Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hidekata Yasuoka is active.

Publication


Featured researches published by Hidekata Yasuoka.


American Journal of Pathology | 2009

The Fibrotic Phenotype Induced by IGFBP-5 Is Regulated by MAPK Activation and Egr-1-Dependent and -Independent Mechanisms

Hidekata Yasuoka; Eileen Hsu; Ximena D. Ruiz; Richard A. Steinman; Augustine M. K. Choi; Carol A. Feghali-Bostwick

We have previously shown that insulin-like growth factor (IGF) binding protein- 5 (IGFBP-5) is overexpressed in lung fibrosis and induces the production of extracellular matrix components, such as collagen and fibronectin, both in vitro and in vivo. The exact mechanism by which IGFBP-5 exerts these novel fibrotic effects is unknown. We thus examined the signaling cascades that mediate IGFBP-5-induced fibrosis. We demonstrate for the first time that IGFBP-5 induction of extracellular matrix occurs independently of IGF-I, and results from IGFBP-5 activation of MAPK signaling, which facilitates the translocation of IGFBP-5 to the nucleus. We examined the effects of IGFBP-5 on early growth response (Egr)-1, a transcription factor that is central to growth factor-mediated fibrosis. Egr-1 was up-regulated by IGFBP-5 in a MAPK-dependent manner and bound to nuclear IGFBP-5. In fibroblasts from Egr-1 knockout mice, induction of fibronectin by IGFBP-5 was abolished. Expression of Egr-1 in these cells rescued the extracellular matrix-promoting effects of IGFBP-5. Moreover, IGFBP-5 induced cell migration in an Egr-1-dependent manner. Notably, Egr-1 levels, similar to IGFBP-5, were increased in vivo in lung tissues and in vitro in primary fibroblasts of patients with pulmonary idiopathic fibrosis. Taken together, our findings suggest that IGFBP-5 induces a fibrotic phenotype via the activation of MAPK signaling and the induction of nuclear Egr-1 that interacts with IGFBP-5 and promotes fibrotic gene transcription.


American Journal of Respiratory Cell and Molecular Biology | 2009

The Pro-Fibrotic Factor IGFBP-5 Induces Lung Fibroblast and Mononuclear Cell Migration

Hidekata Yasuoka; Yukie Yamaguchi; Carol A. Feghali-Bostwick

We have previously shown that insulin-like growth factor-binding protein-5 (IGFBP-5) is overexpressed in fibrotic lung tissues and that it induces production of extracellular matrix components such as collagen and fibronectin both in vitro and in vivo. We recently observed mononuclear cell infiltration in lung tissues of mice expressing IGFBP-5. We therefore examined the role of IGFBP-5 on the migration of immune cells. Migration assays demonstrated that IGFBP-5 induced migration of peripheral blood mononuclear cells (PBMCs) in a dose-dependent manner. Preferential migration of monocytes/macrophages, natural killer cells, and T cells was observed. Moreover, the CD4/CD8 ratio of migrating cells was significantly higher in vitro and in vivo in response to IGFBP-5. IGFBP-5 resulted in preferential migration of activated CD4(+) T cells and monocytes. Interestingly, IGFBP-5 also induced migration of primary human lung fibroblasts. Exogenous administration of IGFBP-5 induced activation of mitogen-activated protein kinase (MAPK) signaling cascade but not PI3K in PBMCs. IGFBP-5-induced migration was blocked by the MEK1/2 inhibitor U0126, suggesting that IGFBP-5-induced migration occurs via MAPK activation. Furthermore, monocytes treated with recombinant IGFBP-5 expressed the mesenchymal markers alpha-smooth muscle actin and fibronectin in vitro and in vivo, suggesting that IGFBP-5 can induce the transformation of monocytes into mesenchymal cells. Collectively, our results suggest that IGFBP-5 induces cell migration via MAPK-dependent and IGF-I-independent mechanisms.


American Journal of Respiratory and Critical Care Medicine | 2009

Role of Insulin-like Growth Factor Binding Protein-3 in Allergic Airway Remodeling

Kristen L. Veraldi; Bethany T. Gibson; Hidekata Yasuoka; Michael M. Myerburg; Elizabeth A. Kelly; Silvana Balzar; Nizar N. Jarjour; Joseph M. Pilewski; Sally E. Wenzel; Carol A. Feghali-Bostwick

RATIONALEnThe hallmarks of allergic asthma are airway inflammation, obstruction, and remodeling. Airway remodeling may lead to irreversible airflow obstruction with increased morbidity and mortality. Despite advances in the treatment of asthma, the mechanisms underlying airway remodeling are still poorly understood. We reported that insulin-like growth factor (IGF) binding proteins (IGFBPs) contribute to extracellular matrix deposition in idiopathic pulmonary fibrosis; however, their contribution to airway remodeling in asthma has not been established.nnnOBJECTIVESnWe hypothesized that IGFBP-3 is overexpressed in asthma and contributes to airway remodeling.nnnMETHODSnWe evaluated levels of IGFBP-3 in tissues and bronchoalveolar lavage fluid from patients with asthma at baseline and 48 hours after allergen challenge, in reparative epithelium in an in vitro wounding assay, and in conditioned media from cytokine- and growth factor-stimulated primary epithelial cells.nnnMEASUREMENTS AND MAIN RESULTSnIGFBP-3 levels and distribution were evaluated by Western blot, ELISA, and immunofluorescence. IGFBP-3 is increased in vivo in the airway epithelium of patients with asthma compared with normal control subjects. The concentration of IGFBP-3 is increased in the bronchoalveolar lavage fluid of patients with asthma after allergen challenge, its levels are increased in reparative epithelium in an in vitro wounding assay and in the conditioned medium of primary airway epithelial cell cultures stimulated with IGF-I.nnnCONCLUSIONSnOur results suggest that one mechanism of allergic airway remodeling is through the secretion of the profibrotic IGFBP-3 from IGF-I-stimulated airway epithelial cells during allergic inflammation.


Journal of Cellular and Molecular Medicine | 2011

Decreased caveolin-1 levels contribute to fibrosis and deposition of extracellular IGFBP-5

Yukie Yamaguchi; Hidekata Yasuoka; Donna B. Stolz; Carol A. Feghali-Bostwick

Our previous studies have demonstrated increased expression of insulin‐like growth factor binding protein‐5 (IGFBP‐5) in fibrotic tissues and IGFBP‐5 induction of extracellular matrix (ECM) components. The mechanism resulting in increased IGFBP‐5 in the extracellular milieu of fibrotic fibroblasts is unknown. Since Caveolin‐1 (Cav‐1) has been implicated to play a role in membrane trafficking and signal transduction in tissue fibrosis, we examined the effect of Cav‐1 on IGFBP‐5 internalization, trafficking and secretion. We demonstrated that IGFBP‐5 localized to lipid rafts in human lung fibroblasts and bound Cav‐1. Cav‐1 was detected in the nucleus in IGFBP‐5‐expressing fibroblasts, within aggregates enriched with IGFBP‐5, suggesting a coordinate trafficking of IGFBP‐5 and Cav‐1 from the plasma membrane to the nucleus. This trafficking was dependent on Cav‐1 as fibroblasts from Cav‐1 null mice had increased extracellular IGFBP‐5, and as fibroblasts in which Cav‐1 was silenced or lipid raft structure was disrupted through cholesterol depletion also had defective IGFBP‐5 internalization. Restoration of Cav‐1 function through administration of Cav‐1 scaffolding peptide dramatically increased IGFBP‐5 uptake. Finally, we demonstrated that IGFBP‐5 in the ECM protects fibronectin from proteolytic degradation. Taken together, our findings identify a novel role for Cav‐1 in the internalization and nuclear trafficking of IGFBP‐5. Decreased Cav‐1 expression in fibrotic diseases likely leads to increased deposition of IGFBP‐5 in the ECM with subsequent reduction in ECM degradation, thus identifying a mechanism by which reduced Cav‐1 and increased IGFBP‐5 concomitantly contribute to the perpetuation of fibrosis.


Arthritis Research & Therapy | 2013

Estradiol promotes the development of a fibrotic phenotype and is increased in the serum of patients with systemic sclerosis

Keiko Aida-Yasuoka; Christine Peoples; Hidekata Yasuoka; Pamela A. Hershberger; Katelynn Thiel; Jane A. Cauley; Thomas A. Medsger; Carol A. Feghali-Bostwick

IntroductionSystemic sclerosis (SSc) is more prevalent in women. Our goal is to determine the effects of 17β-estradiol (E2) on the development of fibrosis and to compare circulating levels of estrogens in SSc patients and healthy controls.MethodsUsing primary human dermal fibroblasts, we evaluated the effect of E2 on fibronectin (FN) expression with and without the estrogen receptor (ER) antagonist ICI 182,780, inhibitors of signaling, propyl-pyrazole-triol, an ERα specific ligand, and genistein, an ERβ selective ligand, to identify the signaling pathways mediating E2s effect. We confirmed the fibrotic effect of E2 in human skin using an ex vivo organ culture model. Lastly, we measured levels of E2 and estrone in serum samples from SSc patients with diffuse cutaneous involvement and healthy controls using mass spectrometry.ResultsE2 increased expression of FN in dermal fibroblasts. ICI 182,780, inositol-1,4,5-triphosphate inhibitor, and p38 mitogen-activated protein kinase inhibitor blocked the effects of E2 on FN. Propyl-pyrazole-triol, but not genistein, significantly increased FN expression. Ex vivo, E2 induced fibrosis of human skin. The effects of E2 were abrogated by ICI 182,780. Circulating levels of E2 and estrone were significantly increased in sera of patients with diffuse cutaneous SSc.ConclusionOur findings implicate estrogens in the fibrotic process and may explain the preponderance of SSc in women. ICI 182,780 or other ER signaling antagonists may be effective agents for the treatment of fibrosis.


PLOS ONE | 2014

The Membrane-Associated Adaptor Protein DOK5 Is Upregulated in Systemic Sclerosis and Associated with IGFBP-5-Induced Fibrosis

Hidekata Yasuoka; Yukie Yamaguchi; Carol A. Feghali-Bostwick

Systemic sclerosis (SSc) is characterized by excessive fibrosis of the skin and internal organs due to fibroblast proliferation and excessive production of extracellular matrix (ECM). We have shown that insulin-like growth factor binding protein (IGFBP)-5 plays an important role in the development of fibrosis in vitro, ex vivo, and in vivo. We identified a membrane-associated adaptor protein, downstream of tyrosine kinase/docking protein (DOK)5, as an IGFBP-5-regulated target gene using gene expression profiling of primary fibroblasts expressing IGFBP-5. DOK5 is a tyrosine kinase substrate associated with intracellular signaling. Our objective was to determine the role of DOK5 in the pathogenesis of SSc and specifically in IGFBP-5-induced fibrosis. DOK5 mRNA and protein levels were increased in vitro by endogenous and exogenous IGFBP-5 in primary human fibroblasts. DOK5 upregulation required activation of the mitogen-activated protein kinase (MAPK) signaling cascade. Further, IGFBP-5 triggered nuclear translocation of DOK5. DOK5 protein levels were also increased in vivo in mouse skin and lung by IGFBP-5. To determine the effect of DOK5 on fibrosis, DOK5 was expressed ex vivo in human skin in organ culture. Expression of DOK5 in human skin resulted in a significant increase in dermal thickness. Lastly, levels of DOK5 were compared in primary fibroblasts and lung tissues of patients with SSc and healthy donors. Both DOK5 mRNA and protein levels were significantly increased in fibroblasts and skin tissues of patients with SSc compared with those of healthy controls, as well as in lung tissues of SSc patients. Our findings suggest that IGFBP-5 induces its pro-fibrotic effects, at least in part, via DOK5. Furthermore, IGFBP-5 and DOK5 are both increased in SSc fibroblasts and tissues and may thus be acting in concert to promote fibrosis.


American Journal of Pathology | 2006

Insulin-Like Growth Factor-Binding Protein-5 Induces Pulmonary Fibrosis and Triggers Mononuclear Cellular Infiltration

Hidekata Yasuoka; Zhihong Zhou; Joseph M. Pilewski; Tim D. Oury; Augustine M. K. Choi; Carol A. Feghali-Bostwick


Arthritis & Rheumatism | 2006

Insulin-like growth factor binding protein 5 induces skin fibrosis: A novel murine model for dermal fibrosis.

Hidekata Yasuoka; Drazen M. Jukic; Zhihong Zhou; Augustine M. K. Choi; Carol A. Feghali-Bostwick


Critical Reviews in Eukaryotic Gene Expression | 2008

Gene expression in pulmonary fibrosis.

Eileen Hsu; Hidekata Yasuoka; Carol A. Feghali-Bostwick


Japanese Journal of Chest Diseases | 2014

Pulmonary Veno-Occlusive Disease (PVOD) Associated with Connective Tissue Disease

Hidekata Yasuoka; Masataka Kuwana

Collaboration


Dive into the Hidekata Yasuoka's collaboration.

Top Co-Authors

Avatar

Carol A. Feghali-Bostwick

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eileen Hsu

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhihong Zhou

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Augustine M. K. Choi

NewYork–Presbyterian Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donna B. Stolz

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge