Network


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

Hotspot


Dive into the research topics where Takuji Mori is active.

Publication


Featured researches published by Takuji Mori.


Journal of Clinical Oncology | 2005

Predictive Utility of Circulating Methylated DNA in Serum of Melanoma Patients Receiving Biochemotherapy

Takuji Mori; Steven J. O’Day; Naoyuki Umetani; Steve R. Martinez; Minoru Kitago; Kazuo Koyanagi; Christine Kuo; Teh-Ling Takeshima; Robert Milford; He-Jing Wang; Vu D. Vu; Sandy L. Nguyen; Dave S.B. Hoon

PURPOSE Currently, no validated blood-based assays accurately predict treatment response or outcome in melanoma patients. We hypothesized that methylation of tumor-related genes detected in serum DNA could predict disease outcome and therapeutic response in patients receiving concurrent biochemotherapy (BC) for metastatic melanoma. PATIENTS AND METHODS American Joint Committee on Cancer stage IV melanoma patients (N = 50) had blood drawn before administration of BC. Patients (n = 47) were classified as BC responders or nonresponders. Responders (n = 23) demonstrated a complete or partial response following BC; nonresponders (n = 24) demonstrated progressive disease. Hypermethylation of Ras association domain family 1 (RASSF1A), retinoic acid receptor-beta2 (RAR-beta2), and O6-methylguanine DNA methyltransferase (MGMT) genes were assessed by methylation-specific polymerase chain reaction. RESULTS Circulating methylated RASSF1A was significantly less frequent for responders (three of 23 patients; 13%) than nonresponders (10 of 24 patients; 42%; P = .028). Patients with RASSF1A, RAR-beta2, or at least one serum methylated gene had significantly worse overall survival than patients with no methylated genes (log-rank, P = .013, .021, and .01, respectively). Methylated RASSF1A was the only factor that significantly correlated with overall survival and BC response (risk ratio, 2.38; 95% CI, 1.16 to 4.86; P = .018; odds ratio = 0.21; 95% CI, 0.05 to 0.90; P = .036). CONCLUSION Detection of circulating methylated DNA in serum can predict response to BC and disease outcome.


Annals of Surgery | 2006

Chemokine Receptor CXCR4 Expression in Patients With Melanoma and Colorectal Cancer Liver Metastases and the Association With Disease Outcome

Joseph Kim; Takuji Mori; Steven L. Chen; Farin Amersi; Steve R. Martinez; Christine Kuo; Roderick R. Turner; Xing Ye; Anton J. Bilchik; Donald L. Morton; Dave S.B. Hoon

Objective:To determine the role of chemokine receptor (CR) expression in patients with melanoma and colorectal cancer (CRC) liver metastases. Summary Background Data:Murine and in vitro models have identified CR as potential factors in organ-specific metastasis of multiple cancers. Chemokines via their respective receptors have been shown to promote cell migration to distant organs. Methods:Patients who underwent hepatic surgery for melanoma or CRC liver metastases were assessed. Screening cDNA microarrays of melanoma/CRC cell lines and tumor specimens were analyzed to identify CR. Microarray data were validated by quantitative real-time RT-PCR (qRT) in paraffin-embedded liver metastases. Migration assays and immunohistochemistry were performed to verify CR function and confirm CR expression, respectively. Results:Microarray analysis identified CXCR4 as the most common CR expressed by both cancers. qRT demonstrated CXCR4 expression in 24 of 27 (89%) melanoma and 28 of 29 (97%) CRC liver metastases. In vitro treatment of melanoma or CRC cells with CXCL12, the ligand for CXCR4, significantly increased cell migration (P < 0.001). Low versus high CXCR4 expression in CRC liver metastases correlated with a significant difference in overall survival (median 27 months vs. 10 months, respectively; P = 0.036). In melanoma, low versus high CXCR4 expression in liver metastases demonstrated no difference in overall survival (median 11 months vs. 8 months, respectively; P = not significant). Conclusions:CXCR4 is expressed and functional on melanoma and CRC cells. The ligand for CXCR4 is highly expressed in liver and may specifically attract melanoma and CRC CXCR4 (+) cells. Quantitative analysis of CXCR4 gene expression in patients with liver metastases has prognostic significance for disease outcome.


Cancer Research | 2005

Epigenetic up-regulation of C-C chemokine receptor 7 and C-X-C chemokine receptor 4 expression in melanoma cells.

Takuji Mori; Joseph Kim; Tomoki Yamano; Hiroya Takeuchi; Sharon K. Huang; Naoyuki Umetani; Kazuo Koyanagi; Dave S.B. Hoon

Histone deacetylation and DNA methylation establish epigenetic modifications, which through chromatin remodeling may result in gene silencing. We hypothesized that chemokine receptors C-C chemokine receptor 7 (CCR7) and C-X-C chemokine receptor 4 (CXCR4) on melanoma cells undergo epigenetic regulation. We investigated whether a histone deacetylase inhibitor and a demethylating agent influence CCR7 and CXCR4 expression on melanoma cells. Initially, microarray analysis was done to screen changes in chemokine receptor expression on melanoma cells after treatment with trichostatin A (TSA) and 5-Aza-2-deoxycytidine (5-Aza). CCR7 and CXCR4 mRNA expression were uniformly altered and selected for further investigation. Quantitative real-time reverse transcription-PCR assay, immunohistochemistry, and Western blot analysis were used to assess changes in mRNA and protein expression induced by TSA and 5-Aza in melanoma lines. Cell migration assays were conducted to assess the effects of altered CCR7 and CXCR4 expression on cell function. Treatment with TSA or 5-Aza increased gene expression of both CCR7 and CXCR4 in melanoma lines. TSA was the strongest enhancer. With combined treatment, CCR7 and CXCR4 mRNA expression was also up-regulated. Immunohistochemistry after combined treatment showed enhanced staining of both CCR7 and CXCR4 compared with control cells. Melanoma cell migration in TSA- and 5-Aza-treated cells was 7- and 2-fold higher than control cells for CCR7 and CXCR4, respectively. In summary, a histone deacetylase inhibitor and a demethylating agent up-regulated CCR7 and CXCR4 expression on melanoma cells. This increase in chemokine receptor expression correlated with functional activity. Most importantly, we have identified an epigenetic mechanism that may endogenously regulate chemokine receptor expression on melanoma cells.


Oncogene | 2005

Aberrant hypermethylation of ID4 gene promoter region increases risk of lymph node metastasis in T1 breast cancer

Naoyuki Umetani; Takuji Mori; Kazuo Koyanagi; Masaru Shinozaki; Joseph Kim; Armando E. Giuliano; Dave S.B. Hoon

ID4 gene is a member of the inhibitor of DNA-binding (ID) family, which inhibits DNA binding of basic helix–loop–helix transcription factors. Certain human primary breast cancers reportedly have low or no expression of ID4 protein, but its role in carcinogenesis and cancer progression is unknown. To determine its possible role, we examined epigenetic inactivation of ID4 gene by promoter hypermethylation in human breast cell lines and T1 breast cancer tissues. Methylation status of ID4 promoter CpG island was assessed by methylation-specific PCR (MSP); ID4 mRNA level was assessed by quantitative real-time RT–PCR. Of eight cell lines, two were fully methylated, four were partially methylated, and two were not methylated. ID4 mRNA level was suppressed in fully methylated cell lines. ID4 hypermethylation was observed in 16 of 24 (67%) node-positive and seven of 36 (19%) node-negative T1 primary breast cancers matched by patient age and tumor diameter. It was a significant risk factor for nodal metastasis (OR 13.1, P=0.0004). ID4 mRNA level was suppressed in hypermethylated cancer specimens (P=0.014). ID4 may play an important suppressive role in tumor progression, and its silencing by hypermethylation may increase the risk of regional lymph node metastasis.


Cancer and Metastasis Reviews | 2006

Molecular mechanisms of metastasis.

Dave S.B. Hoon; Minoru Kitago; Joseph Kim; Takuji Mori; Adriano Piris; Katherine Szyfelbein; Martin C. Mihm; S. David Nathanson; Timothy P. Padera; Ann F. Chambers; Sharon A. Vantyghem; Ian C. MacDonald; Steven C. Shivers; Marwan Alsarraj; Douglas S. Reintgen; Bernward Passlick; Wulf Sienel; Klaus Pantel

A major topic covered at the First International Symposium on Cancer Metastasis and the Lymphovascular System was the molecular mechanisms of metastasis. This has become of major interest in recent years as we have discovered new metastasis-related genes and gained understanding of the molecular events of lymphatic metastasis. The symposium covered new aspects and important questions related to the events of metastasis in both humans and animals. The basic and clinical related research covered in this topic represented many disciplines. The presentations showed novel findings and at the same time, raised many new unanswered questions, indicating the limited knowledge we still have regarding the molecular events of metastasis. The hope is that further unraveling of the direct and indirect molecular events of lymphatic metastasis will lead to new approaches in developing effective therapeutics.


Clinical Cancer Research | 2005

X-Linked Inhibitor of Apoptosis Protein Expression Level in Colorectal Cancer Is Regulated by Hepatocyte Growth Factor/C-Met Pathway via Akt Signaling

Hiroya Takeuchi; Joseph Kim; Akihide Fujimoto; Naoyuki Umetani; Takuji Mori; Anton J. Bilchik; Rod Turner; Andy Tran; Christine Kuo; Dave S.B. Hoon

Purpose: The inhibitor of the apoptosis protein (IAP) family members, such as the X-linked IAP (XIAP), survivin, and livin, are essential for cell survival and antiapoptosis in colorectal cancer cells. We hypothesized that the hepatocyte growth factor (HGF) activation in colorectal cancer via c-Met receptor regulates IAP proteins through Akt signaling. Experimental Design: The level of IAPs and C-Met mRNA expression was assessed using a quantitative real-time reverse transcriptase-PCR (RT-PCR) assay on colorectal normal mucosa (n = 13), adenomas (n = 6), and colorectal cancer tumors (n = 50). The role of HGF/C-Met pathway through Akt and XIAP was investigated by small interfering RNA (siRNA) and quantitative RT-PCR analysis of colorectal cancer lines. Results: Of the IAPs, only XIAP showed significant correlation to tumor development and progression. XIAP mRNA level in primary colorectal cancer was significantly higher than that in colorectal normal mucosa (P = 0.01); liver metastases was significantly higher than primary colorectal cancer tumors (P = 0.04); and primary colorectal cancer N1/N2 cases were significantly higher than N0 cases (P = 0.008). HGF stimulation of colorectal cancer lines enhanced XIAP mRNA expression but not other IAPs. Activation of XIAP expression by HGF was inhibited by siRNA targeting Akt1 and Akt2. Conclusions: Activation of C-MET enhances XIAP through the Akt pathway. XIAP up-regulation was shown to be correlated to colorectal cancer tumor progression. The Akt-XIAP pathway may be a potential molecular target for regulating colorectal cancer progression.


Expert Opinion on Therapeutic Targets | 2007

The role of estrogen receptor in melanoma

Atsushi Tanemura; Anneke Q. van Hoesel; Takuji Mori; Terrance Yu; Dave S.B. Hoon

The estrogen receptor (ER) belongs to the group of sex hormone receptors and binds the biologically active form of estrogen, 17β-estradiol. Expression of ER in tumor tissue is a well-established prognostic marker in breast cancer. The role of ER has been extensively studied in several other types of human cancers. This report investigates the potential role of ER as a surrogate marker for predicting melanoma progression, response to therapy, and patient survival. In addition, the authors review what is known, so far, about ER signaling pathways and their potential role in carcinogenesis and progression of cutaneous melanoma. Possibilities and limitations of using ER as a therapeutic target in the treatment of melanoma is also discussed.


Archive | 2009

Sentinel Lymph Node Chemokine Microenvironment Modulated by Melanoma Metastasis

Steve R. Martinez; Takuji Mori; Kazuo Koyanagi; Naoyuki Umetani; Roderick R. Turner; Dave S.B. Hoon

Primary melanoma tumor-draining lymph nodes (LNs) play a significant role in controlling regional metastasis. Chemokine receptors CXCR4 and CCR7 are expressed on immune cells, whereby their respective ligands are, CXCL12 and CCL21. We hypothesized that melanoma metastasis to the sentinel LN (SLN) suppresses chemokine production, creating a more favorable tumor microenvironment for metastasis. A quantitative real-time reverse transcription PCR (qRT) assay was used to assess CXCL12 and CCL21 expression in paraffin-embedded (PE) SLN from melanoma patients (n = 124). SLN metastases were diagnosed by hematoxylin and eosin (HE CCL21, p = 0.006) or in SLN without metastasis (CXCL12, p = 0.04; CCL21, p < 0.001). IHC analysis of SLN showed that the chemokine mRNA expression correlated with protein expression. Increase in primary tumor burden significantly correlated with suppressed CXCL12 (p < 0.0001) and CCL21 (p < 0.05) expression in the SLN. Progression of nodal metastasis burden and primary tumor burden was shown to significantly suppress both SLN CXCL12 and CCL21. Reversal of this chemokine suppression may improve the immunity of draining LN against tumor invasion.


Clinical Chemistry | 2005

Multimarker Quantitative Real-Time PCR Detection of Circulating Melanoma Cells in Peripheral Blood: Relation to Disease Stage in Melanoma Patients

Kazuo Koyanagi; Christine Kuo; Taku Nakagawa; Takuji Mori; Hideaki Ueno; Arnulfo R. Lorico; He-Jing Wang; Eddie Hseuh; Steven O'Day; Dave S.B. Hoon


Cancer Research | 2006

Estrogen Receptor-α Methylation Predicts Melanoma Progression

Takuji Mori; Steve R. Martinez; Steven O'Day; Donald L. Morton; Naoyuki Umetani; Atsushi Tanemura; Sandy L. Nguyen; Andy Tran; He-Jing Wang; Dave S.B. Hoon

Collaboration


Dive into the Takuji Mori'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

Christine Kuo

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

He-Jing Wang

University of California

View shared research outputs
Top Co-Authors

Avatar

Minoru Kitago

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge