79 results on '"Jian H. Song"'
Search Results
2. Endothelial-to-osteoblast transition in normal mouse bone development
- Author
-
Song-Chang Lin, Guoyu Yu, Yu-Chen Lee, Jian H. Song, Xingzhi Song, Jianhua Zhang, Theocharis Panaretakis, Christopher J. Logothetis, Yoshihiro Komatsu, Li-Yuan Yu-Lee, Guocan Wang, and Sue-Hwa Lin
- Subjects
Biological sciences ,Cell biology ,Cancer ,Science - Abstract
Summary: Metastatic prostate cancer (PCa) in bone induces bone-forming lesions. We have previously shown that PCa-induced bone originates from endothelial cells (ECs) that have undergone EC-to-osteoblast (OSB) transition. Here, we investigated whether EC-to-OSB transition also occurs during normal bone formation. We developed an EC and OSB dual-color reporter mouse (DRM) model that marks EC-OSB hybrid cells with red and green fluorescent proteins. We observed EC-to-OSB transition (RFP and GFP co-expression) in both endochondral and intramembranous bone formation during embryonic development and in adults. Co-expression was confirmed in cells isolated from DRM. Bone marrow– and lung-derived ECs underwent transition to OSBs and mineralization in osteogenic medium. RNA-sequencing revealed GATA family transcription factors were upregulated in EC-OSB hybrid cells and knockdown of GATA3 inhibited BMP4-induced mineralization. Our findings support that EC-to-OSB transition occurs during normal bone development and suggest a new paradigm regarding the endothelial origin of OSBs.
- Published
- 2023
- Full Text
- View/download PDF
3. Multiple pathways coordinating reprogramming of endothelial cells into osteoblasts by BMP4
- Author
-
Guoyu Yu, Pengfei Shen, Yu-Chen Lee, Jing Pan, Jian H. Song, Tianhong Pan, Song-Chang Lin, Xin Liang, Guocan Wang, Theocharis Panaretakis, Christopher J. Logothetis, Gary E. Gallick, Li-Yuan Yu-Lee, and Sue-Hwa Lin
- Subjects
Molecular Biology ,Cell Biology ,Cancer ,Science - Abstract
Summary: Cell type transition occurs during normal development and under pathological conditions. In prostate cancer bone metastasis, prostate cancer-secreted BMP4 induces endothelial cell-to-osteoblast (EC-to-OSB) transition. Such tumor-induced stromal reprogramming supports prostate cancer progression. We delineate signaling pathways mediating EC-to-OSB transition using EC lines 2H11 and SVR. We found that BMP4-activated pSmad1-Notch-Hey1 pathway inhibits EC migration and tube formation. BMP4-activated GSK3β-βcatenin-Slug pathway stimulates Osx expression. In addition, pSmad1-regulated Dlx2 converges with the Smad1 and β-catenin pathways to stimulate osteocalcin expression. By co-expressing Osx, Dlx2, Slug and Hey1, we were able to achieve EC-to-OSB transition, leading to bone matrix mineralization in the absence of BMP4. In human prostate cancer bone metastasis specimens and MDA-PCa-118b and C4-2b-BMP4 osteogenic xenografts, immunohistochemical analysis showed that β-catenin and pSmad1 are detected in activated osteoblasts rimming the tumor-induced bone. Our results elucidated the pathways and key molecules coordinating prostate cancer-induced stromal programming and provide potential targets for therapeutic intervention.
- Published
- 2021
- Full Text
- View/download PDF
4. BIGH3 Promotes Osteolytic Lesions in Renal Cell Carcinoma Bone Metastasis by Inhibiting Osteoblast Differentiation
- Author
-
Tianhong Pan, Song-Chang Lin, Kai-Jie Yu, Guoyu Yu, Jian H. Song, Valerae O. Lewis, Justin E. Bird, Bryan Moon, Patrick P. Lin, Nizar M. Tannir, Eric Jonasch, Christopher G Wood, Gary E. Gallick, Li-Yuan Yu-Lee, Sue-Hwa Lin, and Robert L. Satcher
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
BACKGROUND: Bone metastasis is common in renal cell carcinoma (RCC), and the lesions are mainly osteolytic. The mechanism of bone destruction in RCC bone metastasis is unknown. METHODS: We used a direct intrafemur injection of mice with bone-derived 786-O RCC cells (Bo-786) as an in vivo model to study if inhibition of osteoblast differentiation is involved in osteolytic bone lesions in RCC bone metastasis. RESULTS: We showed that bone-derived Bo-786 cells induced osteolytic bone lesions in the femur of mice. We examined the effect of conditioned medium of Bo-786 cells (Bo-786 CM) on both primary mouse osteoblasts and MC3T3-E1 preosteoblasts and found that Bo-786 CM inhibited osteoblast differentiation. Secretome analysis of Bo-786 CM revealed that BIGH3 (Beta ig h3 protein), also known as TGFBI (transforming growth factor beta-induced protein), is highly expressed. We generated recombinant BIGH3 and found that BIGH3 inhibited osteoblast differentiation in vitro. In addition, CM from Bo-786 BIGH3 knockdown cells (786-BIGH3 KD) reduced the inhibition of osteoblast differentiation compared to CM from vector control. Intrafemural injection of mice with 786-BIGH3 KD cells showed a reduction in osteolytic bone lesions compared to vector control. Immunohistochemical staining of 18 bone metastasis specimens from human RCC showed strong BIGH3 expression in 11/18 (61%) and moderate BIGH3 expression in 7/18 (39%) of the specimens. CONCLUSIONS: These results suggest that suppression of osteoblast differentiation by BIGH3 is one of the mechanisms that enhance osteolytic lesions in RCC bone metastasis, and raise the possibilty that treatments that increase bone formation may improve therapy outcomes.
- Published
- 2018
- Full Text
- View/download PDF
5. Targeting DNA Damage Response in Prostate Cancer by Inhibiting Androgen Receptor-CDC6-ATR-Chk1 Signaling
- Author
-
Styliani Karanika, Theodoros Karantanos, Likun Li, Jianxiang Wang, Sanghee Park, Guang Yang, Xuemei Zuo, Jian H. Song, Sankar N. Maity, Ganiraju C. Manyam, Bradley Broom, Ana M. Aparicio, Gary E. Gallick, Patricia Troncoso, Paul G. Corn, Nora Navone, Wei Zhang, Shuhua Li, and Timothy C. Thompson
- Subjects
androgen receptor ,CDC6 ,Chk1 ,ATR ,TOPBP1 ,DNA damage ,prostate cancer ,enzalutamide ,AZD7762 ,Biology (General) ,QH301-705.5 - Abstract
Cell division cycle 6 (CDC6), an androgen receptor (AR) target gene, is implicated in regulating DNA replication and checkpoint mechanisms. CDC6 expression is increased during prostate cancer (PCa) progression and positively correlates with AR in PCa tissues. AR or CDC6 knockdown, together with AZD7762, a Chk1/2 inhibitor, results in decreased TopBP1-ATR-Chk1 signaling and markedly increased ataxia-telangiectasia-mutated (ATM) phosphorylation, a biomarker of DNA damage, and synergistically increases treatment efficacy. Combination treatment with the AR signaling inhibitor enzalutamide (ENZ) and the Chk1/2 inhibitor AZD7762 demonstrates synergy with regard to inhibition of AR-CDC6-ATR-Chk1 signaling, ATM phosphorylation induction, and apoptosis in VCaP (mutant p53) and LNCaP-C4-2b (wild-type p53) cells. CDC6 overexpression significantly reduced ENZ- and AZD7762-induced apoptosis. Additive or synergistic therapeutic activities are demonstrated in AR-positive animal xenograft models. These findings have important clinical implications, since they introduce a therapeutic strategy for AR-positive, metastatic, castration-resistant PCa, regardless of p53 status, through targeting AR-CDC6-ATR-Chk1 signaling.
- Published
- 2017
- Full Text
- View/download PDF
6. Supplementary Figure from Inhibition of Cell Adhesion by a Cadherin-11 Antibody Thwarts Bone Metastasis
- Author
-
Sue-Hwa Lin, Wilber Huang, Li-Yuan Yu-Lee, Gary E. Gallick, Jian Kuang, Robert L. Satcher, Jian H. Song, Hyojin Cho, Angelica Ortiz, Chih-Fen Huang, Song-Chang Lin, Guoyu Yu, Mehmet Asim Bilen, and Yu-Chen Lee
- Abstract
PDF file, 556K, S1. (A) Expression of Cad11 in C42B4 cells increased the cell migration. However, Cad11-mediated migratory activity of C42B4/Cad11 cells was not Inhibited by mAb 2C7 or 1A5. Another antibody, mAb 1B9, showed inhibition of migration and was used as a positive control. (B) Cad11-mediated invasion was not inhibited by mAb 2C7 or 1A5, but was inhibited by mAb 1B9.
- Published
- 2023
- Full Text
- View/download PDF
7. Figure S2 from Cabozantinib Reverses Renal Cell Carcinoma–mediated Osteoblast Inhibition in Three-dimensional Coculture In Vitro and Reduces Bone Osteolysis In Vivo
- Author
-
Robert L. Satcher, Sue-Hwa Lin, Mary C. Farach-Carson, Daniel A. Harrington, Shi-Ming Tu, Gary E. Gallick, Yu-Chen Lee, Guoyu Yu, Song-Chang Lin, Jian H. Song, Kelsea M. Hubka, Mariane Martinez, and Tianhong Pan
- Abstract
ALP and OSC gene expression in human Bo-786 RCC cells and PC3 prostate cancer cells
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure 2 from RSK Promotes Prostate Cancer Progression in Bone through ING3, CKAP2, and PTK6-Mediated Cell Survival
- Author
-
Sue-Hwa Lin, Jian Kuang, Li-Yuan Yu-Lee, Gary E. Gallick, Jian H. Song, Chuan-Fen Wu, Chien-Jui Cheng, Yu-Chen Lee, and Guoyu Yu
- Abstract
X-ray analysis the effects of RSK1 and RSK2 on PC3-mm2-induced bone changes.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental Figure 1 from RSK Promotes Prostate Cancer Progression in Bone through ING3, CKAP2, and PTK6-Mediated Cell Survival
- Author
-
Sue-Hwa Lin, Jian Kuang, Li-Yuan Yu-Lee, Gary E. Gallick, Jian H. Song, Chuan-Fen Wu, Chien-Jui Cheng, Yu-Chen Lee, and Guoyu Yu
- Abstract
Expression of pRSK(T359/S363) in human bone metastasis PCa specimens.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplemental Table S1 from RSK Promotes Prostate Cancer Progression in Bone through ING3, CKAP2, and PTK6-Mediated Cell Survival
- Author
-
Sue-Hwa Lin, Jian Kuang, Li-Yuan Yu-Lee, Gary E. Gallick, Jian H. Song, Chuan-Fen Wu, Chien-Jui Cheng, Yu-Chen Lee, and Guoyu Yu
- Abstract
Oligonucleotides of primers.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Inhibition of Cell Adhesion by a Cadherin-11 Antibody Thwarts Bone Metastasis
- Author
-
Sue-Hwa Lin, Wilber Huang, Li-Yuan Yu-Lee, Gary E. Gallick, Jian Kuang, Robert L. Satcher, Jian H. Song, Hyojin Cho, Angelica Ortiz, Chih-Fen Huang, Song-Chang Lin, Guoyu Yu, Mehmet Asim Bilen, and Yu-Chen Lee
- Abstract
Cadherin-11 (CDH11) is a member of the cadherin superfamily mainly expressed in osteoblasts but not in epithelial cells. However, prostate cancer cells with a propensity for bone metastasis express high levels of cadherin-11 and reduced levels of E-cadherin. Downregulation of cadherin-11 inhibits interaction of prostate cancer cells with osteoblasts in vitro and homing of prostate cancer cells to bone in an animal model of metastasis. These findings indicate that targeting cadherin-11 may prevent prostate cancer bone metastasis. To explore this possibility, a panel of 21 monoclonal antibodies (mAb) was generated against the extracellular (EC) domain of cadherin-11. Two antibodies, mAbs 2C7 and 1A5, inhibited cadherin-11–mediated cell–cell aggregation in vitro using L-cells transfected with cadherin-11. Both antibodies demonstrated specificity to cadherin-11, and neither antibody recognized E-cadherin or N-cadherin on C4-2B or PC3 cells, respectively. Furthermore, mAb 2C7 inhibited cadherin-11–mediated aggregation between the highly metastatic PC3-mm2 cells and MC3T3-E1 osteoblasts. Mechanistically, a series of deletion mutants revealed a unique motif, aa 343-348, in the cadherin-11 EC3 domain that is recognized by mAb 2C7 and that this motif coordinated cell–cell adhesion. Importantly, administration of mAb 2C7 in a prophylactic setting effectively prevented metastasis of PC3-mm2 cells to bone in an in vivo mouse model. These results show that targeting the extracellular domain of cadherin-11 can limit cellular adhesion and metastatic dissemination of prostate cancer cells.Implications: Monotherapy using a cadherin-11 antibody is a suitable option for the prevention of bone metastases. Mol Cancer Res; 11(11); 1401–11. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
12. CHD1 Promotes Sensitivity to Aurora Kinase Inhibitors by Suppressing Interaction of AURKA with Its Coactivator TPX2
- Author
-
Haoyan Li, Yin Wang, Kevin Lin, Varadha Balaji Venkadakrishnan, Martin Bakht, Wei Shi, Chenling Meng, Jie Zhang, Kaitlyn Tremble, Xin Liang, Jian H. Song, Xu Feng, Vivien Van, Pingna Deng, Jared K. Burks, Ana Aparicio, Khandan Keyomarsi, Junjie Chen, Yue Lu, Himisha Beltran, and Di Zhao
- Subjects
Male ,Cancer Research ,DNA Helicases ,Prostatic Neoplasms ,Antineoplastic Agents ,Cell Cycle Proteins ,Article ,DNA-Binding Proteins ,Mice ,Oncology ,Cell Line, Tumor ,Animals ,Humans ,Microtubule-Associated Proteins ,Protein Kinase Inhibitors ,Aurora Kinase A - Abstract
Clinical studies have shown that subsets of patients with cancer achieve a significant benefit from Aurora kinase inhibitors, suggesting an urgent need to identify biomarkers for predicting drug response. Chromodomain helicase DNA binding protein 1 (CHD1) is involved in chromatin remodeling, DNA repair, and transcriptional plasticity. Prior studies have demonstrated that CHD1 has distinct expression patterns in cancers with different molecular features, but its impact on drug responsiveness remains understudied. Here, we show that CHD1 promotes the susceptibility of prostate cancer cells to inhibitors targeting Aurora kinases, while depletion of CHD1 impairs their efficacy in vitro and in vivo. Pan-cancer drug sensitivity analyses revealed that high expression of CHD1 was associated with increased sensitivity to Aurora kinase A (AURKA) inhibitors. Mechanistically, KPNA2 served as a direct target of CHD1 and suppressed the interaction of AURKA with the coactivator TPX2, thereby rendering cancer cells more vulnerable to AURKA inhibitors. Consistent with previous research reporting that loss of PTEN elevates CHD1 levels, studies in a genetically engineered mouse model, patient-derived organoids, and patient samples showed that PTEN defects are associated with a better response to AURKA inhibition in advanced prostate cancer. These observations demonstrate that CHD1 plays an important role in modulating Aurora kinases and drug sensitivities, providing new insights into biomarker-driven therapies targeting Aurora kinases for future clinical studies. Significance: CHD1 plays a critical role in controlling AURKA activation and promoting Aurora kinase inhibitor sensitivity, providing a potential clinical biomarker to guide cancer treatment.
- Published
- 2022
- Full Text
- View/download PDF
13. Figure S1 from Targeting Src and Tubulin in Mucinous Ovarian Carcinoma
- Author
-
Anil K. Sood, Michael Frumovitz, Robert L. Coleman, David G. Hangauer, Keith A. Baggerly, Gary E. Gallick, Cristina Ivan, Nicholas B. Jennings, Behrouz Zand, Justin Bottsford-Miller, Chad V. Pecot, Chunhua Lu, Yunfei Wen, Jian H. Song, Takahito Miyake, Sunila Pradeep, Yu Kang, Jie Huang, Hyun Jin Choi, Heather J. Dalton, Wei Hu, and Tao Liu
- Abstract
PDF 116K, Figure S1. (A) Western blot results for p-paxillin and p-P130cas for the dose-finding experiment. HeyA8 cells were injected into the peritoneal cavity of 18 nude mice (2.5x105 cells per mouse). When tumors were palpable (4-5 weeks after the injection), mice were given 1 dose of 15mg/kg or 25mg/kg KX-01 orally. At 24, 36, or 48 hours after the treatment, mice were sacrificed and tumor tissues harvested. (B, C) Mouse weight following treatment in the RMUG-S and RMUG-L models (mean {plus-minus} standard deviation). No significant weight loss was observed in any of the groups
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary figures 1-10 from Integrating Murine and Clinical Trials with Cabozantinib to Understand Roles of MET and VEGFR2 as Targets for Growth Inhibition of Prostate Cancer
- Author
-
Gary E. Gallick, Christopher J. Logothetis, Sue-Hwa Lin, Mian Alauddin, Yiping Shao, Bogdan A. Czerniak, Menashe Bar Eli, Sankar N. Maity, Lynnelle Thorpe, Sanchaika Gaur, Anh G. Hoang, Ana Aparicio, Yu-Chen Lee, Jian H. Song, Eleni Efstathiou, Nila U. Parikh, Paul G. Corn, and Andreas Varkaris
- Abstract
Supplemental Figure 1 (fig. S1). Representative whole body NaF-18 PET scan images after 44 days of (A) vehicle-treated and (B) cabozantinib-treated animals. Supplemental Figure 2 (fig. S2). Effect of cabozantinib on PDX tumor growth, animal weight, and survival. Supplemental Figure 3 (fig. S3). Representative images of viable islets of tumor cells in (A) Patients; (B) MDA PCa-118b grown intrafemurally; and (C) MDA PCa- 118b grown subcutaneously. Supplemental Figure 4 (fig. S4). Time-dependent effect of cabozantinib on expression and phosphorylation of primary and MET downstream targets. Supplemental Figure 5 (fig. S5). Relative expression of c-met mRNA in NT and knockdown cells as determined by qRT-PCR. Supplemental Figure 6 (fig. S6). MET expression and activity in NT and knockdown tumors. Supplemental Figure 7 (fig. S7): Quantitation of CD31 staining on bone tumors. Supplemental Figure 8 (fig. S8). Effects of cabozantinib on bone remodeling markers in the phase-2 clinical trial and growth and differentiation of primary osteoblasts. Supplemental Figure S9 (fig S9). Effect of cabozantinib on bone turnover in the Phase 2 trial and PDX grown intrafemurally. Supplemental Figure 10 (fig. S10). Quantitation of phospho-histone H3 staining.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure from CHD1 Promotes Sensitivity to Aurora Kinase Inhibitors by Suppressing Interaction of AURKA with Its Coactivator TPX2
- Author
-
Di Zhao, Himisha Beltran, Yue Lu, Junjie Chen, Khandan Keyomarsi, Ana Aparicio, Jared K. Burks, Pingna Deng, Vivien Van, Xu Feng, Jian H. Song, Xin Liang, Kaitlyn Tremble, Jie Zhang, Chenling Meng, Wei Shi, Martin Bakht, Varadha Balaji Venkadakrishnan, Kevin Lin, Yin Wang, and Haoyan Li
- Abstract
Supplementary Figure from CHD1 Promotes Sensitivity to Aurora Kinase Inhibitors by Suppressing Interaction of AURKA with Its Coactivator TPX2
- Published
- 2023
- Full Text
- View/download PDF
16. Data from CHD1 Promotes Sensitivity to Aurora Kinase Inhibitors by Suppressing Interaction of AURKA with Its Coactivator TPX2
- Author
-
Di Zhao, Himisha Beltran, Yue Lu, Junjie Chen, Khandan Keyomarsi, Ana Aparicio, Jared K. Burks, Pingna Deng, Vivien Van, Xu Feng, Jian H. Song, Xin Liang, Kaitlyn Tremble, Jie Zhang, Chenling Meng, Wei Shi, Martin Bakht, Varadha Balaji Venkadakrishnan, Kevin Lin, Yin Wang, and Haoyan Li
- Abstract
Clinical studies have shown that subsets of patients with cancer achieve a significant benefit from Aurora kinase inhibitors, suggesting an urgent need to identify biomarkers for predicting drug response. Chromodomain helicase DNA binding protein 1 (CHD1) is involved in chromatin remodeling, DNA repair, and transcriptional plasticity. Prior studies have demonstrated that CHD1 has distinct expression patterns in cancers with different molecular features, but its impact on drug responsiveness remains understudied. Here, we show that CHD1 promotes the susceptibility of prostate cancer cells to inhibitors targeting Aurora kinases, while depletion of CHD1 impairs their efficacy in vitro and in vivo. Pan-cancer drug sensitivity analyses revealed that high expression of CHD1 was associated with increased sensitivity to Aurora kinase A (AURKA) inhibitors. Mechanistically, KPNA2 served as a direct target of CHD1 and suppressed the interaction of AURKA with the coactivator TPX2, thereby rendering cancer cells more vulnerable to AURKA inhibitors. Consistent with previous research reporting that loss of PTEN elevates CHD1 levels, studies in a genetically engineered mouse model, patient-derived organoids, and patient samples showed that PTEN defects are associated with a better response to AURKA inhibition in advanced prostate cancer. These observations demonstrate that CHD1 plays an important role in modulating Aurora kinases and drug sensitivities, providing new insights into biomarker-driven therapies targeting Aurora kinases for future clinical studies.Significance:CHD1 plays a critical role in controlling AURKA activation and promoting Aurora kinase inhibitor sensitivity, providing a potential clinical biomarker to guide cancer treatment.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Data from CHD1 Promotes Sensitivity to Aurora Kinase Inhibitors by Suppressing Interaction of AURKA with Its Coactivator TPX2
- Author
-
Di Zhao, Himisha Beltran, Yue Lu, Junjie Chen, Khandan Keyomarsi, Ana Aparicio, Jared K. Burks, Pingna Deng, Vivien Van, Xu Feng, Jian H. Song, Xin Liang, Kaitlyn Tremble, Jie Zhang, Chenling Meng, Wei Shi, Martin Bakht, Varadha Balaji Venkadakrishnan, Kevin Lin, Yin Wang, and Haoyan Li
- Abstract
Supplementary Data from CHD1 Promotes Sensitivity to Aurora Kinase Inhibitors by Suppressing Interaction of AURKA with Its Coactivator TPX2
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Data from Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment
- Author
-
Theocharis Panaretakis, Christopher J. Logothetis, Sue-Hwa Lin, Sumit K. Subudhi, Eleni Efstathiou, Gary E. Gallick, Nora Navone, Patricia Troncoso, Marites P. Melancon, Elmer Santos, Guoyu Yu, Song-Chang Lin, Yu-Chen Lee, Leah Guerra, Nila Parikh, Anh Hoang, Namrata Madan, Jian H. Song, Emanuela Gentile, Paul Corn, and Ioulia Vardaki
- Abstract
Supplementary Figures
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Table from CHD1 Promotes Sensitivity to Aurora Kinase Inhibitors by Suppressing Interaction of AURKA with Its Coactivator TPX2
- Author
-
Di Zhao, Himisha Beltran, Yue Lu, Junjie Chen, Khandan Keyomarsi, Ana Aparicio, Jared K. Burks, Pingna Deng, Vivien Van, Xu Feng, Jian H. Song, Xin Liang, Kaitlyn Tremble, Jie Zhang, Chenling Meng, Wei Shi, Martin Bakht, Varadha Balaji Venkadakrishnan, Kevin Lin, Yin Wang, and Haoyan Li
- Abstract
Supplementary Table from CHD1 Promotes Sensitivity to Aurora Kinase Inhibitors by Suppressing Interaction of AURKA with Its Coactivator TPX2
- Published
- 2023
- Full Text
- View/download PDF
20. Data from Integrating Murine and Clinical Trials with Cabozantinib to Understand Roles of MET and VEGFR2 as Targets for Growth Inhibition of Prostate Cancer
- Author
-
Gary E. Gallick, Christopher J. Logothetis, Sue-Hwa Lin, Mian Alauddin, Yiping Shao, Bogdan A. Czerniak, Menashe Bar Eli, Sankar N. Maity, Lynnelle Thorpe, Sanchaika Gaur, Anh G. Hoang, Ana Aparicio, Yu-Chen Lee, Jian H. Song, Eleni Efstathiou, Nila U. Parikh, Paul G. Corn, and Andreas Varkaris
- Abstract
Purpose: We performed parallel investigations in cabozantinib-treated patients in a phase II trial and simultaneously in patient-derived xenograft (PDX) models to better understand the roles of MET and VEGFR2 as targets for prostate cancer therapy.Experimental Design: In the clinical trial, radiographic imaging and serum markers were examined, as well as molecular markers in tumors from bone biopsies. In mice harboring PDX intrafemurally or subcutaneously, cabozantinib effects on tumor growth, MET, PDX in which MET was silenced, VEGFR2, bone turnover, angiogenesis, and resistance were examined.Results: In responsive patients and PDX, islets of viable pMET-positive tumor cells persisted, which rapidly regrew after drug withdrawal. Knockdown of MET in PDX did not affect tumor growth in mice nor did it affect cabozantinib-induced growth inhibition but did lead to induction of FGFR1. Inhibition of VEGFR2 and MET in endothelial cells reduced the vasculature, leading to necrosis. However, each islet of viable cells surrounded a VEGFR2-negative vessel. Reduction of bone turnover was observed in both cohorts.Conclusions: Our studies demonstrate that MET in tumor cells is not a persistent therapeutic target for metastatic castrate-resistant prostate cancer (CRPC), but inhibition of VEGFR2 and MET in endothelial cells and direct effects on osteoblasts are responsible for cabozantinib-induced tumor inhibition. However, vascular heterogeneity represents one source of primary therapy resistance, whereas induction of FGFR1 in tumor cells suggests a potential mechanism of acquired resistance. Thus, integrated cross-species investigations demonstrate the power of combining preclinical models with clinical trials to understand mechanisms of activity and resistance of investigational agents. Clin Cancer Res; 22(1); 107–21. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Figure from Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition
- Author
-
Sue-Hwa Lin, Li-Yuan Yu-Lee, Christopher J. Logothetis, Maurizio Pacifici, Theocharis Panaretakis, Sandeep K. Agarwal, Jing Pan, Song-Chang Lin, Yu-Chen Lee, Jian H. Song, Pengfei Shen, Paul G. Corn, and Guoyu Yu
- Abstract
Supplementary Figure from Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition
- Published
- 2023
- Full Text
- View/download PDF
22. Data from Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment
- Author
-
Theocharis Panaretakis, Christopher J. Logothetis, Sue-Hwa Lin, Sumit K. Subudhi, Eleni Efstathiou, Gary E. Gallick, Nora Navone, Patricia Troncoso, Marites P. Melancon, Elmer Santos, Guoyu Yu, Song-Chang Lin, Yu-Chen Lee, Leah Guerra, Nila Parikh, Anh Hoang, Namrata Madan, Jian H. Song, Emanuela Gentile, Paul Corn, and Ioulia Vardaki
- Abstract
Purpose:Radium-223 prolongs survival in a fraction of men with bone metastatic prostate cancer (PCa). However, there are no markers for monitoring response and resistance to Radium-223 treatment. Exosomes are mediators of intercellular communication and may reflect response of the bone microenvironment to Radium-223 treatment. We performed molecular profiling of exosomes and compared the molecular profile in patients with favorable and unfavorable overall survival.Experimental Design:We performed exosomal transcriptome analysis in plasma derived from our preclinical models (MDA-PCa 118b tumors, TRAMP-C2/BMP4 PCa) and from the plasma of 25 patients (paired baseline and end of treatment) treated with Radium-223. All samples were run in duplicate, and array data analyzed with fold changes +2 to −2 and P < 0.05.Results:We utilized the preclinical models to establish that genes derived from the tumor and the tumor-associated bone microenvironment (bTME) are differentially enriched in plasma exosomes upon Radium-223 treatment. The mouse transcriptome analysis revealed changes in bone-related and DNA damage repair–related pathways. Similar findings were observed in plasma-derived exosomes from patients treated with Radium-223 detected changes. In addition, exosomal transcripts detected immune-suppressors (e.g., PD-L1) that were associated with shorter survival to Radium-223. Treatment of the Myc-CaP mouse model with a combination of Radium-223 and immune checkpoint therapy (ICT) resulted in greater efficacy than monotherapy.Conclusions:These clinical and coclinical analyses showed that RNA profiling of plasma exosomes may be used for monitoring the bTME in response to treatment and that ICT may be used to increase the efficacy of Radium-223.
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Table from Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition
- Author
-
Sue-Hwa Lin, Li-Yuan Yu-Lee, Christopher J. Logothetis, Maurizio Pacifici, Theocharis Panaretakis, Sandeep K. Agarwal, Jing Pan, Song-Chang Lin, Yu-Chen Lee, Jian H. Song, Pengfei Shen, Paul G. Corn, and Guoyu Yu
- Abstract
Supplementary Table from Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Data from Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition
- Author
-
Sue-Hwa Lin, Li-Yuan Yu-Lee, Christopher J. Logothetis, Maurizio Pacifici, Theocharis Panaretakis, Sandeep K. Agarwal, Jing Pan, Song-Chang Lin, Yu-Chen Lee, Jian H. Song, Pengfei Shen, Paul G. Corn, and Guoyu Yu
- Abstract
Supplementary Data from Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Tables from Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment
- Author
-
Theocharis Panaretakis, Christopher J. Logothetis, Sue-Hwa Lin, Sumit K. Subudhi, Eleni Efstathiou, Gary E. Gallick, Nora Navone, Patricia Troncoso, Marites P. Melancon, Elmer Santos, Guoyu Yu, Song-Chang Lin, Yu-Chen Lee, Leah Guerra, Nila Parikh, Anh Hoang, Namrata Madan, Jian H. Song, Emanuela Gentile, Paul Corn, and Ioulia Vardaki
- Abstract
Supp Tables 1-3
- Published
- 2023
- Full Text
- View/download PDF
26. supplementary materials from Integrating Murine and Clinical Trials with Cabozantinib to Understand Roles of MET and VEGFR2 as Targets for Growth Inhibition of Prostate Cancer
- Author
-
Gary E. Gallick, Christopher J. Logothetis, Sue-Hwa Lin, Mian Alauddin, Yiping Shao, Bogdan A. Czerniak, Menashe Bar Eli, Sankar N. Maity, Lynnelle Thorpe, Sanchaika Gaur, Anh G. Hoang, Ana Aparicio, Yu-Chen Lee, Jian H. Song, Eleni Efstathiou, Nila U. Parikh, Paul G. Corn, and Andreas Varkaris
- Abstract
supplementary materials and methods, supplementary tables and supplementary figure legends Supplemental Table 1: Patient Characteristics. Supplemental Table 2: Targeting sequences for c-met. Supplemental Table 3: Antibodies and Sources. Supplemental Table 4: Oligonucleotides for PCR Amplification of Mouse Alkaline Phosphatase and Osteonectin.
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary information from Targeting Src and Tubulin in Mucinous Ovarian Carcinoma
- Author
-
Anil K. Sood, Michael Frumovitz, Robert L. Coleman, David G. Hangauer, Keith A. Baggerly, Gary E. Gallick, Cristina Ivan, Nicholas B. Jennings, Behrouz Zand, Justin Bottsford-Miller, Chad V. Pecot, Chunhua Lu, Yunfei Wen, Jian H. Song, Takahito Miyake, Sunila Pradeep, Yu Kang, Jie Huang, Hyun Jin Choi, Heather J. Dalton, Wei Hu, and Tao Liu
- Abstract
PDF file 106K, Table S1. Molecules whose expression significantly differed between cells treated with KX-01 and control cells in RMUG-S cells Table S2. Molecules whose expression significantly differed between cells treated with KX-01 and control cells in RMUG-L cells
- Published
- 2023
- Full Text
- View/download PDF
28. Data from Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition
- Author
-
Sue-Hwa Lin, Li-Yuan Yu-Lee, Christopher J. Logothetis, Maurizio Pacifici, Theocharis Panaretakis, Sandeep K. Agarwal, Jing Pan, Song-Chang Lin, Yu-Chen Lee, Jian H. Song, Pengfei Shen, Paul G. Corn, and Guoyu Yu
- Abstract
Metastatic prostate cancer in the bone induces bone-forming lesions that contribute to progression and therapy resistance. Prostate cancer–induced bone formation originates from endothelial cells (EC) that have undergone endothelial-to-osteoblast (EC-to-OSB) transition in response to tumor-secreted BMP4. Current strategies targeting prostate cancer–induced bone formation are lacking. Here, we show that activation of retinoic acid receptor (RAR) inhibits EC-to-OSB transition and reduces prostate cancer–induced bone formation. Treatment with palovarotene, an RARγ agonist being tested for heterotopic ossification in fibrodysplasia ossificans progressiva, inhibited EC-to-OSB transition and osteoblast mineralization in vitro and decreased tumor-induced bone formation and tumor growth in several osteogenic prostate cancer models, and similar effects were observed with the pan-RAR agonist all-trans-retinoic acid (ATRA). Knockdown of RARα, β, or γ isoforms in ECs blocked BMP4-induced EC-to-OSB transition and osteoblast mineralization, indicating a role for all three isoforms in prostate cancer–induced bone formation. Furthermore, treatment with palovarotene or ATRA reduced plasma Tenascin C, a factor secreted from EC-OSB cells, which may be used to monitor treatment response. Mechanistically, BMP4-activated pSmad1 formed a complex with RAR in the nucleus of ECs to activate EC-to-OSB transition. RAR activation by palovarotene or ATRA caused pSmad1 degradation by recruiting the E3-ubiquitin ligase Smad ubiquitination regulatory factor1 (Smurf1) to the nuclear pSmad1/RARγ complex, thus blocking EC-to-OSB transition. Collectively, these findings suggest that palovarotene can be repurposed to target prostate cancer–induced bone formation to improve clinical outcomes for patients with bone metastasis.Significance:This study provides mechanistic insights into how RAR agonists suppress prostate cancer–induced bone formation and offers a rationale for developing RAR agonists for prostate cancer bone metastasis therapy.See related commentary by Bhowmick and Bhowmick, p. 2975
- Published
- 2023
- Full Text
- View/download PDF
29. Data from Targeting Src and Tubulin in Mucinous Ovarian Carcinoma
- Author
-
Anil K. Sood, Michael Frumovitz, Robert L. Coleman, David G. Hangauer, Keith A. Baggerly, Gary E. Gallick, Cristina Ivan, Nicholas B. Jennings, Behrouz Zand, Justin Bottsford-Miller, Chad V. Pecot, Chunhua Lu, Yunfei Wen, Jian H. Song, Takahito Miyake, Sunila Pradeep, Yu Kang, Jie Huang, Hyun Jin Choi, Heather J. Dalton, Wei Hu, and Tao Liu
- Abstract
Purpose: To investigate the antitumor effects of targeting Src and tubulin in mucinous ovarian carcinoma.Experimental Design: The in vitro and in vivo effects and molecular mechanisms of KX-01, which inhibits Src pathway and tubulin polymerization, were examined in mucinous ovarian cancer models.Results:In vitro studies using RMUG-S and RMUG-L cell lines showed that KX-01 inhibited cell proliferation, induced apoptosis, arrested the cell cycle at the G2–M phase, and enhanced the cytotoxicity of oxaliplatin in the KX-01–sensitive cell line, RMUG-S. In vivo studies showed that KX-01 significantly decreased tumor burden in RMUG-S and RMUG-L mouse models relative to untreated controls, and the effects were greater when KX-01 was combined with oxaliplatin. KX-01 alone and in combination with oxaliplatin significantly inhibited tumor growth by reducing cell proliferation and inducing apoptosis in vivo. PTEN knock-in experiments in RMUG-L cells showed improved response to KX-01. Reverse phase protein array analysis showed that in addition to blocking downstream molecules of Src family kinases, KX-01 also activated acute stress-inducing molecules.Conclusion: Our results showed that targeting both the Src pathway and tubulin with KX-01 significantly inhibited tumor growth in preclinical mucinous ovarian cancer models, suggesting that this may be a promising therapeutic approach for patients with mucinous ovarian carcinoma. Clin Cancer Res; 19(23); 6532–43. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
30. Prostate tumor-induced stromal reprogramming generates Tenascin C that promotes prostate cancer metastasis through YAP/TAZ inhibition
- Author
-
Ming Zhu, Darryl J. Pappin, Li-Yuan Yu-Lee, Christopher J. Logothetis, Sue-Hwa Lin, Theocharis Panaretakis, Yu Chen Lee, Guoyu Yu, Jian H. Song, Keith Rivera, Song-Chang Lin, and Guocan Wang
- Subjects
Cancer Research ,Tumor microenvironment ,Stromal cell ,Chemistry ,Tenascin C ,Tenascin ,Cell migration ,Biology ,urologic and male genital diseases ,musculoskeletal system ,medicine.disease ,Metastasis ,Prostate cancer ,Bone morphogenetic protein 4 ,Genetics ,medicine ,Cancer research ,biology.protein ,Immunohistochemistry ,Reprogramming ,Molecular Biology - Abstract
Metastatic prostate cancer (PCa) in bone induces bone-forming lesions that enhance PCa progression. How tumor-induced bone formation enhances PCa progression is not known. We have previously shown that PCa-induced bone originates from endothelial cells (ECs) that have undergone endothelial-to-osteoblast (EC-to-OSB) transition by tumor-secreted bone morphogenetic protein 4 (BMP4). Here, we show that EC-to-OSB transition leads to changes in the tumor microenvironment that increases the metastatic potential of PCa cells. We found that conditioned medium (CM) from EC-OSB hybrid cells increases the migration, invasion, and survival of PC3-mm2 and C4-2B4 PCa cells. Quantitative mass spectrometry (Isobaric Tags for Relative and Absolute Quantitation) identified Tenascin C (TNC) as one of the major proteins secreted from EC-OSB hybrid cells. TNC expression in tumor-induced OSBs was confirmed by immunohistochemistry of MDA PCa-118b xenograft and human bone metastasis specimens. Mechanistically, BMP4 increases TNC expression in EC-OSB cells through the Smad1-Notch/Hey1 pathway. How TNC promotes PCa metastasis was next interrogated by in vitro and in vivo studies. In vitro studies showed that a TNC-neutralizing antibody inhibits EC-OSB-CM-mediated PCa cell migration and survival. TNC knockdown decreased, while the addition of recombinant TNC or TNC overexpression increased migration and anchorage-independent growth of PC3 or C4-2b cells. When injected orthotopically, PC3-mm2-shTNC clones decreased metastasis to bone, while C4-2b-TNC-overexpressing cells increased metastasis to lymph nodes. TNC enhances PCa cell migration through α5β1 integrin-mediated YAP/TAZ inhibition. These studies elucidate that tumor-induced stromal reprogramming generates TNC that enhances PCa metastasis and suggest that TNC may be a target for PCa therapy.
- Published
- 2021
- Full Text
- View/download PDF
31. P4HA2-induced prolyl hydroxylation suppresses YAP1-mediated prostate cancer cell migration, invasion, and metastasis
- Author
-
Jiwon Park, Christopher J. Logothetis, Ailing Huang, Celia Sze Ling Mak, Hiroyuki Katayama, Ming Zhu, Ruiqing Peng, Jian H. Song, Guocan Wang, Ruidong Chen, Xin Liang, Qing Chang, Samir M. Hanash, Ming Tang, Xingdi Ma, Zhengdao Lan, Sue Hwa Lin, Pingping Hou, Abhinav K. Jain, and Shui er Zheng
- Subjects
Male ,Cancer Research ,Biology ,Article ,Prolyl Hydroxylases ,Metastasis ,Hydroxylation ,Mice ,chemistry.chemical_compound ,Prostate cancer ,Cell Movement ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Molecular Biology ,YAP1 ,Hippo signaling pathway ,Microarray analysis techniques ,Prostatic Neoplasms ,YAP-Signaling Proteins ,Cell migration ,medicine.disease ,chemistry ,Tumor progression ,Cancer research - Abstract
Yes-associated protein 1 (YAP1), a key player in the Hippo pathway, has been shown to play a critical role in tumor progression. However, the role of YAP1 in prostate cancer cell invasion, migration, and metastasis is not well defined. Through functional, transcriptomic, epigenomic, and proteomic analyses, we showed that prolyl hydroxylation of YAP1 plays a critical role in the suppression of cell migration, invasion, and metastasis in prostate cancer. Knockdown (KD) or knockout (KO) of YAP1 led to an increase in cell migration, invasion, and metastasis in prostate cancer cells. Microarray analysis showed that the EMT pathway was activated in Yap1-KD cells. ChIP-seq analysis showed that YAP1 target genes are enriched in pathways regulating cell migration. Mass spectrometry analysis identified P4H prolyl hydroxylase in the YAP1 complex and YAP1 was hydroxylated at multiple proline residues. Proline-to-alanine mutations of YAP1 isoform 3 identified proline 174 as a critical residue, and its hydroxylation suppressed cell migration, invasion, and metastasis. KO of P4ha2 led to an increase in cell migration and invasion, which was reversed upon Yap1 KD. Our study identified a novel regulatory mechanism of YAP1 by which P4HA2-dependent prolyl hydroxylation of YAP1 determines its transcriptional activities and its function in prostate cancer metastasis.
- Published
- 2021
- Full Text
- View/download PDF
32. Characterization of prostate cancer adrenal metastases: dependence upon androgen receptor signaling and steroid hormones
- Author
-
Minas J. Sakellakis, Andrew W. Hahn, Sumankalai Ramachandran, Miao Zhang, Anh Hoang, Jian H. Song, Jingjing Liu, Feng Wang, Hirak S. Basu, Peter Sheperd, Xuemei Wang, Daniel E. Frigo, Sue-Hwa Lin, Theocharis Panaretakis, Jianhua Zhang, Nora Navone, Patricia Troncoso, Christopher J. Logothetis, and Mark A. Titus
- Subjects
Cancer Research ,Oncology ,Urology - Abstract
Prostate cancer (PCa) typically spreads to the bone, and this distribution is attributed to the central role of the microenvironment in progression. However, metastasis to the adrenal glands, while not as common, does occur. The biology that accounts for adrenal metastases may be attributed to the unique local steroid metabolome and co-clinical characterization may elucidate the role steroid biosynthesis plays in PCa progression.Three patients with metastatic PCa who had archived tumor tissue from an adrenalectomy were retrospectively identified, and one adrenal metastasis was developed into a xenograft (MDA-PCa-250). The adrenal metastases were characterized by performing somatic DNA whole exome sequencing (WES), RNA-Seq, immunohistochemistry (IHC), and steroid metabolite quantitation. The influence of steroid metabolites on adrenal metastasis cells and tumor growth was tested in vitro and in vivo.Clinically, adrenalectomy was performed during castration-resistant oligometastatic disease, and two men experienced resensitization to leuprolide. Somatic DNA WES revealed heterogeneous alterations in tumor suppressor and DNA damage repair pathway genes. Adrenal metastases had active androgen receptor (AR) signaling by IHC, and RNA-Seq supported a potential role for adrenal androgen precursor metabolism in activating the AR. Steroid quantitation suggested the adrenal androgen precursors were converted into testosterone in these metastases, and stable isotope tracing of an organoid from MDA-PCa-250 confirmed the capability of adrenal metastases to biosynthesize testosterone from adrenal precursors. In vitro testing of a cell line derived from MDA-PCa-250 showed that testosterone and cortisol stimulated tumor cell growth. In vivo experiments demonstrated that MDA-PCa-250 grew in intact mice with circulating testosterone, but not in castrated mice.PCa adrenal metastases depend upon AR signaling driven by androgen precursors, androstenedione and dehydroepiandrosterone, available in the microenvironment, despite the presence of heterogeneous somatic DNA alterations. Moreover, MDA-PCa-250 provides a preclinical model that can recapitulate the unique androgen-dependence of adrenal metastases.This study does not report the clinical results of a clinical trial, but it does use samples from a completed clinical trial that is registered with clinicaltrials.gov (NCT01254864).
- Published
- 2022
- Full Text
- View/download PDF
33. Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment
- Author
-
Elmer B Santos, Yu Chen Lee, Ioulia Vardaki, Marites P. Melancon, Leah D Guerra, Gary E. Gallick, Namrata Madan, Jian H. Song, Theocharis Panaretakis, Paul G. Corn, Anh Hoang, Sumit K. Subudhi, Nila U. Parikh, Nora M. Navone, Emanuela Gentile, Guoyu Yu, Patricia Troncoso, Song-Chang Lin, Eleni Efstathiou, Sue-Hwa Lin, and Christopher J. Logothetis
- Subjects
Male ,0301 basic medicine ,Radium-223 ,Cancer Research ,DNA damage ,Gene Expression ,Bone Neoplasms ,Exosomes ,Article ,Transcriptome ,Extracellular Vesicles ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Gene ,business.industry ,Gene Expression Profiling ,Prostatic Neoplasms ,Immune Checkpoint Proteins ,medicine.disease ,Microvesicles ,Immune checkpoint ,Survival Rate ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,RNA ,Radiopharmaceuticals ,business ,Intracellular ,Radium ,medicine.drug - Abstract
Purpose: Radium-223 prolongs survival in a fraction of men with bone metastatic prostate cancer (PCa). However, there are no markers for monitoring response and resistance to Radium-223 treatment. Exosomes are mediators of intercellular communication and may reflect response of the bone microenvironment to Radium-223 treatment. We performed molecular profiling of exosomes and compared the molecular profile in patients with favorable and unfavorable overall survival. Experimental Design: We performed exosomal transcriptome analysis in plasma derived from our preclinical models (MDA-PCa 118b tumors, TRAMP-C2/BMP4 PCa) and from the plasma of 25 patients (paired baseline and end of treatment) treated with Radium-223. All samples were run in duplicate, and array data analyzed with fold changes +2 to −2 and P < 0.05. Results: We utilized the preclinical models to establish that genes derived from the tumor and the tumor-associated bone microenvironment (bTME) are differentially enriched in plasma exosomes upon Radium-223 treatment. The mouse transcriptome analysis revealed changes in bone-related and DNA damage repair–related pathways. Similar findings were observed in plasma-derived exosomes from patients treated with Radium-223 detected changes. In addition, exosomal transcripts detected immune-suppressors (e.g., PD-L1) that were associated with shorter survival to Radium-223. Treatment of the Myc-CaP mouse model with a combination of Radium-223 and immune checkpoint therapy (ICT) resulted in greater efficacy than monotherapy. Conclusions: These clinical and coclinical analyses showed that RNA profiling of plasma exosomes may be used for monitoring the bTME in response to treatment and that ICT may be used to increase the efficacy of Radium-223.
- Published
- 2021
- Full Text
- View/download PDF
34. Cabozantinib Reverses Renal Cell Carcinoma–mediated Osteoblast Inhibition in Three-dimensional Coculture In Vitro and Reduces Bone Osteolysis In Vivo
- Author
-
Mary C. Farach-Carson, Sue Hwa Lin, Shi Ming Tu, Robert L. Satcher, Yu Chen Lee, Song Chang Lin, Daniel A. Harrington, Gary E. Gallick, Jian H. Song, Tianhong Pan, Kelsea M. Hubka, Mariane Martinez, and Guoyu Yu
- Subjects
0301 basic medicine ,Cancer Research ,Osteolysis ,Chemistry ,Cellular differentiation ,Osteoblast ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Bone morphogenetic protein 4 ,Osteoclast ,In vivo ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Tyrosine kinase ,TGFBI - Abstract
Renal cell carcinoma bone metastases (RCCBM) are typically osteolytic. We previously showed that BIGH3 (beta Ig-h3/TGFBI), secreted by 786-O renal cell carcinoma, plays a role in osteolytic bone lesion in RCCBM through inhibition of osteoblast (OSB) differentiation. To study this interaction, we employed three-dimensional (3D) hydrogels to coculture bone-derived 786-O (Bo-786) renal cell carcinoma cells with MC3T3-E1 pre-OSBs. Culturing pre-OSBs in the 3D hydrogels preserved their ability to differentiate into mature OSB; however, this process was decreased when pre-OSBs were cocultured with Bo-786 cells. Knockdown of BIGH3 in Bo-786 cells recovered OSB differentiation. Furthermore, treatment with bone morphogenetic protein 4, which stimulates OSB differentiation, or cabozantinib (CBZ), which inhibits VEGFR1 and MET tyrosine kinase activities, also increased OSB differentiation in the coculture. CBZ also inhibited pre-osteoclast RAW264.7 cell differentiation. Using RCCBM mouse models, we showed that CBZ inhibited Bo-786 tumor growth in bone. CBZ treatment also increased bone volume and OSB number, and decreased osteoclast number and blood vessel density. When tested in SN12PM6 renal cell carcinoma cells that have been transduced to overexpress BIGH3, CBZ also inhibited SN12PM6 tumor growth in bone. These observations suggest that enhancing OSB differentiation could be one of the therapeutic strategies for treating RCCBM that exhibit OSB inhibition characteristics, and that this 3D coculture system is an effective tool for screening osteoanabolic agents for further in vivo studies.
- Published
- 2020
- Full Text
- View/download PDF
35. Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition
- Author
-
Guoyu Yu, Paul G. Corn, Pengfei Shen, Jian H. Song, Yu-Chen Lee, Song-Chang Lin, Jing Pan, Sandeep K. Agarwal, Theocharis Panaretakis, Maurizio Pacifici, Christopher J. Logothetis, Li-Yuan Yu-Lee, and Sue-Hwa Lin
- Subjects
Male ,Cancer Research ,Osteoblasts ,Oncology ,Receptors, Retinoic Acid ,Ubiquitin-Protein Ligases ,Endothelial Cells ,Humans ,Prostatic Neoplasms ,Bone Neoplasms ,Tretinoin - Abstract
Metastatic prostate cancer in the bone induces bone-forming lesions that contribute to progression and therapy resistance. Prostate cancer–induced bone formation originates from endothelial cells (EC) that have undergone endothelial-to-osteoblast (EC-to-OSB) transition in response to tumor-secreted BMP4. Current strategies targeting prostate cancer–induced bone formation are lacking. Here, we show that activation of retinoic acid receptor (RAR) inhibits EC-to-OSB transition and reduces prostate cancer–induced bone formation. Treatment with palovarotene, an RARγ agonist being tested for heterotopic ossification in fibrodysplasia ossificans progressiva, inhibited EC-to-OSB transition and osteoblast mineralization in vitro and decreased tumor-induced bone formation and tumor growth in several osteogenic prostate cancer models, and similar effects were observed with the pan-RAR agonist all-trans-retinoic acid (ATRA). Knockdown of RARα, β, or γ isoforms in ECs blocked BMP4-induced EC-to-OSB transition and osteoblast mineralization, indicating a role for all three isoforms in prostate cancer–induced bone formation. Furthermore, treatment with palovarotene or ATRA reduced plasma Tenascin C, a factor secreted from EC-OSB cells, which may be used to monitor treatment response. Mechanistically, BMP4-activated pSmad1 formed a complex with RAR in the nucleus of ECs to activate EC-to-OSB transition. RAR activation by palovarotene or ATRA caused pSmad1 degradation by recruiting the E3-ubiquitin ligase Smad ubiquitination regulatory factor1 (Smurf1) to the nuclear pSmad1/RARγ complex, thus blocking EC-to-OSB transition. Collectively, these findings suggest that palovarotene can be repurposed to target prostate cancer–induced bone formation to improve clinical outcomes for patients with bone metastasis. Significance: This study provides mechanistic insights into how RAR agonists suppress prostate cancer–induced bone formation and offers a rationale for developing RAR agonists for prostate cancer bone metastasis therapy. See related commentary by Bhowmick and Bhowmick, p. 2975
- Published
- 2022
36. Yap1 Hydroxylation Suppress Prostate Cancer Metastasis
- Author
-
Ming Zhu, Ruiqing Peng, Xin Liang, Zhengdao Lan, Meng Tang, Pingping Hou, Jian H. Song, Celia Sze Ling Mak, Jiwon Park, Shui-Er Zheng, Ailing Huang, Xingdi Ma, Ruidong Chen, Qing Chang, Christopher J. Logothetis, Abhinav K. Jain, Sue-Hwa Lin, Hiroyuki Katayama, Samir Hanash, and Guocan Wang
- Published
- 2022
- Full Text
- View/download PDF
37. Activation of retinoic acid receptor reduces metastatic prostate cancer bone lesions through blocking endothelial-to-osteoblast transition
- Author
-
Guoyu Yu, Paul G. Corn, Pengfei Shen, Jian H. Song, Yu-Chen Lee, Song-Chang Lin, Jing Pan, Sandeep K. Agarwal, Theocharis Panaretakis, Maurizio Pacifici, Christopher J. Logothetis, Li-Yuan Yu-Lee, and Sue-Hwa Lin
- Abstract
Metastatic prostate cancer (PCa) in bone induces bone-forming lesions that contribute to progression and therapy resistance. Currently strategies targeting PCa-induced bone formation are lacking. We previously showed that PCa-induced bone originates from endothelial cells (EC) that have undergone endothelial-to-osteoblast (EC-to-OSB) transition in response to tumor-secreted BMP4. Here, we show that activation of retinoic acid receptor (RAR) inhibits EC-to-OSB transition and reduces PCa-induced bone formation. We found that palovarotene, a RARγ agonist being tested for heterotopic ossification in fibrodysplasia ossificans progressiva, inhibited EC-to-OSB transition and osteoblast mineralizationin vitro, and decreased tumor-induced bone formation and tumor growth in several osteogenic PCa models. RARα/β/γ isoform knockdown in 2H11 ECs blocked EC-to-OSB transition and osteoblast mineralization. Pan-RAR agonist ATRA inhibited MycCaP-BMP4-induced bone formation and tumor growth under castration. Furthermore, palovarotene or ATRA reduced plasma Tenascin C, a factor secreted by EC-OSB cells, which may be used to monitor treatment response. Mechanistically, BMP4-activated pSmad1 forms a complex with RAR in the nucleus of 2H11 cells. RAR activation by palovarotene or ATRA causes pSmad1 degradation by recruiting E3-ubiquitin ligase Smurf1 into the nuclear pSmad1/RARγ complex. Our findings suggest that palovarotene can be repurposed to target PCa-induced bone formation to improve clinical outcomes for bone metastasis.
- Published
- 2021
- Full Text
- View/download PDF
38. Statins reduce castration-induced bone marrow adiposity and prostate cancer progression in bone
- Author
-
Yu Chen Lee, Christopher J. Logothetis, Li-Yuan Yu-Lee, Tianhong Pan, Robert L. Satcher, Jian H. Song, Guoyu Yu, Mark Titus, Sue-Hwa Lin, Jing Pan, Theocharis Panaretakis, and Song-Chang Lin
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Statin ,Stromal cell ,medicine.drug_class ,adipocytes ,Biology ,urologic and male genital diseases ,Article ,Androgen deprivation therapy ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Molecular Biology ,Adiposity ,bone metastasis ,Leptin ,statin ,Prostatic Neoplasms ,medicine.disease ,prostate cancer ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Primary bone ,030220 oncology & carcinogenesis ,androgen depletion ,Cancer research ,Bone marrow ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,bone marrow stromal cells - Abstract
A fraction of patients undergoing androgen deprivation therapy (ADT) for advanced prostate cancer (PCa) will develop recurrent castrate-resistant PCa (CRPC) in bone. Strategies to prevent CRPC relapse in bone are lacking. Here we show that the cholesterol-lowering drugs statins decrease castration-induced bone marrow adiposity in the tumor microenvironment and reduce PCa progression in bone. Using primary bone marrow stromal cells (BMSC) and M2-10B4 cells, we showed that ADT increases bone marrow adiposity by enhancing BMSC-to-adipocyte transition in vitro. Knockdown of androgen receptor abrogated BMSC-to-adipocyte transition, suggesting an androgen receptor-dependent event. RNAseq analysis showed that androgens reduce the secretion of adipocyte hormones/cytokines including leptin during BMSC-to-adipocyte transition. Treatment of PCa C4-2b, C4-2B4, and PC3 cells with leptin led to an increase in cell cycle progression and nuclear Stat3. RNAseq analysis also showed that androgens inhibit cholesterol biosynthesis pathway, raising the possibility that inhibiting cholesterol biosynthesis may decrease BMSC-to-adipocyte transition. Indeed, statins decreased BMSC-to-adipocyte transition in vitro and castration-induced bone marrow adiposity in vivo. Statin pre-treatment reduced 22RV1 PCa progression in bone after ADT. Our findings with statin may provide one of the mechanisms to the clinical correlations that statin use in patients undergoing ADT seems to delay progression to "lethal" PCa.
- Published
- 2021
39. Multiple pathways coordinating reprogramming of endothelial cells into osteoblasts by BMP4
- Author
-
Christopher J. Logothetis, Li Yuan Yu-Lee, Guoyu Yu, Jing Pan, Theocharis Panaretakis, Gary E. Gallick, Pengfei Shen, Guocan Wang, Song Chang Lin, Tianhong Pan, Xin Liang, Jian H. Song, Yu Chen Lee, and Sue Hwa Lin
- Subjects
0301 basic medicine ,animal structures ,Stromal cell ,Angiogenesis ,Science ,02 engineering and technology ,Article ,03 medical and health sciences ,Prostate cancer ,Prostate ,medicine ,Protein kinase B ,Transcription factor ,Molecular Biology ,Cancer ,Tube formation ,Multidisciplinary ,biology ,Chemistry ,Bone metastasis ,Cell Biology ,021001 nanoscience & nanotechnology ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,embryonic structures ,Osteocalcin ,biology.protein ,Cancer research ,0210 nano-technology ,Reprogramming - Abstract
Summary Cell type transition occurs during normal development and under pathological conditions. In prostate cancer bone metastasis, prostate cancer-secreted BMP4 induces endothelial cell-to-osteoblast (EC-to-OSB) transition. Such tumor-induced stromal reprogramming supports prostate cancer progression. We delineate signaling pathways mediating EC-to-OSB transition using EC lines 2H11 and SVR. We found that BMP4-activated pSmad1-Notch-Hey1 pathway inhibits EC migration and tube formation. BMP4-activated GSK3β-βcatenin-Slug pathway stimulates Osx expression. In addition, pSmad1-regulated Dlx2 converges with the Smad1 and β-catenin pathways to stimulate osteocalcin expression. By co-expressing Osx, Dlx2, Slug and Hey1, we were able to achieve EC-to-OSB transition, leading to bone matrix mineralization in the absence of BMP4. In human prostate cancer bone metastasis specimens and MDA-PCa-118b and C4-2b-BMP4 osteogenic xenografts, immunohistochemical analysis showed that β-catenin and pSmad1 are detected in activated osteoblasts rimming the tumor-induced bone. Our results elucidated the pathways and key molecules coordinating prostate cancer-induced stromal programming and provide potential targets for therapeutic intervention., Graphical abstract, Highlights • BMP4 upregulates several pathways essential for EC-to-OSB transition • BMP4 activates pSmad1-Notch-Hey1 and GSK3β-βcatenin-Slug-OSX pathways • pSmad1-regulated Dlx2 expression connects the pSmad1 and β-catenin pathways • Coexpression of OSX, Dlx2, Slug, and Hey1 is sufficient to induce EC-to-OSB transition, Molecular Biology ; Cell Biology ; Cancer
- Published
- 2020
40. Combined Inhibition of IGF-1R/IR and Src family kinases enhances antitumor effects in prostate cancer by decreasing activated survival pathways.
- Author
-
Farshid Dayyani, Nila U Parikh, Andreas S Varkaris, Jian H Song, Shhyam Moorthy, Tanushree Chatterji, Sankar N Maity, Adam R Wolfe, Joan M Carboni, Marco M Gottardis, Christopher J Logothetis, and Gary E Gallick
- Subjects
Medicine ,Science - Abstract
Treatment of metastatic prostate cancer (PCa) with single agents has shown only modest efficacy. We hypothesized dual inhibition of different pathways in PCa results in improved tumor inhibition. The Src family kinases (SFK) and insulin-like growth factor-1 (IGF-1) signaling axes are aberrantly activated in both primary PCa and bone metastases and regulate distinct and overlapping functions in PCa progression. We examined the antitumor effects of combined inhibition of these pathways.Src andIGF-1 receptor (IGF-1R) inhibition was achieved in vitro by short hairpin (sh)RNA and in vitro and in vivo by small molecule inhibitors (dasatinib and BMS-754807, against SFK and IGF-1R/Insulin Receptor(IR), respectively).In vitro, inhibition of IGF-1 signaling affected cell survival and proliferation. SFK blockade alone had modest effects on proliferation, but significantly enhanced the IGF-1R blockade. These findings correlated with a robust inhibition of IGF-1-induced Akt1 phophorylation by dasatinib, whereas Akt2 phosphorylation was SFK independent and only inhibited by BMS-754807. Thus, complete inhibition of both Akt genes, not seen by either drug alone, is likely a major mechanism for the decreased survival of PCa cells. Furthermore, dasatinib and BMS-754807 inhibited in vivo growth of the primary human xenograft MDA PCa 133, with corresponding inhibition of Akt in tumors. Also, both orthotopic and intratibial tumor growth of PC-3 cells were more potently inhibited by dual SFK and IGF-1R/IR blockade compared to either pathway alone, with a corresponding decrease in bone turnover markers.Dual IGF-1R/IR and SFK inhibition may be a rational therapeutic approach in PCa by blocking both independent and complementary processes critical to tumor growth.
- Published
- 2012
- Full Text
- View/download PDF
41. BIGH3 Promotes Osteolytic Lesions in Renal Cell Carcinoma Bone Metastasis by Inhibiting Osteoblast Differentiation
- Author
-
Valerae O. Lewis, Bryan S. Moon, Christopher G. Wood, Eric Jonasch, Nizar M. Tannir, Guoyu Yu, Robert L. Satcher, Justin E. Bird, Gary E. Gallick, Jian H. Song, Tianhong Pan, Li Yuan Yu-Lee, Song Chang Lin, Kai Jie Yu, Patrick P. Lin, and Sue Hwa Lin
- Subjects
0301 basic medicine ,Cancer Research ,Osteolysis ,biology ,Chemistry ,Cellular differentiation ,Bone metastasis ,Osteoblast ,Transforming growth factor beta ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,3. Good health ,Extracellular matrix ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,biology.protein ,Carcinoma ,Cancer research ,TGFBI - Abstract
BACKGROUND : Bone metastasis is common in renal cell carcinoma (RCC), and the lesions are mainly osteolytic. The mechanism of bone destruction in RCC bone metastasis is unknown. METHODS : We used a direct intrafemur injection of mice with bone-derived 786-O RCC cells (Bo-786) as an in vivo model to study if inhibition of osteoblast differentiation is involved in osteolytic bone lesions in RCC bone metastasis. RESULTS : We showed that bone-derived Bo-786 cells induced osteolytic bone lesions in the femur of mice. We examined the effect of conditioned medium of Bo-786 cells (Bo-786 CM) on both primary mouse osteoblasts and MC3T3-E1 preosteoblasts and found that Bo-786 CM inhibited osteoblast differentiation. Secretome analysis of Bo-786 CM revealed that BIGH3 (Beta ig h3 protein), also known as TGFBI (transforming growth factor beta-induced protein), is highly expressed. We generated recombinant BIGH3 and found that BIGH3 inhibited osteoblast differentiation in vitro . In addition, CM from Bo-786 BIGH3 knockdown cells (786-BIGH3 KD) reduced the inhibition of osteoblast differentiation compared to CM from vector control. Intrafemural injection of mice with 786-BIGH3 KD cells showed a reduction in osteolytic bone lesions compared to vector control. Immunohistochemical staining of 18 bone metastasis specimens from human RCC showed strong BIGH3 expression in 11/18 (61%) and moderate BIGH3 expression in 7/18 (39%) of the specimens. CONCLUSIONS: These results suggest that suppression of osteoblast differentiation by BIGH3 is one of the mechanisms that enhance osteolytic lesions in RCC bone metastasis, and raise the possibilty that treatments that increase bone formation may improve therapy outcomes.
- Published
- 2018
42. Targeting DNA Damage Response in Prostate Cancer by Inhibiting Androgen Receptor-CDC6-ATR-Chk1 Signaling
- Author
-
Theodoros Karantanos, Jianxiang Wang, Styliani Karanika, Paul G. Corn, Guang Yang, Gary E. Gallick, Sanghee Park, Timothy C. Thompson, Xuemei Zuo, Sankar N. Maity, Likun Li, Ana Aparicio, Bradley M. Broom, Wei Zhang, Ganiraju C. Manyam, Shuhua Li, Nora M. Navone, Patricia Troncoso, and Jian H. Song
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,DNA damage ,Chk1 ,Biology ,CDC6 ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,androgen receptor ,medicine ,Enzalutamide ,CHEK1 ,Androgen Receptor Antagonists ,lcsh:QH301-705.5 ,enzalutamide ,medicine.disease ,prostate cancer ,AZD7762 ,3. Good health ,Androgen receptor ,030104 developmental biology ,ATR ,chemistry ,lcsh:Biology (General) ,Cancer research ,Signal transduction ,biological phenomena, cell phenomena, and immunity ,TOPBP1 - Abstract
Cell division cycle 6 (CDC6), an androgen receptor (AR) target gene, is implicated in regulating DNA replication and checkpoint mechanisms. CDC6 expression is increased during prostate cancer (PCa) progression and positively correlates with AR in PCa tissues. AR or CDC6 knockdown, together with AZD7762, a Chk1/2 inhibitor, results in decreased TopBP1-ATR-Chk1 signaling and markedly increased ataxia-telangiectasia-mutated (ATM) phosphorylation, a biomarker of DNA damage, and synergistically increases treatment efficacy. Combination treatment with the AR signaling inhibitor enzalutamide (ENZ) and the Chk1/2 inhibitor AZD7762 demonstrates synergy with regard to inhibition of AR-CDC6-ATR-Chk1 signaling, ATM phosphorylation induction, and apoptosis in VCaP (mutant p53) and LNCaP-C4-2b (wild-type p53) cells. CDC6 overexpression significantly reduced ENZ- and AZD7762-induced apoptosis. Additive or synergistic therapeutic activities are demonstrated in AR-positive animal xenograft models. These findings have important clinical implications, since they introduce a therapeutic strategy for AR-positive, metastatic, castration-resistant PCa, regardless of p53 status, through targeting AR-CDC6-ATR-Chk1 signaling.
- Published
- 2017
43. Chromatin Regulator CHD1 Remodels the Immunosuppressive Tumor Microenvironment in PTEN-Deficient Prostate Cancer
- Author
-
Y. Alan Wang, Chenling Meng, Jun Li, Denise J. Spring, Zhengdao Lan, Peiwen Chen, Jian H. Song, Chang-Jiun Wu, Jiexi Li, Michael Ittmann, Xin Lu, Guocan Wang, Ko Chien Chen, Pingna Deng, Shan Jiang, Di Zhao, Xiaoying Shang, James W. Horner, Qing Chang, Li Cai, Haoyan Li, Xin Liang, Ivonne Flores, and Ronald A. DePinho
- Subjects
0301 basic medicine ,Male ,Regulator ,Mice, Transgenic ,Article ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,Cell Line, Tumor ,Tumor Microenvironment ,Medicine ,PTEN ,Animals ,Humans ,Smad4 Protein ,Tumor microenvironment ,biology ,business.industry ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,medicine.disease ,Immune checkpoint ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Tumor Escape ,business - Abstract
Genetic inactivation of PTEN is common in prostate cancer and correlates with poorer prognosis. We previously identified CHD1 as an essential gene in PTEN-deficient cancer cells. Here, we sought definitive in vivo genetic evidence for, and mechanistic understanding of, the essential role of CHD1 in PTEN-deficient prostate cancer. In Pten and Pten/Smad4 genetically engineered mouse models, prostate-specific deletion of Chd1 resulted in markedly delayed tumor progression and prolonged survival. Chd1 deletion was associated with profound tumor microenvironment (TME) remodeling characterized by reduced myeloid-derived suppressor cells (MDSC) and increased CD8+ T cells. Further analysis identified IL6 as a key transcriptional target of CHD1, which plays a major role in recruitment of immunosuppressive MDSCs. Given the prominent role of MDSCs in suppressing responsiveness to immune checkpoint inhibitors (ICI), our genetic and tumor biological findings support combined testing of anti-IL6 and ICI therapies, specifically in PTEN-deficient prostate cancer. Significance: We demonstrate a critical role of CHD1 in MDSC recruitment and discover CHD1/IL6 as a major regulator of the immunosuppressive TME of PTEN-deficient prostate cancer. Pharmacologic inhibition of IL6 in combination with immune checkpoint blockade elicits robust antitumor responses in prostate cancer. This article is highlighted in the In This Issue feature, p. 1241
- Published
- 2019
44. Cabozantinib Reverses Renal Cell Carcinoma-mediated Osteoblast Inhibition in Three-dimensional Coculture
- Author
-
Tianhong, Pan, Mariane, Martinez, Kelsea M, Hubka, Jian H, Song, Song-Chang, Lin, Guoyu, Yu, Yu-Chen, Lee, Gary E, Gallick, Shi-Ming, Tu, Daniel A, Harrington, Mary C, Farach-Carson, Sue-Hwa, Lin, and Robert L, Satcher
- Subjects
Male ,Osteoblasts ,Pyridines ,Apoptosis ,Bone Neoplasms ,Cell Differentiation ,Mice, SCID ,Osteolysis ,In Vitro Techniques ,Xenograft Model Antitumor Assays ,Coculture Techniques ,Kidney Neoplasms ,Article ,Mice ,Tumor Cells, Cultured ,Animals ,Humans ,Anilides ,Carcinoma, Renal Cell ,Cell Proliferation - Abstract
Renal cell carcinoma (RCC) bone metastases (RCCBM) are typically osteolytic. We previously showed that BIGH3 (beta Ig-h3/TGFbI), secreted by 786-O RCC, plays a role in osteolytic bone lesion in RCCBM through inhibition of osteoblast (OSB) differentiation. To study this interaction, we employed three-dimensional (3D) hydrogels to co-culture bone-derived 786-O RCC cells (Bo-786) with MC3T3-E1 pre-osteoblasts (OSB). Culturing pre-osteoblasts in the 3D hydrogels preserved their ability to differentiate into mature OSB; however, this process was decreased when pre-osteoblasts were co-cultured with Bo-786 cells. Knockdown of BIGH3 in Bo-786 cells recovered OSB differentiation. Further, treatment with bone morphogenetic protein 4 (BMP4), which stimulates osteoblast differentiation, or cabozantinib (CBZ), which inhibits VEGFR1 and MET tyrosine kinase activities, also increased OSB differentiation in the co-culture. CBZ also inhibited pre-osteoclast RAW264.7 cell differentiation. Using RCCBM mouse models, we showed that CBZ inhibited Bo-786 tumor growth in bone. CBZ treatment also increased bone volume and OSB number, and decreased osteoclast number and blood vessel density. When tested in SN12PM6 RCC cells that have been transduced to overexpress BIGH3, CBZ also inhibited SN12PM6 tumor growth in bone. These observations suggest that enhancing OSB differentiation could be one of the therapeutic strategies for treating RCCBM that exhibit OSB inhibition characteristics, and that this 3D co-culture system is an effective tool for screening osteoanabolic agents for further in vivo studies.
- Published
- 2019
45. Androgen receptor inhibitor–induced 'BRCAness' and PARP inhibition are synthetically lethal for castration-resistant prostate cancer
- Author
-
Christopher J. Logothetis, Abul Kalam Azad, Spyridon P. Basourakos, Theodoros Karantanos, Bradley M. Broom, Likun Li, Hyun-Sung Lee, Sanghee Park, Jian H. Song, Jianhua Yin, Yong Luo, Jiangxiang Wang, Wenhui Wu, Particia Troncoso, Ganiraju C. Manyam, Xuemei Zuo, Timothy P. Heffernan, Gary E. Gallick, Carlo Toniatti, Paul G. Corn, Guang Yang, Timothy C. Thompson, Ana Aparicio, Styliani Karanika, Wenjun Chang, Dimitrios Korentzelos, Ju Seog Lee, and Jeri Kim
- Subjects
0301 basic medicine ,Combination therapy ,medicine.drug_class ,Cell Biology ,Biology ,urologic and male genital diseases ,Antiandrogen ,medicine.disease ,Biochemistry ,Olaparib ,Androgen receptor ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,chemistry ,030220 oncology & carcinogenesis ,PARP inhibitor ,Immunology ,Cancer research ,medicine ,Enzalutamide ,Molecular Biology - Abstract
Cancers with loss-of-function mutations in BRCA1 or BRCA2 are deficient in the DNA damage repair pathway called homologous recombination (HR), rendering these cancers exquisitely vulnerable to poly(ADP-ribose) polymerase (PARP) inhibitors. This functional state and therapeutic sensitivity is referred to as “BRCAness” and is most commonly associated with some breast cancer types. Pharmaceutical induction of BRCAness could expand the use of PARP inhibitors to other tumor types. For example, BRCA mutations are present in only ~20% of prostate cancer patients. We found that castration-resistant prostate cancer (CRPC) cells showed increased expression of a set of HR-associated genes, including BRCA1 , RAD54L , and RMI2 . Although androgen-targeted therapy is typically not effective in CRPC patients, the androgen receptor inhibitor enzalutamide suppressed the expression of those HR genes in CRPC cells, thus creating HR deficiency and BRCAness. A “lead-in” treatment strategy, in which enzalutamide was followed by the PARP inhibitor olaparib, promoted DNA damage–induced cell death and inhibited clonal proliferation of prostate cancer cells in culture and suppressed the growth of prostate cancer xenografts in mice. Thus, antiandrogen and PARP inhibitor combination therapy may be effective for CRPC patients and suggests that pharmaceutically inducing BRCAness may expand the clinical use of PARP inhibitors.
- Published
- 2017
- Full Text
- View/download PDF
46. Talin1 phosphorylation activates β1 integrins: a novel mechanism to promote prostate cancer bone metastasis
- Author
-
Pei-Chieh Tien, Jung-Kang Jin, Sue-Hwa Lin, Chien-Jui Cheng, Gary E. Gallick, Cai Huang, and Jian H. Song
- Subjects
Male ,Talin ,Cancer Research ,Cell Cycle Proteins ,Metastasis ,Mice ,0302 clinical medicine ,Cell Movement ,Anoikis ,Phosphorylation ,RNA, Small Interfering ,0303 health sciences ,biology ,Integrin beta1 ,Prostate ,Bone metastasis ,prostate cancer ,3. Good health ,Cell biology ,Integrin alpha M ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,RNA Interference ,Protein Binding ,integrin ,Cdk5 ,Integrin ,Bone Neoplasms ,Article ,Focal adhesion ,03 medical and health sciences ,Cell Line, Tumor ,Cell Adhesion ,Genetics ,medicine ,metastasis ,Animals ,Humans ,Kinase activity ,Cell adhesion ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Focal Adhesions ,Prostatic Neoplasms ,Cyclin-Dependent Kinase 5 ,medicine.disease ,Enzyme Activation ,biology.protein ,Cancer research - Abstract
Talins are adaptor proteins that regulate focal adhesion signaling by conjugating integrins to the cytoskeleton. Talins directly bind integrins and are essential for integrin activation. We previously showed that β1 integrins are activated in metastatic prostate cancer (PCa) cells, increasing PCa metastasis to lymph nodes and bone. However, how β1 integrins are activated in PCa cells is unknown. In this study, we identified a novel mechanism of β1 integrin activation. Using knockdown experiments, we first demonstrated that talin1, but not talin2, is important in β1 integrin activation. We next showed that talin1 S425 phosphorylation, but not total talin1 expression, correlates with metastatic potential of PCa cells. Expressing a non-phosphorylatable mutant, talin1(S425A), in talin1-silenced PC3-MM2 and C4-2B4 PCa cells, decreased activation of β1 integrins, integrin-mediated adhesion, motility and increased the sensitivity of the cells to anoikis. In contrast, reexpression of the phosphorylation-mimicking mutant talin1(S425D) led to increased β1 integrin activation and generated biologic effects opposite to talin1(S425A) expression. In the highly metastatic PC3-MM2 cells, expression of a non-phosphorylatable mutant, talin1(S425A), in talin1-silenced PC3-MM2 cells, abolished their ability to colonize in the bone following intracardiac injection, while reexpression of phosphorylation-mimicking mutant talin1(S425D) restored their ability to metastasize to bone. Immunohistochemical staining demonstrated that talin S425 phosphorylation is significantly increased in human bone metastases when compared with normal tissues, primary tumors or lymph node metastases. We further showed that p35 expression, an activator of Cdk5, and Cdk5 activity were increased in metastatic tumor cells, and that Cdk5 kinase activity is responsible for talin1 phosphorylation and subsequent β1 integrin activation. Together, our study reveals Cdk5-mediated phosphorylation of talin1 leading to β1 integrin activation is a novel mechanism that increases metastatic potential of PCa cells.
- Published
- 2014
- Full Text
- View/download PDF
47. Abstract 385: MTAP gene deficiency creates vulnerability to anti-folate therapy in urothelial bladder carcinoma
- Author
-
Mark Titus, Omar Alhalabi, William F. Benedict, Jianfeng Chen, Guangchun Han, Thai H. Ho, Jennifer Wang, Xin-qiao Zhang, Wei-Lien Wang, Amishi Yogesh Shah, Jian H. Song, Anh Hoang, Charles C. Guo, Shi-Ming Tu, Eleni Efstathiou, Jianjun Gao, Matthew Campbell, Arlene O. Siefker-Radtke, Sumankalai Ramachandran, Lidia Lopez, Gary E. Gallick, Christopher J. Logothetis, Tyrone Garnett, and Linghua Wang
- Subjects
Cancer Research ,Gene knockdown ,Bladder cancer ,Microarray ,business.industry ,Cancer ,Synthetic lethality ,medicine.disease ,Pemetrexed ,Oncology ,Cancer research ,Carcinoma ,medicine ,Immunohistochemistry ,business ,medicine.drug - Abstract
Background: The methylthioadenosine phosphorylase (MTAP) gene encodes an essential enzyme for the salvage pathway of adenosine synthesis and is frequently lost in different types of cancer including urothelial bladder carcinoma. Therefore, MTAP-deficient tumors are theoretically very sensitive to anti-folate agents such as pemetrexed that can effectively block the de novo pathway of adenosine synthesis and as a result, create a state of synthetic lethality. We thus hypothesize that tumor MTAP gene deficiency is associated with response to pemetrexed therapy in bladder cancer. Methods: In this study, we investigated MTAP gene deficiency rates in the TCGA database and confirmed MTAP protein loss by immunohistochemistry using a tumor tissue microarray containing bladder tumor tissues from 151 patients. We then performed in vitro and in vivo studies using MTAP-proficient and MTAP-deficient human bladder cancer cell lines. Functional loss of MTAP was verified with mass spectrometry, which detects its substrate methylthioadenosine (MTA) levels. We also correlated these pre-clinical studies with clinical response data on patients with metastatic bladder cancer treated with pemetrexed. Results: We identified that 27.8% bladder cancer patients have MTAP protein deficiency, which is consistent with exome sequencing data from the TCGA database. In vitro data showed MTAP-deficient human bladder cancer cell lines were significantly more sensitive to pemetrexed, with IC50 at least 40 times lower than MTAP-proficient cell lines. Subsequent knockdown of the MTAP gene in MTAP-proficient cell lines increased sensitivities to pemetrexed treatment. Consistent with the in vitro data, pemetrexed significantly inhibited the growth of MTAP-deficient or knockdown xenograft tumors but not MTAP-proficient tumors. Furthermore, 4 of 4 (100%) patients with MTAP-deficient metastatic bladder cancer responded to pemetrexed treatment, whereas only 1 of 11 (9%) patients with MTAP-proficient metastatic bladder cancer responded to pemetrexed. Conclusion: Our data demonstrate that MTAP gene loss in urothelial bladder cancer leads to a metabolic state of synthetic lethality with pemetrexed therapy. Therefore, bladder tumor MTAP loss should be further investigated as a potential biomarker for selection of patients for anti-folate therapy. Citation Format: Jianfeng Chen, Omar Alhalabi, Guangchun Han, Wei-Lien Wang, Xin-Qiao Zhang, Jian H. Song, Lidia P. Lopez, Sumankalai Ramachandran, Anh G. Hoang, Tyrone Garnett, Matthew Campbell, Amishi Y. Shah, Jennifer Wang, Arlene O. Siefker-Radtke, Shi-Ming Tu, Mark Titus, Charles C. Guo, Gary E. Gallick, Eleni Efstathiou, William F. Benedict, Christopher J. Logothetis, Thai H. Ho, Linghua Wang, Jianjun Gao. MTAP gene deficiency creates vulnerability to anti-folate therapy in urothelial bladder carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 385.
- Published
- 2019
- Full Text
- View/download PDF
48. Correlation of methylthioadenosine phosphorylase (MTAP) loss with response to anti-folate therapy in urothelial bladder carcinoma (UBC)
- Author
-
Omar Alhalabi, Arlene O. Siefker-Radtke, Sumankalai Ramachandran, Lidia Lopez, Gary E. Gallick, Christopher J. Logothetis, Guangchun Han, Rebecca S. S. Tidwell, Eleni Efstathiou, Thai H. Ho, Linghua Wang, Jianfeng Chen, Jianjun Gao, Charles C. Guo, Mark Titus, Anh Hoang, Pavlos Msaouel, Jian H. Song, Matthew T. Campbell, and Wei-Lien Wang
- Subjects
chemistry.chemical_classification ,Cancer Research ,business.industry ,fungi ,Nucleotide synthesis ,medicine.disease ,03 medical and health sciences ,Methylthioadenosine phosphorylase ,0302 clinical medicine ,Enzyme ,Pemetrexed ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Carcinoma ,Medicine ,business ,Gene ,Nucleotide salvage ,030215 immunology ,medicine.drug - Abstract
4521 Background: The MTAP gene encodes an essential enzyme for the salvage pathway of nucleotide synthesis and is frequently deleted in UBC. Anti-folate agents such as pemetrexed can effectively inhibit the de novo pathway of nucleotide synthesis and as a result, create a synthetic lethality in MTAP deficient UBC. We hypothesize that MTAP gene loss correlates with enhanced response to pemetrexed in UBC. Methods: We investigated MTAP gene deletion rates in the TCGA database and determined MTAP protein loss rates by immunohistochemistry (IHC) using a UBC tissue microarray (TMA) from 151 patients (pts). We then performed in vitro and in vivo studies using MTAP proficient and MTAP deficient bladder cancer cell lines. At the clinical level, we performed a retrospective analysis based on MTAP status of pts treated with pemetrexed as 2nd line at our institution between 2014 and 2018. We are now enrolling pts in a single-arm, open-label, phase II clinical trial (NCT02693717) with pemetrexed in pts with MTAP deficient UBC. Results: Per our TCGA and TMA IHC analyses, MTAP deficiency rate was 25.9% and 27.8%, respectively. MTAP deficient UBC cell lines were at least 40 times more sensitive to pemetrexed than MTAP proficient lines. Knockdown of the MTAP gene increased apoptosis rate by pemetrexed from approximately 20% to 60%. Additionally, pemetrexed significantly inhibited the growth of MTAP deficient or knockdown xenograft tumors but not MTAP proficient tumors. Retrospective analysis of 12 pts using RECIST criteria indicated that all 4 MTAP deficient UBC pts responded to pemetrexed whereas only 1 of 8 (12.5%) MTAP proficient UBC pts responded. Of the 6 pts enrolled on the clinical trial, 3 (50%) had complete or partial response, 1 had stable disease, 1 was not evaluable and 1 had disease progression. Combined analysis of the entire experience demonstrates a higher response rate in MTAP deficient UBC (70%) as compared to MTAP proficient UBC (12.5%). Conclusions: Our preclinical and clinical data demonstrate that MTAP loss in UBC leads to a state of synthetic lethality when treated with pemetrexed and should be further investigated as a novel biomarker to predict response to anti-folate agents.
- Published
- 2019
- Full Text
- View/download PDF
49. Targeting Src and Tubulin in Mucinous Ovarian Carcinoma
- Author
-
Anil K. Sood, Chad V. Pecot, Hyun Jin Choi, Jian H. Song, Sunila Pradeep, Tao Liu, Michael Frumovitz, Cristina Ivan, Wei Hu, David G. Hangauer, Nicholas B. Jennings, Takahito Miyake, Behrouz Zand, Heather J. Dalton, Gary E. Gallick, Keith A. Baggerly, Yu Kang, Jie Huang, Chunhua Lu, Yunfei Wen, Justin Bottsford-Miller, and Robert L. Coleman
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Organoplatinum Compounds ,Cell Survival ,Pyridines ,Morpholines ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Biology ,Microtubules ,Article ,Inhibitory Concentration 50 ,Mice ,In vivo ,Cell Line, Tumor ,Internal medicine ,Ovarian carcinoma ,Acetamides ,medicine ,Animals ,Humans ,PTEN ,Cell Proliferation ,Ovarian Neoplasms ,Cell growth ,Cell Cycle ,PTEN Phosphohydrolase ,Cancer ,Drug Synergism ,Cell cycle ,medicine.disease ,Adenocarcinoma, Mucinous ,Xenograft Model Antitumor Assays ,Tubulin Modulators ,Tumor Burden ,Oxaliplatin ,src-Family Kinases ,biology.protein ,Cancer research ,Female ,Protein Multimerization ,Ovarian cancer ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Purpose: To investigate the antitumor effects of targeting Src and tubulin in mucinous ovarian carcinoma. Experimental Design: The in vitro and in vivo effects and molecular mechanisms of KX-01, which inhibits Src pathway and tubulin polymerization, were examined in mucinous ovarian cancer models. Results: In vitro studies using RMUG-S and RMUG-L cell lines showed that KX-01 inhibited cell proliferation, induced apoptosis, arrested the cell cycle at the G2–M phase, and enhanced the cytotoxicity of oxaliplatin in the KX-01–sensitive cell line, RMUG-S. In vivo studies showed that KX-01 significantly decreased tumor burden in RMUG-S and RMUG-L mouse models relative to untreated controls, and the effects were greater when KX-01 was combined with oxaliplatin. KX-01 alone and in combination with oxaliplatin significantly inhibited tumor growth by reducing cell proliferation and inducing apoptosis in vivo. PTEN knock-in experiments in RMUG-L cells showed improved response to KX-01. Reverse phase protein array analysis showed that in addition to blocking downstream molecules of Src family kinases, KX-01 also activated acute stress-inducing molecules. Conclusion: Our results showed that targeting both the Src pathway and tubulin with KX-01 significantly inhibited tumor growth in preclinical mucinous ovarian cancer models, suggesting that this may be a promising therapeutic approach for patients with mucinous ovarian carcinoma. Clin Cancer Res; 19(23); 6532–43. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
50. Inhibition of Cell Adhesion by a Cadherin-11 Antibody Thwarts Bone Metastasis
- Author
-
Song Chang Lin, Guoyu Yu, Gary E. Gallick, Angelica Ortiz, Jian Kuang, Chih-Fen Huang, Wilber Huang, Jian H. Song, Hyojin Cho, Li Yuan Yu-Lee, Sue Hwa Lin, Mehmet Asim Bilen, Robert L. Satcher, and Yu Chen Lee
- Subjects
Cancer Research ,biology ,Cadherin ,medicine.drug_class ,Bone metastasis ,Osteoblast ,Transfection ,medicine.disease ,Monoclonal antibody ,Metastasis ,medicine.anatomical_structure ,Oncology ,biology.protein ,medicine ,Cancer research ,Antibody ,Cell adhesion ,Molecular Biology - Abstract
Cadherin-11 (CDH11) is a member of the cadherin superfamily mainly expressed in osteoblasts but not in epithelial cells. However, prostate cancer cells with a propensity for bone metastasis express high levels of cadherin-11 and reduced levels of E-cadherin. Downregulation of cadherin-11 inhibits interaction of prostate cancer cells with osteoblasts in vitro and homing of prostate cancer cells to bone in an animal model of metastasis. These findings indicate that targeting cadherin-11 may prevent prostate cancer bone metastasis. To explore this possibility, a panel of 21 monoclonal antibodies (mAb) was generated against the extracellular (EC) domain of cadherin-11. Two antibodies, mAbs 2C7 and 1A5, inhibited cadherin-11–mediated cell–cell aggregation in vitro using L-cells transfected with cadherin-11. Both antibodies demonstrated specificity to cadherin-11, and neither antibody recognized E-cadherin or N-cadherin on C4-2B or PC3 cells, respectively. Furthermore, mAb 2C7 inhibited cadherin-11–mediated aggregation between the highly metastatic PC3-mm2 cells and MC3T3-E1 osteoblasts. Mechanistically, a series of deletion mutants revealed a unique motif, aa 343-348, in the cadherin-11 EC3 domain that is recognized by mAb 2C7 and that this motif coordinated cell–cell adhesion. Importantly, administration of mAb 2C7 in a prophylactic setting effectively prevented metastasis of PC3-mm2 cells to bone in an in vivo mouse model. These results show that targeting the extracellular domain of cadherin-11 can limit cellular adhesion and metastatic dissemination of prostate cancer cells. Implications: Monotherapy using a cadherin-11 antibody is a suitable option for the prevention of bone metastases. Mol Cancer Res; 11(11); 1401–11. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.