Network


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

Hotspot


Dive into the research topics where Nobuyuki Nishizawa is active.

Publication


Featured researches published by Nobuyuki Nishizawa.


American Journal of Pathology | 2016

Vascular Endothelial Growth Factor Receptor Type 1 Signaling Prevents Delayed Wound Healing in Diabetes by Attenuating the Production of IL-1β by Recruited Macrophages

Shin-ichiro Okizaki; Yoshiya Ito; Kanako Hosono; Kazuhito Oba; Hirotoki Ohkubo; Ken Kojo; Nobuyuki Nishizawa; Masabumi Shibuya; Masayoshi Shichiri; Masataka Majima

The persistence of proinflammatory macrophages, which are recruited to the granulation tissue, impairs the healing of diabetic wounds. Herein, we examined the role of vascular endothelial growth factor receptor type 1 (VEGFR1) signaling in streptozotocin (STZ)-induced diabetic wound healing. Angiogenesis, lymphangiogenesis, and the healing of full-thickness skin wounds were impaired in STZ-treated wild-type (WT) mice compared with vehicle-treated WT mice, with attenuated recruitment of VEGFR1-positive macrophages expressing vascular endothelial growth factor (VEGF)-A, VEGF-C, and VEGF-D to the wound granulation tissue. These phenomena were even more prevalent in STZ-treated VEGFR1 tyrosine kinase knockout mice (VEGFR1 TK(-/-) mice). STZ-treated WT mice, but not STZ-treated VEGFR1 TK(-/-) mice, showed accelerated wound healing when treated with placenta growth factor. Compared with that of STZ-treated WT mice, the wound granulation tissue of STZ-treated VEGFR1 TK(-/-) mice contained more VEGFR1-positive cells expressing IL-1β [a classic (M1) activated macrophage marker] and fewer VEGFR1-positive cells expressing the mannose receptor [CD206; an alternatively activated (M2) macrophage marker]. Treatment of STZ-treated VEGFR1 TK(-/-) mice with an IL-1β-neutralizing antibody restored impaired wound healing and angiogenesis/lymphangiogenesis and induced macrophages in the wound granulation tissue to switch to an M2 phenotype. Taken together, these results suggest that VEGFR1 signaling plays a role in regulating the balance between macrophage phenotypes in STZ-induced diabetic wounds, prevents impaired diabetic wound healing, and promotes angiogenesis/lymphangiogenesis.


Diseases of The Esophagus | 2016

Promoter DNA methylation of CDO1 gene and its clinical significance in esophageal squamous cell carcinoma.

Hideki Ushiku; Keishi Yamashita; Hiroshi Katoh; Akira Ema; Naoko Minatani; Mariko Kikuchi; Ken Kojo; Keigo Yokoi; Toshimichi Tanaka; Nobuyuki Nishizawa; Satoru Ishii; Kei Hosoda; Hiromitsu Moriya; Hiroaki Mieno; Natsuya Katada; Shiro Kikuchi; Masahiko Watanabe

We have demonstrated that CDO1 methylation is frequently found in various cancers, including esophageal squamous cell carcinoma (ESCC), but its clinical relevance has remained elusive. CDO1 methylation was investigated in 169 ESCC patients who underwent esophagectomy between 1996 and 2007. CDO1 methylation was assessed by Q-MSP (quantitative methylation specific PCR), and its clinical significance, including its relationship to prognosis, was analyzed. (i) The median TaqMeth value of CDO1 methylation was 9.4, ranging from 0 to 279.5. CDO1 methylation was significantly different between cStage I and cStage II/III (Pu2009=u20090.02). (ii) On the log-rank plot, the optimal cut-off value was determined to be 8.9; ESCC patients with high CDO1 methylation showed a significantly worse prognosis than those with low CDO1 methylation (Pu2009=u20090.02). (iii) A multivariate Cox proportional hazards model identified only CDO1 hypermethylation as an independent prognostic factor (HR 2.00, CI 1.09-3.78, Pu2009=u20090.03). (iv) CDO1 hypermethylation stratified ESCC patients prognosis in cStage II/III for both neoadjuvant chemo(radio)therapy (NAC)-positive and NAC-negative cases. Moreover, the CDO1 methylation level was significantly lower in cases with Grade 2/3 than in those with Grade 0/1 (Pu2009=u20090.02) among cStage II/III ESCC patients with NAC. Promoter DNA hypermethylation of CDO1 could be an independent prognostic factor in ESCC; it may also reflect NAC eradication of tumor cells in the primary tumors.


Pancreas | 2016

Homeobox-Only Protein Expression Is a Critical Prognostic Indicator of Pancreatic Neuroendocrine Tumor and Is Regulated by Promoter DNA Hypermethylation.

Hideki Ushiku; Keishi Yamashita; Hiroshi Kawamata; Mina Waraya; Hiroshi Katoh; Keigo Yokoi; Toshimichi Tanaka; Satoru Ishii; Nobuyuki Nishizawa; Mariko Kikuchi; Naoko Minatani; Ken Kojo; Hiroshi Tajima; Ryo Nishiyama; Takashi Kaizu; Yusuke Kumamoto; Masahiko Watanabe

Objectives We have identified homeobox-only protein (HOPX) as a tumor suppressor gene in various human cancer, and its expression was reduced by promoter DNA hypermethylation. Homeobox-only protein is strongly expressed on pancreatic islet cells; however, clinical relevance of HOPX expression has remained elusive in pancreatic neuroendocrine tumor (pNET). Methods We investigated 36 patients with pNET who undertook surgical resection between 1988 and 2012 for HOPX expression and DNA methylation to reveal its clinical significance. Results (1) Homeobox-only protein is strongly expressed on pancreatic islet cells by immunohistochemistry (IHC). Homeobox-only protein expression was recognized on pNET tumor cells for 1+ in 15, for 2+ in 16, and for 3+ in 5. (2) Homeobox-only protein IHC expression was significantly associated with prognosis (P = 0.03), and survival rate was 37.5%, 70.3%, and 100% in HOPX 1+, 2+, and 3+, respectively. (3) Promoter DNA methylation was quantitatively assessed, and HOPX hypermethylation is found in 6.3%, 11.8%, and 66.7% of G1/G2/G3 pNET, respectively (P = 0.02). (4) Multivariate Cox proportional hazards model identified HOPX IHC expression and HOPX promoter DNA hypermethylation as independent prognostic factors in pNET. Conclusions Homeobox-only protein expression is a critical prognostic indicator of pNET, and its regulation may be made through promoter DNA methylation.


PLOS ONE | 2017

Prognostic significance of promoter DNA hypermethylation of the cysteine dioxygenase 1 (CDO1) gene in primary gallbladder cancer and gallbladder disease

Kazuharu Igarashi; Keishi Yamashita; Hiroshi Katoh; Keita Kojima; Yosuke Ooizumi; Nobuyuki Nishizawa; Ryo Nishiyama; Hiroshi Kawamata; Hiroshi Tajima; Takashi Kaizu; Yusuke Kumamoto; Masahiko Watanabe

Background Aberrant promoter DNA methylation of the cysteine dioxygenase 1 (CDO1) gene is found in various human cancers and is associated with clinical outcome. In this study, we assessed for the first time the clinicopathological significance of CDO1 methylation in primary gallbladder cancer (GBC) in comparison with non-malignant gallbladder disease. Methods CDO1 DNA methylation was quantified using quantitative TaqMan methylation specific PCR (Q-MSP) in 99 primary GBC patients together with the 78 corresponding non-tumor tissues and 26 benign gallbladder disease (including 7 patients with xanthogranulomatous cholecystitis) who underwent surgical resection between 1986 and 2014. Results The average CDO1 TaqMeth value of primary GBCs was 23.5±26. These values were significantly higher than those of corresponding non-tumor tissues (average 8±13, p < .0001) and diseased gallbladder tissues from patients with benign gallbladder diseases (average 0.98±1.6, p < .0001). CDO1 hypermethylation is also found in xanthogranulomatous cholecystitis. Using a cut-off value of 17.7, GBC cases with CDO1 hypermethylation (n = 47) showed significantly poorer prognosis than those with CDO1 hypomethylation (n = 52) (p = 0.0023). Multivariate Cox proportional hazards analysis identified that CDO1 hypermethylation was an independent prognostic factor. Notably, CDO1 hypermethylation showed prognostic relevance, especially in stage II GBC, in which it is highly anticipated to work as a predictive marker for candidates of adjuvant therapy. Conclusions Promoter NA methylation of CDO1 was demonstrated for the first time to be a cancer-associated methylation in primary GBC, and it has the potential to be a prognostic biomarker of GBC for high-risk patients with stage II GBC.


Gastric Cancer | 2017

DNA diagnosis of peritoneal fluid cytology test by CDO1 promoter DNA hypermethylation in gastric cancer

Hideki Ushiku; Keishi Yamashita; Akira Ema; Naoko Minatani; Mariko Kikuchi; Ken Kojo; Keigo Yokoi; Toshimichi Tanaka; Nobuyuki Nishizawa; Satoru Ishii; Kei Hosoda; Hiromitsu Moriya; Hiroaki Mieno; Natsuya Katada; Shiro Kikuchi; Hiroshi Katoh; Masahiko Watanabe

BackgroundMinimal residual disease of the peritoneum is challenging for early cancer detection in gastric cancer (GC). Utility of PCR amplification of cancer-derived DNA has been considered feasible due to its molecular stability, however such markers have never been available in GC clinics. We recently discovered cancer-specific methylation of CDO1 gene in GC, and investigated the clinical potential to detect the minimal residual disease.MethodsOne hundred and two GC patients were investigated for peritoneal fluid cytology test (CY), and detection level of the promoter DNA methylation of CDO1 gene was assessed by quantitative methylation specific PCR (Q-MSP) in the sediments (DNA CY).Results(1) CY1 was pathologically confirmed in 8 cases, while DNA CY1 was detected in 18 cases. All 8 CY1 were DNA CY1. (2) DNA CY1 was recognized in 14.3, 25.0, 20.0, and 42.9%, in macroscopic Type II, small type III, large type III, and type IV, respectively, while it was not recognized in Type 0/I/V. (3) DNA CY1 was prognostic relevance in gastric cancer (pxa0=xa00.0004), and its significance was robust among Type III/IV gastric cancer (pxa0=xa00.006 for overall survival and pxa0=xa00.0006 for peritoneal recurrence free survival). (4) The peritoneal recurrence was hardly seen in GC patients with potent perioperative chemotherapy among those with DNA CY1.ConclusionsDNA CY1 detected by Q-MSP for CDO1 gene promoter DNA methylation has a great potential to detect minimal residual disease of the peritoneum in GC clinics as a novel DNA marker.


Surgery Today | 2016

Predictive factors for lymph node metastasis in additional gastrectomy after endoscopic resection of cT1aN0 gastric cancer

Satoru Ishii; Keishi Yamashita; Hiroshi Kato; Nobuyuki Nishizawa; Hideki Ushiku; Hiroaki Mieno; Hiromitu Moriya; Kei Hosoda; Natuya Katada; Shiro Kikuchi; Satoshi Tanabe; Wasaburo Koizumi; Makoto Saegusa; Masahiko Watanabe

PurposeEndoscopic therapy for clinical T1aN0 (cT1aN0) gastric cancer is an excellent therapeutic strategy; however, pathological lymph node metastasis (LNM) occasionally occurs. Patients who have a potential for LNM are subject to additional gastrectomy. Our aim was to identify predictors of LNM in additional gastrectomy.MethodsOne hundred and twelve cT1aN0 gastric cancer patients undergoing additional gastrectomy after endoscopic resection were identified between 1997 and 2013. Predictors for LNM were initially selected by a univariate analysis and applied to a multivariate analysis.Results(1) Twelve patients (10.7xa0%) had LNM following additional gastrectomy. (2) Clinicopathological factors significantly associated with LNM were the depth of invasion (SM2 or deeper, designated as SM2) (pxa0=xa00.0018) and rigorous lymphatic invasion (ly2,3) (pxa0<xa00.001). (3) The univariate predictors for LNM were applied to the multivariate logistic regression model, and SM2 (pxa0=xa00.0027) and ly2,3 (pxa0=xa00.0028) remained significant predictors. (4) When classified into 2xa0×xa02 subgroups, the predictability for LNM was as follows: SM2 plus ly2,3 (46.7xa0%), SM2 plus ly0,1 (10.0xa0%), M,SM1 plus ly2,3 (0xa0%), and M,SM1 plus ly0,1 (0xa0%).ConclusionsIn cT1aN0 gastric cancer patients, both SM2 and ly2,3 are significant predictors for LNM that may be important as references for additional gastrectomy after endoscopic resection.


Oncotarget | 2017

The H19-PEG10/IGF2BP3 axis promotes gastric cancer progression in patients with high lymph node ratios

Satoru Ishii; Keishi Yamashita; Hiroki Harada; Hideki Ushiku; Toshimichi Tanaka; Nobuyuki Nishizawa; Keigo Yokoi; Marie Washio; Akira Ema; Hiroaki Mieno; Hiromitsu Moriya; Kei Hosoda; Mina Waraya; Hiroshi Katoh; Masahiko Watanabe

We previously demonstrated that the lymph node ratio (LNR) is a prognostic factor associated with EGFR expression, among first priority genes amplified or overexpressed in cancer. Here, we investigated the associations between high LNR and second, third, and fourth priority genes. We performed mRNA expression microarray analysis of tumor tissue from patients with stage III gastric cancer and high or low LNRs. Candidate high LNR-associated genes were further evaluated in 39 patients with stage III gastric cancer. The functional relevance of these genes was evaluated in gastric cancer cell lines. We focused on five genes: H19,PEG10, IGF2BP3, CD177, and PGA3. H19 and PEG10 were confirmed as high LNR-associated genes. H19, PEG10, and IGF2BP3 were found to promote each other’s expression. Knocking down H19 or PEG10 using RNAi decreased cell proliferation, invasion, anchorage-independent growth, and chemoresistance. These genes had a mutual relationship in MKN7 cells. H19 knockdown decreased expression of epithelial-mesenchymal transition-associated genes in MKN74 cells to suppress transformation. Thus, H19 promotes epithelial-mesenchymal transition in gastric cancer and is a potential therapeutic target.


PLOS ONE | 2018

Cysteine dioxygenase type 1 (CDO1) gene promoter methylation during the adenoma-carcinoma sequence in colorectal cancer

Keita Kojima; Takatoshi Nakamura; Makoto Ohbu; Hiroshi Katoh; Yosuke Ooizumi; Kazuharu Igarashi; Satoru Ishii; Toshimichi Tanaka; Keigo Yokoi; Nobuyuki Nishizawa; Kazuko Yokota; Yoshimasa Kosaka; Takeo Sato; Masahiko Watanabe; Keishi Yamashita

Background Progression of colorectal cancer (CRC) has been explained by genomic abnormalities along with the adenoma-carcinoma sequence theory (ACS). The aim of our study is to elucidate whether the promoter DNA methylation of the cancer-specific methylation gene, cysteine dioxygenase 1 (CDO1), contributes to the carcinogenic process in CRC. Methods The study group comprised 107 patients with CRC who underwent surgical resection and 90 adenomas treated with endoscopic resection in the Kitasato University Hospital in 2000. We analyzed the extent of methylation in each tissue using quantitative TaqMan methylation-specific PCR for CDO1. Results The methylation level increased along with the ACS process (p < 0.0001), and statistically significant differences were found between normal-appearing mucosa (NAM) and low-grade adenoma (p < 0.0001), and between low-grade adenoma and high-grade adenoma (p = 0.01), but not between high-grade adenoma and cancer with no liver metastasis. Furthermore, primary CRC cancers with liver metastasis harbored significantly higher methylation of CDO1 than those without liver metastasis (p = 0.02). As a result, the area under the curve by CDO1 promoter methylation was 0.96, 0.80, and 0.67 to discriminate cancer from NAM, low-grade adenoma from NAM, and low-grade adenoma from high-grade adenoma, respectively. Conclusions CDO1 methylation accumulates during the ACS process, and consistently contributes to CRC progression.


Journal of Gastroenterology and Hepatology | 2017

RAMP1 suppresses mucosal injury from dextran sodium sulfate-induced colitis in mice.

Noriko Kawashima-Takeda; Yoshiya Ito; Nobuyuki Nishizawa; Rei Kawashima; Kiyoshi Tanaka; Kazutake Tsujikawa; Masahiko Watanabe; Masataka Majima

Calcitonin gene‐related peptide (CGRP) is thought to be involved in the modulation of intestinal motility. CGRP receptor is composed of receptor activity‐modifying protein (RAMP) 1 combined with calcitonin receptor‐like receptor (CRLR) for CGRP. The study investigated the role of CGRP in mice with experimentally induced colitis.


Cancer Letters | 2017

Epigenetic silencing of HOPX contributes to cancer aggressiveness in breast cancer

Mariko Kikuchi; Hiroshi Katoh; Mina Waraya; Yoko Tanaka; Satoru Ishii; Toshimichi Tanaka; Nobuyuki Nishizawa; Keigo Yokoi; Naoko Minatani; Akira Ema; Yoshimasa Kosaka; Hirokazu Tanino; Keishi Yamashita; Masahiko Watanabe

Epigenetic silencing of HOPX has been shown to be frequent and specific in human cancers. HOPX is thought as a tumor suppressor gene and its promoter methylation is the main mechanism of down-regulation. In non-hereditary breast cancer, since roles of epigenetic modifications are more critical than in other cancers, the aim of this study is to seek into the roles and clinical relevance of epigenetic silencing of HOPX. Down-regulation of HOPX was observed in all human breast cancer cell lines tested. The promoter methylation was found in six of seven cell lines, and demethylating agents restored HOPX expression. The promoter methylation was cancer-specific in human breast tissues. Forced expression of HOPX attenuated anchorage-independent growth inxa0vitro. HOPX promoter methylation independently predicted worse prognosis of breast cancer patients. Of note, HOPX promoter methylation was significantly associated with HER2 positivity as well as advanced lymph node metastasis. HOPX promoter methylation is not only frequent and cancer-specific but also associated with aggressive phenotype in breast cancer. Epigenetic silencing of HOPX may have clinical potential as a biomarker in the treatment strategy of breast cancer patients.

Collaboration


Dive into the Nobuyuki Nishizawa'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