (specification) phase (days 8-20), endogenous Runx1+ FGRS-transduced ECs commit to a hematopoietic fate and no longer require exogenous FGRS expression. Finally, the vascular niche drives robust proliferation of rEC-HSPCs during the expansion phase (days 20-28). The resulting converted cells possess a transcriptomic signature and long-term self-renewal capacity indistinguishable from those of adult HSCs. In this protocol, we also describe functional in vitro and in vivo assays that can be used to demonstrate that rEC-HSPCs are competent for clonal engraftment and possess multi-lineage reconstitution potential, including antigen-dependent adaptive immune function. This approach thus provides a tractable strategy for interrogating the generation of engraftable hematopoietic cells, advancing the mechanistic understanding of hematopoietic development and HSC self-renewal.


Asunto(s)
Diferenciación Celular , Técnicas de Reprogramación Celular/métodos , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Animales , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Factores de Transcripción/genética , Transcriptoma
12.
Nat Commun ; 9(1): 2681, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29992946

RESUMEN

GLIS3 mutations are associated with type 1, type 2, and neonatal diabetes, reflecting a key function for this gene in pancreatic ß-cell biology. Previous attempts to recapitulate disease-relevant phenotypes in GLIS3-/- ß-like cells have been unsuccessful. Here, we develop a "minimal component" protocol to generate late-stage pancreatic progenitors (PP2) that differentiate to mono-hormonal glucose-responding ß-like (PP2-ß) cells. Using this differentiation platform, we discover that GLIS3-/- hESCs show impaired differentiation, with significant death of PP2 and PP2-ß cells, without impacting the total endocrine pool. Furthermore, we perform a high-content chemical screen and identify a drug candidate that rescues mutant GLIS3-associated ß-cell death both in vitro and in vivo. Finally, we discovered that loss of GLIS3 causes ß-cell death, by activating the TGFß pathway. This study establishes an optimized directed differentiation protocol for modeling human ß-cell disease and identifies a drug candidate for treating a broad range of GLIS3-associated diabetic patients.


Asunto(s)
Diabetes Mellitus/prevención & control , Descubrimiento de Drogas/métodos , Hipoglucemiantes/farmacología , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Línea Celular , Proteínas de Unión al ADN , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Perfilación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones SCID , Mutación , Pirazoles/farmacología , Quinolinas/farmacología , Proteínas Represoras , Transactivadores , Factores de Transcripción/metabolismo , Trasplante Heterólogo
13.
Mol Cancer ; 17(1): 47, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29455640

RESUMEN

BACKGROUND: Minimal residual disease is the main issue of advanced ovarian cancer treatment. According to the literature and previous results, we hypothesized that Mesenchymal Stromal Cells (MSC) could support this minimal residual disease by protecting ovarian cancer cells (OCC) from chemotherapy. In vitro study confirmed that MSC could induce OCC chemoresistance without contact using transwell setting. Further experiments showed that this induced chemoresistance was dependent on IL-6 OCC stimulation. METHODS: We combined meticulous in vitro profiling and tumor xenograft models to study the role of IL-6 in MSC/OCC intereactions. RESULTS: We demonstrated that Tocilizumab® (anti-IL-6R therapy) in association with chemotherapy significantly reduced the peritoneal carcinosis index (PCI) than chemotherapy alone in mice xenografted with OCCs+MSCs. Further experiments showed that CCL2 and CCL5 are released by MSC in transwell co-culture and induce OCCs IL-6 secretion and chemoresistance. Finally, we found that IL-6 induced chemoresistance was dependent on PYK2 phosphorylation. CONCLUSIONS: These findings highlight the potential key role of the stroma in protecting minimal residual disease from chemotherapy, thus favoring recurrences. Future clinical trials targeting stroma could use anti-IL-6 therapy in association with chemotherapy.


Asunto(s)
Quimiocina CCL2/metabolismo , Quimiocina CCL5/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Interleucina-6/metabolismo , Neoplasias Ováricas/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/genética , Quimiocina CCL5/genética , Técnicas de Cocultivo , Femenino , Quinasa 2 de Adhesión Focal/genética , Humanos , Interleucina-6/genética , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Transducción de Señal/efectos de los fármacos
14.
J Clin Invest ; 127(12): 4242-4256, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29058691

RESUMEN

Angiocrine factors, such as Notch ligands, supplied by the specialized endothelial cells (ECs) within the bone marrow and splenic vascular niche play an essential role in modulating the physiology of adult hematopoietic stem and progenitor cells (HSPCs). However, the relative contribution of various Notch ligands, specifically jagged-2, to the homeostasis of HSPCs is unknown. Here, we show that under steady state, jagged-2 is differentially expressed in tissue-specific vascular beds, but its expression is induced in hematopoietic vascular niches after myelosuppressive injury. We used mice with EC-specific deletion of the gene encoding jagged-2 (Jag2) to demonstrate that while EC-derived jagged-2 was dispensable for maintaining the capacity of HSPCs to repopulate under steady-state conditions, by activating Notch2 it did contribute to the recovery of HSPCs in response to myelosuppressive conditions. Engraftment and/or expansion of HSPCs was dependent on the expression of endothelial-derived jagged-2 following myeloablation. Additionally, jagged-2 expressed in bone marrow ECs regulated HSPC cell cycle and quiescence during regeneration. Endothelial-deployed jagged-2 triggered Notch2/Hey1, while tempering Notch2/Hes1 signaling in HSPCs. Collectively, these data demonstrate that EC-derived jagged-2 activates Notch2 signaling in HSPCs to promote hematopoietic recovery and has potential as a therapeutic target to accelerate balanced hematopoietic reconstitution after myelosuppression.


Asunto(s)
Células Madre Adultas/metabolismo , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Proteína Jagged-2/biosíntesis , Transducción de Señal , Aloinjertos , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Eliminación de Gen , Proteína Jagged-2/genética , Ratones , Ratones Transgénicos , Receptor Notch2/genética , Receptor Notch2/metabolismo , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/metabolismo
15.
J Ovarian Res ; 10(1): 64, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28927438

RESUMEN

BACKGROUND: New data suggests that endothelial cells (ECs) elaborate essential "angiocrine factors". The aim of this study is to investigate the role of activated ovarian endothelial cells in early in-vitro follicular development. METHODS: Mouse ovarian ECs were isolated using magnetic cell sorting or by FACS and cultured in serum free media. After a constitutive activation of the Akt pathway was initiated, early follicles (50-150 um) were mechanically isolated from 8-day-old mice and co-cultured with these activated ovarian endothelial cells (AOEC) (n = 32), gel (n = 24) or within matrigel (n = 27) in serum free media for 14 days. Follicular growth, survival and function were assessed. RESULTS: After 6 passages, flow cytometry showed 93% of cells grown in serum-free culture were VE-cadherin positive, CD-31 positive and CD 45 negative, matching the known EC profile. Beginning on day 4 of culture, we observed significantly higher follicular and oocyte growth rates in follicles co-cultured with AOECs compared with follicles on gel or matrigel. After 14 days of culture, 73% of primary follicles and 83% of secondary follicles co-cultured with AOEC survived, whereas the majority of follicles cultured on gel or matrigel underwent atresia. CONCLUSIONS: This is the first report of successful isolation and culture of ovarian ECs. We suggest that co-culture with activated ovarian ECs promotes early follicular development and survival. This model is a novel platform for the in vitro maturation of early follicles and for the future exploration of endothelial-follicular communication. CAPSULE: In vitro development of early follicles necessitates a complex interplay of growth factors and signals required for development. Endothelial cells (ECs) may elaborate essential "angiocrine factors" involved in organ regeneration. We demonstrate that co-culture with ovarian ECs enables culture of primary and early secondary mouse ovarian follicles.


Asunto(s)
Técnicas de Cultivo de Célula , Células Endoteliales/citología , Folículo Ovárico/citología , Animales , Apoptosis , Comunicación Celular , Proliferación Celular , Células Cultivadas , Estradiol/metabolismo , Femenino , Fibroblastos/citología , Células de la Granulosa/citología , Ratones , Oocitos/citología , Folículo Ovárico/metabolismo
16.
Nat Immunol ; 18(7): 780-790, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28553951

RESUMEN

The acquisition of a protective vertebrate immune system hinges on the efficient generation of a diverse but self-tolerant repertoire of T cells by the thymus through mechanisms that remain incompletely resolved. Here we identified the endosomal-sorting-complex-required-for-transport (ESCRT) protein CHMP5, known to be required for the formation of multivesicular bodies, as a key sensor of thresholds for signaling via the T cell antigen receptor (TCR) that was essential for T cell development. CHMP5 enabled positive selection by promoting post-selection thymocyte survival in part through stabilization of the pro-survival protein Bcl-2. Accordingly, loss of CHMP5 in thymocyte precursor cells abolished T cell development, a phenotype that was 'rescued' by genetic deletion of the pro-apoptotic protein Bim or transgenic expression of Bcl-2. Mechanistically, positive selection resulted in the stabilization of CHMP5 by inducing its interaction with the deubiquitinase USP8. Our results thus identify CHMP5 as an essential component of the post-translational machinery required for T cell development.


Asunto(s)
Diferenciación Celular/inmunología , Complejos de Clasificación Endosomal Requeridos para el Transporte/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Timocitos/inmunología , Animales , Proteína 11 Similar a Bcl2/inmunología , Endopeptidasas/inmunología , Immunoblotting , Inmunoprecipitación , Ratones , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/inmunología , Linfocitos T/citología , Timocitos/citología , Ubiquitina Tiolesterasa/inmunología
17.
Nature ; 545(7655): 439-445, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28514438

RESUMEN

Developmental pathways that orchestrate the fleeting transition of endothelial cells into haematopoietic stem cells remain undefined. Here we demonstrate a tractable approach for fully reprogramming adult mouse endothelial cells to haematopoietic stem cells (rEC-HSCs) through transient expression of the transcription-factor-encoding genes Fosb, Gfi1, Runx1, and Spi1 (collectively denoted hereafter as FGRS) and vascular-niche-derived angiocrine factors. The induction phase (days 0-8) of conversion is initiated by expression of FGRS in mature endothelial cells, which results in endogenous Runx1 expression. During the specification phase (days 8-20), RUNX1+ FGRS-transduced endothelial cells commit to a haematopoietic fate, yielding rEC-HSCs that no longer require FGRS expression. The vascular niche drives a robust self-renewal and expansion phase of rEC-HSCs (days 20-28). rEC-HSCs have a transcriptome and long-term self-renewal capacity similar to those of adult haematopoietic stem cells, and can be used for clonal engraftment and serial primary and secondary multi-lineage reconstitution, including antigen-dependent adaptive immune function. Inhibition of TGFß and CXCR7 or activation of BMP and CXCR4 signalling enhanced generation of rEC-HSCs. Pluripotency-independent conversion of endothelial cells into autologous authentic engraftable haematopoietic stem cells could aid treatment of haematological disorders.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Endotelio/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Inmunidad Adaptativa , Envejecimiento/genética , Animales , Línea Celular , Linaje de la Célula , Autorrenovación de las Células , Células Clonales/citología , Células Clonales/trasplante , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
18.
Nat Commun ; 8: 13963, 2017 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-28091527

RESUMEN

Transplanting vascular endothelial cells (ECs) to support metabolism and express regenerative paracrine factors is a strategy to treat vasculopathies and to promote tissue regeneration. However, transplantation strategies have been challenging to develop, because ECs are difficult to culture and little is known about how to direct them to stably integrate into vasculature. Here we show that only amniotic cells could convert to cells that maintain EC gene expression. Even so, these converted cells perform sub-optimally in transplantation studies. Constitutive Akt signalling increases expression of EC morphogenesis genes, including Sox17, shifts the genomic targeting of Fli1 to favour nearby Sox consensus sites and enhances the vascular function of converted cells. Enforced expression of Sox17 increases expression of morphogenesis genes and promotes integration of transplanted converted cells into injured vessels. Thus, Ets transcription factors specify non-vascular, amniotic cells to EC-like cells, whereas Sox17 expression is required to confer EC function.


Asunto(s)
Células Endoteliales/trasplante , Endotelio Vascular/metabolismo , Factores de Transcripción SOXF/metabolismo , Enfermedades Vasculares/terapia , Amnios/citología , Amnios/embriología , Amnios/metabolismo , Animales , Células Endoteliales/metabolismo , Endotelio Vascular/fisiopatología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Regeneración , Factores de Transcripción SOXF/genética , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/fisiopatología
19.
Nat Med ; 22(2): 154-62, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26779814

RESUMEN

Although the lung can undergo self-repair after injury, fibrosis in chronically injured or diseased lungs can occur at the expense of regeneration. Here we study how a hematopoietic-vascular niche regulates alveolar repair and lung fibrosis. Using intratracheal injection of bleomycin or hydrochloric acid in mice, we show that repetitive lung injury activates pulmonary capillary endothelial cells (PCECs) and perivascular macrophages, impeding alveolar repair and promoting fibrosis. Whereas the chemokine receptor CXCR7, expressed on PCECs, acts to prevent epithelial damage and ameliorate fibrosis after a single round of treatment with bleomycin or hydrochloric acid, repeated injury leads to suppression of CXCR7 expression and recruitment of vascular endothelial growth factor receptor 1 (VEGFR1)-expressing perivascular macrophages. This recruitment stimulates Wnt/ß-catenin-dependent persistent upregulation of the Notch ligand Jagged1 (encoded by Jag1) in PCECs, which in turn stimulates exuberant Notch signaling in perivascular fibroblasts and enhances fibrosis. Administration of a CXCR7 agonist or PCEC-targeted Jag1 shRNA after lung injury promotes alveolar repair and reduces fibrosis. Thus, targeting of a maladapted hematopoietic-vascular niche, in which macrophages, PCECs and perivascular fibroblasts interact, may help to develop therapy to spur lung regeneration and alleviate fibrosis.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Capilares/metabolismo , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lesión Pulmonar/metabolismo , Pulmón/metabolismo , Proteínas de la Membrana/metabolismo , Fibrosis Pulmonar/metabolismo , Receptores CXCR/metabolismo , Regeneración/fisiología , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Proteínas de Unión al Calcio/antagonistas & inhibidores , Capilares/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Fibroblastos/efectos de los fármacos , Fibrosis , Técnica del Anticuerpo Fluorescente , Humanos , Ácido Clorhídrico/toxicidad , Proteína Jagged-1 , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Oligopéptidos/farmacología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Circulación Pulmonar/efectos de los fármacos , Circulación Pulmonar/fisiología , ARN Interferente Pequeño/farmacología , Receptores CXCR/agonistas , Receptores Notch/metabolismo , Regeneración/efectos de los fármacos , Proteínas Serrate-Jagged , Proteína smad3/efectos de los fármacos , Proteína smad3/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Vía de Señalización Wnt
20.
Nat Cell Biol ; 17(2): 123-136, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25621952

RESUMEN

The lung alveoli regenerate after surgical removal of the left lobe by pneumonectomy (PNX). How this alveolar regrowth/regeneration is initiated remains unknown. We found that platelets trigger lung regeneration by supplying stromal-cell-derived factor-1 (SDF-1, also known as CXCL12). After PNX, activated platelets stimulate SDF-1 receptors CXCR4 and CXCR7 on pulmonary capillary endothelial cells (PCECs) to deploy the angiocrine membrane-type metalloproteinase MMP14, stimulating alveolar epithelial cell (AEC) expansion and neo-alveolarization. In mice lacking platelets or platelet Sdf1, PNX-induced alveologenesis was diminished. Reciprocally, infusion of Sdf1(+/+) but not Sdf1-deficient platelets rescued lung regeneration in platelet-depleted mice. Endothelial-specific ablation of Cxcr4 and Cxcr7 in adult mice similarly impeded lung regeneration. Notably, mice with endothelial-specific Mmp14 deletion exhibited impaired expansion of AECs but not PCECs after PNX, which was not rescued by platelet infusion. Therefore, platelets prime PCECs to initiate lung regeneration, extending beyond their haemostatic contribution. Therapeutic targeting of this haemo-vascular niche could enable regenerative therapy for lung diseases.


Asunto(s)
Plaquetas/metabolismo , Capilares/metabolismo , Quimiocina CXCL12/metabolismo , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/fisiología , Regeneración , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/enzimología , Factor de Crecimiento Epidérmico/metabolismo , Eliminación de Gen , Ligandos , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones , Especificidad de Órganos , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Neumonectomía , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4 , Transducción de Señal , Trombopoyetina/deficiencia , Trombopoyetina/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA