Network


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

Hotspot


Dive into the research topics where Hyeongsun Moon is active.

Publication


Featured researches published by Hyeongsun Moon.


Molecular & Cellular Proteomics | 2012

Expression of PTRF in PC-3 Cells Modulates Cholesterol Dynamics and the Actin Cytoskeleton Impacting Secretion Pathways

Kerry L. Inder; Yu Zi Zheng; Melissa J. Davis; Hyeongsun Moon; Dorothy Loo; Hien D. Nguyen; Judith A. Clements; Robert G. Parton; Leonard J. Foster; Michelle M. Hill

Expression of caveolin-1 is up-regulated in prostate cancer metastasis and is associated with aggressive recurrence of the disease. Intriguingly, caveolin-1 is also secreted from prostate cancer cell lines and has been identified in secreted prostasomes. Caveolin-1 is the major structural component of the plasma membrane invaginations called caveolae. Co-expression of the coat protein Polymerase I and transcript release factor (PTRF) is required for caveolae formation. We recently found that expression of caveolin-1 in the aggressive prostate cancer cell line PC-3 is not accompanied by PTRF, leading to noncaveolar caveolin-1 lipid rafts. Moreover, ectopic expression of PTRF in PC-3 cells sequesters caveolin-1 into caveolae. Here we quantitatively analyzed the effect of PTRF expression on the PC-3 proteome using stable isotope labeling by amino acids in culture and subcellular proteomics. We show that PTRF reduced the secretion of a subset of proteins including secreted proteases, cytokines, and growth regulatory proteins, partly via a reduction in prostasome secretion. To determine the cellular mechanism accounting for the observed reduction in secreted proteins we analyzed total membrane and the detergent-resistant membrane fractions. Our data show that PTRF expression selectively impaired the recruitment of actin cytoskeletal proteins to the detergent-resistant membrane, which correlated with altered cholesterol distribution in PC-3 cells expressing PTRF. Consistent with this, modulating cellular cholesterol altered the actin cytoskeleton and protein secretion in PC-3 cells. Intriguingly, several proteins that function in ER to Golgi trafficking were reduced by PTRF expression. Taken together, these results suggest that the noncaveolar caveolin-1 found in prostate cancer cells generates a lipid raft microenvironment that accentuates secretion pathways, possibly at the step of ER sorting/exit. Importantly, these effects could be modulated by PTRF expression.


Trends in Endocrinology and Metabolism | 2014

Statins: protectors or pretenders in prostate cancer?

Hyeongsun Moon; Michelle M. Hill; Matthew J. Roberts; Robert A. Gardiner; Andrew J. Brown

The role of statin therapy in prostate cancer (PCa) prevention and treatment is plagued by controversy. This critical review of published clinical series reveals several caveats in earlier studies, which reported no benefit. Recent studies that adjust for confounding factors have demonstrated statin therapy to be associated with PCa prevention and favorable clinical outcomes. Developed as inhibitors of cholesterol synthesis, the expected mechanism of statin action is systemic cholesterol reduction. By lowering circulating cholesterol, statins indirectly reduce cellular cholesterol levels in multiple cell types, impacting on membrane microdomains and steroidogenesis. Although non-cholesterol mechanisms of statin action have been proposed, they are limited by the uncertainties surrounding in vivo tissue statin concentrations.


Oncogene | 2014

PTRF/cavin-1 neutralizes non-caveolar caveolin-1 microdomains in prostate cancer

Hyeongsun Moon; Cheok Soon Lee; Kerry L. Inder; Sowmya Sharma; Eunju Choi; Debra Black; Kim-Anh Lê Cao; Clay Winterford; Jermaine Coward; Ming-Tat Ling; David J. Craik; Robert G. Parton; Pamela J. Russell; Michelle M. Hill

Caveolin-1 has a complex role in prostate cancer and has been suggested to be a potential biomarker and therapeutic target. As mature caveolin-1 resides in caveolae, invaginated lipid raft domains at the plasma membrane, caveolae have been suggested as a tumor-promoting signaling platform in prostate cancer. However, caveola formation requires both caveolin-1 and cavin-1 (also known as PTRF; polymerase I and transcript release factor). Here, we examined the expression of cavin-1 in prostate epithelia and stroma using tissue microarray including normal, non-malignant and malignant prostate tissues. We found that caveolin-1 was induced without the presence of cavin-1 in advanced prostate carcinoma, an expression pattern mirrored in the PC-3 cell line. In contrast, normal prostate epithelia expressed neither caveolin-1 nor cavin-1, while prostate stroma highly expressed both caveolin-1 and cavin-1. Utilizing PC-3 cells as a suitable model for caveolin-1-positive advanced prostate cancer, we found that cavin-1 expression in PC-3 cells inhibits anchorage-independent growth, and reduces in vivo tumor growth and metastasis in an orthotopic prostate cancer xenograft mouse model. The expression of α-smooth muscle actin in stroma along with interleukin-6 (IL-6) in cancer cells was also decreased in tumors of mice bearing PC-3-cavin-1 tumor cells. To determine whether cavin-1 acts by neutralizing caveolin-1, we expressed cavin-1 in caveolin-1-negative prostate cancer LNCaP and 22Rv1 cells. Caveolin-1 but not cavin-1 expression increased anchorage-independent growth in LNCaP and 22Rv1 cells. Cavin-1 co-expression reversed caveolin-1 effects in caveolin-1-positive LNCaP cells. Taken together, these results suggest that caveolin-1 in advanced prostate cancer is present outside of caveolae, because of the lack of cavin-1 expression. Cavin-1 expression attenuates the effects of non-caveolar caveolin-1 microdomains partly via reduced IL-6 microenvironmental function. With circulating caveolin-1 as a potential biomarker for advanced prostate cancer, identification of the molecular pathways affected by cavin-1 could provide novel therapeutic targets.


Journal of extracellular vesicles | 2014

Cavin-1/PTRF alters prostate cancer cell-derived extracellular vesicle content and internalization to attenuate extracellular vesicle-mediated osteoclastogenesis and osteoblast proliferation

Kerry L. Inder; Jayde E. Ruelcke; Lara Petelin; Hyeongsun Moon; Eunju Choi; James Rae; Antje Blumenthal; Dietmar W. Hutmacher; Nicholas A. Saunders; Jennifer L. Stow; Robert G. Parton; Michelle M. Hill

Background Tumour-derived extracellular vesicles (EVs) play a role in tumour progression; however, the spectrum of molecular mechanisms regulating EV secretion and cargo selection remain to be fully elucidated. We have reported that cavin-1 expression in prostate cancer PC3 cells reduced the abundance of a subset of EV proteins, concomitant with reduced xenograft tumour growth and metastasis. Methods We examined the functional outcomes and mechanisms of cavin-1 expression on PC3-derived EVs (PC3-EVs). Results PC3-EVs were internalized by osteoclast precursor RAW264.7 cells and primary human osteoblasts (hOBs) in vitro, stimulating osteoclastogenesis 37-fold and hOB proliferation 1.5-fold, respectively. Strikingly, EVs derived from cavin-1-expressing PC3 cells (cavin-1-PC3-EVs) failed to induce multinucleate osteoblasts or hOB proliferation. Cavin-1 was not detected in EVs, indicating an indirect mechanism of action. EV morphology, size and quantity were also not affected by cavin-1 expression, suggesting that cavin-1 modulated EV cargo recruitment rather than release. While cavin-1-EVs had no osteoclastogenic function, they were internalized by RAW264.7 cells but at a reduced efficiency compared to control EVs. EV surface proteins are required for internalization of PC3-EVs by RAW264.7 cells, as proteinase K treatment abolished uptake of both control and cavin-1-PC3-EVs. Removal of sialic acid modifications by neuraminidase treatment increased the amount of control PC3-EVs internalized by RAW264.7 cells, without affecting cavin-1-PC3-EVs. This suggests that cavin-1 expression altered the glycosylation modifications on PC3-EV surface. Finally, cavin-1 expression did not affect EV in vivo tissue targeting as both control and cavin-1-PC3-EVs were predominantly retained in the lung and bone 24 hours after injection into mice. Discussion Taken together, our results reveal a novel pathway for EV cargo sorting, and highlight the potential of utilizing cavin-1-mediated pathways to attenuate metastatic prostate cancer.


Cancer Research | 2014

Abstract 4950: Hypercholesterolemia promotes prostate cancer PC-3 metastases in orthotopic xenograft mice

Hyeongsun Moon; Laura J. Sharpe; Eunju Choi; Helle Bielefeldt-Ohmann; Zeyad D. Nassar; Marie-Odile Parat; Mathias Francois; C. Soon Lee; Andrew J. Brown; Pamela J. Russell; Kerry L. Inder; Michelle M. Hill

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Hypercholesterolemia has been proposed as a potential risk factor for advanced prostate cancer, and use of cholesterol-lowering drugs, statins, inversely correlates with advanced prostate cancer risk. Hypercholesterolemia increases growth of androgen-sensitive LNCaP xenograft in vivo, potentially via androgen signaling. The aim of this study was to determine if hypercholesterolemia affects castration-resistant prostate tumor progression using an androgen receptor-negative prostate cancer cell line PC-3. Compared to control media, cholesterol-deficient media reduced PC-3 proliferation, migration and anchorage-independent growth in vitro. While adding cholesterol did not significantly increase proliferation or anchorage-independent growth, cholesterol replacement in cholesterol-deficient media significantly increased PC-3 transmigration. In order to determine the in vivo effect, mice were randomly assigned to normal or hypercholesterolemic, isocaloric diet groups (N=14 and 15, respectively). After two weeks, hypercholesterolemic diet significantly increased circulating cholesterol but did not increase body weight. PC-3 cells stably expressing luciferase were orthotopically injected into the dorsolateral prostate. Tumor progression and metastases were monitored by in vivo and ex vivo optical bioluminescence imaging for 6 weeks. Strikingly, the results show that diet-induced hypercholesterolemia accelerated tumor metastases to lymph nodes, lung, proximal and distant bones without significantly affecting primary tumor growth. The metastases were confirmed histopathologcally. Hypercholesterolemia was not associated with elevated weight or circulating testosterone. This is the first study to directly demonstrate a causal relationship between hypercholesterolemia and prostate tumor metastases mediated through androgen-independent mechanisms, highlighting the potential clinical benefit of cholesterol lowering therapy such as statins in advanced, castration-resistant, prostate cancer patients. Citation Format: Hyeongsun Moon, Laura Sharpe, Eunju Choi, Helle Bielefeldt-Ohmann, Zeyad Nassar, Marie-Odile Parat, Mathias Francois, C Soon Lee, Andrew Brown, Pamela Russell, Kerry Inder, Michelle Hill. Hypercholesterolemia promotes prostate cancer PC-3 metastases in orthotopic xenograft mice. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4950. doi:10.1158/1538-7445.AM2014-4950


Cancer Research | 2013

Abstract 3040: Cavin-1 alters oncogenic effects of caveolin-1 microdomains in prostate cancer.

Hyeongsun Moon; C. Soon Lee; Sowmya Sharma; Kerry L. Inder; Debra Black; Kim-Anh Lê Cao; Clay Winterford; Patrick Ling; David J. Craik; Robert G. Parton; Pamela J. Russell; Michelle M. Hill

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Caveolin-1 is associated with prostate cancer progression and has been suggested to be a biomarker and therapeutic target. Mature caveolin-1 resides in lipid raft domains at the plasma membrane, where it forms caveolae upon co-expression of cavin-1 (also known as PTRF; polymerase I and transcript release factor). In the absence of cavin-1, caveolin-1 does not form caveolae but are found on flat membrane. To determine if oncogenic caveolin-1 in prostate cancer is present in caveolae, we examined the relative expression of caveolin-1 and cavin-1 in normal, non-malignant and malignant prostate tissues. We found that caveolin-1 is induced in prostate cancer without cavin-1, an expression pattern mirror in the PC3 cell line. Previously we showed that expression of cavin-1 in PC3 cells recruits flat membrane caveolin-1 to caveolae and reduced transmigration. Here we report that cavin-1 expression reduces tumour size and metastasis of PC3 cells in vivo, using an orthotopic prostate cancer xenograft mouse model. To determine if cavin-1 acts by neutralizing oncogenic caveolin-1, we expressed cavin-1 in caveolin-1 negative LNCaP and 22Rv1 cells. While caveolin-1 over-expression increased anchorage-independent growth of LNCaP and 22Rv1 cells, cavin-1 over-expression had no effect. Furthermore, co-expression of cavin-1 in LNCaP+caveolin-1 cells reversed caveolin-1 effect. Taken together, these results suggest that caveolin-1 in prostate cancer is present outside of caveolae, and caveola formation by cavin-1 co-expression alters the oncogenic effect of non-caveolar caveolin-1 microdomains. Citation Format: Hyeongsun Moon, C Soon Lee, Sowmya Sharma, Kerry Inder, Debra Black, Kim-Anh Le Cao, Clay Winterford, Patrick Ling, the Australian Prostate Cancer BioResource, David Craik, Robert Parton, Pamela Russell, Michelle Hill. Cavin-1 alters oncogenic effects of caveolin-1 microdomains in prostate cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3040. doi:10.1158/1538-7445.AM2013-3040


Oncotarget | 2013

PTRF/Cavin-1 decreases prostate cancer angiogenesis and lymphangiogenesis

Zeyad D. Nassar; Hyeongsun Moon; Tam Duong; LiQi Neo; Michelle M. Hill; Mathias Francois; Robert G. Parton; Marie-Odile Parat


Cancer Research | 2018

Abstract 150: Elucidating the molecular mechanisms of cutaneous melanoma development from melanocyte stem cells

Hyeongsun Moon; Leanne Donahue; Jerry Zhu; Andrew J. P. White


Faculty of Health; Institute of Health and Biomedical Innovation | 2015

Diet-induced hypercholesterolemia promotes androgen-independent prostate cancer metastasis via IQGAP1 and caveolin-1

Hyeongsun Moon; Jayde E. Ruelcke; Eunju Choi; Laura J. Sharpe; Zeyad D. Nassar; Helle Bieledeldt-Ohmann; Marie-Odile Parat; Anup Shah; Mathias Francois; Kerry L. Inder; Andrew J. Brown; Pamela J. Russell; Robert G. Parton; Michelle M. Hill


Archive | 2014

Dissecting the roles of cholesterol and caveolin in castration-resistant prostate cancer

Hyeongsun Moon

Collaboration


Dive into the Hyeongsun Moon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kerry L. Inder

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eunju Choi

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Pamela J. Russell

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Brown

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Soon Lee

Royal Prince Alfred Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge