Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nat Med ; 12(6): 657-64, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16715089

RESUMEN

Here we investigated the potential role of bone-resorbing osteoclasts in homeostasis and stress-induced mobilization of hematopoietic progenitors. Different stress situations induced activity of osteoclasts (OCLs) along the stem cell-rich endosteum region of bone, secretion of proteolytic enzymes and mobilization of progenitors. Specific stimulation of OCLs with RANKL recruited mainly immature progenitors to the circulation in a CXCR4- and MMP-9-dependent manner; however, RANKL did not induce mobilization in young female PTPepsilon-knockout mice with defective OCL bone adhesion and resorption. Inhibition of OCLs with calcitonin reduced progenitor egress in homeostasis, G-CSF mobilization and stress situations. RANKL-stimulated bone-resorbing OCLs also reduced the stem cell niche components SDF-1, stem cell factor (SCF) and osteopontin along the endosteum, which was associated with progenitor mobilization. Finally, the major bone-resorbing proteinase, cathepsin K, also cleaved SDF-1 and SCF. Our findings indicate involvement of OCLs in selective progenitor recruitment as part of homeostasis and host defense, linking bone remodeling with regulation of hematopoiesis.


Asunto(s)
Resorción Ósea , Huesos/anatomía & histología , Movimiento Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Osteoclastos/metabolismo , Animales , Proteínas Portadoras/metabolismo , Catepsina K , Catepsinas/genética , Catepsinas/metabolismo , Línea Celular , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Femenino , Células Madre Hematopoyéticas/citología , Homeostasis , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Osteoclastos/citología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Receptores CXCR4/metabolismo , Factor de Células Madre/metabolismo
2.
Blood ; 111(10): 4934-43, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18334674

RESUMEN

Heparanase is involved in tumor growth and metastasis. Because of its unique cleavage of heparan sulfate, which binds cytokines, chemokines and proteases, we hypothesized that heparanase is also involved in regulation of early stages of hematopoiesis. We report reduced numbers of maturing leukocytes but elevated levels of undifferentiated Sca-1(+)/c-Kit(+)/Lin(-) cells in the bone marrow (BM) of mice overexpressing heparanase (hpa-Tg). This resulted from increased proliferation and retention of the primitive cells in the BM microenvironment, manifested in increased SDF-1 turnover. Furthermore, heparanase overexpression in mice was accompanied by reduced protease activity of MMP-9, elastase, and cathepsin K, which regulate stem and progenitor cell mobilization. Moreover, increased retention of the progenitor cells also resulted from up-regulated levels of stem cell factor (SCF) in the BM, in particular in the stem cell-rich endosteum and endothelial regions. Increased SCF-induced adhesion of primitive Sca-1(+)/c-Kit(+)/Lin(-) cells to osteoblasts was also the result of elevation of the receptor c-Kit. Regulation of these phenomena is mediated by hyperphosphorylation of c-Myc in hematopoietic progenitors of hpa-Tg mice or after exogenous heparanase addition to wildtype BM cells in vitro. Altogether, our data suggest that heparanase modification of the BM microenvironment regulates the retention and proliferation of hematopoietic progenitor cells.


Asunto(s)
Médula Ósea , Proliferación Celular , Glucuronidasa/fisiología , Células Madre Hematopoyéticas/citología , Animales , Células de la Médula Ósea , Adhesión Celular , Movimiento Celular , Quimiocina CXCL12/metabolismo , Inmunofenotipificación , Ratones , Ratones Transgénicos , Proteínas de Neoplasias , Péptido Hidrolasas/metabolismo
3.
Nat Commun ; 11(1): 3547, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669546

RESUMEN

Neutrophils provide first line of host defense against bacterial infections utilizing glycolysis for their effector functions. How glycolysis and its major byproduct lactate are triggered in bone marrow (BM) neutrophils and their contribution to neutrophil mobilization in acute inflammation is not clear. Here we report that bacterial lipopolysaccharides (LPS) or Salmonella Typhimurium triggers lactate release by increasing glycolysis, NADPH-oxidase-mediated reactive oxygen species and HIF-1α levels in BM neutrophils. Increased release of BM lactate preferentially promotes neutrophil mobilization by reducing endothelial VE-Cadherin expression, increasing BM vascular permeability via endothelial lactate-receptor GPR81 signaling. GPR81-/- mice mobilize reduced levels of neutrophils in response to LPS, unless rescued by VE-Cadherin disrupting antibodies. Lactate administration also induces release of the BM neutrophil mobilizers G-CSF, CXCL1 and CXCL2, indicating that this metabolite drives neutrophil mobilization via multiple pathways. Our study reveals a metabolic crosstalk between lactate-producing neutrophils and BM endothelium, which controls neutrophil mobilization under bacterial infection.


Asunto(s)
Células de la Médula Ósea/inmunología , Ácido Láctico/metabolismo , Neutrófilos/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Infecciones por Salmonella/inmunología , Animales , Médula Ósea/irrigación sanguínea , Células de la Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Femenino , Humanos , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Receptores Acoplados a Proteínas G/genética , Infecciones por Salmonella/microbiología , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología
4.
Brain Behav Immun ; 23(8): 1059-65, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19341792

RESUMEN

The nervous system regulates immunity through hormonal and neuronal routes as part of host defense and repair mechanism. Here, we review the emerging evidence for regulation of human hematopoietic stem and progenitor cells (HSPC) by the nervous system both directly and indirectly via their bone marrow (BM) niche-supporting stromal cells. Functional expression of several neurotransmitter receptors was demonstrated on HSPC, mainly on the more primitive CD34(+)/CD38(-/low) fraction. The myeloid cytokines, G-CSF and GM-CSF, dynamically upregulate neuronal receptor expression on human HSPC. This is followed by an increased response to neurotransmitters, leading to enhanced proliferation and motility of human CD34(+) progenitors, repopulation of the murine BM and their egress to the circulation. Importantly, recent observations showed rapid mobilization of human HSPC to high SDF-1 expressing ischemic tissues of stroke individuals followed by neoangiogenesis, neurological and functional recovery. Along with decreased levels of circulating immature CD34(+) cells and SDF-1 blood levels found in patients with early-stage Alzheimer's disease, these findings suggest a possible involvement of human HSPC in brain homeostasis and thus their potential clinical applications in neuropathology.


Asunto(s)
Antígenos CD34/metabolismo , Células Madre Hematopoyéticas/metabolismo , Sistema Nervioso/metabolismo , Receptores de Neurotransmisores/metabolismo , Médula Ósea/metabolismo , Hematopoyesis , Humanos , Neurotransmisores/metabolismo , Células del Estroma/metabolismo
5.
J Leukoc Biol ; 84(4): 1130-40, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18653785

RESUMEN

The chemokine CXCL12 promotes migration of human leukocytes, hematopoietic progenitors, and tumor cells. The binding of CXCL12 to its receptor CXCR4 triggers Gi protein signals for motility and integrin activation in many cell types. CXCR7 is a second, recently identified receptor for CXCL12, but its role as an intrinsic G-protein-coupled receptor (GPCR) has been debated. We report that CXCR7 fails to support on its own any CXCL12-triggered integrin activation or motility in human T lymphocytes or CD34(+) progenitors. CXCR7 is also scarcely expressed on the surface of both cell types and concentrates right underneath the plasma membrane with partial colocalization in early endosomes. Nevertheless, various specific CXCR7 blockers get access to this pool and attenuate the ability of CXCR4 to properly rearrange by surface-bound CXCL12, a critical step in the ability of the GPCR to trigger optimal CXCL12-mediated stimulation of integrin activation in T lymphocytes as well as in CD34(+) cells. In contrast, CXCL12-triggered CXCR4 signaling to early targets, such as Akt as well as CXCR4-mediated chemotaxis, is insensitive to identical CXCR7 blocking. Our findings suggest that although CXCR7 is not an intrinsic signaling receptor for CXCL12 on lymphocytes or CD34(+) cells, its blocking can be useful for therapeutic interference with CXCR4-mediated activation of integrins.


Asunto(s)
Antígenos CD34/fisiología , Movimiento Celular/fisiología , Quimiocina CXCL12/fisiología , Quimiotaxis de Leucocito/fisiología , Integrinas/fisiología , Receptores CXCR4/fisiología , Receptores CXCR/fisiología , Linfocitos T/fisiología , Citometría de Flujo , Humanos , Receptor Cross-Talk , Receptores CXCR/antagonistas & inhibidores , Receptores CXCR4/antagonistas & inhibidores
6.
J Clin Invest ; 115(1): 168-76, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15630457

RESUMEN

The chemokine stromal cell-derived factor-1 (SDF-1) and its receptor, CXCR4, play a major role in migration, retention, and development of hematopoietic progenitors in the bone marrow. We report the direct involvement of atypical PKC-zeta in SDF-1 signaling in immature human CD34(+)-enriched cells and in leukemic pre-B acute lymphocytic leukemia (ALL) G2 cells. Chemotaxis, cell polarization, and adhesion of CD34(+) cells to bone marrow stromal cells were found to be PKC-zeta dependent. Overexpression of PKC-zeta in G2 and U937 cells led to increased directional motility to SDF-1. Interestingly, impaired SDF-1-induced migration of the pre-B ALL cell line B1 correlated with reduced PKC-zeta expression. SDF-1 triggered PKC-zeta phosphorylation, translocation to the plasma membrane, and kinase activity. Furthermore we identified PI3K as an activator of PKC-zeta, and Pyk-2 and ERK1/2 as downstream targets of PKC-zeta. SDF-1-induced proliferation and MMP-9 secretion also required PKC-zeta activation. Finally, we showed that in vivo engraftment, but not homing, of human CD34(+)-enriched cells to the bone marrow of NOD/SCID mice was PKC-zeta dependent and that injection of mice with inhibitory PKC-zeta pseudosubstrate peptides resulted in mobilization of murine progenitors. Our results demonstrate a central role for PKC-zeta in SDF-1-dependent regulation of hematopoietic stem and progenitor cell motility and development.


Asunto(s)
Antígenos CD34/metabolismo , Diferenciación Celular , Movimiento Celular , Quimiocinas CXC/metabolismo , Proteína Quinasa C/metabolismo , Células Madre/citología , Células Madre/enzimología , Actinas/metabolismo , Animales , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Activación Enzimática , Sangre Fetal/citología , Sangre Fetal/efectos de los fármacos , Sangre Fetal/metabolismo , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/metabolismo
7.
Nat Cell Biol ; 20(9): 1084-1097, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30154549

RESUMEN

Lack of insight into mechanisms governing breast cancer metastasis has precluded the development of curative therapies. Metastasis-initiating cancer cells (MICs) are uniquely equipped to establish metastases, causing recurrence and therapeutic resistance. Using various metastasis models, we discovered that certain primary tumours elicit a systemic inflammatory response involving interleukin-1ß (IL-1ß)-expressing innate immune cells that infiltrate distant MIC microenvironments. At the metastatic site, IL-1ß maintains MICs in a ZEB1-positive differentiation state, preventing MICs from generating highly proliferative E-cadherin-positive progeny. Thus, when the inherent plasticity of MICs is impeded, overt metastases cannot be established. Ablation of the pro-inflammatory response or inhibition of the IL-1 receptor relieves the differentiation block and results in metastatic colonization. Among patients with lymph node-positive breast cancer, high primary tumour IL-1ß expression is associated with better overall survival and distant metastasis-free survival. Our data reveal complex interactions that occur between primary tumours and disseminated MICs that could be exploited to improve patient survival.


Asunto(s)
Neoplasias de la Mama/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Neoplasias Pulmonares/metabolismo , Células Mieloides/metabolismo , Microambiente Tumoral , Animales , Antiinflamatorios/farmacología , Antígenos CD/genética , Antígenos CD/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Cadherinas/genética , Cadherinas/metabolismo , Comunicación Celular , Diferenciación Celular , Línea Celular Tumoral , Plasticidad de la Célula , Proliferación Celular , Femenino , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Interleucina-1beta/genética , Interleucina-1beta/farmacología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Metástasis Linfática , Ratones Desnudos , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/patología , Transducción de Señal , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
8.
J Clin Invest ; 112(2): 160-9, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12865405

RESUMEN

Hematopoietic stem cells rarely contribute to hepatic regeneration, however, the mechanisms governing their homing to the liver, which is a crucial first step, are poorly understood. The chemokine stromal cell-derived factor-1 (SDF-1), which attracts human and murine progenitors, is expressed by liver bile duct epithelium. Neutralization of the SDF-1 receptor CXCR4 abolished homing and engraftment of the murine liver by human CD34+ hematopoietic progenitors, while local injection of human SDF-1 increased their homing. Engrafted human cells were localized in clusters surrounding the bile ducts, in close proximity to SDF-1-expressing epithelial cells, and differentiated into albumin-producing cells. Irradiation or inflammation increased SDF-1 levels and hepatic injury induced MMP-9 activity, leading to both increased CXCR4 expression and SDF-1-mediated recruitment of hematopoietic progenitors to the liver. Unexpectedly, HGF, which is increased following liver injury, promoted protrusion formation, CXCR4 upregulation, and SDF-1-mediated directional migration by human CD34+ progenitors, and synergized with stem cell factor. Thus, stress-induced signals, such as increased expression of SDF-1, MMP-9, and HGF, recruit human CD34+ progenitors with hematopoietic and/or hepatic-like potential to the liver of NOD/SCID mice. Our results suggest the potential of hematopoietic CD34+/CXCR4+cells to respond to stress signals from nonhematopoietic injured organs as an important mechanism for tissue targeting and repair.


Asunto(s)
Antígenos CD34/biosíntesis , Quimiocinas CXC/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Hígado/metabolismo , Metaloproteinasa 9 de la Matriz/fisiología , Células Madre/citología , Animales , Conductos Biliares/metabolismo , Movimiento Celular , Células Cultivadas , Quimiocina CXCL12 , ADN/metabolismo , Células Madre Hematopoyéticas , Humanos , Inmunohistoquímica , Hígado/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones SCID , Microscopía Fluorescente , ARN Mensajero/metabolismo , Receptores CXCR4/metabolismo , Distribución Tisular , Regulación hacia Arriba
9.
Cancer Discov ; 6(6): 630-49, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27072748

RESUMEN

UNLABELLED: Immune cells promote the initial metastatic dissemination of carcinoma cells from primary tumors. In contrast to their well-studied functions in the initial stages of metastasis, the specific roles of immunocytes in facilitating progression through the critical later steps of the invasion-metastasis cascade remain poorly understood. Here, we define novel functions of neutrophils in promoting intraluminal survival and extravasation at sites of metastatic dissemination. We show that CD11b(+)/Ly6G(+) neutrophils enhance metastasis formation via two distinct mechanisms. First, neutrophils inhibit natural killer cell function, which leads to a significant increase in the intraluminal survival time of tumor cells. Thereafter, neutrophils operate to facilitate extravasation of tumor cells through the secretion of IL1ß and matrix metalloproteinases. These results identify neutrophils as key regulators of intraluminal survival and extravasation through their cross-talk with host cells and disseminating carcinoma cells. SIGNIFICANCE: This study provides important insights into the systemic contributions of neutrophils to cancer metastasis by identifying how neutrophils facilitate intermediate steps of the invasion-metastasis cascade. We demonstrate that neutrophils suppress natural killer cell activity and increase extravasation of tumor cells. Cancer Discov; 6(6); 630-49. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 561.


Asunto(s)
Carcinoma/inmunología , Carcinoma/patología , Células Asesinas Naturales/inmunología , Neutrófilos/inmunología , Traslado Adoptivo , Animales , Biomarcadores , Carcinoma/genética , Carcinoma/metabolismo , Comunicación Celular , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Xenoinjertos , Humanos , Inmunidad Innata , Inmunofenotipificación , Células Asesinas Naturales/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Noqueados , Invasividad Neoplásica , Metástasis de la Neoplasia , Neutrófilos/metabolismo , Fenotipo
10.
Cell Stem Cell ; 3(5): 484-92, 2008 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-18983964

RESUMEN

Physiological interactions between the nervous and immune systems with components of the local microenvironment are needed to maintain homeostasis throughout the body. Dynamic regulation of bone remodeling, hematopoietic stem cells, and their evolving niches via neurotransmitter signaling are part of the host defense and repair mechanisms. This crosstalk links activated leukocytes, neuronal, and stromal cells, which combine to directly and indirectly regulate hematopoietic stem cells. Together, interactions between diverse systems create a regulatory "brain-bone-blood triad," contributing an additional dimension to the concept of the hematopoietic stem cell niche.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Homeostasis/fisiología , Neuroinmunomodulación/fisiología , Comunicación Paracrina/fisiología , Nicho de Células Madre/fisiología , Animales , Diferenciación Celular , Movimiento Celular , Ritmo Circadiano/fisiología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Estrés Fisiológico
11.
Nat Immunol ; 8(10): 1123-31, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17828268

RESUMEN

Catecholamines are important regulators of homeostasis, yet their functions in hematopoiesis are poorly understood. Here we report that immature human CD34+ cells dynamically expressed dopamine and beta2-adrenergic receptors, with higher expression in the primitive CD34+CD38(lo) population. The myeloid cytokines G-CSF and GM-CSF upregulated neuronal receptor expression on immature CD34+ cells. Treatment with neurotransmitters increased the motility, proliferation and colony formation of human progenitor cells, correlating with increased polarity, expression of the metalloproteinase MT1-MMP and activity of the metalloproteinase MMP-2. Treatment with catecholamines enhanced human CD34+ cell engraftment of NOD-SCID mice through Wnt signaling activation and increased cell mobilization and bone marrow Sca-1+c-Kit+Lin- cell numbers. Our results identify new functions for neurotransmitters and myeloid cytokines in the direct regulation of human and mouse progenitor cell migration and development.


Asunto(s)
Antígenos CD34/análisis , Catecolaminas/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Neurotransmisores/farmacología , Transducción de Señal/fisiología , Proteínas Wnt/fisiología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dopamina/farmacología , Agonistas de Dopamina/farmacología , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Norepinefrina/farmacología , Receptores Dopaminérgicos/análisis
12.
Blood ; 103(8): 2900-7, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15070661

RESUMEN

The mechanisms governing migration and extramedullary dissemination of leukemic cells remain obscure. In this study the migration and in vivo homing to the bone marrow of nonobese diabetic severe combined immunodeficient (NOD/SCID) mice injected with human precursor-B acute lymphoblastic leukemia (ALL) cells in comparison to normal CD34+ progenitors (both cord blood and mobilized peripheral blood) was investigated. Although migration and homing of both cell populations was dependent on stromal cell-derived factor 1 (SDF-1)/CXCR4 interactions, major differences in receptor expression as well as the migratory capacity toward various concentrations of SDF-1 were found. Furthermore, unlike normal CD34+ progenitors, in vivo homing of the leukemic cells was superior when recipient NOD/SCID mice were not irradiated prior to transplantation. In addition, we report differences in the adhesion molecules activated following SDF-1 stimulation, documenting a major role for very late antigen 4 (VLA-4), but not VLA-5 and lymphocyte function-associated antigen-1 (LFA-1), in homing of precursor-B ALL cells. Interestingly, Toxin-B and pertussis toxin inhibited the homing of the leukemic cells but not that of normal CD34+ progenitors or normal CD10+/CD19+ precursor-B cells, revealing differences in CXCR4 signaling pathways that are based on changes that acquired by the leukemic cells. Altogether, our data provide new insights into different SDF-1-induced signaling, activation, and consequent motility between normal CD34+ and precursor-B ALL progenitors, which may lead to improved clinical protocols.


Asunto(s)
Linfoma de Burkitt/inmunología , Linfoma de Burkitt/fisiopatología , Quimiocinas CXC/farmacología , Receptores CXCR4/fisiología , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/fisiología , Linfoma de Burkitt/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12 , Quimiotaxis de Leucocito/efectos de los fármacos , Sangre Fetal/efectos de los fármacos , Sangre Fetal/inmunología , Sangre Fetal/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/fisiología , Humanos , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Transducción de Señal/efectos de los fármacos , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA