Network


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

Hotspot


Dive into the research topics where Yingyu Ma is active.

Publication


Featured researches published by Yingyu Ma.


Endocrinology | 2010

CYP24A1 inhibition enhances the antitumor activity of calcitriol

Josephia R. Muindi; Wei-Dong Yu; Yingyu Ma; Kristie L. Engler; Rui-Xian Kong; Donald L. Trump; Candace S. Johnson

High systemic exposures to calcitriol are necessary for optimal antitumor effects. Human prostate cancer PC3 cells are insensitive to calcitriol treatment. Therefore, we investigated whether the inhibition of 24-hydroxylase (CYP24A1), the major calcitriol inactivating enzyme, by ketoconazole (KTZ) or RC2204 modulates calcitriol serum pharmacokinetics and biologic effects. Dexamethasone (Dex) was added to minimize calcitriol-induced hypercalcemia and as a steroid replacement for the KTZ inhibition of steroid biosynthesis cytochrome P450 enzymes. KTZ effectively inhibited time-dependent calcitriol-inducible CYP24A1 protein expression and enzyme activity in PC3 cells and C3H/HeJ mouse kidney tissues. Systemic calcitriol exposure area under the curve was higher in mice treated with a combination of calcitriol and KTZ than with calcitriol alone. KTZ and Dex synergistically potentiated calcitriol-mediated antiproliferative effects in PC3 cells in vitro; this effect was associated with enhanced apoptosis. After treatment with calcitriol and KTZ/Dex, although caspase-9 and caspase-3 were not activated and cytochrome c was not released by mitochondria, caspase-8 was activated and the truncated Bid protein level was increased. Translocation of apoptosis-inducing factor to the nucleus was observed, indicating a role of the apoptosis-inducing factor-mediated and caspase-independent apoptotic pathways. Calcitriol and KTZ/Dex combination suppressed the clonogenic survival and enhanced the growth inhibition observed with calcitriol alone in PC3 human prostate cancer xenograft mouse model. Our results show that the administration of calcitriol in combination with CYP24A1 inhibitor enhances antiproliferative effects, increases systemic calcitriol exposure, and promotes the activation of caspase-independent apoptosis pathway.


Cancer | 2010

1,25D3 Enhances antitumor activity of gemcitabine and cisplatin in human bladder cancer models†

Yingyu Ma; Wei-Dong Yu; Donald L. Trump; Candace S. Johnson

1,25 dihydroxyvitamin D3 (1,25D3) potentiates the cytotoxic effects of several common chemotherapeutic agents. The combination of gemcitabine and cisplatin is a current standard chemotherapy regimen for bladder cancer. The authors investigated whether 1,25D3 could enhance the antitumor activity of gemcitabine and cisplatin in bladder cancer model systems.


Molecular Cancer Therapeutics | 2008

1α,25-Dihydroxyvitamin D3 potentiates cisplatin antitumor activity by p73 induction in a squamous cell carcinoma model

Yingyu Ma; Wei-Dong Yu; Pamela A. Hershberger; Geraldine Flynn; Rui-Xian Kong; Donald L. Trump; Candace S. Johnson

1α,25-Dihydroxyvitamin D3 (1,25D3) exhibits antitumor activity in a variety of cancers including squamous cell carcinoma (SCC). Intrinsic resistance of SCC cells to cisplatin was observed and led to the investigation into whether 1,25D3 sensitizes SCC cells to cisplatin. Pretreatment with 1,25D3 followed by cisplatin enhanced growth inhibition in SCC cells compared with 1,25D3 alone as assessed by cytotoxicity and in vitro clonogenic assays. In addition, 1,25D3 sensitized SCC cells to cisplatin-mediated apoptosis. Treatment of tumor-bearing C3H mice with 1,25D3 before cisplatin reduced clonogenic survival using in vivo excision clonogenic assay. These results were not observed in a 1,25D3-resistant SCC variant, indicating the critical role of 1,25D3 in sensitizing SCC cells to cisplatin. Further, a marked decrease in fractional tumor volume was observed when SCC tumor-bearing mice were treated with 1,25D3 before cisplatin compared with either agent administered alone. Cisplatin has been shown to modulate p73 protein level in certain cancer cells. Our data showed that p73 level was not affected by cisplatin but increased by 1,25D3 in SCC cells. Knocking down p73 by small interfering RNA protected SCC cells against 1,25D3 and cisplatin-mediated clonogenic cell kill and apoptosis. Increasing p73 protein level by knocking down UFD2a, which mediates p73 degradation, promoted 1,25D3 and cisplatin-mediated clonogenic cell kill. These results suggest that 1,25D3 potentiates cisplatin antitumor activity in vitro and in vivo in a SCC model system possibly through p73 induction and apoptosis. The combination treatment may provide a more effective therapeutic regimen in cancer treatment. [Mol Cancer Ther 2008;7(9):3047–55]


Cell Cycle | 2010

Calcitriol enhances gemcitabine antitumor activity in vitro and in vivo by promoting apoptosis in a human pancreatic carcinoma model system

Wei-Dong Yu; Yingyu Ma; Geraldine Flynn; Josephia R. Muindi; Rui-Xian Kong; Donald L. Trump; Candace S. Johnson

Gemcitabine is the standard care chemotherapeutic agent to treat pancreatic cancer. Previously we demonstrated that calcitriol (1, 25-dihydroxycholecalciferol) has significant anti-proliferative effects in vitro and in vivo in multiple tumor models and enhances the activity of a variety of chemotherapeutic agents. We therefore investigated whether calcitriol could potentiate the cytotoxic activity of gemcitabine in the human pancreatic cancer Capan-1 model system. Isobologram analysis revealed that calcitriol and gemcitabine had synergistic antiproliferative effect over a wide range of drug concentrations. Calcitriol did not reduce the CDDase activity in Capan-1 tumors nor in the livers of Capan-1 tumor bearing mice. Calcitriol and gemcitabine combination promoted apoptosis in Capan-1 cells compared with either agent alone. The combination treatment also increased the activation of caspases-8, -9, -6, and -3 in Capan-1 cells. This result was confirmed by substrate-based caspase activity assay. Akt phosphorylation was reduced by calcitriol and gemcitabine combination treatment compared to single agent treatment. However, ERK1/2 phosphorylation was not modulated by either agent alone or by the combination. Tumor regrowth delay studies showed that calcitriol in combination with gemcitabine resulted in a significant reduction of Capan-1 tumor volume compared to single agent treatment. Our study suggests that calcitriol and gemcitabine in combination promotes caspase-dependent apoptosis, which may contribute to increased anti-tumor activity compared to either agent alone.


Cell Cycle | 2013

Inecalcitol, an analog of 1,25D₃, displays enhanced antitumor activity through the induction of apoptosis in a squamous cell carcinoma model system

Yingyu Ma; Wei-Dong Yu; Alejandro A. Hidalgo; Wei Luo; Remi Delansorne; Candace S. Johnson; Donald L. Trump

Epidemiological data suggest an important role of vitamin D signaling in cancer development and progression, and experimental studies demonstrate that the active vitamin D metabolite 1α, 25-dihydroxyvitamin D₃ (1,25D₃) has broad spectrum antitumor activity. Hypercalcemia has often been suggested to limit the clinical application of these data. The 14-epi-analog of 1,25D₃, inecalcitol [19-nor-14-epi-23-yne-1,25-(OH)₂D₃; TX522], was developed to have superagonistic antitumor activities but low hypercalcemia potential. We examined the antitumor activity of inecalcitol and the underlying mechanisms in a murine squamous cell carcinoma (SCC) model system. In vitro, compared with 1,25D₃, inecalcitol showed enhanced vitamin D receptor (VDR)-mediated transcriptional activity. Inecalcitol suppressed SCC cell proliferation in a dose-dependent manner with an IC₅₀ value 30 times lower than that of 1,25D₃. Both inecalcitol and 1,25D₃ induced a comparable level of G₀/G₁ cell cycle arrest in SCC cells. The level of apoptosis induced by inecalcitol was markedly higher than that of 1,25D₃. Apoptosis was mediated through the activation of the caspase 8/10- caspase 3 pathway. Further, inecalcitol markedly inhibited the mRNA and protein expression of c-IAP1 and XIAP compared with 1,25D₃. In vivo, inecalcitol inhibits SCC tumor growth in a dose-dependent fashion. Notably, inecalcitol induced a significantly higher level of apoptosis in the SCC xenograft model. While in vitro inecalcitol demonstrates apparent enhanced VDR binding and antiproliferative effects compared to 1,25D₃, in vivo these advantages disappear; at doses of inecalcitol that have equivalent antitumor effects, similar hypercalcemia is seen. This may be explained by the pharmacokinetics of 1,25D₃ vs. inecalcitol and attributed to the much shorter serum half-life of inecalcitol.We show that inecalcitol has potent antitumor activity in the SCC model system, and this is associated with a strong induction of apoptosis. These findings support the further development of inecalcitol in cancer treatment.


Cancer | 2013

Regulation of motility, invasion, and metastatic potential of squamous cell carcinoma by 1α,25-dihydroxycholecalciferol.

Yingyu Ma; Wei-Dong Yu; Bing Su; Mukund Seshadri; Wei Luo; Donald L. Trump; Candace S. Johnson

The active metabolite of vitamin D 1α,25‐dihydroxycholecalciferol (1,25D3) has exhibited broad‐spectrum antitumor activity in xenograft animal models. However, its activity against metastatic disease has not been extensively investigated.


Vitamins and Hormones Series | 2016

Mechanistic Insights of Vitamin D Anticancer Effects

Yingyu Ma; Candace S. Johnson; Donald L. Trump

Vitamin D is a secosteroid hormone that regulates many biological functions in addition to its classical role in maintaining calcium homeostasis and bone metabolism. Vitamin D deficiency appears to predispose individuals to increased risk of developing a number of cancers. Compelling epidemiological and experimental evidence supports a role for vitamin D in cancer prevention and treatment in many types of cancers. Preclinical studies show that 1,25D3, the active metabolite of vitamin D, and its analogs have antitumor effects in vitro and in vivo through multiple mechanisms including the induction of cell cycle arrest, apoptosis, differentiation and the suppression of inflammation, angiogenesis, invasion, and metastasis. 1,25D3 also potentiates the effect of chemotherapeutic agents and other agents in the combination treatment. In this review, the antitumor effects of 1,25D3 and the potential underlying mechanisms will be discussed. The current findings support the application of 1,25D3 in cancer prevention and treatment.


Cancer Research | 2013

Inhibition of protein kinase CK2 reduces CYP24A1 expression and enhances 1,25-dihydroxyvitamin D3 anti-tumor activity in human prostate cancer cells

Wei Luo; Wei-Dong Yu; Yingyu Ma; Mikhail Chernov; Donald L. Trump; Candace S. Johnson

Vitamin D has broad range of physiological functions and antitumor effects. 24-Hydroxylase, encoded by the CYP24A1 gene, is the key enzyme for degrading many forms of vitamin D including the most active form, 1,25D(3). Inhibition of CYP24A1 enhances 1,25D(3) antitumor activity. To isolate regulators of CYP24A1 expression in prostate cancer cells, we established a stable prostate cancer cell line PC3 with CYP24A1 promoter driving luciferase expression to screen a small molecular library for compounds that inhibit CYP24A1 promoter activity. From this screening, we identified, 4,5,6,7-tetrabromobenzimidazole (TBBz), a protein kinase CK2 selective inhibitor as a disruptor of CYP24A1 promoter activity. We show that TBBz inhibits CYP24A1 promoter activity induced by 1,25D(3) in prostate cancer cells. In addition, TBBz downregulates endogenous CYP24A1 mRNA level in TBBz-treated PC3 cells. Furthermore, siRNA-mediated CK2 knockdown reduces 1,25D(3)-induced CYP24A1 mRNA expression in PC3 cells. These results suggest that CK2 contributes to 1,25D(3)-mediated target gene expression. Finally, inhibition of CK2 by TBBz or CK2 siRNA significantly enhances 1,25D(3)-mediated antiproliferative effect in vitro and in vivo in a xenograft model. In summary, our findings reveal that protein kinase CK2 is involved in the regulation of CYP24A1 expression by 1,25D(3) and CK2 inhibitor enhances 1,25D(3)-mediated antitumor effect.


Current Gene Therapy | 2014

Vitamin D and miRNAs in cancer.

Yingyu Ma; Donald L. Trump; Candace S. Johnson

Vitamin D is a steroid hormone that regulates mineral homeostasis, bone metabolism and many other physiological processes. The active metabolite of vitamin D, 1α, 25-dihydroxyvitamin D (1,25D(3)), has broad spectrum antitumor activities and potentiates the effects of a number of chemotherapeutic agents. 1,25D(3) exerts its anti-tumor effects mainly through genomic mechanisms involving the regulation of gene transcription through vitamin D response elements (VDREs). More recently, miRNAs have been shown to be regulated by 1,25D(3). miRNAs are short non-coding RNAs that post-transcriptionally modulate the expression of a wide range of genes. Therefore, they have important regulatory roles in the development and progression of many diseases including cancer. This review focuses on the regulation of miRNA expression by 1,25D(3) in cancer model systems and the contribution of the regulated miRNAs to the anti-tumor effect of 1,25D(3). In addition, the impact of miRNAs on 1,25D(3) signaling is discussed.


Archive | 2011

Vitamin D and Angiogenesis

Yingyu Ma; Candace S. Johnson; Donald L. Trump

Angiogenesis is a physiological process involving the formation of new blood vessels from existing vessels. It is essential for the growth of primary tumor and local tumor invasion and metastasis. This chapter reviews the general angiogenesis process, the endogenous factors that regulate angiogenesis, and therapeutic angiogenesis inhibitors. It also reviews the effect of vitamin D on angiogenesis. Vitamin D receptor is detected on endothelial cells and vascular smooth muscle cells (VSMCs). 1,25D3 has anti-proliferative effects on tumor-derived endothelial cells through the induction of cell cycle arrest and apoptosis. Increasing evidence supports an anti-angiogenic role of 1,25D3 in a number of in vivo tumor model systems. However, vitamin D promotes angiogenesis in more physiological settings. Besides endothelial cells, vitamin D affects the physiological functions and pathology of VSMCs, including cell growth, contractility, motility, and the evolution of vascular calcification, which are involved in cardiovascular diseases. In summary, vitamin D plays important roles in vasculature and angiogenesis. Preclinical studies support the anti-angiogenic effect and the use of 1,25D3 in cancer therapy.

Collaboration


Dive into the Yingyu Ma's collaboration.

Top Co-Authors

Avatar

Candace S. Johnson

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Donald L. Trump

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Wei Luo

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Wei-Dong Yu

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Rui-Xian Kong

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Song Liu

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Bing Su

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Mukund Seshadri

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Brittany L. Bunch

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Gissou Azabdaftari

Roswell Park Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge