Network


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

Hotspot


Dive into the research topics where Yozo Mitsui is active.

Publication


Featured researches published by Yozo Mitsui.


BJUI | 2012

Prediction of survival benefit using an automated bone scan index in patients with castration-resistant prostate cancer.

Yozo Mitsui; Hiroaki Shiina; Yasushi Yamamoto; Masuo Haramoto; Naoko Arichi; Hiroaki Yasumoto; Hajime Kitagaki; Mikio Igawa

Study Type – Prognosis (case series)


Cuaj-canadian Urological Association Journal | 2011

Identification of lymphatic pathway involved in the spreading of prostate cancer by fluorescence navigation approach with intraoperatively injected indocyanine green

Shogo Inoue; Hiroaki Shiina; Naoko Arichi; Yozo Mitsui; Takeo Hiraoka; Koji Wake; Masahiro Sumura; Satoshi Honda; Hiroaki Yasumoto; Shinji Urakami; Akio Matsubara; Mikio Igawa

OBJECTIVE : The objective of this study was to identify lymphatic vessels draining from the prostate by using a fluorescence navigation (FN) system. METHODS : Fourteen subjects were candidates for radical retropubic prostatectomy (RRP) and pelvic lymph node dissection (PLND). After an indocyanine green solution was injected into the prostate during RRP, lymphatic vessels draining from the prostate were analyzed using a FN system. After PLND based on lymphatic mapping by the FN system (in vivo probing) was performed in the external iliac, obturator and internal iliac regions; the fluorescence of the removed lymph nodes (LNs) was analyzed on the bench (ex vivo probing). RESULTS : Under in vivo and ex vivo probing, the fluorescence intensity of internal iliac nodes was greater than that of external iliac or obturator nodes. CONCLUSION : The current study suggests that using a FN system after injecting indocyanine green is a safe and rational approach for detecting the lymphatic channel draining from the prostate. The major lymphatic pathway involved in the spreading of prostate cancer appears to relate to internal iliac LNs, which would mean that the standard PLND covering external iliac and obturator regions would not keep the cancer from spreading.


Oncotarget | 2016

Oncogenic microRNA-4534 regulates PTEN pathway in prostate cancer

Hannah Nip; Altaf A. Dar; Melissa Colden; Shahryari Varahram; Harshika Chowdhary; Soichiro Yamamura; Yozo Mitsui; Yuichiro Tanaka; Taku Kato; Yutaka Hashimoto; Marisa Shiina; Priyanka Kulkarni; Pritha Dasgupta; Mitsuho Imai-Sumida; Z. Laura Tabatabai; Kirsten L. Greene; Guoren Deng; Rajvir Dahiya; Shahana Majid

Prostate carcinogenesis involves alterations in several signaling pathways, the most prominent being the PI3K/AKT pathway. This pathway is constitutively active and drives prostate cancer (PCa) progression to advanced metastatic disease. PTEN, a critical tumor and metastasis suppressor gene negatively regulates cell survival, proliferation, migration and angiogenesis via the PI3K/Akt pathway. PTEN is mutated, downregulated/dysfunctional in many cancers and its dysregulation correlates with poor prognosis in PCa. Here, we demonstrate that microRNA-4534 (miR-4534) is overexpressed in PCa and show that miR-4534 is hypermethylated in normal tissues and cell lines compared to PCa tissues/cells. miR-4534 exerts its oncogenic effects partly by downregulating the tumor suppressor PTEN gene. Knockdown of miR-4534 impaired cell proliferation, migration/invasion and induced G0/G1 cell cycle arrest and apoptosis in PCa. Suppression of miR-4534 and its effects on tumor growth was confirmed in a xenograft mouse model. We performed parallel experiments in non-cancer RWPE1 cells by overexpessing miR-4534 followed by functional assays. Overexpression of miR-4534 induced pro-cancerous characteristics in this non-cancer cell line. Statistical analyses revealed that miR-4534 has potential to independently distinguish malignant from normal tissues and positively correlated with poor overall and PSA recurrence free survival. Taken together, our results show that depletion of miR-4534 in PCa induces a tumor suppressor phenotype partly through induction of PTEN. These results have important implications for identifying and defining the role of new PTEN regulators such as microRNAs in prostate tumorigenesis. Understanding aberrantly overexpressed miR-4534 and its downregulation of PTEN will provide mechanistic insight and therapeutic targets for PCa therapy.


International Urology and Nephrology | 2012

Indocyanine green (ICG)-based fluorescence navigation system for discrimination of kidney cancer from normal parenchyma: application during partial nephrectomy

Yozo Mitsui; Hiroaki Shiina; Naoko Arichi; Takeo Hiraoka; Shogo Inoue; Masahiro Sumura; Satoshi Honda; Hiroaki Yasumoto; Mikio Igawa

PurposeTo determine the definite border between normal and tumor kidney tissues during partial nephrectomy (PN) procedures using intraoperative indocyanine green (ICG)-based fluorescence imaging.MethodsSixteen potential candidates for PN with organ-confined, small renal masses treated between July 2008 and June 2011 at Shimane University Hospital were enrolled. An ICG-based fluorescence navigation (FN) system was used to evaluate the border between the tumor and normal kidney parenchyma (step 1), the cavity following tumor excision (step 2), and the negative surgical margin of resected tissues (step 3). The R.E.N.A.L nephrometry score (RNS) was applied to evaluate the correlation between tumor anatomy and ICG-based fluorescence imaging.ResultsIn step 1, in vivo probing revealed 14 tumors with a mean RNS of 7 points that showed quite low ICG fluorescence signals in the tumor mass as compared with normal kidney parenchyma. In step 2, in vivo probing around the bed revealed highly fluorescent signals with no remnant tumor residing in 10 cases with a mean RNS of 6 points. In step 3, ex vivo probing revealed cancer tissues involving normal parenchyma that were completely excised with minimum amounts of normal parenchyma in all 16 resected specimens.ConclusionsICG-based FN system was very helpful for confirming negative margin status in even the most complex cases. Further evaluations may open the door for widespread use of this ICG-based FN system as a feasible and attractive alternative during a PN procedure.


Oncogene | 2017

MicroRNA-383 located in frequently deleted chromosomal locus 8p22 regulates CD44 in prostate cancer

Nathan Bucay; Kirandeep Sekhon; Thao Yang; Shahana Majid; C Hsieh; Yozo Mitsui; Guoren Deng; Z L Tabatabai; Soichiro Yamamura; G A Calin; Rajvir Dahiya; Yuichiro Tanaka

A major genomic alteration in prostate cancer (PCa) is frequent loss of chromosome (chr) 8p with a common region of loss of heterozygosity (LOH) at chr8p22 locus. Genomic studies implicate this locus in the initiation of clinically significant PCa and with progression to metastatic disease. However, the genes within this region have not been fully characterized to date. Here we demonstrate for the first time that a microRNA component of this region—miR-383—is frequently downregulated in prostate cancer, has a critical role in determining tumor-initiating potential and is involved in prostate cancer metastasis via direct regulation of CD44, a ubiquitous marker of PCa tumor-initiating cells (TICs)/stem cells. Expression analyses of miR-383 in PCa clinical tissues established that low miR-383 expression is associated with poor prognosis. Functional data suggest that miR-383 regulates PCa tumor-initiating/stem-like cells via CD44 regulation. Ectopic expression of miR-383 inhibited tumor-initiating capacity of CD44+ PCa cells. Also, ‘anti-metastatic’ effects of ectopic miR-383 expression were observed in a PCa experimental metastasis model. In view of our results, we propose that frequent loss of miR-383 at chr8p22 region leads to tumor initiation and prostate cancer metastasis. Thus, we have identified a novel finding that associates a long observed genomic alteration to PCa stemness and metastasis. Our data suggest that restoration of miR-383 expression may be an effective therapeutic modality against PCa. Importantly, we identified miR-383 as a novel PCa tissue diagnostic biomarker with a potential that outperforms that of serum PSA.


Molecular Cancer Research | 2017

Versican Promotes Tumor Progression, Metastasis and Predicts Poor Prognosis in Renal Carcinoma

Yozo Mitsui; Hiroaki Shiina; Taku Kato; Shigekatsu Maekawa; Yutaka Hashimoto; Marisa Shiina; Mitsuho Imai-Sumida; Priyanka Kulkarni; Pritha Dasgupta; Ryan Kenji Wong; Miho Hiraki; Naoko Arichi; Shinichiro Fukuhara; Soichiro Yamamura; Shahana Majid; Guoren Deng; Rajvir Dahiya; Koichi Nakajima; Yuichiro Tanaka

The proteoglycan versican (VCAN) promotes tumor progression and enhances metastasis in several cancers; however, its role in clear cell renal cell carcinoma (ccRCC) remains unknown. Recent evidence suggests that VCAN is an important target of chromosomal 5q gain, one of the most prevalent genetic abnormalities in ccRCC. Thus, we investigated whether VCAN expression is associated with the pathogenesis of ccRCC. VCAN expression was analyzed using three RCC and normal kidney cell lines as well as a clinical cohort of 84 matched ccRCC and normal renal tissues. Functional analyses on growth and progression properties were performed using VCAN-depleted ccRCC cells. Microarray expression profiling was employed to investigate the target genes and biologic pathways involved in VCAN-mediated ccRCC carcinogenesis. ccRCC had elevated VCAN expression in comparison with normal kidney in both cell lines and clinical specimens. The elevated expression of VCAN was significantly correlated with metastasis (P < 0.001) and worse 5-year overall survival after radical nephrectomy (P = 0.014). In vitro, VCAN knockdown significantly decreased cell proliferation and increased apoptosis in Caki-2 and 786-O cells, and this was associated with alteration of several TNF signaling–related genes such as TNFα, BID, and BAK. Furthermore, VCAN depletion markedly decreased cell migration and invasion which correlated with reduction of MMP7 and CXCR4. These results demonstrate that VCAN promotes ccRCC tumorigenesis and metastasis and thus is an attractive target for novel diagnostic, prognostic, and therapeutic strategies. Implications: This study highlights the oncogenic role of VCAN in renal cell carcinogenesis and suggests that this gene has therapeutic and/or biomarker potential for renal cell cancer. Mol Cancer Res; 15(7); 884–95. ©2017 AACR.


International Urology and Nephrology | 2012

Current chemotherapeutic strategies against bladder cancer

Yozo Mitsui; Hiroaki Yasumoto; Naoko Arichi; Satoshi Honda; Hiroaki Shiina; Mikio Igawa

Urothelial cancer is a chemotherapy-sensitive malignancy, with the regimen of methotrexate, vinblastine, doxorubicin, and cisplatin (M-VAC) until recently considered to be the first choice for chemotherapy. Poor survival and substantial toxicity associated with M-VAC have led to investigations into alternative chemotherapy strategies, and the combination of gemcitabine and cisplatin (GC) may be promising. In addition, combination chemotherapy of taxanes along with gemcitabine and/or platinum-based agents is also considered to provide clinical benefits as second-line chemotherapy following M-VAC or GC therapy. In the near future, results of trials using molecular target therapies may bring improved outcomes for patients with bladder cancer.


BMC Cancer | 2015

CYP1B1 promotes tumorigenesis via altered expression of CDC20 and DAPK1 genes in renal cell carcinoma

Yozo Mitsui; Inik Chang; Shinichiro Fukuhara; Miho Hiraki; Naoko Arichi; Hiroaki Yasumoto; Hiroshi Hirata; Soichiro Yamamura; Guoren Deng; Darryn K. Wong; Shahana Majid; Hiroaki Shiina; Rajvir Dahiya; Yuichiro Tanaka

BackgroundCytochrome P450 1B1 (CYP1B1) has been shown to be up-regulated in many types of cancer including renal cell carcinoma (RCC). Several reports have shown that CYP1B1 can influence the regulation of tumor development; however, its role in RCC has not been well investigated. The aim of the present study was to determine the functional effects of CYP1B1 gene on tumorigenesis in RCC.MethodsExpression of CYP1B1 was determined in RCC cell lines, and tissue microarrays of 96 RCC and 25 normal tissues. To determine the biological significance of CYP1B1 in RCC progression, we silenced the gene in Caki-1 and 769-P cells by RNA interference and performed various functional analyses.ResultsFirst, we confirmed that CYP1B1 protein expression was significantly higher in RCC cell lines compared to normal kidney tissue. This trend was also observed in RCC samples (p < 0.01). Interestingly, CYP1B1 expression was associated with tumor grade and stage. Next, we silenced the gene in Caki-1 and 769-P cells by RNA interference and performed various functional analyses to determine the biological significance of CYP1B1 in RCC progression. Inhibition of CYP1B1 expression resulted in decreased cell proliferation, migration and invasion of RCC cells. In addition, reduction of CYP1B1 induced cellular apoptosis in Caki-1. We also found that these anti-tumor effects on RCC cells caused by CYP1B1 depletion may be due to alteration of CDC20 and DAPK1 expression based on gene microarray and confirmed by real-time PCR. Interestingly, CYP1B1 expression was associated with CDC20 and DAPK1 expression in clinical samples.ConclusionsCYP1B1 may promote RCC development by inducing CDC20 expression and inhibiting apoptosis through the down-regulation of DAPK1. Our results demonstrate that CYP1B1 can be a potential tumor biomarker and a target for anticancer therapy in RCC.


Cancer Epidemiology, Biomarkers & Prevention | 2012

Tumor Suppressor Function of PGP9.5 Is Associated with Epigenetic Regulation in Prostate Cancer—Novel Predictor of Biochemical Recurrence after Radical Surgery

Yozo Mitsui; Hiroaki Shiina; Miho Hiraki; Naoko Arichi; Takeo Hiraoka; Masahiro Sumura; Satoshi Honda; Hiroaki Yasumoto; Mikio Igawa

Background: The expression level of protein G product 9.5 (PGP9.5) is downregulated because of promoter CpG hypermethylation in several tumors. We speculated that impaired regulation of PGP9.5 through epigenetic pathways is associated with the pathogenesis of prostate cancer. Methods: CpG methylation of the PGP9.5 gene was analyzed in cultured prostate cancer cell lines, 226 localized prostate cancer samples from radical prostatectomy cases, and 80 benign prostate hyperplasia (BPH) tissues. Results: Following 5-aza-2′-deoxycytidune treatment, increased PGP9.5 mRNA transcript expression was found in the LNCaP and PC3 cell lines. With bisulfite DNA sequencing, partial methylation of the PGP9.5 promoter was shown in LNCaP whereas complete methylation was found in PC3 cells. After transfection of PGP9.5 siRNA, cell viability was significantly accelerated in LNCaP but not in PC3 cells as compared with control siRNA transfection. Promoter methylation of PGP9.5 was extremely low in only one of 80 BPH tissues, whereas it was found in 37 of 226 prostate cancer tissues. Expression of the mRNA transcript of PGP9.5 was significantly lower in methylation (+) than methylation (−) prostate cancer tissues. Multivariate analysis of biochemical recurrence (BCR) after an radical prostatectomy revealed pT category and PGP9.5 methylation as prognostically relevant. Further stratification with the pT category in addition to methylation status identified a stepwise reduction of BCR-free probability. Conclusion: This is the first clinical and comprehensive study of inactivation of the PGP9.5 gene via epigenetic pathways in primary prostate cancer. Impact: CpG methylation of PGP9.5 in primary prostate cancer might become useful as a molecular marker for early clinical prediction of BCR after radical prostatectomy. Cancer Epidemiol Biomarkers Prev; 21(3); 487–96. ©2012 AACR.


Journal of Visualized Experiments | 2015

miRNA Expression Analyses in Prostate Cancer Clinical Tissues

Nathan Bucay; Shahana Majid; Soichiro Yamamura; Yozo Mitsui; Z. Laura Tabatabai; Kirsten L. Greene; Guoren Deng; Rajvir Dahiya; Yuichiro Tanaka

A critical challenge in prostate cancer (PCa) clinical management is posed by the inadequacy of currently used biomarkers for disease screening, diagnosis, prognosis and treatment. In recent years, microRNAs (miRNAs) have emerged as promising alternate biomarkers for prostate cancer diagnosis and prognosis. However, the development of miRNAs as effective biomarkers for prostate cancer heavily relies on their accurate detection in clinical tissues. miRNA analyses in prostate cancer clinical specimens is often challenging owing to tumor heterogeneity, sampling errors, stromal contamination etc. The goal of this article is to describe a simplified workflow for miRNA analyses in archived FFPE or fresh frozen prostate cancer clinical specimens using a combination of quantitative real-time PCR (RT-PCR) and in situ hybridization (ISH). Within this workflow, we optimize the existing methodologies for miRNA extraction from FFPE and frozen prostate tissues and expression analyses by Taqman-probe based miRNA RT-PCR. In addition, we describe an optimized method for ISH analyses formiRNA detection in prostate tissues using locked nucleic acid (LNA)- based probes. Our optimized miRNA ISH protocol can be applied to prostate cancer tissue slides or prostate cancer tissue microarrays (TMA).

Collaboration


Dive into the Yozo Mitsui's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rajvir Dahiya

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Soichiro Yamamura

San Francisco VA Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shahana Majid

San Francisco VA Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guoren Deng

San Francisco VA Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge