Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 168(3): 442-459.e20, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28111071

RESUMEN

Oct4, Sox2, Klf4, and cMyc (OSKM) reprogram somatic cells to pluripotency. To gain a mechanistic understanding of their function, we mapped OSKM-binding, stage-specific transcription factors (TFs), and chromatin states in discrete reprogramming stages and performed loss- and gain-of-function experiments. We found that OSK predominantly bind active somatic enhancers early in reprogramming and immediately initiate their inactivation genome-wide by inducing the redistribution of somatic TFs away from somatic enhancers to sites elsewhere engaged by OSK, recruiting Hdac1, and repressing the somatic TF Fra1. Pluripotency enhancer selection is a stepwise process that also begins early in reprogramming through collaborative binding of OSK at sites with high OSK-motif density. Most pluripotency enhancers are selected later in the process and require OS and other pluripotency TFs. Somatic and pluripotency TFs modulate reprogramming efficiency when overexpressed by altering OSK targeting, somatic-enhancer inactivation, and pluripotency enhancer selection. Together, our data indicate that collaborative interactions among OSK and with stage-specific TFs direct both somatic-enhancer inactivation and pluripotency-enhancer selection to drive reprogramming.


Asunto(s)
Reprogramación Celular , Factores de Transcripción/metabolismo , Animales , Cromatina/metabolismo , Fibroblastos/metabolismo , Código de Histonas , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Elementos Reguladores de la Transcripción , Factores de Transcripción SOXB1/metabolismo , Elementos Silenciadores Transcripcionales
2.
EMBO J ; 39(23): e105606, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33433018

RESUMEN

Chromosomes have an intrinsic tendency to segregate into compartments, forming long-distance contacts between loci of similar chromatin states. How genome compartmentalization is regulated remains elusive. Here, comparison of mouse ground-state embryonic stem cells (ESCs) characterized by open and active chromatin, and advanced serum ESCs with a more closed and repressed genome, reveals distinct regulation of their genome organization due to differential dependency on BAZ2A/TIP5, a component of the chromatin remodeling complex NoRC. On ESC chromatin, BAZ2A interacts with SNF2H, DNA topoisomerase 2A (TOP2A) and cohesin. BAZ2A associates with chromatin sub-domains within the active A compartment, which intersect through long-range contacts. We found that ground-state chromatin selectively requires BAZ2A to limit the invasion of active domains into repressive compartments. BAZ2A depletion increases chromatin accessibility at B compartments. Furthermore, BAZ2A regulates H3K27me3 genome occupancy in a TOP2A-dependent manner. Finally, ground-state ESCs require BAZ2A for growth, differentiation, and correct expression of developmental genes. Our results uncover the propensity of open chromatin domains to invade repressive domains, which is counteracted by chromatin remodeling to establish genome partitioning and preserve cell identity.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Genoma , Células Madre Pluripotentes/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , ADN-Topoisomerasas de Tipo II/metabolismo , Epigenómica , Regulación de la Expresión Génica , Histonas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Cohesinas
3.
Nucleic Acids Res ; 49(1): 145-157, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33290556

RESUMEN

Mammalian de novo DNA methyltransferases (DNMT) are responsible for the establishment of cell-type-specific DNA methylation in healthy and diseased tissues. Through genome-wide analysis of de novo methylation activity in murine stem cells we uncover that DNMT3A prefers to methylate CpGs followed by cytosines or thymines, while DNMT3B predominantly methylates CpGs followed by guanines or adenines. These signatures are further observed at non-CpG sites, resembling methylation context observed in specialised cell types, including neurons and oocytes. We further show that these preferences result from structural differences in the catalytic domains of the two de novo DNMTs and are not a consequence of differential recruitment to the genome. Molecular dynamics simulations suggest that, in case of human DNMT3A, the preference is due to favourable polar interactions between the flexible Arg836 side chain and the guanine that base-pairs with the cytosine following the CpG. By exchanging arginine to a lysine, the corresponding side chain in DNMT3B, the sequence preference is reversed, confirming the requirement for arginine at this position. This context-dependent enzymatic activity provides additional insights into the complex regulation of DNA methylation patterns.


Asunto(s)
Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Ratones/genética , Sustitución de Aminoácidos , Animales , Arginina/química , Secuencia de Bases , Cristalografía por Rayos X , Citosina/química , ADN Metiltransferasa 3A , Conjuntos de Datos como Asunto , Células Madre Embrionarias/metabolismo , Técnicas de Inactivación de Genes , Guanina/química , Humanos , Lisina/química , Simulación de Dinámica Molecular , Especificidad por Sustrato , Sulfitos , Secuenciación Completa del Genoma , ADN Metiltransferasa 3B
4.
EMBO J ; 37(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30297530

RESUMEN

Endothelial cells line blood and lymphatic vessels and form intercellular junctions, which preserve vessel structure and integrity. The vascular endothelial cadherin, VE-cadherin, mediates endothelial adhesion and is indispensible for blood vessel development and permeability regulation. However, its requirement for lymphatic vessels has not been addressed. During development, VE-cadherin deletion in lymphatic endothelial cells resulted in abortive lymphangiogenesis, edema, and prenatal death. Unexpectedly, inducible postnatal or adult deletion elicited vessel bed-specific responses. Mature dermal lymph vessels resisted VE-cadherin loss and maintained button junctions, which was associated with an upregulation of junctional molecules. Very different, mesenteric lymphatic collectors deteriorated and formed a strongly hyperplastic layer of lymphatic endothelial cells on the mesothelium. This massive hyperproliferation may have been favored by high mesenteric VEGF-C expression and was associated with VEGFR-3 phosphorylation and upregulation of the transcriptional activator TAZ Finally, intestinal lacteals fragmented into cysts or became highly distended possibly as a consequence of the mesenteric defects. Taken together, we demonstrate here the importance of VE-cadherin for lymphatic vessel development and maintenance, which is however remarkably vessel bed-specific.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Dermis/embriología , Regulación del Desarrollo de la Expresión Génica , Linfangiogénesis , Vasos Linfáticos/metabolismo , Mesenterio/embriología , Animales , Antígenos CD/genética , Cadherinas/genética , Células Endoteliales/metabolismo , Eliminación de Gen , Ratones , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 40(2): 378-393, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31826650

RESUMEN

OBJECTIVE: Vascular endothelial (VE)-cadherin is of dominant importance for the formation and stability of endothelial junctions, yet induced gene inactivation enhances vascular permeability in the lung but does not cause junction rupture. This study aims at identifying the junctional adhesion molecule, which is responsible for preventing endothelial junction rupture in the pulmonary vasculature in the absence of VE-cadherin. Approach and Results: We have compared the relevance of ESAM (endothelial cell-selective adhesion molecule), JAM (junctional adhesion molecule)-A, PECAM (platelet endothelial cell adhesion molecule)-1, and VE-cadherin for vascular barrier integrity in various mouse tissues. Gene inactivation of ESAM enhanced vascular permeability in the lung but not in the heart, skin, and brain. In contrast, deletion of JAM-A or PECAM-1 did not affect barrier integrity in any of these organs. Blocking VE-cadherin with antibodies caused lethality in ESAM-/- mice within 30 minutes but had no such effect in JAM-A-/-, PECAM-1-/- or wild-type mice. Likewise, induced gene inactivation of VE-cadherin caused rapid lethality only in the absence of ESAM. Ultrastructural analysis revealed that only combined interference with VE-cadherin and ESAM disrupted endothelial junctions and caused massive blood coagulation in the lung. Mechanistically, we could exclude a role of platelet ESAM in coagulation, changes in the expression of other junctional proteins or a contribution of cytoplasmic signaling domains of ESAM. CONCLUSIONS: Despite well-documented roles of JAM-A and PECAM-1 for the regulation of endothelial junctions, only for ESAM, we detected an essential role for endothelial barrier integrity in a tissue-specific way. In addition, we found that it is ESAM which prevents endothelial junction rupture in the lung when VE-cadherin is absent.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/fisiología , Moléculas de Adhesión Celular/metabolismo , Muerte Celular/fisiología , Endotelio Vascular/metabolismo , Pulmón/metabolismo , Animales , Coagulación Sanguínea/fisiología , Adhesión Celular , Células Cultivadas , Cricetinae , Endotelio Vascular/ultraestructura , Femenino , Immunoblotting , Pulmón/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Modelos Animales , Transducción de Señal
6.
Haematologica ; 105(12): 2746-2756, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33256374

RESUMEN

Bone marrow endothelium plays an important role in the homing of hematopoietic stem and progenitor cells upon transplantation, but surprisingly little is known on how the bone marrow endothelial cells regulate local permeability and hematopoietic stem and progenitor cells transmigration. We show that temporal loss of vascular endothelial-cadherin function promotes vascular permeability in BM, even upon low-dose irradiation. Loss of vascular endothelial-cadherin function also enhances homing of transplanted hematopoietic stem and progenitor cells to the bone marrow of irradiated mice although engraftment is not increased. Intriguingly, stabilizing junctional vascular endothelial-cadherin in vivo reduced bone marrow permeability, but did not prevent hematopoietic stem and progenitor cells migration into the bone marrow, suggesting that hematopoietic stem and progenitor cells use the transcellular migration route to enter the bone marrow. Indeed, using an in vitro migration assay, we show that human hematopoietic stem and progenitor cells predominantly cross bone marrow endothelium in a transcellular manner in homeostasis by inducing podosome-like structures. Taken together, vascular endothelial-cadherin is crucial for BM vascular homeostasis but dispensable for the homing of hematopoietic stem and progenitor cells. These findings are important in the development of potential therapeutic targets to improve hematopoietic stem and progenitor cell homing strategies.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Podosomas , Animales , Médula Ósea , Células de la Médula Ósea , Movimiento Celular , Células Endoteliales , Endotelio , Células Madre Hematopoyéticas , Ratones , Ratones Endogámicos C57BL
7.
Blood ; 128(4): 529-41, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27235139

RESUMEN

Growth differentiation factor 15 (GDF-15) is the first cytokine known to counteract chemokine-induced activation of leukocyte integrins. We showed recently that this activity dampens neutrophil recruitment into inflamed tissue and is required for survival of myocardial infarction in mice. The receptor responsible for this GDF-15-triggered anti-inflammatory mechanism on myeloid cells is not known. Here, we identify this receptor as transforming growth factor ß receptor I (TGF-ßRI) (activin receptor-like kinase 5 [ALK-5]) and TGF-ß receptor II (TGF-ßRII). We show that interference with these receptors by small-molecule inhibitors, antibodies, or small interfering RNA, blocked the GDF-15 effect on leukocyte integrin activation. Likewise, gene inactivation of each of the 2 receptors in neutrophils isolated from conditional gene-deficient mice abolished the inhibitory effect of GDF-15 on CXCL1-induced ß2-integrin activation and neutrophil diapedesis. Rapid neutrophil arrest induced by CXCL1 in vivo was inhibited by GDF-15 in an ALK-5 and TGF-ßRII dependent way. As for GDF-15 gene-deficient mice, we found that extravasation of neutrophils deficient for ALK-5 or TGF-ßRII was strongly increased in the interleukin-1ß inflamed cremaster. The inhibitory effects of GDF-15 on neutrophil integrin activation and in vivo neutrophil arrest were also found for TGF-ß1. Mechanistically, GDF-15 and TGF-ß1 interfered with integrin activation by inhibiting the activation of Ras-related protein 1 (Rap-1), an effect that depended on CalDAG- guanine nucleotide exchange factor 1 (GEF1) and cell division control protein 42 homolog. We conclude that both GDF-15 and TGF-ß1 counteract chemokine-induced integrin activation on neutrophils via the ALK-5/TGF-ßRII heterodimer. This represents a novel, rapid anti-inflammatory activity of the 2 TGF-ß receptors and of TGF-ß1.


Asunto(s)
Antígenos CD18/metabolismo , Factor 15 de Diferenciación de Crecimiento/metabolismo , Neutrófilos/metabolismo , Multimerización de Proteína/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Antígenos CD18/genética , Factor 15 de Diferenciación de Crecimiento/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Neutrófilos/citología , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
8.
Circ Res ; 115(6): 581-90, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25057127

RESUMEN

RATIONALE: Endothelial cell-specific molecule 1 (Esm1) is a secreted protein thought to play a role in angiogenesis and inflammation. However, there is currently no direct in vivo evidence supporting a function of Esm1 in either of these processes. OBJECTIVE: To determine the role of Esm1 in vivo and the underlying molecular mechanisms. METHODS AND RESULTS: We generated and analyzed Esm1 knockout (Esm1(KO)) mice to study its role in angiogenesis and inflammation. Esm1 expression is induced by the vascular endothelial growth factor A (VEGF-A) in endothelial tip cells of the mouse retina. Esm1(KO) mice showed delayed vascular outgrowth and reduced filopodia extension, which are both VEGF-A-dependent processes. Impairment of Esm1 function led to a decrease in phosphorylated Erk1/2 (extracellular-signal regulated kinases 1/2) in sprouting vessels. We also found that Esm1(KO) mice displayed a 40% decrease in leukocyte transmigration. Moreover, VEGF-induced vascular permeability was decreased by 30% in Esm1(KO) mice and specifically on stimulation with VEGF-A165 but not VEGF-A121. Accordingly, cerebral edema attributable to ischemic stroke-induced vascular permeability was reduced by 50% in the absence of Esm1. Mechanistically, we show that Esm1 binds directly to fibronectin and thereby displaces fibronectin-bound VEGF-A165 leading to increased bioavailability of VEGF-A165 and subsequently enhanced levels of VEGF-A signaling. CONCLUSIONS: Esm1 is simultaneously a target and modulator of VEGF signaling in endothelial cells, playing a role in angiogenesis, inflammation, and vascular permeability, which might be of potential interest for therapeutic applications.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Membrana Celular/fisiología , Células Endoteliales/fisiología , Proteoglicanos/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Disponibilidad Biológica , Fibronectinas/metabolismo , Inflamación/fisiopatología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Neovascularización Fisiológica/fisiología , Proteoglicanos/deficiencia , Proteoglicanos/genética , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
J Immunol ; 190(3): 892-6, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23293350

RESUMEN

CD99-like 2 (CD99L2) is a membrane protein with moderate sequence homology to CD99, which initiates cell aggregation of transfected cells and that is strongly expressed on endothelial cells, neutrophils, and lymphocytes. We showed recently that Abs against CD99L2 inhibit neutrophil, but not T lymphocyte, recruitment into inflamed tissues. In this study, we have generated conditional gene-deficient mice for CD99L2 and show by analyzing them in various inflammation models several results. First, gene ablation of CD99L2 impairs neutrophil recruitment into inflamed cremaster and peritoneum. Second, despite the strong expression of CD99L2 on peripheral neutrophils, only gene ablation on endothelial cells but not on myeloid cells affects neutrophil extravasation. Third, in contrast to our previous Ab-based results, recruitment of activated T cells into inflamed skin was impaired in mice lacking CD99L2 on endothelial cells. We conclude that CD99L2 is an essential endothelial Ag for leukocyte extravasation, which does not require homophilic interactions with CD99L2 on leukocytes.


Asunto(s)
Antígenos CD/fisiología , Quimiotaxis de Leucocito/fisiología , Migración Transendotelial y Transepitelial/fisiología , Antígeno 12E7 , Animales , Anticuerpos/farmacología , Antígenos CD/genética , Antígenos CD/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/inmunología , Células Endoteliales/patología , Técnicas de Silenciamiento del Gen , Inflamación/inmunología , Pulmón/irrigación sanguínea , Masculino , Ratones , Microcirculación , Células Mieloides/inmunología , Miositis/inmunología , Neutrófilos/fisiología , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Peritonitis/inducido químicamente , Peritonitis/inmunología , Quimera por Radiación , Linfocitos T/inmunología
10.
Sci Rep ; 14(1): 14471, 2024 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-38914623

RESUMEN

Circulating leukocytes enter tissue either through endothelial junctions (paracellular) or via a pore through the body of endothelial cells (transcellular). We have previously shown that genetically replacing VE-cadherin with a VE-cadherin-α-catenin (VEC-αC) fusion construct-which binds constitutively to actin-obstructs junctions, and blocks leukocyte extravasation in lung, skin and postcapillary venules of cremaster muscle. However, neutrophil recruitment into the inflamed peritoneal cavity was unimpaired. Investigating reasons for this, here, we visualized neutrophil diapedesis by 3D intravital video microscopy in the cremaster muscle and omentum, the major site of neutrophil recruitment into the peritoneal cavity. We found that 80% of neutrophil-extravasation occurred through HEVs in the omentum, which was unimpaired by VEC-αC. In addition, in larger venules (60-85 µm) of both tissues, less than 15% of neutrophils extravasated transcellularly in WT mice. However, in VEC-α-C mice, transcellular diapedesis increased severalfold in the omentum, but not in the cremaster. In line with this, omental venules expressed higher levels of ICAM-1 and atypical chemokine receptor 1. Furthermore, only in the omentum, VEC-αC expression caused reduced elongation of venular endothelium in flow-direction, suggesting different biomechanical properties. Collectively, VEC-αC does not inhibit paracellular transmigration in all types of venules and can modulate the diapedesis route.


Asunto(s)
Neutrófilos , Animales , Neutrófilos/metabolismo , Ratones , Migración Transendotelial y Transepitelial , Epiplón/metabolismo , Cadherinas/metabolismo , Vénulas/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Células Endoteliales/metabolismo , Antígenos CD/metabolismo , Antígenos CD/genética , Infiltración Neutrófila , Ratones Endogámicos C57BL , Migración Transcelular de la Célula
11.
J Exp Med ; 204(10): 2349-62, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17846148

RESUMEN

Recirculation of fluid and cells through lymphatic vessels plays a key role in normal tissue homeostasis, inflammatory diseases, and cancer. Despite recent advances in understanding lymphatic function (Alitalo, K., T. Tammela, and T.V. Petrova. 2005. Nature. 438:946-953), the cellular features responsible for entry of fluid and cells into lymphatics are incompletely understood. We report the presence of novel junctions between endothelial cells of initial lymphatics at likely sites of fluid entry. Overlapping flaps at borders of oak leaf-shaped endothelial cells of initial lymphatics lacked junctions at the tip but were anchored on the sides by discontinuous button-like junctions (buttons) that differed from conventional, continuous, zipper-like junctions (zippers) in collecting lymphatics and blood vessels. However, both buttons and zippers were composed of vascular endothelial cadherin (VE-cadherin) and tight junction-associated proteins, including occludin, claudin-5, zonula occludens-1, junctional adhesion molecule-A, and endothelial cell-selective adhesion molecule. In C57BL/6 mice, VE-cadherin was required for maintenance of junctional integrity, but platelet/endothelial cell adhesion molecule-1 was not. Growing tips of lymphatic sprouts had zippers, not buttons, suggesting that buttons are specialized junctions rather than immature ones. Our findings suggest that fluid enters throughout initial lymphatics via openings between buttons, which open and close without disrupting junctional integrity, but most leukocytes enter the proximal half of initial lymphatics.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/inmunología , Vasos Linfáticos/citología , Vasos Linfáticos/inmunología , Animales , Cadherinas/metabolismo , Movimiento Celular , Células Endoteliales/metabolismo , Vasos Linfáticos/metabolismo , Linfocitos/citología , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
12.
Blood ; 117(3): 942-52, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21030556

RESUMEN

The endothelium actively participates in neutrophil migration out of the vasculature via dynamic, cytoskeleton-dependent rearrangements leading to the formation of transmigratory cups in vitro, and to domes that completely surround the leukocyte in vivo. Leukocyte-specific protein 1 (LSP1), an F-actin-binding protein recently shown to be in the endothelium, is critical for effective transmigration, although the mechanism has remained elusive. Herein we show that endothelial LSP1 is expressed in the nucleus and cytosol of resting endothelial cells and associates with the cytoskeleton upon endothelial activation. Two-photon microscopy revealed that endothelial LSP1 was crucial for the formation of endothelial domes in vivo in response to neutrophil chemokine keratinocyte-derived chemokine (KC) as well as in response to endogenously produced chemokines stimulated by cytokines (tumor necrosis factor α [TNFα] or interleukin-1ß [IL-1ß]). Endothelial domes were significantly reduced in Lsp1(-/-) compared with wild-type (WT) mice. Lsp1(-/-) animals not only showed impaired neutrophil emigration after KC and TNFα stimulation, but also had disproportionate increases in vascular permeability. We demonstrate that endothelial LSP1 is recruited to the cytoskeleton in inflammation and plays an important role in forming endothelial domes thereby regulating neutrophil transendothelial migration. The permeability data may underscore the physiologic relevance of domes and the role for LSP1 in endothelial barrier integrity.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Permeabilidad Capilar/fisiología , Endotelio/metabolismo , Neutrófilos/fisiología , Migración Transendotelial y Transepitelial , Animales , Western Blotting , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Citoesqueleto/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio/citología , Endotelio/efectos de los fármacos , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos , Microscopía Confocal/instrumentación , Microscopía Electrónica de Transmisión , Microscopía de Fluorescencia por Excitación Multifotónica/instrumentación , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Neutrófilos/citología , Neutrófilos/ultraestructura , Factor de Necrosis Tumoral alfa/farmacología
13.
J Exp Med ; 203(7): 1671-7, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16818677

RESUMEN

Endothelial cell-selective adhesion molecule (ESAM) is specifically expressed at endothelial tight junctions and on platelets. To test whether ESAM is involved in leukocyte extravasation, we have generated mice carrying a disrupted ESAM gene and analyzed them in three different inflammation models. We found that recruitment of lymphocytes into inflamed skin was unaffected by the gene disruption. However, the migration of neutrophils into chemically inflamed peritoneum was inhibited by 70% at 2 h after stimulation, recovering at later time points. Analyzing neutrophil extravasation directly by intravital microscopy in the cremaster muscle revealed that leukocyte extravasation was reduced (50%) in ESAM(-/-) mice without affecting leukocyte rolling and adhesion. Depletion of >98% of circulating platelets did not abolish the ESAM deficiency-related inhibitory effect on neutrophil extravasation, indicating that it is only ESAM at endothelial tight junctions that is relevant for the extravasation process. Knocking down ESAM expression in endothelial cells resulted in reduced levels of activated Rho, a GTPase implicated in the destabilization of tight junctions. Indeed, vascular permeability stimulated by vascular endothelial growth factor was reduced in ESAM(-/-) mice. Collectively, ESAM at endothelial tight junctions participates in the migration of neutrophils through the vessel wall, possibly by influencing endothelial cell contacts.


Asunto(s)
Permeabilidad Capilar/inmunología , Moléculas de Adhesión Celular/fisiología , Movimiento Celular/inmunología , Neutrófilos/patología , Factor A de Crecimiento Endotelial Vascular/fisiología , Proteínas de Unión al GTP rho/metabolismo , Animales , Permeabilidad Capilar/genética , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Comunicación Celular/genética , Comunicación Celular/inmunología , Movimiento Celular/genética , Activación Enzimática/genética , Activación Enzimática/inmunología , Femenino , Masculino , Ratones , Ratones Noqueados , Neutrófilos/inmunología
14.
Blood ; 116(7): 1172-84, 2010 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-20479283

RESUMEN

Leukocyte extravasation depends on various adhesion receptors at endothelial cell contacts. Here we have analyzed how mouse CD99 and CD99L2 cooperate with PECAM-1. We found that antibodies against mouse CD99 and PECAM-1 trap neutrophils between endothelial cells in in vitro transmigration assays. A sequential function, as has been suggested for human PECAM-1 and CD99, could not be demonstrated. In contrast to these in vitro results, blocking CD99 or CD99L2 or gene disruption of PECAM-1 trapped neutrophils in vivo between endothelial cells and the underlying basement membrane as revealed by electron microscopy and by 3-dimensional confocal fluorescence microscopy in the inflamed cremaster tissue. Leukocyte extravasation was inhibited in interleukin-1beta-inflamed peritoneum and in the cremaster by PECAM-1 gene disruption and was further attenuated by blocking antibodies against CD99 and CD99L2. In addition, CD99 and CD99L2 were required for leukocyte extravasation in the cremaster after stimulation with tumor necrosis factor-alpha, where the need for PECAM-1 is known to be bypassed. We conclude that CD99 and CD99L2 act independently of PECAM-1 in leukocyte extravasation and cooperate in an independent way to help neutrophils overcome the endothelial basement membrane.


Asunto(s)
Antígenos CD/fisiología , Endotelio Vascular/metabolismo , Leucocitos/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Antígeno 12E7 , Animales , Membrana Basal/inmunología , Membrana Basal/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Inflamación , Leucocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/metabolismo , Peritoneo/inmunología
15.
Nat Genet ; 54(11): 1702-1710, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36333500

RESUMEN

Genomic imprinting is regulated by parental-specific DNA methylation of imprinting control regions (ICRs). Despite an identical DNA sequence, ICRs can exist in two distinct epigenetic states that are memorized throughout unlimited cell divisions and reset during germline formation. Here, we systematically study the genetic and epigenetic determinants of this epigenetic bistability. By iterative integration of ICRs and related DNA sequences to an ectopic location in the mouse genome, we first identify the DNA sequence features required for maintenance of epigenetic states in embryonic stem cells. The autonomous regulatory properties of ICRs further enabled us to create DNA-methylation-sensitive reporters and to screen for key components involved in regulating their epigenetic memory. Besides DNMT1, UHRF1 and ZFP57, we identify factors that prevent switching from methylated to unmethylated states and show that two of these candidates, ATF7IP and ZMYM2, are important for the stability of DNA and H3K9 methylation at ICRs in embryonic stem cells.


Asunto(s)
Metilación de ADN , Impresión Genómica , Ratones , Animales , Secuencia de Bases , Metilación de ADN/genética , Epigenómica , Cromatina/genética , Proteínas Represoras/genética
16.
Blood ; 113(13): 2914-23, 2009 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-19096010

RESUMEN

Although recent advances have enabled hematopoietic stem cells (HSCs) to be enriched to near purity, more information about their characteristics will improve our understanding of their development and stage-related functions. Here, using microarray technology, we identified endothelial cell-selective adhesion molecule (ESAM) as a novel marker for murine HSCs in fetal liver. Esam was expressed at high levels within a Rag1(-) c-kit(Hi) Sca1(+) HSC-enriched fraction, but sharply down-regulated with activation of the Rag1 locus, a valid marker for the most primitive lymphoid progenitors in E14.5 liver. The HSC-enriched fraction could be subdivided into 2 on the basis of ESAM levels. Among endothelial antigens on hematopoietic progenitors, ESAM expression showed intimate correlation with HSC activity. The ESAM(Hi) population was highly enriched for multipotent myeloid-erythroid progenitors and primitive progenitors with lymphopoietic activity, and exclusively reconstituted long-term lymphohematopoiesis in lethally irradiated recipients. Tie2(+) c-kit(+) lymphohematopoietic cells in the E9.5-10.5 aorta-gonad-mesonephros region also expressed high levels of ESAM. Furthermore, ESAM was detected on primitive hematopoietic progenitors in adult bone marrow. Interestingly, ESAM expression in the HSC-enriched fraction was up-regulated in aged mice. We conclude that ESAM marks HSC in murine fetal liver and will facilitate studies of hematopoiesis throughout life.


Asunto(s)
Envejecimiento/genética , Moléculas de Adhesión Celular/genética , Células Madre Hematopoyéticas/metabolismo , Envejecimiento/metabolismo , Animales , Biomarcadores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Embrión de Mamíferos , Células Endoteliales/metabolismo , Femenino , Feto/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos
17.
Stem Cells ; 27(3): 653-61, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19074415

RESUMEN

Hematopoietic stem cells (HSCs) have been highly enriched using combinations of 12-14 surface markers. Genes specifically expressed by HSCs as compared with other multipotent progenitors may yield new stem cell enrichment markers, as well as elucidate self-renewal and differentiation mechanisms. We previously reported that multiple cell surface molecules are enriched on mouse HSCs compared with more differentiated progeny. Here, we present a definitive expression profile of the cell adhesion molecule endothelial cell-selective adhesion molecule (Esam1) in hematopoietic cells using reverse transcription-quantitative polymerase chain reaction and flow cytometry studies. We found Esam1 to be highly and selectively expressed by HSCs from mouse bone marrow (BM). Esam1 was also a viable positive HSC marker in fetal, young, and aged mice, as well as in mice of several different strains. In addition, we found robust levels of Esam1 transcripts in purified human HSCs. Esam1(-/-) mice do not exhibit severe hematopoietic defects; however, Esam1(-/-) BM has a greater frequency of HSCs and fewer T cells. HSCs from Esam1(-/-) mice give rise to more granulocyte/monocytes in culture and a higher T cell:B cell ratio upon transplantation into congenic mice. These studies identify Esam1 as a novel, widely applicable HSC-selective marker and suggest that Esam1 may play roles in both HSC proliferation and lineage decisions.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante de Células Madre
18.
Cell Stem Cell ; 26(3): 431-440.e8, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32142663

RESUMEN

Forward genetic screens with genome-wide CRISPR libraries are powerful tools for resolving cellular circuits and signaling pathways. Applying this technology to organoids, however, has been hampered by technical limitations. Here we report improved accuracy and robustness for pooled-library CRISPR screens by capturing sgRNA integrations in single organoids, substantially reducing required cell numbers for genome-scale screening. We applied our approach to wild-type and APC mutant human intestinal organoids to identify genes involved in resistance to TGF-ß-mediated growth restriction, a key process during colorectal cancer progression, and validated hits including multiple subunits of the tumor-suppressive SWI/SNF chromatin remodeling complex. Mutations within these genes require concurrent inactivation of APC to promote TGF-ß resistance and attenuate TGF-ß target gene transcription. Our approach can be applied to a variety of assays and organoid types to facilitate biological discovery in primary 3D tissue models.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Organoides , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Pruebas Genéticas , Humanos , Intestinos , Factor de Crecimiento Transformador beta
19.
Nat Biotechnol ; 38(6): 728-736, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32123383

RESUMEN

Chromatin modifications regulate genome function by recruiting proteins to the genome. However, the protein composition at distinct chromatin modifications has yet to be fully characterized. In this study, we used natural protein domains as modular building blocks to develop engineered chromatin readers (eCRs) selective for DNA methylation and histone tri-methylation at H3K4, H3K9 and H3K27 residues. We first demonstrated their utility as selective chromatin binders in living cells by stably expressing eCRs in mouse embryonic stem cells and measuring their subnuclear localization, genomic distribution and histone-modification-binding preference. By fusing eCRs to the biotin ligase BASU, we established ChromID, a method for identifying the chromatin-dependent protein interactome on the basis of proximity biotinylation, and applied it to distinct chromatin modifications in mouse stem cells. Using a synthetic dual-modification reader, we also uncovered the protein composition at bivalently modified promoters marked by H3K4me3 and H3K27me3. These results highlight the ability of ChromID to obtain a detailed view of protein interaction networks on chromatin.


Asunto(s)
Cromatina , Histonas , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas/genética , Proteómica/métodos , Animales , Células Cultivadas , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN/genética , Células Madre Embrionarias , Histonas/química , Histonas/genética , Histonas/metabolismo , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA