Network


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

Hotspot


Dive into the research topics where Fumihito Hosoi is active.

Publication


Featured researches published by Fumihito Hosoi.


Cancer Research | 2005

HER2 Overexpression Increases Sensitivity to Gefitinib, an Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor, through Inhibition of HER2/HER3 Heterodimer Formation in Lung Cancer Cells

Akira Hirata; Fumihito Hosoi; Miho Miyagawa; Shu-ichi Ueda; Seiji Naito; Teruhiko Fujii; Michihiko Kuwano; Mayumi Ono

Gefitinib (Iressa), an epidermal growth factor receptor targeting drug, has been clinically useful for the treatment of patients with non-small cell lung cancer (NSCLC). Gefitinib is currently being applied in clinical studies as either a monotherapy, or as part of a combination therapy against prostate, head and neck, gastric, breast, and colorectal tumors. However, success rates vary between different tumor types, and thus it is important to understand which molecular target(s) are responsible for limiting the therapeutic efficacy of the drug. In this study, we ask whether expression of HER2 affects sensitivity to gefitinib in human lung cancer cells. We established two clones, LK2/HER2-32 and LK2/HER2-57, by transfecting HER2 cDNA into LK2, a NSCLC line with a low expression level of HER2. We observed no mutations in exons 18, 19, and 21 of EGFR gene in LK2, LK2/mock- and two HER2-trasfectants when we observed in-frame deletion mutations (E746-A750) adjacent to K745 in a gefitinib-sensitive NSCLC cell line, PC9. These LK2/HER2-32 and LK2/HER2-57 were much more sensitive to the cytotoxic effects of gefitinib than the parental LK2 lines. Treatment with 0.5 to 1 micromol/L gefitinib specifically blocked Akt activation in both HER2-transfectant lines, but not in the parental LK2 cells. Extracellular signal-regulated kinase-1/2 activation, however, was not blocked by gefitinib up to 10 micromol/L in either the parent or transfectant lines. Gefitinib was also shown to induce cell cycle arrest in the G1-S phase, and an accompanying increase of p27Kip1 was observed. LK2/HER2 transfectants showed constitutive formation of HER2/HER3 heterodimer, which were seen to associate with a regulatory subunit of phosphoinositide-3-kinase, p85alpha, when active. Treatment of LK2/HER2 cells with gefitinib markedly decreased the formation of HER2/HER3 heterodimers, HER3 basal phosphorylation, and the association of p85alpha with HER3. This study is the first to show that under basal growth conditions, HER2 sensitizes low-EGFR NSCLC cell lines to growth inhibition by gefitinib.


Cancer Science | 2007

Inflammatory stimuli from macrophages and cancer cells synergistically promote tumor growth and angiogenesis.

Yusuke N. Kimura; Kosuke Watari; Abbas Fotovati; Fumihito Hosoi; Kosei Yasumoto; Hiroto Izumi; Kimitoshi Kohno; Kazuo Umezawa; Haruo Iguchi; Sinzo Takamori; Michihiko Kuwano; Mayumi Ono

The focus of the present study was whether and how infiltrating macrophages play a role in angiogenesis and the growth of cancer cells in response to the inflammatory cytokine interleukin (IL)‐1β. Lewis lung carcinoma cells overexpressing IL‐1β grew faster and induced greater neovascularization than a low IL‐1β‐expressing counterpart in vivo. When macrophages were depleted using clodronate liposomes, both neovascularization and tumor growth were reduced in the IL‐1β‐expressing tumors. Co‐cultivation of IL‐1β‐expressing cancer cells with macrophages synergistically augmented neovascularization and the migration of vascular endothelial cells. In these co‐cultures, production of the angiogenic factors vascular endothelial growth factor‐A and IL‐8, monocyte chemoattractant protein‐1, and matrix metalloproteinase‐9 were increased markedly. The production of these factors, induced by IL‐1β‐stimulated lung cancer cells, was blocked by a nuclear factor (NF)‐κB inhibitor, and also by the knockdown of p65 (NF‐κB) and c‐Jun using small interference RNA, suggesting involvement of the transcription factors NF‐κB and AP‐1. These results demonstrated that macrophages recruited into tumors by monocyte chemoattractant protein‐1 and other chemokines could play a critical role in promoting tumor growth and angiogenesis, through interactions with cancer cells mediated by inflammatory stimuli. (Cancer Sci 2007; 98: 2009–2018)


Oncogene | 2007

Akt-dependent nuclear localization of Y-box-binding protein 1 in acquisition of malignant characteristics by human ovarian cancer cells

Yuji Basaki; Fumihito Hosoi; Yoshinao Oda; Abbas Fotovati; Y Maruyama; Shinji Oie; Mayumi Ono; Hiroto Izumi; Kimitoshi Kohno; Kazuko Sakai; Tatsu Shimoyama; Kazuto Nishio; Michihiko Kuwano

Y-box-binding protein 1 (YB-1), which is a member of the DNA-binding protein family containing a cold-shock domain, has pleiotropic functions in response to various environmental stimuli. As we previously showed that YB-1 is a global marker of multidrug resistance in ovarian cancer and other tumor types. To identify YB-1-regulated genes in ovarian cancers, we investigated the expression profile of YB-1 small-interfering RNA (siRNA)-transfected ovarian cancer cells using a high-density oligonucleotide array. YB-1 knockdown by siRNA upregulated 344 genes, including MDR1, thymidylate synthetase, S100 calcium binding protein and cyclin B, and downregulated 534 genes, including CXCR4, N-myc downstream regulated gene 1, E-cadherin and phospholipase C. Exogenous serum addition stimulated YB-1 translocation from the cytoplasm to the nucleus, and treatment with Akt inhibitors as well as Akt siRNA and integrin-linked kinase (ILK) siRNA specifically blocked YB-1 nuclear localization. Inhibition of Akt activation downregulated CXCR4 and upregulated MDR1 (ABCB1) gene expression. Administration of Akt inhibitor resulted in decrease in nuclear YB-1-positive cancer cells in a xenograft animal model. Akt activation thus regulates the nuclear translocation of YB-1, affecting the expression of drug-resistance genes and other genes associated with the malignant characteristics in ovarian cancer cells. Therefore, the Akt pathway could be a novel target of disrupting the nuclear translocation of YB-1 that has important implications for further development of therapeutic strategy against ovarian cancers.


Biochemical and Biophysical Research Communications | 2008

Role of macrophages in inflammatory lymphangiogenesis: Enhanced production of vascular endothelial growth factor C and D through NF-κB activation ☆

Kosuke Watari; Shintaro Nakao; Abbas Fotovati; Yuji Basaki; Fumihito Hosoi; Biborka Bereczky; Ryuichi Higuchi; Tomofumi Miyamoto; Michihiko Kuwano; Mayumi Ono

The close association of inflammation, angiogenesis and cancer progression is now highlighted, and in this study we especially focused on a close association of inflammation and lymphangiogenesis. We found that proinflammatory cytokine, interleukin-1beta (IL-1beta), could induce lymphangiogenesis in mouse cornea through enhanced production of potent lymphangiogenic factors, VEGF-A, VEGF-C and VEGF-D. IL-1beta-induced lymphangiogenesis, but not angiogenesis, was inhibited by administration of a selective anti-VEGF receptor-3 (VEGFR-3) neutralizing antibody. And in mouse cornea we observed recruitment of monocyte/macrophages and neutrophils by IL-1beta implanted cornea. Depletion of macrophages by a bisphosphonate encapsulated in liposomes inhibited this IL-1beta-induced lymphangiogenesis and also up-regulation of VEGF-A, VEGF-C, and VEGF-D. Furthermore, IL-1beta-induced lymphangiogenesis and angiogenesis were suppressed by NF-kappaB inhibition with marked suppression of VEGF-A, VEGF-C, and VEGF-D expression.


Cancer Research | 2009

N-myc Downstream Regulated Gene 1/Cap43 Suppresses Tumor Growth and Angiogenesis of Pancreatic Cancer through Attenuation of Inhibitor of κB Kinase β Expression

Fumihito Hosoi; Hiroto Izumi; Akihiko Kawahara; Yuichi Murakami; Hisafumi Kinoshita; Masayoshi Kage; Kazuto Nishio; Kimitoshi Kohno; Michihiko Kuwano; Mayumi Ono

N-myc downstream regulated gene 1 (NDRG1)/Cap43 expression is a predictive marker of good prognosis in patients with pancreatic cancer as we reported previously. In this study, NDRG1/Cap43 decreased the expression of various chemoattractants, including CXC chemokines for inflammatory cells, and the recruitment of macrophages and neutrophils with suppression of both angiogenesis and growth in mouse xenograft models. We further found that NDRG1/Cap43 induced nuclear factor-kappaB (NF-kappaB) signaling attenuation through marked decreases in inhibitor of kappaB kinase (IKK) beta expression and IkappaBalpha phosphorylation. Decreased IKKbeta expression in cells overexpressing NDRG1/Cap43 resulted in reduction of both nuclear translocation of p65 and p50 and their binding to the NF-kappaB motif. The introduction of an exogenous IKKbeta gene restored NDRG1/Cap43-suppressed expression of melanoma growth-stimulating activity alpha/CXCL1, epithelial-derived neutrophil activating protein-78/CXCL5, interleukin-8/CXCL8 and vascular endothelial growth factor-A, accompanied by increased phosphorylation of IkappaBalpha in NDRG1/Cap43-expressing cells. In patients with pancreatic cancer, NDRG1/Cap43 expression levels were also inversely correlated with the number of infiltrating macrophages in the tumor stroma. This study suggests a novel mechanism by which NDRG1/Cap43 modulates tumor angiogenesis/growth and infiltration of macrophages/neutrophils through attenuation of NF-kappaB signaling.


Biochemical and Biophysical Research Communications | 2010

Identification of sites subjected to serine/threonine phosphorylation by SGK1 affecting N-myc downstream-regulated gene 1 (NDRG1)/Cap43-dependent suppression of angiogenic CXC chemokine expression in human pancreatic cancer cells

Yuichi Murakami; Fumihito Hosoi; Hiroto Izumi; Yuichiro Maruyama; Hiroki Ureshino; Kosuke Watari; Kimitoshi Kohno; Michihiko Kuwano; Mayumi Ono

We have recently reported that N-myc downstream-regulated gene 1 (NDRG1)/Ca(2+)-associated protein with a molecular mass of 43kDa (Cap43) suppresses angiogenesis and tumor growth of pancreatic cancer through marked decreases in both the expression of CXC chemokines and phosphorylation of a NF-kappaB signaling molecule, inhibitor of kappaB kinase (IkappaBalpha). NDRG1/Cap43 is phosphorylated at serine/threonine sites in its C-terminal domain by serum- and glucocorticoid-regulated kinase 1 (SGK1). In this study, we attempted to clarify the domain or site of NDRG1/Cap43 responsible for its suppression of CXC chemokine expression in pancreatic cancer cells. Expression of the deletion constructs CapDelta2 [deletion of amino acids (AA) 130-142] and CapDelta4 [deletion of AA 180-294] as well as the wild-type full sequence of NDRG1/Cap43 (F-Cap), suppressed the production of CXC chemokines such as Groalpha/CXCL1 and ENA-78/CXCL5, whereas no or low suppression was observed in cell expressing the CapDelta5 mutant [deletion of AA 326-350] and CapDelta6 mutant [deletion of AA 326-394]. We further introduced mutations at the serine and threonine sites at 328 [T328A], 330 [S330A] and 346 [T346A], which are susceptible to phosphorylation by SGK1, and also constructed double mutants [T328A, S330A], [T328A, T346A] and [S330A, T346A]. Expression of all these mutants, with the exception of [S330A, T346A], suppressed the production of CXC chemokine to similar levels as their wild-type counterpart. IkappaBalpha was found to be specifically phosphorylated by this double mutant [S330A, T346A] and the CapDelta5 mutant at levels comparable to that induced in their wild-type counterpart. Phosphorylation of NDRG1/Cap43 at both serine330 and threonine346 is required for its suppressive action on the NF-kappaB signaling pathway and CXC chemokine expression in pancreatic cancer cells.


Human Pathology | 2010

N-myc downstream regulated gene-1/Cap43 may play an important role in malignant progression of prostate cancer, in its close association with E-cadherin

YooHyun Song; Yoshinao Oda; Mikifumi Hori; Kentaro Kuroiwa; Mayumi Ono; Fumihito Hosoi; Yuji Basaki; Shoji Tokunaga; Michihiko Kuwano; Seiji Naito; Masazumi Tsuneyoshi

N-myc downstream regulated gene-1 (NDRG1)/Cap43 plays an important role in tumor progression and metastases in many kinds of cancers. Recently, it was reported that NDRG1/Cap43 is involved in the aggressiveness of prostate cancer and also that its expression is associated with the expression of E-cadherin in prostate carcinoma cell lines. In the current study, to elucidate the functional and pathologic roles of NDRG1/Cap43 in prostate cancer, we investigated whether the expression of NDRG1/Cap43 is associated with the clinicopathologic parameters of prostate cancer or E-cadherin expression. NDRG1/Cap43 expression and E-cadherin expression were examined immunohistochemically in 148 patients with prostate cancer. We investigated the correlation between membranous or cytoplasmic expression of NDRG1/Cap43 and E-cadherin and evaluated the prognostic or clinicopathologic significance of the expression of NDRG1/Cap43. The patients with decreased NDRG1/Cap43 membranous expression showed significantly lower disease-free survival rates compared with the patients with preserved NDRG1/Cap43 membranous expression. Decreased membranous and high cytoplasmic NDRG1/Cap43 expression was also correlated with a higher Gleason score. A significant correlation was observed between NDRG1/Cap43 membranous expression and E-cadherin membranous expression (r = 0.7130; P < .0001) and between NDRG1/Cap43 cytoplasmic expression and E-cadherin cytoplasmic expression (r = 0.5847; P < .0001). Decreased NDRG1/Cap43 membranous expression had a significant impact on patient disease-free survival in multivariate analysis (P = .0175). NDRG1/Cap43 could be a novel marker for malignant progression and poor prognosis in prostate cancer, plausibly in its close association with the down-regulation of E-cadherin expression.


European Journal of Cancer | 2010

Y-box binding protein-1 (YB-1) promotes cell cycle progression through CDC6-dependent pathway in human cancer cells

Yuji Basaki; Ken Ichi Taguchi; Hiroto Izumi; Yuichi Murakami; Takuya Kubo; Fumihito Hosoi; Kosuke Watari; Kenji Nakano; Hidetoshi Kawaguchi; Shinji Ohno; Kimitoshi Kohno; Mayumi Ono; Michihiko Kuwano

Y-box binding protein-1 (YB-1) plays pivotal roles in acquisition of global drug resistance and cell growth promotion through transcriptional activation of genes for both drug resistance and growth factor receptors. In this study, we investigated whether YB-1 is involved in regulation of the cell cycle and cell proliferation of human cancer cells. Treatment with YB-1 siRNA caused a marked suppression of cell proliferation and expression of a cell cycle related gene, CDC6 by cancer cells. Of cell cycle of cancer cells, S phase content was specifically reduced by knockdown of YB-1. The overexpression of CDC6 abrogated this inhibition of both cell proliferation and S phase entry. ChIP assay demonstrated that YB-1 binds to a Y-box located in the promoter region of the CDC6 gene. Expression of cyclin D1, CDK1 and CDK2 was also reduced with increased expression of p21(Cip1) and p16(INK4A) when treated with YB-1 siRNA. Furthermore, the nuclear YB-1 expression was significantly associated with the level of CDC6 nuclear expression in patients with breast cancer. In conclusion, YB-1 plays an important role in cell cycle progression at G1/S of human cancer cells. YB-1 thus could be a potent biomarker for tumour growth and cell cycle in its close association with CDC6.


Cancer Science | 2006

High expression of insulin-like growth factor binding protein-3 is correlated with lower portal invasion and better prognosis in human hepatocellular carcinoma

Shinichi Aishima; Yuji Basaki; Yoshinao Oda; Yousuke Kuroda; Yunosuke Nishihara; Kenichi Taguchi; Akinobu Taketomi; Yoshihiko Maehara; Fumihito Hosoi; Yuichiro Maruyama; Abbas Fotovati; Shinji Oie; Mayumi Ono; Takato Ueno; Michio Sata; Hirohisa Yano; Masamichi Kojiro; Michihiko Kuwano; Masazumi Tsuneyoshi

Insulin‐like growth factor binding protein‐3 (IGFBP‐3) modulates cell proliferation of various cancer cell types. However, it remains unclear how IGF–IGFBP‐3‐signaling is involved in growth and progression of hepatocellular carcinoma (HCC). The aim of the present study was to evaluate the role of IGFBP‐3 in HCC. Type 1 receptor for IGF (IGF‐1R) was expressed at various levels in the seven lines examined, but IGF‐2R was not expressed. Of the seven lines, the growth of HAK‐1B, KIM‐1, KYN‐2 and HepG2 cells was stimulated in a dose‐dependent manner by the exogenous addition of IGF‐I or IGF‐II, but the HAK‐1A, KYN‐1 and KYN‐3 cell lines showed no growth. Exogenous addition of IGFBP‐3 markedly blocked IGF‐I and IGF‐II‐stimulated cell growth of KYN‐2 and HepG2 cells, and moderately stimulated that of KIM‐1 and HAK‐1B cells, but no growth of the KYN‐1, KYN‐3 and HAK‐1A cell lines was observed. IGF‐I enhanced the phosphorylation of IGF‐1R, Akt and Erk1/2 in KYN‐2 cells, and coadministration of IGFBP‐3 blocked all types of activation by IGF‐I investigated here. In contrast, no such activation by IGF‐I was detected in KYN‐3 cells. IGFBP‐3 also suppressed IGF‐I‐induced cell invasion by KYN‐2 cells. Moreover, we were able to observe the apparent expression of IGFBP‐3 in KYN‐3 cells, but not in the other six cell lines. Furthermore reduced expression of IGFBP‐3, but not that of IGF‐1R, was significantly correlated with tumor size, histological differentiation, capsular invasion and portal venous invasion. Low expression of IGFBP‐3 was independently associated with poor survival. IGFBP‐3 could be a molecular target of intrinsic importance for further development of novel therapeutic strategy against HCC. (Cancer Sci 2006; 97: 1182–1190)


Oncology Reports | 2010

Infiltration of thymidine phosphorylase-positive macrophages is closely associated with tumor angiogenesis and survival in intestinal type gastric cancer

Akihiko Kawahara; Satoshi Hattori; Jun Akiba; Kazutaka Nakashima; Tomoki Taira; Kosuke Watari; Fumihito Hosoi; Manami Uba; Yuji Basaki; Kikuo Koufuji; Shin-ichi Akiyama; Michihiko Kuwano; Masayoshi Kage; Mayumi Ono

Thymidine phosphorylase (TP), an enzyme catalyzing the reversible phospholysis of thymidine, deoxyuridine and their analogs at their respective bases and 2-deoxyribose-1-phosphate, thus promoting angiogenesis, is often expressed in macrophages present in tumor stroma. In this study, we investigated whether infiltration of TP-positive macrophages as well as tumor-associated macrophages affected tumor angiogenesis. TP was expressed in human macrophage-like cells, but not in gastric cancer cells in culture. The expression level of TP, the number of infiltrating CD68+ and CD163+ macrophages, and microvessel density (MVD) in the tumor were further analyzed by immunohistochemistry in 111 patients with gastric cancer. Biostatistical analysis of digitized data obtained by image analysis showed that TP expression was significantly correlated with the number of infiltrating macrophages and MVD in intestinal type gastric cancer (p<0.05). The number of infiltrating macrophages was also correlated with MVD in both the intestinal and diffuse types (p<0.05). An increased number of CD68+ macrophages was significantly associated with poor outcome in patients with intestinal type (p<0.001), but not diffuse type cancer. TP could be a specific marker enzyme that is expressed in tumor-infiltrating macrophages, being associated with tumor angiogenesis and poor prognosis in patients with intestinal-type gastric cancer.

Collaboration


Dive into the Fumihito Hosoi'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

Abbas Fotovati

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge