Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
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.
Ultrasound Med Biol ; 49(2): 460-472, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36335054

RESUMEN

Previously, we developed an instrument to evaluate the heel fat pad (HFP) two-layer structure, under varying loading conditions, with ultrasonography from the plantar surface through a polymethylpentene resin plate; the measured values were equivalent to those obtained without this plate. The study described here aimed to determine the intra- and inter-examiner reliabilities of the HFP thickness measurements and the agreement between long- and short-axis measured values using this instrument. Two examiners successively recorded the HFPs of 40 healthy adults twice under the no loading and loading conditions on the long- and short-axis scans. The HFPs were classified into two layers, and their thicknesses were measured. Short-term intra- and inter-examiner reliabilities were determined using the intraclass correlation coefficients. Measurements were repeated 1 mo later to determine the long-term intra-examiner reliability. The agreement between the measured long- and short-axis values was investigated by calculating the minimal detectable changes. The determined short- and long-term intra-examiner reliabilities ranged from 0.750 to 0.999 and from 0.765 to 0.952, respectively. Inter-examiner reliability ranged from 0.765 to 0.997. Differences may occur between the values measured at different axes. The measurements using this evaluation instrument were reliable, and it is best to unify the measurement axis for quantitative research.


Asunto(s)
Talón , Ultrasonido , Adulto , Humanos , Talón/diagnóstico por imagen , Reproducibilidad de los Resultados , Ultrasonografía , Tejido Adiposo/diagnóstico por imagen
3.
J Comp Neurol ; 530(11): 2033-2055, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35368102

RESUMEN

The structural plasticity of dendritic spines serves as the adaptive capabilities of the central nervous system to various stimuli. Among these stimuli, cerebral ischemia induces dynamic alterations in neuronal network activity. Arcadlin/Paraxial protocadherin/Protocadherin-8 (Acad), a regulator of dendritic spine density, is strongly induced by activating stimuli to the neurons. However, the detailed distribution of Acad in normal and ischemic adult brains remains unclear. We comprehensively described Acad expression patterns in normal and ischemic adult brains by in situ hybridization histochemistry. We found that intact adult brains expressed Acad in the piriform cortex, dentate gyrus, hippocampal CA3, entorhinal cortex, amygdala, and hypothalamus. Acad expression was dramatically upregulated in the piriform cortex, olfactory area, dentate gyrus, entorhinal cortex, prefrontal cortex, insular cortex, amygdala, and septohippocampal nucleus 4 h after cerebral ischemia. Cerebral ischemia induced widespread neuronal activation, which was required for Acad upregulation. Our data suggested the involvement of Acad in the adaptive plasticity and remodeling of the neuronal network in the limbic and paralimbic systems.


Asunto(s)
Isquemia Encefálica , Protocadherinas , ARN Mensajero , Animales , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Ratones , Protocadherinas/genética , Protocadherinas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Ultrasound Med Biol ; 48(2): 358-372, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34823945

RESUMEN

To evaluate the two-layer structure of the heel fat pad (HFP) from non-weight-bearing to full-weight-bearing conditions, we developed an instrument that assesses these changes from the sole through a polymethylpentene resin plate (PMP) with ultrasonography. For actual use, we investigated the influence on measured values and ultrasonogram appearance by interposing the PMP. Additionally, as the PMP may be bent under weight-bearing conditions, its influence on the measured values was investigated. First, two examiners measured the distances inside the phantom with and without a PMP. Second, ultrasonograms were obtained from 40 healthy adults with and without a PMP, and the thicknesses of the whole layer and the two layers of the HFP were measured using the same ultrasonogram. For each experiment, reproducibility was investigated. Third, the distances inside the phantom were measured and compared through the bent PMP, which models the weight-bearing condition, and the flat PMP. The reproducibility of the measurements was equivalent with and without the PMP interposed. Potential bias in measured values arising from deformation of PMP under weight-bearing conditions was not detected. Overall, the PMP's interposition and the bending of the PMP might not influence the measured values and reproducibility of the measurements.


Asunto(s)
Talón , Ultrasonido , Tejido Adiposo , Adulto , Talón/diagnóstico por imagen , Humanos , Reproducibilidad de los Resultados , Soporte de Peso
5.
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
6.
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
7.
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
8.
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
9.
FEBS J ; 286(18): 3701-3717, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31120172

RESUMEN

Development of the salivary gland is characterized by extensive branching morphogenesis. Although various molecules have been implicated in salivary gland development, the role of the mammalian target of rapamycin (mTOR) signalling pathway, including both mTOR complexes 1 and 2 (mTORC1 and 2), in salivary gland development is unknown. Here, we examined protein expression levels related to the mTOR signalling pathway using an ex vivo submandibular salivary gland (SMG) organ culture. We showed that branching buds in the salivary glands were substantially decreased and phosphorylation of mTORC1 signalling pathway related proteins (mTOR, p70 ribosomal protein S6 kinase 1 and eukaryotic initiation factor 4E-binding protein 1) was inhibited by rapamycin (an mTOR inhibitor). In addition, AKT, which is an upstream protein kinase of mTORC1 and is downstream of mTORC2, is inhibited by LY294002 (a phosphatidylinositol 3-kinase inhibitor), but not by rapamycin. Moreover, rapamycin-treated ICR neonatal mice exhibited a reduction in both body weight and salivary glands compared with vehicle-treated neonatal mice. The present data indicate that the mTOR signalling pathway, including both mTORC1 and mTORC2, plays a critical role in salivary gland development both in ex vivo SMG organ culture and ICR neonatal mice in vivo.


Asunto(s)
Desarrollo Embrionario/genética , Glándulas Salivales/metabolismo , Glándula Submandibular/metabolismo , Serina-Treonina Quinasas TOR/genética , Animales , Animales Recién Nacidos , Cromonas/farmacología , Embrión de Mamíferos , Diana Mecanicista del Complejo 1 de la Rapamicina/efectos de los fármacos , Diana Mecanicista del Complejo 2 de la Rapamicina/efectos de los fármacos , Ratones , Morfogénesis/genética , Morfolinas/farmacología , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Glándulas Salivales/crecimiento & desarrollo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Glándula Submandibular/crecimiento & desarrollo
10.
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
11.
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
12.
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
13.
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
14.
Neuroscience ; 346: 43-51, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28077279

RESUMEN

The state of microglial activation provides important information about the central nervous system. However, a reliable index of microglial activation in histological samples has yet to be established. Here, we show that microglial activation induces topological changes of Iba1 localization that can be detected by analysis based on homology theory. Analysis of homology was applied to images of Iba1-stained tissue sections, and the 0-dimentional Betti number (b0: the number of solid components) and the 1-dimentional Betti number (b1: the number of windows surrounded by solid components) were obtained. We defined b1/b0 as the Homology Value (HV), and investigated its validity as an index of microglial activation using cerebral ischemia model mice. Microglial activation was accompanied by changes to Iba1 localization and morphology of microglial processes. In single microglial cells, the change of Iba1 localization increased b1. Conversely, thickening or retraction of microglial processes decreased b0. Consequently, microglial activation increased the HV. The HV of a tissue area increased with proximity to the ischemic core and showed a high degree of concordance with the number of microglia expressing activation makers. Furthermore, the HV of human metastatic brain tumor tissue also increased with proximity to the tumor. These results suggest that our index, based on homology theory, can be used to correctly evaluate microglial activation in various tissue images.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Inmunohistoquímica/métodos , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Animales , Isquemia Encefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Femenino , Humanos , Masculino , Ratones
15.
J Cell Sci ; 129(19): 3620-3634, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27550519

RESUMEN

The function of ARHGEF10, a known guanine nucleotide exchange factor (GEF) for RhoA with proposed roles in various diseases, is poorly understood. To understand the precise function of this protein, we raised a monoclonal antibody against ARHGEF10 and determined its localization in HeLa cells. ARHGEF10 was found to localize to vesicles containing Rab6 (of which there are three isoforms, Rab6a, Rab6b and Rab6c), Rab8 (of which there are two isoforms, Rab8a and Rab8b), and/or the secretion marker neuropeptide Y (NPY)-Venus in a Rab6-dependent manner. These vesicles were known to originate from the Golgi and contain secreted or membrane proteins. Ectopic expression of an N-terminal-truncated ARHGEF10 mutant led to the generation of large vesicle-like structures containing both Rab6 and Rab8. Additionally, small interfering (si)RNA-mediated knockdown of ARHGEF10 impaired the localization of Rab8 to these exocytotic vesicles. Furthermore, the invasiveness of MDA-MB231 cells was markedly decreased by knockdown of ARHGEF10, as well as of Rab8. From these results, we propose that ARHGEF10 acts in exocytosis and tumor invasion in a Rab8-dependent manner.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Vesículas Secretoras/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Citoesqueleto de Actina/metabolismo , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos/inmunología , Línea Celular Tumoral , Exocitosis , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Mutantes/metabolismo , Invasividad Neoplásica , Neuropéptido Y/metabolismo , Polimerizacion , Dominios Proteicos , Transporte de Proteínas
16.
Neurosci Res ; 108: 6-11, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26752319

RESUMEN

Sema4D, originally identified as a negative regulator of axon guidance during development, is involved in various physiological and pathological responses. In this study, we evaluated the effect of Sema4D-deficiency on oligodendrocyte restoration after the cerebral ischemia/reperfusion using direct ligation of the middle cerebral artery followed by reperfusion. In both Sema4D(+/+) wild-type and Sema4D(-/-) null mutant mice, the peri-infarct area showed a decrease in the number of oligodendrocytes at 3 days post-reperfusion. Subsequently, the number of oligodendrocytes was observed to gradually recover in both groups. Sema4D-deficient mice, however, showed an enhanced recovery of oligodendrocytes and an upregulation of oligodendrocyte progenitor cells at days 14 and 28 of reperfusion. Cell proliferation identified by incorporation of bromodeoxyuridine was enhanced in Sema4D(-/-) mice from days 3 to 14 post-reperfusion compared to the Sema4D(+/+) mice. Furthermore, apoptotic cell death of oligodendrocytes was reduced at days 7 post-reperfusion in Sema4D(-/-) mice compared to Sema4D(+/+) mice. These findings indicate that enhanced proliferation of progenitor cells and survival of oligodendrocytes resulted in improved oligodendrocyte recovery in Sema4D(-/-) mice. This may provide a new approach for neurorestorative treatment in patients with stroke, which aims to manipulate endogenous oligodendrogenesis and thereby to promote brain repair after stroke.


Asunto(s)
Isquemia Encefálica/patología , Corteza Cerebral/patología , Oligodendroglía/patología , Daño por Reperfusión/patología , Semaforinas/metabolismo , Animales , Isquemia Encefálica/metabolismo , Proliferación Celular , Supervivencia Celular , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/metabolismo , Semaforinas/genética
17.
Glia ; 63(12): 2249-59, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26202989

RESUMEN

Cerebral ischemia evokes neuroinflammatory response. Inflammatory stimulation induces microglial activation, such as changes of their morphology from ramified to ameboid, expression of iNOS and cytokines, and the elevation of proliferative activity. Activated microglia play important roles in pathogenesis of cerebral ischemia. A previous study indicated that Sema4D promoted iNOS expression in cultured microglia; however, roles of Sema4D on microglial activation in ischemic injury remains unclear. We investigated the effect of Sema4D-deficiency on microglial activation by using permanent middle cerebral artery occlusion (MCAO) in mice. In this study, ischemia-induced activated microglia were classified into activated-ramified microglia and ameboid microglia based on their morphology. We demonstrated that the rate of iNOS expression in activated-ramified microglia was lower than that in ameboid microglia, while the most proliferating microglia were activated-ramified microglia but not ameboid microglia after cerebral ischemia. Sema4D-deficiency decreased the number of ameboid microglia and iNOS-expressing activated-ramified microglia in the peri-ischemic cortex. These changes by Sema4D-deficiency contributed to the reduction of NO production that was estimated by nitrite concentration in ischemic cortex. On the other hand, Sema4D-deficiency promoted proliferation of microglia in the peri-ischemic cortex. Importantly, ischemia-induced apoptosis and postischemic behavioral abnormality were moderated in Sema4D(-/-) mice. These findings suggest that Sema4D promotes cytotoxic activation of microglia and inhibits functional recovery after cerebral ischemia.


Asunto(s)
Corteza Cerebral/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Microglía/metabolismo , Semaforinas/metabolismo , Animales , Apoptosis/fisiología , Proteínas de Unión al Calcio/metabolismo , Proliferación Celular/fisiología , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Microglía/patología , Actividad Motora/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Recuperación de la Función/fisiología
18.
PLoS One ; 9(1): e86606, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24466168

RESUMEN

BACKGROUND: Alkaptonuria, caused by a deficiency of homogentisate 1,2-dioxygenase, results in the accumulation of homogentisic acid (2,5-dihydroxyphenylacetic acid, HGA) in the urine. Alkaptonuria is suspected when the urine changes color after it is left to stand at room temperature for several hours to days; oxidation of homogentisic acid to benzoquinone acetic acid underlies this color change, which is accelerated by the addition of alkali. In an attempt to develop a facile screening test for alkaptonuria, we added alkali to urine samples obtained from patients with alkaptonuria and measured the absorbance spectra in the visible light region. METHODS: We evaluated the characteristics of the absorption spectra of urine samples obtained from patients with alkaptonuria (n = 2) and compared them with those of urine specimens obtained from healthy volunteers (n = 5) and patients with phenylketonuria (n = 3), and also of synthetic homogentisic acid solution after alkalization. Alkalization of the urine samples and HGA solution was carried out by the addition of NaOH, KOH or NH4OH. The sample solutions were incubated at room temperature for 1 min, followed by measurement of the absorption spectra. RESULTS: Addition of alkali to alkaptonuric urine yielded characteristic absorption peaks at 406 nm and 430 nm; an identical result was obtained from HGA solution after alkalization. The absorbance values at both 406 nm and 430 nm increased in a time-dependent manner. In addition, the absorbance values at these peaks were greater in strongly alkaline samples (NaOH- KOH-added) as compared with those in weakly alkaline samples (NH4OH-added). In addition, the peaks disappeared following the addition of ascorbic acid to the samples. CONCLUSIONS: We found two characteristic peaks at 406 nm and 430 nm in both alkaptonuric urine and HGA solution after alkalization. This new quick and easy method may pave the way for the development of an easy method for the diagnosis of alkaptonuria.


Asunto(s)
Alcaptonuria/orina , Ácido Homogentísico/orina , Hidróxidos/farmacología , Fenilcetonurias/orina , Compuestos de Potasio/farmacología , Hidróxido de Sodio/farmacología , Adulto , Alcaptonuria/diagnóstico , Estudios de Casos y Controles , Femenino , Voluntarios Sanos , Humanos , Luz , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Fenilcetonurias/diagnóstico , Espectrofotometría , Adulto Joven
19.
Genes Cells ; 17(9): 758-67, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22845550

RESUMEN

Several reports have suggested that Foxo1, a key regulator in differentiation, growth and metabolism, is involved in pancreatic ß-cell function. However, detailed analyses have been hampered by a lack of Foxo1-deficient ß cells. To elucidate Foxo1's function in ß cells, we produced a ß-cell line with inducible Foxo1 deletion. We generated a conditional knockout mouse line, in which Cre recombinase deletes the Foxo1 gene. We then established a ß-cell line from an insulinoma induced in this knockout mouse by the ß-cell-specific expression of simian virus 40 T antigen. In this cell line, designated MIN6-Foxo1flox/flox, adenovirus-mediated Cre expression ablates the Foxo1 gene, generating MIN6-Foxo1-KO cells. Using these knockout and floxed cell lines, we found that Foxo1 ablation enhanced the glucose-stimulated insulin secretion (GSIS) at high glucose concentrations and enhanced ß-cell proliferation. We also conducted DNA microarray analyses of MIN6-Foxo1-KO cells infected with either an adenovirus vector expressing a constitutively active FOXO1 or a control vector and identified several Foxo1-regulated genes, including some known to be related to ß-cell function. These cells should be useful for further studies on Foxo1's roles in ß-cells and may lead to novel strategies for treating the impaired insulin secretion in type 2 diabetes mellitus.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Alelos , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Apoptosis , Línea Celular , Proliferación Celular , Quimera/genética , Quimera/metabolismo , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glucosa/farmacología , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Virus 40 de los Simios/genética , Virus 40 de los Simios/metabolismo
20.
Mol Cell Neurosci ; 49(3): 290-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22198439

RESUMEN

The specific functions of intrinsic regulators of OL differentiation are poorly understood. Sema4D, originally found as a negative regulator of axon guidance, is mainly expressed by oligodendrocytes in the postnatal brain, and our previous study revealed that the lack of Sema4D induced an increase in the number of oligodendrocytes in the cerebral cortex, suggesting that Sema4D may function as an intrinsic regulator of oligodendrocyte development. In this study, we assessed the effects of Sema4D deficiency and of the exogenous addition of Sema4D on oligodendrocyte differentiation. Sema4D deficiency induced an increase in the number of oligodendrocytes in the cerebral cortex at postnatal day 14 and later, without increase in the number of oligodendrocyte progenitor cells. This increase was also observed in cultured oligodendrocytes obtained from Sema4D-deficient mice. Then we investigated whether Sema4D deficiency can increase the proliferation of the progenitor cells or influence the apoptosis. Apoptotic oligodendrocytes were markedly reduced in number in the developing cerebral cortex and in cultured oligodendrocytes obtained from Sema4D-deficient mice, although no significant change was found in proliferation of oligodendrocyte progenitor cells. Exogenous addition of Sema4D prevented the oligodendrocytes from this reduction of apoptosis, and further enhanced apoptosis in oligodendrocytes. Thus, Sema4D may act as an intrinsic inhibitory regulator of oligodendrocyte differentiation by promoting apoptosis.


Asunto(s)
Apoptosis/fisiología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Semaforinas/metabolismo , Animales , Apoptosis/genética , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/genética , Células Cultivadas , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Ratones , Ratones Noqueados , Células Madre/citología , Células Madre/metabolismo , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA