Network


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

Hotspot


Dive into the research topics where Yasuro Sugimoto is active.

Publication


Featured researches published by Yasuro Sugimoto.


The Journal of Steroid Biochemistry and Molecular Biology | 2005

Translational studies on aromatase, cyclooxygenases, and enzyme inhibitors in breast cancer☆

Robert W. Brueggemeier; Edgar S. Diaz-Cruz; Pui-Kai Li; Yasuro Sugimoto; Young C. Lin; Charles L. Shapiro

Aromatase expression and enzyme activity in breast cancer patients is greater in or near the tumor tissue compared with the normal breast tissue. Regulation of aromatase expression in human tissues is quite complex, involving alternative promoter sites that provide tissue-specific control. Previous studies in our laboratories suggested a strong association between aromatase (CYP19) gene expression and the expression of cyclooxygenase (COX) genes. Our hypothesis is that higher levels of COX expression result in higher levels of prostaglandin E2 (PGE2), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels. This biochemical mechanism may explain the beneficial effects of non-steroidal anti-inflammatory drugs (NSAIDs) on reducing the risks of breast cancer. The effects of NSAIDs (ibuprofen, piroxicam, and indomethacin), a COX-1 selective inhibitor (SC-560), and COX-2 selective inhibitors (celecoxib, niflumic acid, nimesulide, NS-398, and SC-58125) on aromatase activity and CYP19 expression were investigated in breast cancer cell culture systems. Dose-dependent decreases in aromatase activity were observed following treatment with an NSAID or COX inhibitor, with the most effective agents being COX selective inhibitors. Real time PCR analysis of aromatase gene expression showed a significant decrease in mRNA levels in treated cells when compared to vehicle control. These results suggest that the effect of COX inhibitors on aromatase occurs at the transcriptional level. To further probe these interactions, short interfering RNAs (siRNA) were designed against either human CYP19 mRNA or human COX-2 mRNA. Treatment of breast cancer cells with aromatase siRNAs suppressed CYP19 mRNA and aromatase enzyme activity. Finally, treatment with COX-2 siRNAs downregulated the expression of COX-2 mRNA; furthermore, the siCOX-2-mediated suppression of COX-2 also resulted in suppression of aromatase mRNA. In summary, pharmacological regulation of aromatase and cyclooxygenases can act locally in an autocrine fashion to decrease the biosynthesis of estrogen and may provide additional therapy options for patients with hormone-dependent breast cancer.


The Journal of Steroid Biochemistry and Molecular Biology | 2007

Aromatase and COX in breast cancer: Enzyme inhibitors and beyond ☆

Robert W. Brueggemeier; Bin Su; Yasuro Sugimoto; Edgar S. Diaz-Cruz; Danyetta D. Davis

Aromatase expression and enzyme activity in breast cancer patients is greater in or near the tumor tissue compared with the normal breast tissue. Complex regulation of aromatase expression in human tissues involves alternative promoter sites that provide tissue-specific control. Previous studies in our laboratories suggested a strong association between aromatase (CYP19) gene expression and the expression of cyclooxygenase (COX) genes. Additionally, nonsteroidal anti-inflammatory drugs (NSAIDs) and COX selective inhibitors can suppress CYP19 gene expression and decrease aromatase activity. Our current hypothesis is that pharmacological regulation of aromatase and/or cyclooxygenases can act locally to decrease the biosynthesis of estrogen and may provide additional therapy options for patients with hormone-dependent breast cancer. Two pharmacological approaches are being developed, one involving mRNA silencing by selective short interfering RNAs (siRNA) molecules and the second utilizing small molecule drug design. In the first approach, short interfering RNAs were designed against either human aromatase mRNA or human COX-2 mRNA. Treatment of breast cancer cells with siAROMs completely masked the aromatase enzyme activity. Treatment with COX-2 siRNAs decreased the expression of COX-2 mRNA; furthermore, the siCOX-2-mediated decrease also resulted in suppression of CYP19 mRNA. The small molecule drug design approach focuses on the synthesis and biological evaluation of a novel series of sulfonanilide analogs derived from the COX-2 selective inhibitors. The compounds suppress aromatase enzyme activity in SK-BR-3 breast cancer cells in a dose and time-dependent manner, and structure activity analysis does not find a correlation between aromatase suppression and COX inhibition. Real-time PCR analysis demonstrates that the sulfonanilide analogs decrease aromatase gene transcription in breast cells. Thus, these results suggest that the siRNAs and novel sulfonanilides targeting aromatase expression may be valuable tools for selective regulation of aromatase in breast cancer.


Molecular and Cellular Endocrinology | 1999

Parathyroid hormone-related protein is a positive regulator of keratinocyte growth factor expression by normal dermal fibroblasts.

Eric A.G. Blomme; Yasuro Sugimoto; Young C. Lin; Charles C. Capen; Thomas J. Rosol

Parathyroid hormone-related protein (PTHrP), an important factor in the pathogenesis of humoral hypercalcemia of malignancy, is produced by many normal tissues, including the skin, where it regulates keratinocyte growth and differentiation and dermal fibroblast function. Keratinocyte growth factor (KGF), a member of the fibroblast growth factor (FGF) family, is a secretory product of stromal cells and functions as a mediator of epithelial cell growth and differentiation. Phenotypes of the skin in several transgenic mouse models, in which the KGF and PTHrP genes have been overexpressed or disrupted, suggest that these two factors interact in vivo to regulate homeostasis of the skin. In this study, we investigated the effects of KGF on PTHrP secretion and expression by normal human foreskin keratinocytes (NHFK) and the effects of PTHrP on KGF secretion and expression by normal human dermal fibroblasts (NHDF) in vitro. N-terminal PTHrP(1-36) increased KGF secretion, protein expression and mRNA expression by NHDF in a dose-dependent manner, however, KGF did not regulate PTHrP expression and secretion by NHFK. By flow cytometry, PTHrP also increased the percentage of NHDF producing KGF. Our results indicate that PTHrP produced by keratinocytes is a potential paracrine regulator of KGF expression by dermal fibroblasts in vivo. This paracrine regulation may explain, in part, the epidermal atrophy seen in the PTHrP null mice and epidermal hyperplasia seen in transgenic mice overexpressing PTHrP in their basal keratinocytes. Our results also suggest that PTHrP is an important mediator for the healing of skin wounds and growth of neoplasms of squamous origin.


Cancer Letters | 1996

Inhibition of human prostate cancer cells growth by gossypol is associated with stimulation of transforming growth factor-β

Falah Shidaifat; Halit Canatan; Samuel K. Kulp; Yasuro Sugimoto; William Y. Chang; Yuan Zhang; Robert W. Brueggemeier; William J. Somers; Young C. Lin

Gossypol (GP), an antifertility agent in males, is also capable of inhibiting the proliferation of a wide range of cancer cells in vivo and in vitro. Thus, in this study we investigated the effect of GP on the growth of human androgen-independent prostate cancer cell line (PC3). The results showed that GP acts as a potent inhibitor of PC3 cells as determined by thymidine incorporation assay and flow cytometric analysis. Flow cytometry revealed that treatment of PC3 cells with GP resulted in a dose- and time-dependent accumulation of cells in the GO/GI phase with a concomitant decrease in cells progressing to the S and G2/M phase. These data support our thymidine incorporation results which indicated that GP is a potent inhibitor of PC3 cells. By ribonuclease protection assay, we also investigated the effect of GP on transforming growth factor-beta 1 (TGF-beta 1) gene expression in PC3 cells. Interestingly, the stimulatory effect of GP on TGF-beta 1 gene expression correlates well with its inhibitory effect on PC3 cell DNA synthesis and its ability to arrest cells in GO/G1 phase. Based on these data, it can be concluded that GP is a potent inhibitor of prostate cancer cell growth that acts by arresting cells in GO/G1 phase and that this inhibitory effect may be mediated by TGF-beta 1.


Oncogene | 2004

Function analysis of estrogenically regulated protein tyrosine phosphatase gamma (PTPgamma) in human breast cancer cell line MCF-7

Suling Liu; Yasuro Sugimoto; Claudio Sorio; Cristina Tecchio; Young C. Lin

Protein tyrosine phosphatase γ (PTPγ) is a member of the receptor-like family of tyrosine phosphatases and has been implicated as a tumor suppressor gene in kidney and lung cancers. Based on our previous findings, we hypothesize that PTPγ is a potential estrogen-regulated tumor suppressor gene in human breast cancer. To examine the effects of PTPγ on growth of MCF-7 human breast cancer cells and compare the estrogenic responses of human breast cells with different PTPγ expression levels, we established several stably transfected MCF-7 cell lines expressing different levels of PTPγ, which were confirmed by RT–PCR and immunostaining. In our work, we used the antisense construct to breakdown endogenous PTPγ level in MCF-7 cells. The results from doubling time assay suggested that PTPγ is capable of inhibiting MCF-7 breast cancer cell growth. We further demonstrated that PTPγ is able to inhibit anchorage-independent growth of breast cancer cells in soft agar and reduce the estrogenic responses of MCF-7 cell proliferation to estradiol-17β (E2) and zeranol (Z, a nonsteroidal growth promoter with estrogenic activity). Our data suggest that PTPγ may function as an important modulator in regulating the process of tumorigenesis in human breast.


Breast Cancer Research and Treatment | 2002

Involvement of breast epithelial-stromal interactions in the regulation of protein tyrosine phosphatase-γ (PTPγ) mRNA expression by estrogenically active agents

Suling Liu; Samuel K. Kulp; Yasuro Sugimoto; Jiahua Jiang; Hsiang-Lin Chang; Young C. Lin

AbstractBackground. Protein tyrosine phosphatase γ (PTPγ) has been implicated as a tumor suppressor gene in kidney and lung cancers. Our previous results indicate that estradiol-17β (E2)-induced suppression of PTPγ may play a role in mammary tumorigenesis. Zeranol (Z), a nonsteroidal growth promoter with estrogenic activity that is used by the US meat industry, induces estrogenic responses in primary cultured breast cells and breast cancer cell lines. Methods. PTPγ mRNA expression in human breast tissues and cells isolated from surgical specimens of mammoplasty and breast cancer patients were detected and quantified by RT-PCR. Immunohistochemical staining was used to localize PTPγ in human breast tissues. Breast epithelial and stromal cells were isolated and co-cultured to determine the involvement of cell–cell interaction in the regulation of PTPγ mRNA expression by E2 and Z. Results. PTPγ mRNA expression was lower in cancerous than in normal breast tissues. Both E2 and Z suppressed PTPγ mRNA levels in cultured normal breast tissues by ∼80%, but had a lesser effect in cultured epithelial cells isolated from normal breast tissues. In the co-culture system, both E2 and Z suppressed PTPγ mRNA to a greater degree in epithelial cells than in stromal cells. In whole breast tissues, PTPγ was immunolocalized to the epithelium. Treatment with E2 or Z diminished PTPγ staining indicating reductions in PTPγ at the protein level. Conclusions. The results indicate that both E2 and Z regulate PTPγ expression in human breast and that epithelial–stromal cells interaction is important in the regulation of PTPγ expression by estrogenically active agents.


Molecular Pharmaceutics | 2012

SPANosomes as delivery vehicles for small interfering RNA (siRNA)

Chenguang Zhou; Yicheng Mao; Yasuro Sugimoto; Yue Zhang; Naveen Kanthamneni; Bo Yu; Robert W. Brueggemeier; L. James Lee; Robert J. Lee

Nonionic surfactant vesicles, or SPANosomes (SPs), comprised of cationic lipid and sorbitan monooleate (Span 80) were synthesized and evaluated as small interfering RNA (siRNA) vectors. The SPs had a mean diameter of less than 100 nm and exhibited excellent colloidal stability. The SP/siRNA complexes possessed a slightly positive zeta potential of 12 mV and demonstrated a high siRNA incorporation efficiency of greater than 80%. Cryogenic transmission electron microscopy (cryo-TEM) imaging of the SP/siRNA indicated a predominantly core-shell structure. The SP/siRNA complexes were shown to efficiently and specifically silence expression of both green fluorescent protein (GFP) (66% knockdown) and aromatase (77% knockdown) genes in breast cancer cell lines. In addition, the cellular trafficking pathway of the SP/siRNA was investigated by confocal microscopy using molecular beacons as probes for cytosolic delivery. The results showed efficient endosomal escape and cytosolic delivery of the siRNA cargo following internalization of the SP/siRNA complexes. In conclusion, Span 80 is a potent helper lipid, and the SPs are promising vehicles for siRNA delivery.


Nanomedicine: Nanotechnology, Biology and Medicine | 2013

Human serum albumin-coated lipid nanoparticles for delivery of siRNA to breast cancer.

Longzhu Piao; Hong Li; Lesheng Teng; Bryant C. Yung; Yasuro Sugimoto; Robert W. Brueggemeier; Robert J. Lee

UNLABELLED Human serum albumin (HSA)-coated lipid nanoparticles (HSA-LNPs) loaded with phrGFP-targeted siRNA (HSA-LNPs-siRNA) were prepared and evaluated for gene downregulation effect in phrGFP-transfected breast cancer cells and the corresponding xenograft tumor model. HSA-LNPs-siRNA were successfully prepared with a particle size of 79.5±5.5 nm. In phrGFP-transfected MCF-7 cells, HSA-LNPs-siRNA significantly decreased cell fluorescence even in the presence of fetal bovine serum (FBS). Moreover, cell fluorescence and phrGFP mRNA expression were significantly downregulated by HSA-LNPs-siRNA in phrGFP-transfected MCF-7, MDA-MB-231, and SK-BR-3 cells in comparison with control or HSA-LNPs-siRNA (scrambled). In phrGFP-transfected MCF-7 xenograft tumor model, tumor fluorescence was significantly decreased after three IV administrations of HSA-LNPs-siRNA at a dose of 3 mg/kg in comparison with siRNA alone. HSA-LNPs-siRNA demonstrated a superior pharmacokinetic profile in comparison with siRNA at a dose of 1mg/kg. These results show that the novel nonviral carrier, HSA-LNPs, may be used for the delivery of siRNA to breast cancer cells. FROM THE CLINICAL EDITOR Targeted delivery of siRNA to cancer cells may be a viable anti-cancer strategy with low toxicity. In this study the novel nonviral carrier, human serum albumin-coated lipid nanoparticles (HSA-LNP) were demonstrated as an efficient delivery agent of siRNA to breast cancer cells.


Clinical Breast Cancer | 2011

Phase II trial of neoadjuvant exemestane in combination with celecoxib in postmenopausal women who have breast cancer.

Maryam B. Lustberg; Stephen P. Povoski; Weiqiang Zhao; Rebecca Ziegler; Yasuro Sugimoto; Amy S. Ruppert; Amy Lehman; Donna Shiels; Ewa Mrozek; Bhuvaneswari Ramaswamy; Rachel Layman; Robert W. Brueggemeier; Charles L. Shapiro

PURPOSE Within breast tissue, aromatase expression and activity is increased by prostaglandin E2, providing a rationale for combining the COX-2 inhibitor celecoxib with an aromatase inhibitor. To evaluate the effect of these drugs on aromatase and other biomarkers, a phase II trial of neoadjuvant exemestane followed sequentially by celecoxib plus exemestane was performed. METHODS Postmenopausal women with estrogen receptor (ER) and/or progesterone (PR) positive stages II-III breast cancers received 8 weeks of exemestane 25 mg daily, followed by 8 weeks of exemestane 25 mg daily and celecoxib 400 mg twice daily. Core biopsies were collected pretreatment, after 8 weeks of exemestane, and at definitive breast cancer surgery. A tissue microarray was constructed and immunohistochemistry (IHC) for aromatase, ER, PR, HER-2, Ki-67, and COX-2 was performed. RESULTS Twenty-two women were enrolled. Celecoxib was discontinued in 4 (18%) women for toxicity (all grade 1 and 2) and 2 (9%) developed serious cardiac events occurring at 1 and 4 months after completing treatment. By US, there were 8 (36%)-partial responses and 12 (55%)-stable disease. There were no pathological complete responses (pCR). There were statistically significant decreases in ER (P = .003), PR (P = .002), Ki-67 (P < .001), and COX-2 (P = .004) expression. No significant differences in aromatase or HER-2 expression were observed (P = .13 and P = .39, respectively). CONCLUSION The addition of celecoxib to exemestane was tolerated by the majority of women and anti-tumor response was observed. Additional studies, including gene expression, are required to more fully understand the basis for the decreased expression of ER, PR, Ki-67, and COX-2.


The Prostate | 1998

Stromal and epithelial cells of the canine prostate express parathyroid hormone‐related protein, but not the PTH/PTHrP receptor

Eric A.G. Blomme; Yasuro Sugimoto; Laurie K. McCauley; Young C. Lin; Charles C. Capen; Thomas J. Rosol

Parathyroid hormone‐related protein (PTHrP), a principal factor in the pathogenesis of humoral hypercalcemia of malignancy, is also widely expressed in many normal tissues, including human prostatic epithelial cells. The role of PTHrP in the prostate is not known, but may include regulation of cell growth and differentiation or calcium secretion into prostatic fluid. The dog is a valuable animal model for human prostatic diseases. The objective was to investigate the expression of PTHrP and the PTH/PTHrP (type 1) receptor in primary cultures of canine stromal and epithelial prostatic cells.

Collaboration


Dive into the Yasuro Sugimoto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Suling Liu

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Li-Shu Wang

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yi-Wen Huang

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge