Network


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

Hotspot


Dive into the research topics where Yuji Shirakata is active.

Publication


Featured researches published by Yuji Shirakata.


Journal of Immunology | 2005

Induction of Keratinocyte Migration via Transactivation of the Epidermal Growth Factor Receptor by the Antimicrobial Peptide LL-37

Sho Tokumaru; Koji Sayama; Yuji Shirakata; Hitoshi Komatsuzawa; Kazuhisa Ouhara; Yasushi Hanakawa; Yoko Yahata; Xiuju Dai; Mikiko Tohyama; Hiroshi Nagai; Lujun Yang; Shigeki Higashiyama; Akihiko Yoshimura; Motoyuki Sugai; Koji Hashimoto

The closure of skin wounds is essential for resistance against microbial pathogens, and keratinocyte migration is an important step in skin wound healing. Cathelicidin hCAP18/LL-37 is an innate antimicrobial peptide that is expressed in the skin and acts to eliminate microbial pathogens. Because hCAP18/LL-37 is up-regulated at skin wound sites, we hypothesized that LL-37 induces keratinocyte migration. In this study, we found that 1 μg/ml LL-37 induced the maximum level of keratinocyte migration in the Boyden chamber assay. In addition, LL-37 phosphorylated the epidermal growth factor receptor (EGFR) after 10 min, which suggests that LL-37-induced keratinocyte migration occurs via EGFR transactivation. To test this assumption, we used inhibitors that block the sequential steps of EGFR transactivation, such as OSU8-1, CRM197, anti-EGFR no. 225 Ab, and AG1478. All of these inhibitors completely blocked LL-37-induced keratinocyte migration, which indicates that migration occurs via HB-EGF-mediated EGFR transactivation. Furthermore, CRM197, anti-EGFR no. 225, and AG1478 blocked the LL-37-induced phosphorylation of STAT3, and transfection with a dominant-negative mutant of STAT3 abolished LL-37-induced keratinocyte migration, indicating the involvement of the STAT3 pathway downstream of EGFR transactivation. Finally, we tested whether the suppressor of cytokine signaling (SOCS)/cytokine-inducible Src homology 2-containing protein (CIS) family of negative regulators of STAT3 regulates LL-37-induced keratinocyte migration. Transfection with SOCS1/Jak2 binding protein or SOCS3/CIS3 almost completely abolished LL-37-induced keratinocyte migration. In conclusion, LL-37 induces keratinocyte migration via heparin-binding-EGF-mediated transactivation of EGFR, and SOCS1/Jak 2 binding and SOCS3/CIS3 negatively regulate this migration. The results of this study suggest that LL-37 closes skin wounds by the induction of keratinocyte migration.


Journal of Cell Science | 2005

Heparin-binding EGF-like growth factor accelerates keratinocyte migration and skin wound healing

Yuji Shirakata; Rina Kimura; Daisuke Nanba; Ryo Iwamoto; Sho Tokumaru; Chie Morimoto; Koichi Yokota; Masanori Nakamura; Koji Sayama; Eisuke Mekada; Shigeki Higashiyama; Koji Hashimoto

Members of the epidermal growth factor (EGF) family are the most important growth factors involved in epithelialization during cutaneous wound healing. Heparin-binding EGF-like growth factor (HB-EGF), a member of the EGF family, is thought to play an important role in skin wound healing. To investigate the in vivo function of HB-EGF in skin wound healing, we generated keratinocyte-specific HB-EGF-deficient mice using Cre/loxP technology in combination with the keratin 5 promoter. Studies of wound healing revealed that wound closure was markedly impaired in keratinocyte-specific HB-EGF-deficient mice. HB-EGF mRNA was upregulated at the migrating epidermal edge, although cell growth was not altered. Of the members of the EGF family, HB-EGF mRNA expression was induced the most rapidly and dramatically as a result of scraping in vitro. Combined, these findings clearly demonstrate, for the first time, that HB-EGF is the predominant growth factor involved in epithelialization in skin wound healing in vivo and that it functions by accelerating keratinocyte migration, rather than proliferation.


Journal of Biological Chemistry | 2006

A Novel Function of Angiotensin II in Skin Wound Healing INDUCTION OF FIBROBLAST AND KERATINOCYTE MIGRATION BY ANGIOTENSIN II VIA HEPARIN-BINDING EPIDERMAL GROWTH FACTOR (EGF)-LIKE GROWTH FACTOR-MEDIATED EGF RECEPTOR TRANSACTIVATION

Yoko Yahata; Yuji Shirakata; Sho Tokumaru; Lujun Yang; Xiuju Dai; Mikiko Tohyama; Teruko Tsuda; Koji Sayama; Masaru Iwai; Masatsugu Horiuchi; Koji Hashimoto

The role of angiotensin II (Ang II) in the control of systemic blood pressure and volume homeostasis is well known and has been extensively studied. Recently, Ang II was suggested to also have a function in skin wound healing. In the present study, the in vivo function of Ang II in skin wound healing was investigated using Ang II type 1 receptor (AT1R) knock-out mice. Wound healing in these mice was found to be markedly delayed. Keratinocytes and fibroblasts play important roles in wound healing, and thus the effect of Ang II on the migration of these cells was examined. Ang II stimulated keratinocyte and fibroblast migration in a dose-dependent manner. It has been reported that G protein-coupled receptor (GPCR) activation induces epidermal growth factor (EGF) receptor (EGFR) transactivation through the shedding of heparin-binding EGF-like growth factor (HB-EGF). As AT1R is a GPCR, it was hypothesized that Ang II-induced keratinocyte and fibroblast migration is mediated by EGFR transactivation. Ang II induced EGFR phosphorylation, which was inhibited by an AT1R antagonist, HB-EGF neutralizing antibody, and an HB-EGF antagonist in both keratinocytes and in fibroblasts. Moreover, Ang II-induced migration of keratinocytes and fibroblasts was also prevented by these inhibitors. Taken together, these findings clearly demonstrate, for the first time, that Ang II plays an important role in skin wound healing and that it functions by accelerating keratinocyte and fibroblast migration in a process mediated by HB-EGF shedding.


European Journal of Immunology | 2005

New mechanisms of skin innate immunity: ASK1‐mediated keratinocyte differentiation regulates the expression of β‐defensins, LL37, and TLR2

Koji Sayama; Hitoshi Komatsuzawa; Kenshi Yamasaki; Yuji Shirakata; Yasushi Hanakawa; Kazuhisa Ouhara; Sho Tokumaru; Xiuju Dai; Mikiko Tohyama; Peter ten Dijke; Motoyuki Sugai; Hidenori Ichijo; Koji Hashimoto

Epidermal keratinocytes differentiate and form a multilayered epidermis, which is the primary barrier between the body and the outer environment. As the epidermis is constantly exposed to a variety of microbial pathogens, its function of resisting microbial pathogens is vital. This characteristic feature is formed during differentiation. Immunohistochemical analysis revealed that the upper epidermis of normal human skin expresses β‐defensinsu20041–3 and LL37. We hypothesized that epidermal keratinocytes develop an innate immune barrier based on human β‐defensins (hBD) and LL37 during differentiation. To prove this, we introduced an active form of the apoptosis signal‐regulating kinase‐1 (ASK1), an intracellular regulator of keratinocyte differentiation, into cultured normal human keratinocytes. Transfection of this active form, ASK1‐ΔN, significantly enhanced the expression of hBD1–3 and LL37. In addition, a p38 inhibitor abolished this induction, indicating that the ASK1‐p38 cascade regulates the expression of hBD1–3 and LL37. Furthermore, the ASK1‐p38 pathway also regulated the expression of Toll‐like receptor (TLR)2 in keratinocytes. Contact between S.u2004aureus and keratinocytes resulted in the phosphorylation of p38 and induced the expression of hBD2 and hBD3. Moreover, the p38 inhibitor reduced this induction. In conclusion, the ASK1‐p38 cascade regulates the innate immunity of the skin by forming an immune barrier consisting of hBD, LL37, and TLR2 during epidermal differentiation.


Journal of Biological Chemistry | 2002

Phosphatidylinositol 3-kinase is a key regulator of early phase differentiation in keratinocytes.

Koji Sayama; Kenshi Yamasaki; Yasushi Hanakawa; Yuji Shirakata; Sho Tokumaru; Takeshi Ijuin; Tadaomi Takenawa; Koji Hashimoto

The survival and growth of epithelial cells depend on adhesion to the extracellular matrix. Because epidermal keratinocytes differentiate as they leave the basement membrane, an adhesion signal may regulate the initiation of differentiation. Phosphatidylinositol 3-kinase (PI3K) is a fundamental signaling molecule that regulates the adhesion signal. Transfection of a dominant negative form of PI3K into keratinocytes using an adenovirus vector resulted in significant morphological changes comparable to differentiation and the induction of differentiation markers, keratin (K) 1 and K10. In turn, transfection with the constitutively active form of PI3K almost completely abolished the induction of K1 and K10 by differentiation in suspension cultures using polyhydroxyethylmethacrylate-coated dishes. PI3K activity was lost in suspension culture, except by cells bearing the constitutively active form of PI3K. These data demonstrate that blockade of PI3K results in differentiation and that activation of PI3K prevents differentiation. Furthermore, expression of the dominant negative form of PI3K significantly inhibited keratinocyte adhesion to the extracellular matrix and reduced the surface expression of α6 and β1 integrins in suspension culture. Moreover, expression of the active form of PI3K restored the mRNA levels of adhesion molecules that were reduced in suspension culture, including α3, α6, and β1 integrins, BP180, and BP230. In conclusion, loss of PI3K activity results in keratinocytes leaving the basement membrane and the initiation of a “default” differentiation mechanism.


Oncogene | 2007

Pre-B-cell leukemia transcription factor 1 is a major target of promyelocytic leukemia zinc-finger-mediated melanoma cell growth suppression

Ken Shiraishi; Kenshi Yamasaki; Daisuke Nanba; Hirofumi Inoue; Yasushi Hanakawa; Yuji Shirakata; Koji Hashimoto; Shigeki Higashiyama

Promyelocytic leukemia zinc-finger (PLZF) is a transcriptional repressor and tumor suppressor. PLZF is expressed in melanocytes but not in melanoma cells, and recovery of PLZF expression markedly suppresses melanoma cell growth. Several target genes regulated by PLZF have been identified, but the precise function of PLZF remains uncertain. Here, we searched for candidate target genes of PLZF by DNA microarray analysis. Pre-B-cell leukemia transcription factor 1 (Pbx1) was one of the prominently suppressed genes. Pbx1 was highly expressed in melanoma cells, and its expression was reduced by transduction with the PLZF gene. Moreover, the growth suppression mediated by PLZF was reversed by enforced expression of Pbx1. Knockdown of Pbx1 by specific small interfering RNAs suppressed melanoma cell growth. We also found that Pbx1 binds HoxB7. Reverse transcription–polymerase chain reaction analysis demonstrated that repression of Pbx1 by PLZF reduces the expression of HoxB7 target genes, including tumor-associated neoangiogenesis factors such as basic fibroblast growth factor, angiopoietin-2 and matrix metalloprotease 9. These findings suggest that deregulation of Pbx1 expression owing to loss of PLZF expression contributes to the progression and/or pathogenesis of melanoma.


Journal of Dermatological Science | 2003

Lack of evidence for TARC/CCL17 production by normal human keratinocytes in vitro

Teruko Tsuda; Mikiko Tohyama; Kenshi Yamasaki; Yuji Shirakata; Yoko Yahata; Sho Tokumaru; Koji Sayama; Koji Hashimoto

BACKGROUNDnthymus and activation-regulated chemokine (TARC)/CCL17 is a CC chemokine that selectively attracts Th2-type lymphocytes. Immunohistochemical analyses have revealed that TARC is expressed in the epidermal keratinocytes of atopic dermatitis (AD), suggesting TARC involvement in the pathogenesis of the disease. However, keratinocyte TARC production has been described only in the transformed keratinocyte cell line HaCaT.nnnOBJECTIVEnto examine TARC production in normal human epidermal keratinocytes (NHEK) in vitro.nnnMETHODSnthe expression of TARC mRNA and protein were examined in NHEK and HaCaT cells stimulated with various cytokines.nnnRESULTSnstimulation with inflammatory cytokines, including interleukin (IL)-1, IL-4, IL-6, IL-10, interferon (IFN)-alpha, IFN-beta, IFN-gamma, and tumor necrosis factor (TNF)-alpha failed to induce TARC mRNA expression in NHEK. However, stimulation with IFN-gamma and TNF-alpha together enhanced expression slightly. ELISA analysis failed to detect TARC protein in NHEK culture supernatant, even following stimulation with IFN-gamma and TNF-alpha. In contrast, HaCaT cells produced TARC protein even without stimulation of cytokines.nnnCONCLUSIONnthese results indicate that production of TARC by HaCaT cells is a phenomenon specific to the cell line and the observation on TARC in HaCaT cells can not be generalized. NHEK do not produce TARC protein in vitro.


Journal of Cellular Physiology | 1999

Possible involvement of p21 but not of p16 or p53 in keratinocyte senescence.

Koji Sayama; Yuji Shirakata; Kazushige Midorikawa; Yasushi Hanakawa; Koji Hashimoto

It has been reported that p21, p53, and p16 affect the cell cycle and cell senescence. However, their roles in keratinocyte senescence are not clear. We established primary keratinocyte strains from 15 donors and maintained them until replicative senescence; their population doublings ranged from 5.7–45.2. These strains were classified based on their population doublings as short (5.7–10.4), intermediate (13.9–17.4), and long (21.5–45.2). To investigate the roles of p21, p53, and p16 in the cellular senescence of the cultured keratinocytes, we quantitatively analyzed p21, p53, and p16 levels of keratinocyte strains with different life spans by Western blot with FluoroImager. p21 levels increased in the senescent phase but not in the nonsenescent phase in all of the short, intermediate, and long life‐span strains. Northern blot analysis also revealed induction of p21 mRNA was similar to that of p21 protein levels. There were no apparent differences in p53 levels between senescent and nonsenescent cells. The short life‐span strains exhibited a significant increase in p16 levels in the senescent phase (eighth or tenth passage). However, in two long life‐span strains, p16 levels were increased in the nonsenescent phase (eighth passage) but then declined as the cells reached senescence (twenty‐seventh passage). Therefore, induction of p16 appeared not to be associated with senescence in long life‐span strains. In conclusion, p21 but not p16 or p53 may play roles in keratinocyte senescence. J. Cell. Physiol. 179:40–44, 1999.


Journal of Dermatological Science | 2003

So-called biological dressing effects of cultured epidermal sheets are mediated by the production of EGF family, TGF-β and VEGF

Yuji Shirakata; Sho Tokumaru; Kenshi Yamasaki; Koji Sayama; Koji Hashimoto

BACKGROUNDnCultured epidermal sheet (CES) grafts accelerate wound healing as a result of so-called biological dressing effect, which is thought to be mediated by various growth factors. However, the profile of growth factor expression in CESs is unclear.nnnOBJECTIVEnTo investigate whether CESs produce growth factors along with stratification we investigated the production of growth factors and their regulation in CESs.nnnMETHODSnCESs conditioned medium was harvested and the concentration of TGF-alpha, TGF-beta1, TGF-beta2, and VEGF was measured using ELISA. The mRNA of EGF family, TGF-beta family and VEGF was detected by Northern blot or RNase protection assay.nnnRESULTSnThe concentration of TGF-alpha was 100 pg/ml in the monolayer culture, but dramatically increased to 600 pg/ml 2 days after stratification. It decreased to baseline, and then gradually increased to 300 pg/ml in the presence of EGF and remained at that level until day 20. TGF-beta1 increased from 50 to 400 pg/ml after stratification, and remained at that level day 20. TGF-beta2 was undetectable in the monolayer culture, but dramatically increased to 200 pg/ml 2 days after stratification. Unlike TGF-beta1, TGF-beta2 gradually increased over time after stratification. VEGF increased with stratification from 500 to 1500 pg/ml. The addition of EGF upregulated EGF family, TGF-beta, and VEGF production in CESs, as confirmed by ELISA, Northern blot, and RNase protection assay.nnnCONCLUSIONnThese results indicate that so-called biological dressing effect of CESs is mediated by production of the EGF family, TGF-beta, and VEGF. Our results also demonstrate the ability of EGF to enhance growth factor production in CESs.


Journal of Dermatological Science | 1999

Expression of vitamin D receptor in cultured human keratinocytes and fibroblasts is not altered by corticosteroids

Kazushige Midorikawa; Koji Sayama; Yuji Shirakata; Yasushi Hanakawa; Ling Sun; Koji Hashimoto

Topical vitamin D3 therapy is one of the mainstays of psoriasis treatment. However, the effectiveness of combination therapy with topical vitamin D3 and corticosteroids is still controversial. It has been reported that topical vitamin D3 treatment following topical corticosteroids is less effective than that without preceding corticosteroid treatment. We hypothesized that vitamin D receptor (VDR) in the skin is down-regulated by topical corticosteroids. To obtain support for this hypothesis, we determined VDR protein levels in cultured keratinocytes and fibroblasts after corticosteroid treatment. VDR levels were quantified by Western blot analysis with a Fluorolmager. Keratinocytes and fibroblasts were obtained from four psoriasis patients and four normal controls. VDR levels were altered in neither normal nor psoriatic keratinocytes by 2-day incubation with dexamethasone (1x10(-9)-1x10(-6) M) or clobetasol propionate (1x10(-9)-1x10(-6) M). Similarly, VDR levels in normal and psoriatic fibroblasts were not affected by 2-day incubation with dexamethasone (1x10(-6) M). These findings suggest that down-regulation of VDR by topical corticosteroids in keratinocytes and fibroblasts of psoriasis is unlikely.

Collaboration


Dive into the Yuji Shirakata's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge