Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Development ; 151(18)2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39166965

RESUMEN

The microvascular system consists of two cell types: endothelial and mural (pericytes and vascular smooth muscle cells; VSMCs) cells. Communication between endothelial and mural cells plays a pivotal role in the maintenance of vascular homeostasis; however, in vivo molecular and cellular mechanisms underlying mural cell development remain unclear. In this study, we found that macrophages played a crucial role in TGFß-dependent pericyte-to-VSMC differentiation during retinal vasculature development. In mice with constitutively active Foxo1 overexpression, substantial accumulation of TGFß1-producing macrophages and pericytes around the angiogenic front region was observed. Additionally, the TGFß-SMAD pathway was activated in pericytes adjacent to macrophages, resulting in excess ectopic α-smooth muscle actin-positive VSMCs. Furthermore, we identified endothelial SEMA3C as an attractant for macrophages. In vivo neutralization of SEMA3C rescued macrophage accumulation and ectopic VSMC phenotypes in the mice, as well as drug-induced macrophage depletion. Therefore, macrophages play an important physiological role in VSMC development via the FOXO1-SEMA3C pathway.


Asunto(s)
Proteína Forkhead Box O1 , Macrófagos , Músculo Liso Vascular , Miocitos del Músculo Liso , Semaforinas , Animales , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citología , Ratones , Semaforinas/metabolismo , Semaforinas/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/citología , Pericitos/metabolismo , Pericitos/citología , Diferenciación Celular , Transducción de Señal , Vasos Retinianos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Factor de Crecimiento Transformador beta1/metabolismo , Ratones Endogámicos C57BL
2.
Development ; 147(2)2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31852686

RESUMEN

Lymphangiogenesis plays important roles in normal fetal development and postnatal growth. However, its molecular regulation remains unclear. Here, we have examined the function of forkhead box protein O1 (FOXO1) transcription factor, a known angiogenic factor, in developmental dermal lymphangiogenesis using endothelial cell-specific FOXO1-deficient mice. FOXO1-deficient mice showed disconnected and dilated lymphatic vessels accompanied with increased proliferation and decreased apoptosis in the lymphatic capillaries. Comprehensive DNA microarray analysis of the causes of in vivo phenotypes in FOXO1-deficient mice revealed that the gene encoding C-X-C chemokine receptor 4 (CXCR4) was the most drastically downregulated in FOXO1-deficient primary lymphatic endothelial cells (LECs). CXCR4 was expressed in developing dermal lymphatic capillaries in wild-type mice but not in FOXO1-deficient dermal lymphatic capillaries. Furthermore, FOXO1 suppression impaired migration toward the exogenous CXCR4 ligand, C-X-C chemokine ligand 12 (CXCL12), and coordinated proliferation in LECs. These results suggest that FOXO1 serves an essential role in normal developmental lymphangiogenesis by promoting LEC migration toward CXCL12 and by regulating their proliferative activity. This study provides valuable insights into the molecular mechanisms underlying developmental lymphangiogenesis.


Asunto(s)
Dermis/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Linfangiogénesis/genética , Receptores CXCR4/genética , Cola (estructura animal)/metabolismo , Regulación hacia Arriba/genética , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Apoptosis , Secuencia de Bases , Cadherinas/metabolismo , Muerte Celular , Proliferación Celular , Quimiocina CXCL12/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Elementos de Facilitación Genéticos/genética , Eliminación de Gen , Integrasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Regiones Promotoras Genéticas/genética , Unión Proteica , Receptores CXCR4/metabolismo
3.
Biochem Biophys Res Commun ; 521(4): 827-832, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31708102

RESUMEN

Interactions between Sema4D and its receptors, PlexinB1 and CD72, induce various functions, including axon guidance, angiogenesis, and immune activation. Our previous study revealed that Sema4D is involved in the upregulation of nitric oxide production in microglia after cerebral ischemia. In this study, we investigated the underlying mechanisms of the enhancement of microglial nitric oxide production by Sema4D. Primary microglia expressed PlexinB1 and CD72, and cortical microglia expressed CD72. Sema4D promoted nitric oxide production and slightly inhibited Erk1/2 phosphorylation in microglia. Partial Erk1/2 inhibition enhanced microglial nitric oxide production. Inhibition of Erk1/2 phosphorylation induced the expression of Ifn-ß mRNA, and IFN-ß promoted nitric oxide production in microglia. In the ischemic cortex, the expression of Ifn-ß mRNA was downregulated by Sema4D deficiency. These findings indicated that the enhancement of nitric oxide production by Sema4D is involved in partial Erk1/2 inhibition and upregulation of IFN-ß.


Asunto(s)
Interferón beta/metabolismo , Microglía/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Óxido Nítrico/metabolismo , Semaforinas/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Isquemia Encefálica/metabolismo , Células Cultivadas , Flavonoides/farmacología , Interferón beta/genética , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Receptores de Superficie Celular/metabolismo , Semaforinas/genética , Regulación hacia Arriba
4.
Biochem Biophys Res Commun ; 520(2): 304-310, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31601422

RESUMEN

Pericytes are mural cells that cover small blood vessels. While defects in pericyte coverage are known to be involved in various vessel related pathologies, including diabetic retinopathy, the molecular mechanisms underlying pericyte coverage are not fully understood. In this study, we investigated the contribution of the forkhead transcription factor FOXO1 in endothelial cells to pericyte coverage in the developing retina. We observed retinal pericytes in tamoxifen-inducible endothelium-specific Foxo1 deletion mice. Tamoxifen was injected at postnatal day 1-3 and the retinas were harvested at P21. Our results demonstrated that Foxo1 deletion in the endothelium affected arteriole pericyte morphology without altering pericyte number, proliferation, and apoptosis. We hypothesized that abnormal pericyte morphogenesis in the knockout retina was caused by impaired pericyte differentiation. FOXO1 silencing by siRNA in the primary artery endothelium further revealed that THBS1 (thrombospondin 1), which promotes pericyte differentiation via TGFß activation, was reduced in the FOXO1-deficient endothelium. Immunohistochemistry of FOXO1 knockout mice showed reduced numbers of phospho-Smad3+ arteriole pericytes compared with wild-type mice. In addition, endothelium-pericyte co-culture analysis revealed that pericytes cultured with FOXO1-deficient endothelial cells failed to differentiate sufficiently; this failure was partially rescued by the addition of recombinant THBS1 to the supernatant. The findings suggest that endothelial FOXO1 contributes to pericyte differentiation via regulation of THBS1 expression. This study provides new insights into the molecular mechanism of pericyte coverage in the context of endothelium-derived regulation and highlights a new therapeutic target for pericyte-related pathology.


Asunto(s)
Proteína Forkhead Box O1/genética , Pericitos/patología , Retina/citología , Retina/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Diferenciación Celular , Células Cultivadas , Células Endoteliales , Proteína Forkhead Box O1/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/efectos de los fármacos , Tamoxifeno/farmacología , Trombospondinas/metabolismo , Factor de Crecimiento Transformador beta , Arterias Umbilicales/citología
5.
Angiogenesis ; 21(2): 203-214, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29185141

RESUMEN

Forkhead box protein O1 (FoxO1) is a transcription factor and a critical regulator of angiogenesis. Various environmental stimuli, including growth factors, nutrients, shear stress, oxidative stress and hypoxia, affect FoxO1 subcellular localization and strongly influence its transcriptional activity; however, FoxO1-localization patterns in endothelial cells (ECs) during development have not been clarified in vivo. Here, we reported that FoxO1 expression was observed in three layers of angiogenic vessels in developing mouse retinas and that among these layers, the front layer showed high levels of FoxO1 expression in the nuclei of most tip ECs. Because tip ECs migrate toward the avascular hypoxic area, we focused on hypoxia as a major stimulus regulating FoxO1 subcellular localization in tip cells. In cultured ECs, FoxO1 accumulated into the nucleus under hypoxic conditions, with hypoxia also inducing expression of tip-cell-specific genes, including endothelial-specific molecule 1 (ESM1), which was suppressed by FoxO1 knockdown. Additionally, in murine models, EC-specific FoxO1 deletion resulted in reduced ESM1 expression and suppressed tip-cell migration during angiogenesis. These findings indicated roles for FoxO1 in tip-cell migration and that its transcriptional activity is regulated by hypoxia.


Asunto(s)
Células Endoteliales/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Hipoxia/metabolismo , Retina/crecimiento & desarrollo , Neovascularización Retiniana/metabolismo , Animales , Células Endoteliales/patología , Proteína Forkhead Box O1/genética , Técnicas de Silenciamiento del Gen , Humanos , Hipoxia/genética , Hipoxia/patología , Ratones , Ratones Transgénicos , Retina/patología , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología
6.
Biochem Biophys Res Commun ; 489(4): 413-419, 2017 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-28559138

RESUMEN

Sprouting migration of lymphatic endothelial cell (LEC) is a pivotal step in lymphangiogenic process. However, its molecular mechanism remains unclear including effective migratory attractants. Meanwhile, forkhead transcription factor FOXO1 highly expresses in LEC nuclei, but its significance in LEC migratory activity has not been researched. In this study, we investigated function of FOXO1 transcription factor associated with LEC migration toward exogenous ATP which has recently gathered attentions as a cell migratory attractant. The transwell membrane assay indicated that LECs migrated toward exogenous ATP, which was impaired by FOXO1 knockdown. RT-PCR analysis showed that P2Y1, a purinergic receptor, expression was markedly reduced by FOXO1 knockdown in LECs. Moreover, P2Y1 blockage impaired LEC migration toward exogenous ATP. Western blot analysis revealed that Akt phosphorylation contributed to FOXO1-dependent LEC migration toward exogenous ATP and its blockage affected LEC migratory activity. Furthermore, luciferase reporter assay and ChIP assay suggested that FOXO1 directly bound to a conserved binding site in P2RY1 promoter and regulated its activity. These results indicated that FOXO1 serves a pivotal role in LEC migration toward exogenous ATP via direct transcriptional regulation of P2Y1 receptor.


Asunto(s)
Adenosina Trifosfato/metabolismo , Movimiento Celular , Células Endoteliales/metabolismo , Proteína Forkhead Box O1/metabolismo , Receptores Purinérgicos P2Y1/genética , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Receptores Purinérgicos P2Y1/metabolismo
7.
Cell Rep ; 37(9): 110048, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34852224

RESUMEN

Intraluminal lymphatic valves (LVs) contribute to the prevention of lymph backflow and maintain circulatory homeostasis. Several reports have investigated the molecular mechanisms which promote LV formation; however, the way in which they are suppressed is not completely clear. We show that the forkhead transcription factor FOXO1 is a suppressor of LV formation and maintenance in lymphatic endothelial cells. Oscillatory shear stress by bidirectional flow inactivates FOXO1 via Akt phosphorylation, resulting in the upregulation of a subset of LV-specific genes mediated by downregulation of a transcriptional repressor, PRDM1. Mice with an endothelial-specific Foxo1 deletion have an increase in LVs, and overexpression of Foxo1 in mice produces a decrease in LVs. Genetic reduction of PRDM1 rescues the decrease in LV by Foxo1 overexpression. In conclusion, FOXO1 plays a critical role in lymph flow homeostasis by preventing excess LV formation. This gene might be a therapeutic target for lymphatic circulatory abnormalities.


Asunto(s)
Proteína Forkhead Box O1/fisiología , Linfangiogénesis , Vasos Linfáticos/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Animales , Femenino , Humanos , Vasos Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Transducción de Señal
8.
Small GTPases ; 10(3): 169-177, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-28448737

RESUMEN

Small GTPases play crucial roles in the maintenance of a homeostatic environment and appropriate movements of the cell. In these processes, the direct or indirect interaction between distinct small GTPases could be required for regulating mutual signaling pathways. In our recent study, ARHGEF10, known as a guanine nucleotide exchange factor (GEF) for RhoA, was indicated to interact with Rab6A and Rab8A, which are known to function in the exocytotic pathway, and colocalized with these Rabs at exocytotic vesicles. Moreover, it was suggested that ARHGEF10 is involved in the regulation of Rab6A and Rab8A localization and invasion of breast carcinoma cells, in which Rab8 also acts via regulation of membrane trafficking. These results may reveal the existence of a novel small GTPase cascade which connects the signaling of these Rabs with RhoA during membrane trafficking. In this mini-review, we consider the possible functions of ARHGEF10 and RhoA in the Rab6- and Rab8-mediated membrane trafficking pathway.


Asunto(s)
Neoplasias de la Mama/metabolismo , Membrana Celular/metabolismo , Exocitosis , Proteínas de Neoplasias/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Transporte Biológico Activo/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Membrana Celular/genética , Membrana Celular/patología , Femenino , Humanos , Proteínas de Neoplasias/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Proteínas de Unión al GTP rab/genética , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
9.
Neuroscience ; 406: 420-431, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30922994

RESUMEN

Cerebral ischemia induces neuroinflammation and microglial activation, in which activated microglia upregulate their proliferative activity and change their metabolic states. In activated microglia, l-arginine is metabolized competitively by inducible nitric oxide synthase (iNOS) and arginase (Arg), which then synthesize NO or polyamines, respectively. Our previous study demonstrated that Sema4D deficiency inhibits iNOS expression and promotes proliferation of ionized calcium-binding adaptor molecule 1 (Iba1)-positive (Iba1+) microglia in the ischemic cortex, although the underlying mechanisms were unclear. Using middle cerebral artery occlusion, we tested the hypothesis that Sema4D deficiency alters the balance of l-arginine metabolism between iNOS and Arg, leading to an increase in the production of polyamines, which are an essential factor for cell proliferation. In the peri-ischemic cortex, almost all iNOS+ and/or Arg1+ cells were Iba1+ microglia. In the peri-ischemic cortex of Sema4D-deficient (Sema4D-/-) mice, the number of iNOS+ Arg1- Iba1+ microglia was smaller and that of iNOS- Arg1+ Iba1+ microglia was greater than those of wild-type (WT) mice. In addition, urea and polyamine levels in the ischemic cortex of Sema4D-/- mice were higher than those of WT mice; furthermore, the presence of Sema4D inhibited polyamine production in primary microglia obtained from Sema4D-/- mice. Finally, microglia cultured under polyamine putrescine-supplemented conditions demonstrated increased proliferation rates over non-supplemented controls. These findings indicate that Sema4D regulates microglial proliferation at least in part by regulating the competitive balance of l-arginine metabolism.


Asunto(s)
Arginina/metabolismo , Isquemia Encefálica/metabolismo , Proliferación Celular/fisiología , Corteza Cerebral/metabolismo , Microglía/metabolismo , Semaforinas/deficiencia , Animales , Isquemia Encefálica/patología , Células Cultivadas , Corteza Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología
10.
J Vet Med Sci ; 73(6): 801-3, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21224528

RESUMEN

An 11-year-old male Labrador retriever presented with chronic oliguria. Ultrasonography findings revealed a protruding mass at the neck of the urinary bladder. A cystotomy was performed, and the mass was removed by ligation with surgical sutures. Histopathological examination revealed conspicuous foci with a variable number of ganglion cells in the tumor and abundant interwoven bundles of schwannian cells with fine fibers. The ganglion cells were positive for neuron-specific enolase and neurofilament. The schwannian cells were positive for vimentin, S-100 protein, and glial fibrillary acidic protein. Thus, according to the classification of tumor with neuronal cell differentiation, the urinary tumor was diagnosed as a ganglioneuroma.


Asunto(s)
Enfermedades de los Perros/patología , Ganglioneuroma/patología , Ganglioneuroma/veterinaria , Neoplasias de la Vejiga Urinaria/veterinaria , Animales , Cistotomía/veterinaria , Enfermedades de los Perros/metabolismo , Enfermedades de los Perros/cirugía , Perros , Ganglioneuroma/metabolismo , Ganglioneuroma/cirugía , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica/veterinaria , Filamentos Intermedios/metabolismo , Masculino , Fosfopiruvato Hidratasa/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/cirugía , Vimentina/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda