Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 139(4): 791-801, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19914171

RESUMEN

Understanding how cells polarize and coordinate tubulogenesis during organ formation is a central question in biology. Tubulogenesis often coincides with cell-lineage specification during organ development. Hence, an elementary question is whether these two processes are independently controlled, or whether proper cell specification depends on formation of tubes. To address these fundamental questions, we have studied the functional role of Cdc42 in pancreatic tubulogenesis. We present evidence that Cdc42 is essential for tube formation, specifically for initiating microlumen formation and later for maintaining apical cell polarity. Finally, we show that Cdc42 controls cell specification non-cell-autonomously by providing the correct microenvironment for proper control of cell-fate choices of multipotent progenitors. For a video summary of this article, see the PaperFlick file with the Supplemental Data available online.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Organogénesis , Páncreas/embriología , Animales , Polaridad Celular , Células Epiteliales/metabolismo , Laminina/metabolismo , Ratones , Ratones Noqueados , Páncreas/citología , Páncreas/metabolismo , Páncreas Exocrino/citología , Páncreas Exocrino/embriología , Páncreas Exocrino/metabolismo , Células Madre/metabolismo , Quinasas Asociadas a rho/metabolismo
2.
PLoS Genet ; 17(2): e1009283, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33539340

RESUMEN

Activating transcription factor 3 (ATF3) is a key transcription factor involved in regulating cellular stress responses, with different expression levels and functions in different tissues. ATF3 has also been shown to play crucial roles in regulating tumor development and progression, however its potential role in oral squamous cell carcinomas has not been fully explored. In this study, we examined biopsies of tongue squamous cell carcinomas (TSCCs) and found that the nuclear expression level of ATF3 correlated negatively with the differentiation status of TSCCs, which was validated by analysis of the ATGC database. By using gain- or loss- of function analyses of ATF3 in four different TSCC cell lines, we demonstrated that ATF3 negatively regulates the growth and migration of human TSCC cells in vitro. RNA-seq analysis identified two new downstream targets of ATF3, interferon alpha inducible proteins 6 (IFI6) and 27 (IFI27), which were upregulated in ATF3-deleted cells and were downregulated in ATF3-overexpressing cells. Chromatin immunoprecipitation assays showed that ATF3 binds the promoter regions of the IFI6 and IFI27 genes. Both IFI6 and IFI27 were highly expressed in TSCC biopsies and knockdown of either IFI6 or IFI27 in TSCC cells blocked the cell growth and migration induced by the deletion of ATF3. Conversely, overexpression of either IFI6 or IFI27 counteracted the inhibition of TSCC cell growth and migration induced by the overexpression of ATF3. Finally, an in vivo study in mice confirmed those in vitro findings. Our study suggests that ATF3 plays an anti-tumor function in TSCCs through the negative regulation of its downstream targets, IFI6 and IFI27.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Carcinoma de Células Escamosas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias de la Lengua/metabolismo , Factor de Transcripción Activador 3/genética , Animales , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular/genética , Inmunoprecipitación de Cromatina , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Proteínas Mitocondriales/genética , Clasificación del Tumor , Regiones Promotoras Genéticas , ARN Interferente Pequeño , RNA-Seq , Neoplasias de la Lengua/genética , Neoplasias de la Lengua/patología , Regulación hacia Arriba
3.
Gut ; 72(2): 275-294, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35241625

RESUMEN

OBJECTIVE: Increased apoptotic shedding has been linked to intestinal barrier dysfunction and development of inflammatory bowel diseases (IBD). In contrast, physiological cell shedding allows the renewal of the epithelial monolayer without compromising the barrier function. Here, we investigated the role of live cell extrusion in epithelial barrier alterations in IBD. DESIGN: Taking advantage of conditional GGTase and RAC1 knockout mice in intestinal epithelial cells (Pggt1b iΔIEC and Rac1 iΔIEC mice), intravital microscopy, immunostaining, mechanobiology, organoid techniques and RNA sequencing, we analysed cell shedding alterations within the intestinal epithelium. Moreover, we examined human gut tissue and intestinal organoids from patients with IBD for cell shedding alterations and RAC1 function. RESULTS: Epithelial Pggt1b deletion led to cytoskeleton rearrangement and tight junction redistribution, causing cell overcrowding due to arresting of cell shedding that finally resulted in epithelial leakage and spontaneous mucosal inflammation in the small and to a lesser extent in the large intestine. Both in vivo and in vitro studies (knockout mice, organoids) identified RAC1 as a GGTase target critically involved in prenylation-dependent cytoskeleton dynamics, cell mechanics and epithelial cell shedding. Moreover, inflamed areas of gut tissue from patients with IBD exhibited funnel-like structures, signs of arrested cell shedding and impaired RAC1 function. RAC1 inhibition in human intestinal organoids caused actin alterations compatible with arresting of cell shedding. CONCLUSION: Impaired epithelial RAC1 function causes cell overcrowding and epithelial leakage thus inducing chronic intestinal inflammation. Epithelial RAC1 emerges as key regulator of cytoskeletal dynamics, cell mechanics and intestinal cell shedding. Modulation of RAC1 might be exploited for restoration of epithelial integrity in the gut of patients with IBD.


Asunto(s)
Citoesqueleto , Enfermedades Inflamatorias del Intestino , Animales , Humanos , Ratones , Células Epiteliales , Inflamación , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/fisiología , Ratones Noqueados , Proteína de Unión al GTP rac1
4.
Development ; 145(13)2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29853619

RESUMEN

Formation and homeostasis of the vascular system requires several coordinated cellular functions, but their precise interplay during development and their relative importance for vascular pathologies remain poorly understood. Here, we investigated the endothelial functions regulated by Cdc42 and their in vivo relevance during angiogenic sprouting and vascular morphogenesis in the postnatal mouse retina. We found that Cdc42 is required for endothelial tip cell selection, directed cell migration and filopodia formation, but dispensable for cell proliferation or apoptosis. Although the loss of Cdc42 seems generally compatible with apical-basal polarization and lumen formation in retinal blood vessels, it leads to defective endothelial axial polarization and to the formation of severe vascular malformations in capillaries and veins. Tracking of Cdc42-depleted endothelial cells in mosaic retinas suggests that these capillary-venous malformations arise as a consequence of defective cell migration, when endothelial cells that proliferate at normal rates are unable to re-distribute within the vascular network.


Asunto(s)
Capilares/anomalías , Movimiento Celular , Células Endoteliales/metabolismo , Vena Retiniana/anomalías , Malformaciones Vasculares/embriología , Proteína de Unión al GTP cdc42/deficiencia , Animales , Capilares/embriología , Polaridad Celular/genética , Células Endoteliales/patología , Ratones , Ratones Noqueados , Seudópodos/genética , Seudópodos/metabolismo , Vena Retiniana/embriología , Malformaciones Vasculares/genética , Malformaciones Vasculares/patología
5.
Circ Res ; 124(8): 1240-1252, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30732528

RESUMEN

RATIONALE: Aberrant formation of blood vessels precedes a broad spectrum of vascular complications; however, the cellular and molecular events governing vascular malformations are not yet fully understood. OBJECTIVE: Here, we investigated the role of CDC42 (cell division cycle 42) during vascular morphogenesis and its relative importance for the development of cerebrovascular malformations. METHODS AND RESULTS: To avoid secondary systemic effects often associated with embryonic gene deletion, we generated an endothelial-specific and inducible knockout approach to study postnatal vascularization of the mouse brain. Postnatal endothelial-specific deletion of Cdc42 elicits cerebrovascular malformations reminiscent of cerebral cavernous malformations (CCMs). At the cellular level, loss of CDC42 function in brain endothelial cells (ECs) impairs their sprouting, branching morphogenesis, axial polarity, and normal dispersion within the brain tissue. Disruption of CDC42 does not alter EC proliferation, but malformations occur where EC proliferation is the most pronounced during brain development-the postnatal cerebellum-indicating that a high, naturally occurring EC proliferation provides a permissive state for the appearance of these malformations. Mechanistically, CDC42 depletion in ECs elicited increased MEKK3 (mitogen-activated protein kinase kinase kinase 3)-MEK5 (mitogen-activated protein kinase kinase 5)-ERK5 (extracellular signal-regulated kinase 5) signaling and consequent detrimental overexpression of KLF (Kruppel-like factor) 2 and KLF4, recapitulating the hallmark mechanism for CCM pathogenesis. Through genetic approaches, we demonstrate that the coinactivation of Klf4 reduces the severity of vascular malformations in Cdc42 mutant mice. Moreover, we show that CDC42 interacts with CCMs and that CCM3 promotes CDC42 activity in ECs. CONCLUSIONS: We show that endothelial-specific deletion of Cdc42 elicits CCM-like cerebrovascular malformations and that CDC42 is engaged in the CCM signaling network to restrain the MEKK3-MEK5-ERK5-KLF2/4 pathway.


Asunto(s)
Vasos Sanguíneos/anomalías , Proliferación Celular , Células Endoteliales/fisiología , Eliminación de Gen , Hemangioma Cavernoso del Sistema Nervioso Central/etiología , Proteína de Unión al GTP cdc42/genética , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/genética , Encéfalo/irrigación sanguínea , Ciclo Celular/fisiología , Proteína KRIT1/genética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , MAP Quinasa Quinasa 5/metabolismo , MAP Quinasa Quinasa Quinasa 3/metabolismo , Ratones , Proteínas de Microfilamentos/genética , Proteína de Unión al GTP cdc42/metabolismo
6.
Gastroenterology ; 157(5): 1293-1309, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31302143

RESUMEN

BACKGROUND & AIMS: It is not clear how regulation of T-cell function is altered during development of inflammatory bowel diseases (IBD). We studied the mechanisms by which geranylgeranyltransferase-mediated prenylation controls T-cell localization to the intestine and chronic inflammation. METHODS: We generated mice with T-cell-specific disruption of the geranylgeranyltransferase type I, beta subunit gene (Pggt1b), called Pggt1bΔCD4 mice, or the ras homolog family member A gene (Rhoa), called RhoaΔCD4 mice. We also studied mice with knockout of CDC42 or RAC1 and wild-type mice (controls). Intestinal tissues were analyzed by histology, multiphoton and confocal microscopy, and real-time polymerase chain reaction. Activation of CDC42, RAC1, and RHOA were measured with G-LISA, cell fractionation, and immunoblots. T cells and lamina propria mononuclear cells from mice were analyzed by flow cytometry or transferred to Rag1-/- mice. Mice were given injections of antibodies against integrin alpha4beta7 or gavaged with the RORC antagonist GSK805. We obtained peripheral blood and intestinal tissue samples from patients with and without IBD and analyzed them by flow cytometry. RESULTS: Pggt1bΔCD4 mice developed spontaneous colitis, characterized by thickening of the intestinal wall, edema, fibrosis, accumulation of T cells in the colon, and increased expression of inflammatory cytokines. Compared with control CD4+ T cells, PGGT1B-deficient CD4+ T cells expressed significantly higher levels of integrin alpha4beta7, which regulates their localization to the intestine. Inflammation induced by transfer of PGGT1B-deficient CD4+ T cells to Rag1-/- mice was blocked by injection of an antibody against integrin alpha4beta7. Lamina propria of Pggt1bΔCD4 mice had increased numbers of CD4+ T cells that expressed RORC and higher levels of cytokines produced by T-helper 17 cells (granulocyte-macrophage colony-stimulating factor, interleukin [IL]17A, IL17F, IL22, and tumor necrosis factor [TNF]). The RORC inverse agonist GSK805, but not antibodies against IL17A or IL17F, prevented colitis in Pggt1bΔCD4 mice. PGGT1B-deficient CD4+ T cells had decreased activation of RHOA. RhoAΔCD4 mice had a similar phenotype to Pggt1bΔCD4 mice, including development of colitis, increased numbers of CD4+ T cells in colon, increased expression of integrin alpha4beta7 by CD4+ T cells, and increased levels of IL17A and other inflammatory cytokines in lamina propria. T cells isolated from intestinal tissues from patients with IBD had significantly lower levels of PGGT1B than tissues from individuals without IBD. CONCLUSION: Loss of PGGT1B from T cells in mice impairs RHOA function, increasing CD4+ T-cell expression of integrin alpha4beta7 and localization to colon, resulting in increased expression of inflammatory cytokines and colitis. T cells isolated from gut tissues from patients with IBD have lower levels of PGGT1B than tissues from patients without IBD.


Asunto(s)
Transferasas Alquil y Aril/deficiencia , Quimiotaxis de Leucocito , Colitis/enzimología , Colon/enzimología , Integrinas/metabolismo , Linfocitos T/enzimología , Proteínas de Unión al GTP rho/metabolismo , Inmunidad Adaptativa , Transferasas Alquil y Aril/genética , Animales , Estudios de Casos y Controles , Células Cultivadas , Colitis/genética , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Ratones Noqueados , Neuropéptidos/genética , Neuropéptidos/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/patología , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/deficiencia , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA
7.
Mamm Genome ; 31(1-2): 49-53, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32088735

RESUMEN

Design and production of genetically engineered mouse strains by individual research laboratories, research teams, large-scale consortia, and the biopharmaceutical industry have magnified the need for qualified personnel to identify, annotate, and validate (phenotype) these potentially new mouse models of human disease. The PATHBIO project has been recently established and funded by the European Union's ERASMUS+ Knowledge Alliance program to address the current shortfall in formally trained personnel. A series of teaching workshops will be given by experts on anatomy, histology, embryology, imaging, and comparative pathology to increase the availability of individuals with formal training to contribute to this important niche of Europe's biomedical research enterprise. These didactic and hands-on workshops are organized into three modules: (1) embryology, anatomy, histology, and the anatomical basis of imaging, (2) image-based phenotyping, and (3) pathology. The workshops are open to all levels of participants from recent graduates to Ph.D., M.D., and veterinary scientists. Participation is available on a competitive basis at no cost for attending. The first series of Workshop Modules was held in 2019 and these will continue for the next 2 years.


Asunto(s)
Investigación Biomédica/educación , Fenotipo , Animales , Animales Modificados Genéticamente , Investigación Biomédica/organización & administración , Curriculum , Modelos Animales de Enfermedad , Humanos , Ratones , Investigadores/educación
8.
J Biol Chem ; 293(24): 9358-9369, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29700112

RESUMEN

Mesenchymal stem cells (MSC) are suggested to be important progenitors of myofibroblasts in fibrosis. To understand the role of Rho GTPase signaling in TGFß-induced myofibroblast differentiation of MSC, we generated a novel MSC line and its descendants lacking functional Rho GTPases and Rho GTPase signaling components. Unexpectedly, our data revealed that Rho GTPase signaling is required for TGFß-induced expression of α-smooth muscle actin (αSMA) but not of collagen I α1 (col1a1). Whereas loss of RhoA and Cdc42 reduced αSMA expression, ablation of the Rac1 gene had the opposite effect. Although actin polymerization and MRTFa were crucial for TGFß-induced αSMA expression, neither Arp2/3-dependent actin polymerization nor cofilin-dependent severing and depolymerization of F-actin were required. Instead, F-actin levels were dependent on cell contraction, and TGFß-induced actin polymerization correlated with increased cell contraction mediated by RhoA and Cdc42. Finally, we observed impaired collagen I secretion in MSC lacking RhoA or Cdc42. These data give novel molecular insights into the role of Rho GTPases in TGFß signaling and have implications for our understanding of MSC function in fibrosis.


Asunto(s)
Actinas/genética , Colágeno Tipo I/genética , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Neuropéptidos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Actinas/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Colágeno Tipo I/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Miofibroblastos/citología , Miofibroblastos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteína de Unión al GTP rhoA
9.
Genesis ; 56(9): e23238, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30010246

RESUMEN

Development of human hematopoietic stem cells and differentiation of embryonic stem (ES) cells/induced pluripotent stem (iPS) cells to hematopoietic stem cells are poorly understood. NOD (Non-obese diabetic)-derived mouse strains, such as NSG (NOD-Scid-il2Rg) or NRG (NOD-Rag1-il2Rg), are the best available models for studying the function of fetal and adult human hematopoietic cells as well as ES/iPS cell-derived hematopoietic stem cells. Unfortunately, engraftment of human hematopoietic stem cells is very variable in these models. Introduction of additional permissive mutations into these complex genetic backgrounds of the NRG/NSG mice by natural breeding is a very demanding task in terms of time and resources. Specifically, since the genetic elements defining the NSG/NRG phenotypes have not yet been fully characterized, intense backcrossing is required to ensure transmission of the full phenotype. Here we describe the derivation of embryonic stem cell (ESC) lines from NRG pre-implantation embryos generated by in vitro fertilization followed by the CRISPR/CAS9 targeting of the Gata-2 locus. After injection into morula stage embryos, cells from three tested lines gave rise to chimeric adult mice showing high contribution of the ESCs (70%-100%), assessed by coat color. Moreover, these lines have been successfully targeted using Cas9/CRISPR technology, and the mutant cells have been shown to remain germ line competent. Therefore, these new NRG ESC lines combined with genome editing nucleases bring a powerful genetic tool that facilitates the generation of new NOD-based mouse models with the aim to improve the existing xenograft models.


Asunto(s)
Sistemas CRISPR-Cas , Línea Celular , Células Madre Embrionarias , Ratones Endogámicos NOD/genética , Animales , Fertilización In Vitro , Factor de Transcripción GATA2/genética , Marcación de Gen , Huésped Inmunocomprometido/genética , Ratones , Ratones Endogámicos NOD/inmunología , Modelos Biológicos
10.
Blood ; 127(10): 1297-306, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26747246

RESUMEN

Increasing evidence suggests that Rho family GTPases could have a critical role in the biology of T-cell lymphoma. In ALK-rearranged anaplastic large cell lymphoma (ALCL), a specific subtype of T-cell lymphoma, the Rho family GTPases Cdc42 and Rac1 are activated by the ALK oncogenic activity. In vitro studies have shown that Cdc42 and Rac1 control rather similar phenotypes of ALCL biology such as the proliferation, survival, and migration of lymphoma cells. However, their role and possible redundancy in ALK-driven lymphoma development in vivo are still undetermined. We genetically deleted Cdc42 or Rac1 in a mouse model of ALK-rearranged ALCL to show that either Cdc42 or Rac1 deletion impaired lymphoma development, modified lymphoma morphology, actin filament distribution, and migration properties of lymphoma cells. Cdc42 or Rac1 deletion primarily affected survival rather than proliferation of lymphoma cells. Apoptosis of lymphoma cells was equally induced following Cdc42 or Rac1 deletion, was associated with upregulation of the proapoptotic molecule Bid, and was blocked by Bcl2 overexpression. Remarkably, Cdc42/Rac1 double deletion, but not Cdc42 or Rac1 single deletions, completely prevented NPM-ALK lymphoma dissemination in vivo. Thus, Cdc42 and Rac1 have nonredundant roles in controlling ALK-rearranged lymphoma survival and morphology but are redundant for lymphoma dissemination, suggesting that targeting both GTPases could represent a preferable therapeutic option for ALCL treatment.


Asunto(s)
Linfoma de Células B Grandes Difuso/metabolismo , Neuropéptidos/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Supervivencia Celular/genética , Eliminación de Gen , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Neuropéptidos/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas/genética , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/genética
11.
J Cell Sci ; 128(23): 4293-305, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26490995

RESUMEN

The Rho GTPase Cdc42 regulates key signaling pathways required for multiple cell functions, including maintenance of shape, polarity, proliferation, migration, differentiation and morphogenesis. Although previous studies have shown that Cdc42 is required for proper epithelial development and maintenance, its exact molecular function in kidney development is not well understood. In this study, we define the specific role of Cdc42 during murine kidney epithelial tubulogenesis by deleting it selectively at the initiation of ureteric bud or metanephric mesenchyme development. Deletion in either lineage results in abnormal tubulogenesis, with profound defects in polarity, lumen formation and the actin cytoskeleton. Ultimately, these defects lead to renal failure. Additionally, in vitro analysis of Cdc42-null collecting duct cells shows that Cdc42 controls these processes by regulating the polarity Par complex (Par3-Par6-aPKC-Cdc42) and the cytoskeletal proteins N-Wasp and ezrin. Thus, we conclude that the principal role of Cdc42 in ureteric bud and metanephric mesenchyme development is to regulate epithelial cell polarity and the actin cytoskeleton.


Asunto(s)
Polaridad Celular/fisiología , Citoesqueleto/metabolismo , Células Epiteliales/metabolismo , Túbulos Renales/embriología , Proteína de Unión al GTP cdc42/metabolismo , Animales , Citoesqueleto/genética , Células Epiteliales/citología , Ratones , Proteína de Unión al GTP cdc42/genética
12.
J Immunol ; 195(9): 4244-56, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26408665

RESUMEN

Tissues accommodate defined numbers of dendritic cells (DCs) in highly specific niches where different intrinsic and environmental stimuli control DC life span and numbers. DC homeostasis in tissues is important, because experimental changes in DC numbers influence immunity and tolerance toward various immune catastrophes and inflammation. However, the precise molecular mechanisms regulating DC life span and homeostasis are unclear. We report that the GTPase RhoA controls homeostatic proliferation, cytokinesis, survival, and turnover of cDCs. Deletion of RhoA strongly decreased the numbers of CD11b(-)CD8(+) and CD11b(+)Esam(hi) DC subsets, whereas CD11b(+)Esam(lo) DCs were not affected in conditional RhoA-deficient mice. Proteome analyses revealed a defective prosurvival pathway via PI3K/protein kinase B (Akt1)/Bcl-2-associated death promoter in the absence of RhoA. Taken together, our findings identify RhoA as a central regulator of DC homeostasis, and its deletion decreases DC numbers below critical thresholds for immune protection and homeostasis, causing aberrant compensatory DC proliferation.


Asunto(s)
Apoptosis/inmunología , Células Dendríticas/inmunología , Homeostasis/inmunología , Proteína de Unión al GTP rhoA/inmunología , Animales , Apoptosis/genética , Western Blotting , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Proliferación Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Citocinesis/genética , Citocinesis/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Homeostasis/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/inmunología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Proteína Letal Asociada a bcl/inmunología , Proteína Letal Asociada a bcl/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
13.
J Anat ; 228(5): 826-37, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26889750

RESUMEN

Keratinocytes, in response to irritants, secrete pro-inflammatory mediators which recruit and activate immune and mesenchymal cells, including fibroblasts, to repair the skin. Fibroblasts respond by synthesising collagen and promoting the crosslinking extracellular matrix (ECM). We recently showed that the deletion of Rac1 in keratinocytes causes heightened inflammation due to aberrant crosstalk with immune cells. Indeed, the skin of these mice shows a higher inflammatory response to the induction of irritant contact dermatitis (ICD), and also even to treatment with a vehicle alone, compared with controls. As inflammation is intimately linked with fibrotic disease in the skin, this raised the question as to whether this deletion may also affect the deposition and arrangement of the dermal ECM. This study assessed the effects of Rac1 deletion in keratinocytes and of the heightened inflammatory status by induction of ICD on the tissue localisation and arrangements of dermal collagen. Qualitative analysis did not reveal evidence for the formation of pathologies in the dermis. However, quantitative analysis did reveal some perturbations in the dermal matrix, namely that only the combination of the lack of Rac1 and ICD affects the architectural organisation of the dermal collagen, and that a higher inflammatory state in the tissue (i.e. when Rac1 is deleted in the keratinocytes or ICD is induced in the skin, or a combination of both) influences the diameter of the collagen fibrils. It is proposed that this increase in the diameter of collagen fibrils due to inflammation may serve as pre-fibrotic marker enabling earlier determination of fibrosis and earlier treatment. This study has revealed previously unknown effects on the ECM due to the deletion of Rac1 in keratinocytes.


Asunto(s)
Dermis/patología , Matriz Extracelular/patología , Queratinocitos/patología , Neuropéptidos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Dermatitis por Contacto/patología , Modelos Animales de Enfermedad , Fibroblastos/patología , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Neuropéptidos/deficiencia , Proteína de Unión al GTP rac1/deficiencia
14.
Blood ; 123(23): 3635-45, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24782506

RESUMEN

Neutrophil responses are central to host protection and inflammation. Neutrophil activation follows a 2-step process in which priming amplifies responses to activating stimuli. Priming is essential for life span extension, chemotaxis, and respiratory burst activity. Here we show that the cytoskeletal organizer RhoA suppresses neutrophil priming via formins. Premature granule exocytosis in Rho-deficient neutrophils activated numerous signaling pathways and amplified superoxide generation. Deletion of Rho altered front-to-back coordination by simultaneously increasing uropod elongation, leading edge formation, and random migration. Concomitant negative and positive regulation of ß2 integrin-independent and ß2 integrin-dependent migration, respectively, reveal Rho as a key decision point in the neutrophil response to discrete chemotactic agents. Although even restricted influx of Rho-deficient hyperactive neutrophils exacerbated lipopolysaccharide-mediated lung injury, deleting Rho in innate immune cells was highly protective in influenza A virus infection. Hence, Rho is a key regulator of disease progression by maintaining neutrophil quiescence and suppressing hyperresponsiveness.


Asunto(s)
Enfermedades del Sistema Inmune , Gripe Humana/inmunología , Trastornos Leucocíticos , Neutrófilos/inmunología , Proteínas de Unión al GTP rho/fisiología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Animales , Células Cultivadas , Progresión de la Enfermedad , Femenino , Humanos , Virus de la Influenza A/inmunología , Masculino , Ratones , Ratones Transgénicos , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Neumonía Viral/genética , Neumonía Viral/inmunología , Proteína de Unión al GTP rhoA
15.
J Biol Chem ; 289(44): 30772-30784, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25213860

RESUMEN

RhoA is thought to be essential for coordination of the membrane protrusions and retractions required for immune cell motility and directed migration. Whether the subfamily of Rho (Ras homolog) GTPases (RhoA, RhoB, and RhoC) is actually required for the directed migration of primary cells is difficult to predict. Macrophages isolated from myeloid-restricted RhoA/RhoB (conditional) double knock-out (dKO) mice did not express RhoC and were essentially "pan-Rho"-deficient. Using real-time chemotaxis assays, we found that retraction of the trailing edge was dissociated from the advance of the cell body in dKO cells, which developed extremely elongated tails. Surprisingly, velocity (of the cell body) was increased, whereas chemotactic efficiency was preserved, when compared with WT macrophages. Randomly migrating RhoA/RhoB dKO macrophages exhibited multiple small protrusions and developed large "branches" due to impaired lamellipodial retraction. A mouse model of peritonitis indicated that monocyte/macrophage recruitment was, surprisingly, more rapid in RhoA/RhoB dKO mice than in WT mice. In comparison with dKO cells, the phenotypes of single RhoA- or RhoB-deficient macrophages were mild due to mutual compensation. Furthermore, genetic deletion of RhoB partially reversed the motility defect of macrophages lacking the RhoGAP (Rho GTPase-activating protein) myosin IXb (Myo9b). In conclusion, the Rho subfamily is not required for "front end" functions (motility and chemotaxis), although both RhoA and RhoB are involved in pulling up the "back end" and resorbing lamellipodial membrane protrusions. Macrophages lacking Rho proteins migrate faster in vitro, which, in the case of the peritoneum, translates to more rapid in vivo monocyte/macrophage recruitment.


Asunto(s)
Macrófagos Peritoneales/enzimología , Seudópodos/patología , Proteínas ras/genética , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoB/genética , Animales , Polaridad Celular , Células Cultivadas , Quimiotaxis , Femenino , Expresión Génica , Macrófagos Peritoneales/patología , Ratones , Ratones Noqueados , Miosinas/genética , Peritonitis/enzimología , Peritonitis/patología , Seudópodos/enzimología , Proteínas ras/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA , Proteína de Unión al GTP rhoB/metabolismo , Proteína rhoC de Unión a GTP
16.
J Cell Sci ; 126(Pt 20): 4572-88, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23902686

RESUMEN

Cell migration is commonly accompanied by protrusion of membrane ruffles and lamellipodia. In two-dimensional migration, protrusion of these thin sheets of cytoplasm is considered relevant to both exploration of new space and initiation of nascent adhesion to the substratum. Lamellipodium formation can be potently stimulated by Rho GTPases of the Rac subfamily, but also by RhoG or Cdc42. Here we describe viable fibroblast cell lines genetically deficient for Rac1 that lack detectable levels of Rac2 and Rac3. Rac-deficient cells were devoid of apparent lamellipodia, but these structures were restored by expression of either Rac subfamily member, but not by Cdc42 or RhoG. Cells deficient in Rac showed strong reduction in wound closure and random cell migration and a notable loss of sensitivity to a chemotactic gradient. Despite these defects, Rac-deficient cells were able to spread, formed filopodia and established focal adhesions. Spreading in these cells was achieved by the extension of filopodia followed by the advancement of cytoplasmic veils between them. The number and size of focal adhesions as well as their intensity were largely unaffected by genetic removal of Rac1. However, Rac deficiency increased the mobility of different components in focal adhesions, potentially explaining how Rac - although not essential - can contribute to focal adhesion assembly. Together, our data demonstrate that Rac signaling is essential for lamellipodium protrusion and for efficient cell migration, but not for spreading or filopodium formation. Our findings also suggest that Rac GTPases are crucial to the establishment or maintenance of polarity in chemotactic migration.


Asunto(s)
Movimiento Celular/fisiología , Adhesiones Focales/fisiología , Proteínas de Unión al GTP rac/metabolismo , Actinas/metabolismo , Animales , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones Transgénicos , Neuropéptidos/metabolismo , Seudópodos/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo
17.
Blood ; 122(18): 3178-87, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23861250

RESUMEN

Blood platelets are anuclear cell fragments that are essential for blood clotting. Platelets are produced by bone marrow megakaryocytes (MKs), which extend protrusions, or so-called proplatelets, into bone marrow sinusoids. Proplatelet formation requires a profound reorganization of the MK actin and tubulin cytoskeleton. Rho GTPases, such as RhoA, Rac1, and Cdc42, are important regulators of cytoskeletal rearrangements in platelets; however, the specific roles of these proteins during platelet production have not been established. Using conditional knockout mice, we show here that Rac1 and Cdc42 possess redundant functions in platelet production and function. In contrast to a single-deficiency of either protein, a double-deficiency of Rac1 and Cdc42 in MKs resulted in macrothrombocytopenia, abnormal platelet morphology, and impaired platelet function. Double-deficient bone marrow MKs matured normally in vivo but displayed highly abnormal morphology and uncontrolled fragmentation. Consistently, a lack of Rac1/Cdc42 virtually abrogated proplatelet formation in vitro. Strikingly, this phenotype was associated with severely defective tubulin organization, whereas actin assembly and structure were barely affected. Together, these results suggest that the combined action of Rac1 and Cdc42 is crucial for platelet production, particularly by regulating microtubule dynamics.


Asunto(s)
Células Progenitoras de Megacariocitos/metabolismo , Megacariocitos/metabolismo , Tubulina (Proteína)/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/genética , Animales , Western Blotting , Citoesqueleto/metabolismo , Hemostasis/genética , Células Progenitoras de Megacariocitos/citología , Megacariocitos/citología , Megacariocitos/ultraestructura , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Microtúbulos/metabolismo , Seudópodos/genética , Seudópodos/metabolismo , Trombocitopenia/sangre , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombosis/sangre , Trombosis/genética , Trombosis/metabolismo , Proteína de Unión al GTP cdc42/deficiencia , Proteína de Unión al GTP rac1/deficiencia
18.
J Immunol ; 190(1): 27-35, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23209325

RESUMEN

Epidermal Langerhans cells (LCs) of the skin represent the prototype migratory dendritic cell (DC) subtype. In the skin, they take up Ag, migrate to the draining lymph nodes, and contribute to Ag transport and immunity. Different depletion models for LCs have revealed contrasting roles and contributions of this cell type. To target the migratory properties of DCs, we generated mice lacking the Rho-family GTPase Cdc42 specifically in DCs. In these animals, the initial seeding of the epidermis with LCs is functional, resulting in slightly reduced Langerhans cell numbers. However, Cdc42-deficient LCs fail to leave the skin in steady state as well as upon stimulation, as they do not enter the skin-draining afferent lymph vessels. Similarly, also other Cdc42-deficient migratory DC subsets fail to home properly to the corresponding draining lymph nodes. We used this novel mouse model, in which LCs are locked out, to demonstrate that these cells contribute substantially to priming of Ag-specific CD4 and CD8 T cell responses upon epicutaneous immunization, but could not detect a role in the induction of contact hypersensitivity to various doses of hapten.


Asunto(s)
Inhibición de Migración Celular/inmunología , Movimiento Celular/inmunología , Células de Langerhans/inmunología , Proteína de Unión al GTP cdc42/fisiología , Animales , Inhibición de Migración Celular/genética , Movimiento Celular/genética , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Células Dendríticas/patología , Dermatitis por Contacto/genética , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/patología , Modelos Animales de Enfermedad , Epidermis/enzimología , Epidermis/inmunología , Epidermis/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Células de Langerhans/enzimología , Células de Langerhans/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Quimera por Radiación/genética , Quimera por Radiación/inmunología , Proteína de Unión al GTP cdc42/deficiencia , Proteína de Unión al GTP cdc42/genética
19.
Proc Natl Acad Sci U S A ; 109(26): 10474-9, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22689994

RESUMEN

T cells spend the majority of their time perusing lymphoid organs in search of cognate antigen presented by antigen presenting cells (APCs) and then quickly recirculate through the bloodstream to another lymph node. Therefore, regulation of a T-cell response is dependent upon the ability of cells to arrive in the correct location following chemokine gradients ("go" signal) as well as to receive appropriate T-cell receptor (TCR) activation signals upon cognate antigen recognition ("stop" signal). However, the mechanisms by which T cells regulate these go and stop signals remain unclear. We found that overexpression of the hematopoietic-specific RhoH protein in the presence of chemokine signals resulted in decreased Rap1-GTP and LFA-1 adhesiveness to ICAM-1, thus impairing T-cell chemotaxis; while in the presence of TCR signals, there were enhanced and sustained Rap1-GTP and LFA-1 activation as well as prolonged T:APC conjugates. RT-PCR analyses of activated CD4(+) T cells and live images of T-cell migration and immunological synapse (IS) formation revealed that functions of RhoH took place primarily at the levels of transcription and intracellular distribution. Thus, we conclude that RhoH expression provides a key molecular determinant that allows T cells to switch between sensing chemokine-mediated go signals and TCR-dependent stop signals.


Asunto(s)
Activación de Linfocitos , Linfocitos T/citología , Factores de Transcripción/fisiología , Proteínas de Unión al GTP rho/fisiología , Humanos , Receptores de Antígenos de Linfocitos T/fisiología , Receptores de Quimiocina/fisiología
20.
J Neurosci ; 33(3): 1179-89, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23325254

RESUMEN

The molecular mechanisms underlying the generation, maturation, and integration of new granule cells generated throughout life in the mammalian hippocampus remain poorly understood. Small Rho GTPases, such as Cdc42 and Rac1, have been implicated previously in neural stem/progenitor cell (NSPC) proliferation and neuronal maturation during embryonic development. Here we used conditional genetic deletion and virus-based loss-of-function approaches to identify temporally distinct functions for Cdc42 and Rac1 in adult hippocampal neurogenesis. We found that Cdc42 is involved in mouse NSPC proliferation, initial dendritic development, and dendritic spine maturation. In contrast, Rac1 is dispensable for early steps of neuronal development but is important for late steps of dendritic growth and spine maturation. These results establish cell-autonomous and stage-specific functions for the small Rho GTPases Cdc42 and Rac1 in the course of adult hippocampal neurogenesis.


Asunto(s)
Hipocampo/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Linaje de la Célula/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Dendritas/metabolismo , Espinas Dendríticas/metabolismo , Femenino , Ratones , Ratones Transgénicos , Ratas , Ratas Endogámicas F344 , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA