1
|
Zhang X, Jiang P, Wang C. The role of prostate-specific antigen in the osteoblastic bone metastasis of prostate cancer: a literature review. Front Oncol 2023; 13:1127637. [PMID: 37746292 PMCID: PMC10513387 DOI: 10.3389/fonc.2023.1127637] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Prostate cancer is the only human malignancy that generates predominantly osteoblastic bone metastases, and osteoblastic bone metastases account for more than 90% of osseous metastases of prostate cancer. Prostate-specific antigen (PSA) plays an important role in the osteoblastic bone metastasis of prostate cancer, which can promote osteomimicry of prostate cancer cells, suppress osteoclast differentiation, and facilitate osteoblast proliferation and activation at metastatic sites. In the meantime, it can activate osteogenic factors, including insulin-like growth factor, transforming growth factor β2 and urokinase-type plasminogen activator, and meanwhile suppress osteolytic factors such as parathyroid hormone-related protein. To recapitulate, PSA plays a significant role in the osteoblastic predominance of prostate cancer bone metastasis and bone remodeling by regulating multiple cells and factors involved in osseous metastasis.
Collapse
Affiliation(s)
| | | | - Chaojun Wang
- Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Provera MD, Straign DM, Karimpour P, Ihle CL, Owens P. Bone morphogenetic protein pathway responses and alterations of osteogenesis in metastatic prostate cancers. Cancer Rep (Hoboken) 2023; 6:e1707. [PMID: 36054271 PMCID: PMC9940003 DOI: 10.1002/cnr2.1707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/07/2022] [Accepted: 07/27/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Prostate cancer is a common cancer in men that annually results in more than 33 000 US deaths. Mortality from prostate cancer is largely from metastatic disease, reflecting on the great strides in the last century of treatments in care for the localized disease. Metastatic castrate resistant prostate cancer (mCRPC) will commonly travel to the bone, creating unique bone pathology that requires nuanced treatments in those sites with surgical, radio and chemotherapeutic interventions. The bone morphogenetic protein (BMP) pathway has been historically studied in the capacity to regulate the osteogenic nature of new bone. New mineralized bone generation is a frequent and common observation in mCRPC and referred to as blastic bone lesions. Less common are bone destructive lesions that are termed lytic. METHODS We queried the cancer genome atlas (TCGA) prostate cancer databases for the expression of the BMP pathway and found that distinct gene expression of the ligands, soluble antagonists, receptors, and intracellular mediators were altered in localized versus metastatic disease. Human prostate cancer cell lines have an innate ability to promote blastic- or lytic-like bone lesions and we hypothesized that inhibiting BMP signaling in these cell lines would result in a distinct change in osteogenesis gene expression with BMP inhibition. RESULTS We found unique and common changes by comparing these cell lines response and unique BMP pathway alterations. We treated human PCa cell lines with distinct bone pathologic phenotypes with the BMP inhibitor DMH1 and found distinct osteogenesis responses. We analyzed distinct sites of metastatic PCa in the TCGA and found that BMP signaling was selectively altered in commons sites such as lymph node, bone and liver compared to primary tumors. CONCLUSIONS Overall we conclude that BMPs in metastatic prostate cancer are important signals and functional mediators of diverse processes that have potential for individualized precision oncology in mCRPC.
Collapse
Affiliation(s)
- Meredith D. Provera
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | - Desiree M. Straign
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | | | - Claire L. Ihle
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | - Philip Owens
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
- Department of Veterans Affairs, Research Service, Eastern Colorado Health Care SystemAuroraColoradoUSA
| |
Collapse
|
3
|
Chen C, Huang R, Zhou J, Guo L, Xiang S. Formation of pre-metastatic bone niche in prostate cancer and regulation of traditional chinese medicine. Front Pharmacol 2022; 13:897942. [PMID: 36059977 PMCID: PMC9428453 DOI: 10.3389/fphar.2022.897942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Prostate cancer with bone metastasis has a high cancer-specific mortality. Thus, it is essential to delineate the mechanism of bone metastasis. Pre-metastatic niche (PMN) is a concept in tumor metastasis, which is characterized by tumor-secreted factors, reprogramming of stromal cells, and immunosuppression by myeloid-derived suppressor cells (MDSC), which is induced by bone marrow-derived cells (BMDC) in the target organ. However, PMN does not explain the predilection of prostate cancer towards bone metastasis. In this review, we discuss the initiation of bone metastasis of prostate cancer from the perspective of PMN and tumor microenvironment in a step-wise manner. Furthermore, we present a new concept called pre-metastatic bone niche, featuring inherent BMDC, to interpret bone metastasis. Moreover, we illustrate the regulation of traditional Chinese medicine on PMN.
Collapse
|
4
|
Elshafae SM, Dirksen WP, Alasonyalilar-Demirer A, Breitbach J, Yuan S, Kantake N, Supsavhad W, Hassan BB, Attia Z, Rosol TJ. Canine prostatic cancer cell line (LuMa) with osteoblastic bone metastasis. Prostate 2020; 80:698-714. [PMID: 32348616 PMCID: PMC7291846 DOI: 10.1002/pros.23983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Osteoblastic bone metastasis represents the most common complication in men with prostate cancer (PCa). During progression and bone metastasis, PCa cells acquire properties similar to bone cells in a phenomenon called osteomimicry, which promotes their ability to metastasize, proliferate, and survive in the bone microenvironment. The mechanism of osteomimicry resulting in osteoblastic bone metastasis is unclear. METHODS We developed and characterized a novel canine prostatic cancer cell line (LuMa) that will be useful to investigate the relationship between osteoblastic bone metastasis and osteomimicry in PCa. The LuMa cell line was established from a primary prostate carcinoma of a 13-year old mixed breed castrated male dog. Cell proliferation and gene expression of LuMa were measured and compared to three other canine prostatic cancer cell lines (Probasco, Ace-1, and Leo) in vitro. The effect of LuMa cells on calvaria and murine preosteoblastic (MC3T3-E1) cells was measured by quantitative reverse-transcription polymerase chain reaction and alkaline phosphatase assay. LuMa cells were transduced with luciferase for monitoring in vivo tumor growth and metastasis using different inoculation routes (subcutaneous, intratibial [IT], and intracardiac [IC]). Xenograft tumors and metastases were evaluated using radiography and histopathology. RESULTS After left ventricular injection, LuMa cells metastasized to bone, brain, and adrenal glands. IT injections induced tumors with intramedullary new bone formation. LuMa cells had the highest messenger RNA levels of osteomimicry genes (RUNX2, RANKL, and Osteopontin [OPN]), CD44, E-cadherin, and MYOF compared to Ace-1, Probasco, and Leo cells. LuMa cells induced growth in calvaria defects and modulated gene expression in MC3T3-E1 cells. CONCLUSIONS LuMa is a novel canine PCa cell line with osteomimicry and stemness properties. LuMa cells induced osteoblastic bone formation in vitro and in vivo. LuMa PCa cells will serve as an excellent model for studying the mechanisms of osteomimicry and osteoblastic bone and brain metastasis in prostate cancer.
Collapse
Affiliation(s)
- Said M. Elshafae
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Pathology, Faculty of Veterinary medicine, Benha University, Benha, Egypt
- Dept. of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Wessel P. Dirksen
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Aylin Alasonyalilar-Demirer
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Department of Pathology, Faculty of Veterinary Medicine, Bursa Uludag University, Turkey
| | - Justin Breitbach
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Shiyu Yuan
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Noriko Kantake
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Wachiraphan Supsavhad
- Dept. of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Bardes B. Hassan
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Zayed Attia
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Animal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Sadat City University, Sadat City, Egypt
| | - Thomas J. Rosol
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Correspondence to: Dr. Thomas Rosol, Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, 225 Irvine Hall, Athens, OH 45701, USA. , Phone: 740.593.2405
| |
Collapse
|
5
|
Oba E, Aung NY, Ohe R, Sadahiro M, Yamakawa M. The distribution of macrophage subtypes and their relationship to bone morphogenetic protein 2 in calcified aortic valve stenosis. Am J Transl Res 2020; 12:1728-1740. [PMID: 32509172 PMCID: PMC7270035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
Activation of the osteogenic signaling cascade (OSC) is thought to be involved in aortic valve stenosis. The aim of this study was to clarify the distribution of macrophage (M) subtypes in the calcified aortic valve and to clarify the relationship between osteoblast-like cells (OLC) and OSC activation. Thirty-six cases of calcified aortic valve were set as the calcification group, and six autopsy cases of aortic valve without pathological calcification comprised the noncalcification group. Aortic valve tissues were used in histological studies including single and double immunostaining to identify M subtypes, bone morphogenetic protein 2 (BMP2) and osteopontin, reverse transcription polymerase chain reaction (RT-PCR) for CD206, heme oxygenase-1 (HO-1), and BMP2 mRNAs and in situ RT-PCR for BMP2 mRNA. Ms positive for CD68, CD163, CD206, and HO-1 were significantly higher in the calcification group than in the noncalcification group (P < 0.01). Comparison of the positive cells in each section of the calcification group showed that cells of all M subtypes were found around calcifications. Osteopontin+ cells were also observed around calcifications. CD163+/CD206+ M2 and CD163+/HO-1+ Mox were significantly higher in the sponge layer in both groups. In double immunofluorescence, CD206+ and a portion of HO-1+ Ms expressed BMP2, and in RT-PCR, CD206 or HO-1 mRNA was expressed in cases in which BMP2 was expressed. In in situ RT-PCR, expression of BMP2 mRNA was observed around calcifications. This work clarifies the distribution of M subtypes in calcified aortic valves. In addition, the results suggest that CD206+ M2 and HO-1+ Mox, which express BMP2 in calcified aortic valves, are OLC candidates.
Collapse
Affiliation(s)
- Eiichi Oba
- Second Department of Surgery, Yamagata University Faculty of MedicineYamagata, Japan
| | - Naing Ye Aung
- Department of Pathological Diagnostics, Yamagata University Faculty of MedicineYamagata, Japan
| | - Rintaro Ohe
- Department of Pathological Diagnostics, Yamagata University Faculty of MedicineYamagata, Japan
| | - Mitsuaki Sadahiro
- Second Department of Surgery, Yamagata University Faculty of MedicineYamagata, Japan
| | - Mitsunori Yamakawa
- Department of Pathological Diagnostics, Yamagata University Faculty of MedicineYamagata, Japan
| |
Collapse
|
6
|
Shupp AB, Kolb AD, Mukhopadhyay D, Bussard KM. Cancer Metastases to Bone: Concepts, Mechanisms, and Interactions with Bone Osteoblasts. Cancers (Basel) 2018; 10:E182. [PMID: 29867053 PMCID: PMC6025347 DOI: 10.3390/cancers10060182] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
The skeleton is a unique structure capable of providing support for the body. Bone resorption and deposition are controlled in a tightly regulated balance between osteoblasts and osteoclasts with no net bone gain or loss. However, under conditions of disease, the balance between bone resorption and deposition is upset. Osteoblasts play an important role in bone homeostasis by depositing new bone osteoid into resorption pits. It is becoming increasingly evident that osteoblasts additionally play key roles in cancer cell dissemination to bone and subsequent metastasis. Our laboratory has evidence that when osteoblasts come into contact with disseminated breast cancer cells, the osteoblasts produce factors that initially reduce breast cancer cell proliferation, yet promote cancer cell survival in bone. Other laboratories have demonstrated that osteoblasts both directly and indirectly contribute to dormant cancer cell reactivation in bone. Moreover, we have demonstrated that osteoblasts undergo an inflammatory stress response in late stages of breast cancer, and produce inflammatory cytokines that are maintenance and survival factors for breast cancer cells and osteoclasts. Advances in understanding interactions between osteoblasts, osteoclasts, and bone metastatic cancer cells will aid in controlling and ultimately preventing cancer cell metastasis to bone.
Collapse
Affiliation(s)
- Alison B Shupp
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Alexus D Kolb
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Dimpi Mukhopadhyay
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Karen M Bussard
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
7
|
Mancini A, Colapietro A, Pompili S, Del Fattore A, Delle Monache S, Biordi LA, Angelucci A, Mattei V, Liang C, Gravina GL, Festuccia C. Dual PI3 K/mTOR inhibition reduces prostate cancer bone engraftment altering tumor-induced bone remodeling. Tumour Biol 2018; 40:1010428318771773. [PMID: 29687745 DOI: 10.1177/1010428318771773] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Morbidity in advanced prostate cancer patients is largely associated with bone metastatic events. The development of novel therapeutic strategies is imperative in order to effectively treat this incurable stage of the malignancy. In this context, Akt signaling pathway represents a promising therapeutic target able to counteract biochemical recurrence and metastatic progression in prostate cancer. We explored the therapeutic potential of a novel dual PI3 K/mTOR inhibitor, X480, to inhibit tumor growth and bone colonization using different in vivo prostate cancer models including the subcutaneous injection of aggressive and bone metastatic (PC3) and non-bone metastatic (22rv1) cell lines and preclinical models known to generate bone lesions. We observed that X480 both inhibited the primary growth of subcutaneous tumors generated by PC3 and 22rv1 cells and reduced bone spreading of PCb2, a high osteotropic PC3 cell derivative. In metastatic bone, X480 inhibited significantly the growth and osteolytic activity of PC3 cells as observed by intratibial injection model. X480 also increased the bone disease-free survival compared to untreated animals. In vitro experiments demonstrated that X480 was effective in counteracting osteoclastogenesis whereas it stimulated osteoblast activity. Our report provides novel information on the potential activity of PI3 K/Akt inhibitors on the formation and progression of prostate cancer bone metastases and supports a biological rationale for the use of these inhibitors in castrate-resistant prostate cancer patients at high risk of developing clinically evident bone lesions.
Collapse
Affiliation(s)
- Andrea Mancini
- 1 Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Colapietro
- 1 Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Simona Pompili
- 2 Department of Biotechnological and Applied Clinical Sciences, Human Anatomy, University of L'Aquila, L'Aquila, Italy
| | | | - Simona Delle Monache
- 4 Department of Biotechnological and Applied Clinical Sciences, Laboratory of Applied Biology, University of L'Aquila, L'Aquila, Italy
| | - Leda Assunta Biordi
- 5 Department of Biotechnological and Applied Clinical Sciences, Laboratory of Experimental Oncology, University of L'Aquila, L'Aquila, Italy
| | - Adriano Angelucci
- 6 Department of Biotechnological and Applied Clinical Sciences, Laboratory of General Pathology, University of L'Aquila, L'Aquila, Italy
| | - Vincenzo Mattei
- 7 Laboratory of Experimental Medicine and Environmental Pathology, Rieti University Hub "Sabina Universitas," Rieti, Italy
| | | | - Giovanni Luca Gravina
- 9 Department of Biotechnological and Applied Clinical Sciences, Division of Radiology Oncology; University of L'Aquila, L'Aquila, Italy
| | - Claudio Festuccia
- 1 Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
8
|
Wang MH, Zhou XM, Zhang MY, Shi L, Xiao RW, Zeng LS, Yang XZ, Zheng XFS, Wang HY, Mai SJ. BMP2 promotes proliferation and invasion of nasopharyngeal carcinoma cells via mTORC1 pathway. Aging (Albany NY) 2018; 9:1326-1340. [PMID: 28455969 PMCID: PMC5425130 DOI: 10.18632/aging.101230] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/23/2017] [Indexed: 12/11/2022]
Abstract
Bone morphogenetic protein-2 (BMP2) is a secreted protein that highly expressed in a variety of cancers and contributes to cell proliferation, migration, invasiveness, mobility, metastasis and EMT. However, its clinical significance and biological function in nasopharyngeal carcinoma (NPC) remain unknown up to now. Up-regulation of BMP2 was first observed in NPC cell lines by a genome-wide transcriptome analysis in our previous study. In this study, BMP2 mRNA was detected by qRT-PCR and data showed that it was upregulated in NPC compared with non-cancerous nasopharynx samples. Immunohistochemistry (IHC) analysis in NPC specimens revealed that high BMP2 expression was significantly associated with clinical stage, distant metastasis and shorter survival of NPC patients. Moreover, overexpression of BMP2 in NPC cells promoted cell proliferation, migration, invasiveness and epithelial-mesenchymal transition (EMT). Mechanistically, BMP2 overexpression increase phosphorylated protein level of mTOR, S6K and 4EBP1. Correspondingly, mTORC1 inhibitor rapamycin blocked the effect of BMP2 on NPC cell proliferation and invasion. In conclusion, our results suggest that BMP2 overexpression in NPC enhances proliferation, invasion and EMT of tumor cells through the mTORC1 signaling pathway.
Collapse
Affiliation(s)
- Meng-He Wang
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiao-Min Zhou
- Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou, China
| | - Mei-Yin Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Lu Shi
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ruo-Wen Xiao
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Si Zeng
- Cancer Center of Guangzhou Medical University, Guangzhou, China
| | - Xian-Zi Yang
- Cancer Center of Guangzhou Medical University, Guangzhou, China
| | - X F Steven Zheng
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Shi-Juan Mai
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| |
Collapse
|
9
|
Yang Y, Bai Y, He Y, Zhao Y, Chen J, Ma L, Pan Y, Hinten M, Zhang J, Karnes RJ, Kohli M, Westendorf JJ, Li B, Zhu R, Huang H, Xu W. PTEN Loss Promotes Intratumoral Androgen Synthesis and Tumor Microenvironment Remodeling via Aberrant Activation of RUNX2 in Castration-Resistant Prostate Cancer. Clin Cancer Res 2018; 24:834-846. [PMID: 29167276 PMCID: PMC5816982 DOI: 10.1158/1078-0432.ccr-17-2006] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/06/2017] [Accepted: 11/16/2017] [Indexed: 01/10/2023]
Abstract
Purpose: Intratumoral androgen synthesis (IAS) is a key mechanism promoting androgen receptor (AR) reactivation and antiandrogen resistance in castration-resistant prostate cancer (CRPC). However, signaling pathways driving aberrant IAS remain poorly understood.Experimental Design: The effect of components of the AKT-RUNX2-osteocalcin (OCN)-GPRC6A-CREB signaling axis on expression of steroidogenesis genes CYP11A1 and CYP17A1 and testosterone level were examined in PTEN-null human prostate cancer cell lines. Pten knockout mice were used to examine the effect of Runx2 heterozygous deletion or abiraterone acetate (ABA), a prodrug of the CYP17A1 inhibitor abiraterone on Cyp11a1 and Cyp17a1 expression, testosterone level and tumor microenvironment (TME) remodeling in vivoResults: We uncovered that activation of the AKT-RUNX2-OCN-GPRC6A-CREB signaling axis induced expression of CYP11A1 and CYP17A1 and testosterone production in PTEN-null prostate cancer cell lines in culture. Deletion of Runx2 in Pten homozygous knockout prostate tumors decreased Cyp11a1 and Cyp17a1 expression, testosterone level, and tumor growth in castrated mice. ABA treatment also inhibited testosterone synthesis and alleviated Pten loss-induced tumorigenesis in vivoPten deletion induced TME remodeling, but Runx2 heterozygous deletion or ABA treatment reversed the effect of Pten loss by decreasing expression of the collagenase Mmp9.Conclusions: Abnormal RUNX2 activation plays a pivotal role in PTEN loss-induced IAS and TME remodeling, suggesting that the identified signaling cascade represents a viable target for effective treatment of PTEN-null prostate cancer, including CRPC. Clin Cancer Res; 24(4); 834-46. ©2017 AACR.
Collapse
Affiliation(s)
- Yinhui Yang
- Department of Urology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Yang Bai
- Department of Urology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Yundong He
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Yu Zhao
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jiaxiang Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Linlin Ma
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas
| | - Yunqian Pan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Michael Hinten
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - R Jeffrey Karnes
- Department of Urology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Manish Kohli
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jennifer J Westendorf
- Mayo Clinic Cancer Center, Mayo Clinic College of Medicine, Rochester, Minnesota
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Benyi Li
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas
| | - Runzhi Zhu
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas.
- Department for Cell Therapy, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota.
- Department of Urology, Mayo Clinic College of Medicine, Rochester, Minnesota
- Mayo Clinic Cancer Center, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Wanhai Xu
- Department of Urology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
10
|
Overexpression of p54 nrb/NONO induces differential EPHA6 splicing and contributes to castration-resistant prostate cancer growth. Oncotarget 2018. [PMID: 29535823 PMCID: PMC5828187 DOI: 10.18632/oncotarget.24063] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The non-POU domain-containing octamer binding protein p54nrb/NONO is a multifunctional nuclear protein involved in RNA splicing, processing, and transcriptional regulation of nuclear hormone receptors. Through chromosome copy number analysis via whole-exome sequencing, we revealed amplification of the chromosome Xq11.22-q21.33 locus containing the androgen receptor (AR) and NONO genes in androgen-independent, castration-resistant prostate cancer (CRPC)-like LNCaP-SF cells. Moreover, NONO was frequently amplified and overexpressed in patients with CRPC. RNA sequencing data revealed that a truncated ephrin type-A receptor 6 (EPHA6) splice variant (EPHA6-001) was overexpressed in LNCaP-SF cells, and knockdown of NONO or EPHA6-001 prevented EPHA6-001 expression and reduced proliferation and invasion by LNCaP-SF cells grown under androgen deprivation conditions. Growth inhibition and differential splicing of EPHA6 mRNA by p54nrb/NONO were confirmed in gene silencing experiments in 22Rv1 PCa cells. Importantly, NONO knockdown in LNCaP-SF cells led to reduced tumor growth in castrated mice. These findings indicate that p54nrb/NONO is amplified and overexpressed in CRPC cells and clinical samples, and facilitates CRPC growth by mediating aberrant EPHA6 splicing. We therefore propose that p54nrb/NONO constitutes a novel and attractive therapeutic target for CRPC.
Collapse
|
11
|
Tian H, Zhao J, Brochmann EJ, Wang JC, Murray SS. Bone morphogenetic protein-2 and tumor growth: Diverse effects and possibilities for therapy. Cytokine Growth Factor Rev 2017; 34:73-91. [PMID: 28109670 DOI: 10.1016/j.cytogfr.2017.01.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/14/2016] [Accepted: 01/09/2017] [Indexed: 11/19/2022]
Abstract
Concern regarding safety with respect to the clinical use of human bone morphogenetic protein-2 (BMP-2) has become an increasingly controversial topic. The role of BMP-2 in carcinogenesis is of particular concern. Although there have been many studies of this topic, the results have been contradictory and confusing. We conducted a systematic review of articles that are relevant to the relationship or effect of BMP-2 on all types of tumors and a total of 97 articles were included. Studies reported in these articles were classified into three major types: "expression studies", "in vitro studies", and "in vivo studies". An obvious pattern was that those works that hypothesize an inhibitory effect for BMP-2 most often examined only the proliferative properties of the tumor cells. This subset of studies also contained an extraordinary number of contradictory findings which made drawing a reliable general conclusion impossible. In general, we support a pro-tumorigenesis role for BMP-2 based on the data from these in vitro cell studies and in vivo animal studies, however, more clinical studies should be carried out to help make a firm conclusion.
Collapse
Affiliation(s)
- Haijun Tian
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Elsa J Brochmann
- Research Service, VA Greater Los Angeles Healthcare System, North Hills, CA, United States; Geriatric Research, Education and Clinical Center, VA Greater Los Angeles Healthcare System, North Hills, CA, United States; Department of Medicine, University of California, Los Angeles, CA, United States
| | - Jeffrey C Wang
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, United States
| | - Samuel S Murray
- Research Service, VA Greater Los Angeles Healthcare System, North Hills, CA, United States; Geriatric Research, Education and Clinical Center, VA Greater Los Angeles Healthcare System, North Hills, CA, United States; Department of Medicine, University of California, Los Angeles, CA, United States
| |
Collapse
|
12
|
Effect of PI3K/Akt Signaling Pathway on the Process of Prostate Cancer Metastasis to Bone. Cell Biochem Biophys 2016; 72:171-7. [PMID: 27040945 DOI: 10.1007/s12013-014-0433-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We sought to study the effects of PI3K/Akt pathway and its downstream substrate NF-κB on prostate cancer bone metastatic process. Expression level of active p-Akt in PC3 cells was upregulated by transient expression with constitutively active plasmid CA-Akt or, alternatively, suppressed by dominant negative construct DN-Akt. NF-κB activity was determined by luciferase reporter assays. mRNA and protein expressions of receptor activator of NF-κB ligand (RANKL), parathyroid hormone-related protein (PTHrP), and bone morphogenetic protein 2 (BMP-2) were evaluated using RT-PCR and Western blotting. The effect of cross-talk between PC3 and SaOS2 cells on cell proliferation was analyzed using a co-culture system. Stimulation of p-Akt promoted NF-κB activity, and led to an increase in mRNA and protein expressions of RANKL, PTHrP, and BMP-2 in PC3 PCa cells through NF-κB. Co-culturing PC3 and SaOS2 cells significantly increased the expression of p-Akt and the activity of NF-κB, and promoted proliferation of both PC3 and SaOS2 cells. Increasing expression levels of p-Akt by transfection with CA-Akt led to further increase in cells proliferation, whereas NF-κB inhibitor PDTC partially blocked this effect. PI3K/Akt pathway stimulates the expressions of RANKL, PTHrP, and BMP-2 partly through NF-κB, suggesting its importance for bone metastasis of prostate carcinoma. Interaction of prostate cancer cells with bone cells has a stimulatory effect on cell proliferation.
Collapse
|
13
|
Bone morphogenetic protein signaling in musculoskeletal cancer. J Cancer Res Clin Oncol 2016; 142:2061-72. [PMID: 27043154 DOI: 10.1007/s00432-016-2149-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 03/17/2016] [Indexed: 02/08/2023]
Abstract
PURPOSE Bone morphogenetic proteins (BMPs) belong to the transforming growth factor-β (TGF-β) superfamily of proteins; they were initially named after their ability to induce ectopic bone formation. Published studies have proved BMPs' role in a variety of biological processes such as embryogenesis and patterning of body axes, and maintaining adult tissue homeostasis. Other studies have focused on BMPs properties, functions and possible involvement in skeletal diseases, including cancer. METHODS A literature search mainly paying attention to the role of BMPs in musculoskeletal tumors was performed in electronic databases. RESULTS This article discusses BMPs synthesis and signaling, and summarizes their prominent roles in the skeletal system for the differentiation of osteoblasts, osteocytes and chondrocytes. CONCLUSIONS The review emphasizes on the role of BMP signaling in the initiation and progression of musculoskeletal cancer.
Collapse
|
14
|
Abstract
STUDY DESIGN Literature review. OBJECTIVE To evaluate the association between recombinant human bone morphogenetic protein-2 (rhBMP-2) and malignancy. SUMMARY OF BACKGROUND DATA The use of rhBMP-2 in spine surgery has been the topic of much debate as studies assessing the association between rhBMP-2 and malignancy have come to conflicting conclusions. METHODS A systematic review of the literature was performed using the PubMed-National Library of Medicine/National Institute of Health databases. Only non-clinical studies directly addressing BMP-2 and cancer were included. Articles were categorized by study type (animal, in vitro cell line/human/animal), primary malignancy, cancer attributes, and whether BMP-2 was pro-malignancy or not. RESULTS A total of 4,131 articles were reviewed. Of those, 515 articles made reference to both BMP-2 and cancer, 99 of which were found to directly examine the role of BMP-2 in cancer. Seventy-five studies were in vitro and 24 were animal studies. Forty-three studies concluded that BMP-2 enhanced cancer function, whereas 18 studies found that BMP-2 suppressed malignancy. Thirty-six studies did not examine whether BMP-2 enhanced or suppressed cancer function. Fifteen studies demonstrated BMP-2 dose dependence (9 enhancement, 6 suppression) and one study demonstrated no dose dependence. Nine studies demonstrated BMP-2 time dependence (6 enhancement, 3 suppression). However, no study demonstrated that BMP-2 caused cancer de novo. CONCLUSION Currently, conflicting data exist with regard to the effect of exogenous BMP-2 on cancer. The majority of studies addressed the role of BMP-2 in prostate (17%), breast (17%), and lung (15%) cancers. Most were in vitro studies (75%) and examined cancer invasiveness and metastatic potential (37%). Of 99 studies, there was no demonstration of BMP-2 causing cancer de novo. However, 43% of studies suggested that BMP-2 enhances tumor function, motivating more definitive research on the topic that also includes clinically meaningful dose- and time-dependence. LEVEL OF EVIDENCE 2.
Collapse
|
15
|
Abstract
Cancer metastasis is highly inefficient and complex. Common features of metastatic cancer cells have been observed using cancer cell lines and genetically reconstituted mouse and human tumor xenograft models. These include cancer cell interaction with the tumor microenvironment and the ability of cancer cells to sense extracellular stimuli and adapt to adverse growth conditions. This review summarizes the coordinated response of cancer cells to soluble growth factors, such as RANKL, by a unique feed forward mechanism employing coordinated upregulation of RANKL and c-Met with downregulation of androgen receptor. The RANK-mediated signal network was found to drive epithelial to mesenchymal transition in prostate cancer cells, promote osteomimicry and the ability of prostate cancer cells to assume stem cell and neuroendocrine phenotypes, and confer the ability of prostate cancer cells to home to bone. Prostate cancer cells with activated RANK-mediated signal network were observed to recruit and even transform the non-tumorigenic prostate cancer cells to participate in bone and soft tissue colonization. The coordinated regulation of cancer cell invasion and metastasis by the feed forward mechanism involving RANKL, c-Met, transcription factors, and VEGF-neuropilin could offer new therapeutic opportunities to target prostate cancer bone and soft tissue metastases.
Collapse
Affiliation(s)
- Gina Chia-Yi Chu
- Departments of Medicine and Surgery, Samuel Orchin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA,
| | | |
Collapse
|
16
|
Wang Z, Ma F, Wang J, Zhou Z, Liu B, He X, Fu L, He W, Cooper PR. Extracellular Signal-regulated Kinase Mitogen-activated Protein Kinase and Phosphatidylinositol 3-Kinase/Akt Signaling Are Required for Lipopolysaccharide-mediated Mineralization in Murine Odontoblast-like Cells. J Endod 2015; 41:871-6. [PMID: 25720983 DOI: 10.1016/j.joen.2015.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/06/2015] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Odontoblasts play an important role in post-developmental control of mineralization in response to external stimuli in the tooth. The present study investigated whether lipopolysaccharide (LPS), a major bacterial cell wall component, influenced mineralization in a murine odontoblast-like cell (OLC) line and the related intracellular signaling pathways involved. METHODS Alizarin red S staining was used to assess mineralized nodule formation in OLCs in response to LPS. The effects of LPS on gene expression of odontoblastic markers were investigated by using quantitative real-time reverse-transcriptase polymerase chain reaction. The potential involvement of toll-like receptor 4 (TLR4), nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK), or phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways in the mineralized nodule formation, and mRNA expression of several odontoblastic markers of OLCs induced by LPS was assessed by using alizarin red S staining and quantitative real-time reverse-transcriptase polymerase chain reaction. Moreover, LPS stimulation resulted in phosphorylation of protein that was determined by Western blot analysis. RESULTS OLCs showed reduced mineralized nodule formation and several odontoblastic markers expression in response to LPS exposure. Furthermore, inhibition of TLR4, extracellular signal-regulated kinase (ERK), and PI3K/Akt signaling noticeably antagonized LPS-mediated mineralization in OLCs. However, p38 MAPK, c-Jun N-terminal kinase, and NF-κB signaling inhibitors did not affect LPS-mediated mineralization in OLCs. Notably, LPS treatment resulted in a time-dependent phosphorylation of ERK and PI3K/Akt in OLCs, which was abrogated by their specific inhibitors. CONCLUSIONS LPS decreased mineralization in OLCs via TLR4, ERK MAPK, and PI3K/Akt signaling pathways, but not p38, c-Jun N-terminal kinase, or NF-κB signaling.
Collapse
Affiliation(s)
- Zhihua Wang
- State Key Laboratory of Military Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Fengle Ma
- State Key Laboratory of Military Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Juan Wang
- State Key Laboratory of Military Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Zeyuan Zhou
- State Key Laboratory of Military Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Baogang Liu
- Department of Stomatology, Lishilu Outpatient Department, Chinese PLA Second Artillery Corps, Beijing, PR China
| | - Xinyao He
- State Key Laboratory of Military Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Lei Fu
- State Key Laboratory of Military Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; Department of Stomatology, NingXia People's Hospital, NingXia, Yinchuan, PR China
| | - Wenxi He
- State Key Laboratory of Military Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Fourth Military Medical University, Xi'an, PR China.
| | - Paul R Cooper
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham, West Midlands, United Kingdom
| |
Collapse
|
17
|
Abd Elmageed ZY, Yang Y, Thomas R, Ranjan M, Mondal D, Moroz K, Fang Z, Rezk BM, Moparty K, Sikka SC, Sartor O, Abdel-Mageed AB. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells 2015; 32:983-97. [PMID: 24715691 DOI: 10.1002/stem.1619] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 11/19/2013] [Accepted: 11/21/2013] [Indexed: 12/19/2022]
Abstract
Emerging evidence suggests that mesenchymal stem cells (MSCs) are often recruited to tumor sites but their functional significance in tumor growth and disease progression remains elusive. Herein we report that prostate cancer (PC) cell microenvironment subverts PC patient adipose-derived stem cells (pASCs) to undergo neoplastic transformation. Unlike normal ASCs, the pASCs primed with PC cell conditioned media (CM) formed prostate-like neoplastic lesions in vivo and reproduced aggressive tumors in secondary recipients. The pASC tumors acquired cytogenetic aberrations and mesenchymal-to-epithelial transition and expressed epithelial, neoplastic, and vasculogenic markers reminiscent of molecular features of PC tumor xenografts. Our mechanistic studies revealed that PC cell-derived exosomes are sufficient to recapitulate formation of prostate tumorigenic mimicry generated by CM-primed pASCs in vivo. In addition to downregulation of the large tumor suppressor homolog2 and the programmed cell death protein 4, a neoplastic transformation inhibitor, the tumorigenic reprogramming of pASCs was associated with trafficking by PC cell-derived exosomes of oncogenic factors, including H-ras and K-ras transcripts, oncomiRNAs miR-125b, miR-130b, and miR-155 as well as the Ras superfamily of GTPases Rab1a, Rab1b, and Rab11a. Our findings implicate a new role for PC cell-derived exosomes in clonal expansion of tumors through neoplastic reprogramming of tumor tropic ASCs in cancer patients.
Collapse
Affiliation(s)
- Zakaria Y Abd Elmageed
- Department of Urology, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Bone is the most common site for metastasis in human prostate cancer patients. Skeletal metastases are a significant cause of morbidity and mortality and overall greatly affect the quality of life of prostate cancer patients. Despite advances in our understanding of the biology of primary prostate tumors, our knowledge of how and why secondary tumors derived from prostate cancer cells preferentially localize bone remains limited. The physiochemical properties of bone, and signaling molecules including specific chemokines and their receptors, are distinct in nature and function, yet play intricate and significant roles in prostate cancer bone metastasis. Examining the impact of these facets of bone metastasis in vivo remains a significant challenge, as animal models that mimic the natural history and malignant progression clinical prostate cancer are rare. The goals of this article are to discuss (1) characteristics of bone that most likely render it a favorable environment for prostate tumor cell growth, (2) chemokine signaling that is critical in the recruitment and migration of prostate cancer cells to the bone, and (3) current animal models utilized in studying prostate cancer bone metastasis. Further research is necessary to elucidate the mechanisms underlying the extravasation of disseminated prostate cancer cells into the bone and to provide a better understanding of the basis of cancer cell survival within the bone microenvironment. The development of animal models that recapitulate more closely the human clinical scenario of prostate cancer will greatly benefit the generation of better therapies.
Collapse
|
19
|
Cox RF, Morgan MP. Microcalcifications in breast cancer: Lessons from physiological mineralization. Bone 2013; 53:437-50. [PMID: 23334083 DOI: 10.1016/j.bone.2013.01.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 02/02/2023]
Abstract
Mammographic mammary microcalcifications are routinely used for the early detection of breast cancer, however the mechanisms by which they form remain unclear. Two species of mammary microcalcifications have been identified; calcium oxalate and hydroxyapatite. Calcium oxalate is mostly associated with benign lesions of the breast, whereas hydroxyapatite is associated with both benign and malignant tumors. The way in which hydroxyapatite forms within mammary tissue remains largely unexplored, however lessons can be learned from the process of physiological mineralization. Normal physiological mineralization by osteoblasts results in hydroxyapatite deposition in bone. This review brings together existing knowledge from the field of physiological mineralization and juxtaposes it with our current understanding of the genesis of mammary microcalcifications. As an increasing number of breast cancers are being detected in their non-palpable stage through mammographic microcalcifications, it is important that future studies investigate the underlying mechanisms of their formation in order to fully understand the significance of this unique early marker of breast cancer.
Collapse
Affiliation(s)
- Rachel F Cox
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | | |
Collapse
|
20
|
Paradoxical roles of tumour necrosis factor-alpha in prostate cancer biology. Prostate Cancer 2012; 2012:128965. [PMID: 23326670 PMCID: PMC3543804 DOI: 10.1155/2012/128965] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 11/19/2012] [Indexed: 01/08/2023] Open
Abstract
Tumour necrosis factor (TNF) is a pleiotropic cytokine with dual roles in cancer biology including prostate cancer (PCa). On the one hand, there is evidence that it stimulates tumour angiogenesis, is involved in the initiation of PCa from an androgen-dependent to a castrate resistant state, plays a role in epithelial to mesenchymal plasticity, and may contribute to the aberrant regulation of eicosanoid pathways. On the other hand, TNF has also been reported to inhibit neovascularisation, induce apoptosis of PCa cells, and stimulate antitumour immunity. Much of the confusion surrounding its seemingly paradoxical roles in cancer biology stems from the dependence of its effects on the biological model within which TNF is investigated. This paper will address some of these issues and also discuss the therapeutic implications.
Collapse
|
21
|
Hernandez LL, Gregerson KA, Horseman ND. Mammary gland serotonin regulates parathyroid hormone-related protein and other bone-related signals. Am J Physiol Endocrinol Metab 2012; 302:E1009-15. [PMID: 22318950 PMCID: PMC3774078 DOI: 10.1152/ajpendo.00666.2011] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cells drive bone demineralization during lactation and metastatic cancers. A shared mechanism among these physiological and pathological states is endocrine secretion of parathyroid hormone-related protein (PTHrP), which acts through osteoblasts to stimulate osteoclastic bone demineralization. The regulation of PTHrP has not been accounted for fully by any conventional mammotropic stimuli or tumor growth factors. Serotonin (5-HT) synthesis within breast epithelial cells is induced during lactation and in advancing breast cancer. Here we report that serotonin deficiency (knockout of tryptophan hydroxylase-1) results in a reduction of mammary PTHrP expression during lactation, which is rescued by restoring 5-HT synthesis. 5-HT induced PTHrP expression in lactogen-primed mammary epithelial cells from either mouse or cow. In human breast cancer cells 5-HT induced both PTHrP and the metastasis-associated transcription factor Runx2/Cbfa1. Based on receptor expression and pharmacological evidence, the 5-HT2 receptor type was implicated as being critical for induction of PTHrP and Runx2. These results connect 5-HT synthesis to the induction of bone-regulating factors in the normal mammary gland and in breast cancer cells.
Collapse
Affiliation(s)
- Laura L Hernandez
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| | | | | |
Collapse
|
22
|
Abstract
Bone morphogenetic protein (BMP) signaling in diseases is the subject of an overwhelming array of studies. BMPs are excellent targets for treatment of various clinical disorders. Several BMPs have already been shown to be clinically beneficial in the treatment of a variety of conditions, including BMP-2 and BMP-7 that have been approved for clinical application in nonunion bone fractures and spinal fusions. With the use of BMPs increasingly accepted in spinal fusion surgeries, other therapeutic approaches targeting BMP signaling are emerging beyond applications to skeletal disorders. These approaches can further utilize next-generation therapeutic tools such as engineered BMPs and ex vivo- conditioned cell therapies. In this review, we focused to provide insights into such clinical potentials of BMPs in metabolic and vascular diseases, and in cancer. [BMB reports 2011; 44(10): 619-634].
Collapse
Affiliation(s)
- Meejung Kim
- Joint Center for Biosciences at Lee Gil Ya Cancer and Diabetes Research Institute, Gachon University of Medicine and Science, IncheonKorea
| | | |
Collapse
|
23
|
Li Y, Kong D, Ahmad A, Bao B, Sarkar FH. Targeting bone remodeling by isoflavone and 3,3'-diindolylmethane in the context of prostate cancer bone metastasis. PLoS One 2012; 7:e33011. [PMID: 22412975 PMCID: PMC3296768 DOI: 10.1371/journal.pone.0033011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 02/02/2012] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer (PCa) bone metastases have long been believed to be osteoblastic because of bone remodeling leading to the formation of new bone. However, recent studies have shown increased osteolytic activity in the beginning stages of PCa bone metastases, suggesting that targeting both osteolytic and osteoblastic mediators would likely inhibit bone remodeling and PCa bone metastasis. In this study, we found that PCa cells could stimulate differentiation of osteoclasts and osteoblasts through the up-regulation of RANKL, RUNX2 and osteopontin, promoting bone remodeling. Interestingly, we found that formulated isoflavone and 3,3′-diindolylmethane (BR-DIM) were able to inhibit the differentiation of osteoclasts and osteoblasts through the inhibition of cell signal transduction in RANKL, osteoblastic, and PCa cell signaling. Moreover, we found that isoflavone and BR-DIM down-regulated the expression of miR-92a, which is known to be associated with RANKL signaling, EMT and cancer progression. By pathway and network analysis, we also observed the regulatory effects of isoflavone and BR-DIM on multiple signaling pathways such as AR/PSA, NKX3-1/Akt/p27, MITF, etc. Therefore, isoflavone and BR-DIM with their multi-targeted effects could be useful for the prevention of PCa progression, especially by attenuating bone metastasis mechanisms.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Dejuan Kong
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Aamir Ahmad
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Bin Bao
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Fazlul H. Sarkar
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
24
|
Morgenroth VH, Hache LP, Clemens PR. Insights into bone health in Duchenne muscular dystrophy. BONEKEY REPORTS 2012; 1:9. [PMID: 23951421 DOI: 10.1038/bonekey.2012.5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Accepted: 11/05/2011] [Indexed: 12/22/2022]
Abstract
Poor bone health is a significant problem for patients with Duchenne muscular dystrophy (DMD), a progressive, disabling disease. Although the primary focus of DMD disease pathogenesis is degeneration of striated muscle, impairment of bone health likely has a role in the disease that has only been superficially examined to date. Deficiency of bone mineral density and increased incidence of bone fractures are well-recognized clinical components of the DMD phenotype. Furthermore, therapy with corticosteroids, an approved treatment for DMD that prolongs ambulation, may have multiple effects on bone health in DMD patients. This review examines the evidence in preclinical models and in human DMD disease that provides insight into the role performed by bone in the disease pathogenesis and phenotype of DMD. The information reviewed here points toward the need for mechanistic and therapeutic studies to optimize bone health in DMD patients.
Collapse
|
25
|
Yang W, Levine AC. Androgens and prostate cancer bone metastases: effects on both the seed and the soil. Endocrinol Metab Clin North Am 2011; 40:643-53, x. [PMID: 21889726 DOI: 10.1016/j.ecl.2011.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Androgens are essential for normal prostate development and are necessary, but not sufficient, for the development of prostate cancer (PCa). Androgen deprivation therapy has long been the mainstay of treatment for PCa bone metastases, providing palliation of symptoms in the majority of patients, followed by relapse and progression. The majority of published preclinical studies demonstrate a stimulatory effect of androgens and androgen receptor signaling on the multistep process of PCa bone metastases, including androgenic promotion of local PCa growth, angiogenesis, invasion, bone targeting, stimulation of PCa growth factors that enhance osteoclastogenesis, and enhancement of Wnt signaling in osteoblasts.
Collapse
Affiliation(s)
- Wei Yang
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
26
|
Li Y, Su J, DingZhang X, Zhang J, Yoshimoto M, Liu S, Bijian K, Gupta A, Squire JA, Alaoui Jamali MA, Bismar TA. PTEN deletion and heme oxygenase-1 overexpression cooperate in prostate cancer progression and are associated with adverse clinical outcome. J Pathol 2011; 224:90-100. [PMID: 21381033 DOI: 10.1002/path.2855] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 12/23/2010] [Accepted: 01/04/2011] [Indexed: 12/29/2022]
Abstract
Overexpression of the pro-survival protein heme oxygenase-1 (HO-1) and loss of the pro-apoptotic tumour suppressor PTEN are common events in prostate cancer (PCA). We assessed the occurrence of both HO-1 expression and PTEN deletion in two cohorts of men with localized and castration-resistant prostate cancer (CRPC). The phenotypic cooperation of these markers was examined in preclinical and clinical models. Overall, there was a statistically significant difference in HO-1 epithelial expression between benign, high-grade prostatic intraepithelial neoplasia (HGPIN), localized PCA, and CRPC (p < 0.0001). The highest epithelial HO-1 expression was noted in CRPC (2.00 ± 0.89), followed by benign prostate tissue (1.49 ± 1.03) (p = 0.0003), localized PCA (1.20 ± 0.95), and HGPIN (1.07 ± 0.87) (p < 0.0001). However, the difference between HGPIN and PCA was not statistically significant (p = 0.21). PTEN deletions were observed in 35/55 (63.6%) versus 68/183 (37.1%) cases of CRPC and localized PCA, respectively. Although neither HO-1 overexpression nor PTEN deletions alone in localized PCA showed a statistically significant association with PSA relapse, the combined status of both markers correlated with disease progression (log-rank test, p = 0.01). In a preclinical model, inhibition of HO-1 by shRNA in PTEN-deficient PC3M cell line and their matched cells where PTEN is restored strongly reduced cell growth and invasion in vitro and inhibited tumour growth and lung metastasis formation in mice compared to cells where only HO-1 is inhibited or PTEN is restored. In summary, we provide clinical and experimental evidence for cooperation between epithelial HO-1 expression and PTEN deletions in relation to the PCA patient's outcome. These findings could potentially lead to the discovery of novel therapeutic modalities for advanced PCA.
Collapse
Affiliation(s)
- Yunru Li
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zolochevska O, Figueiredo ML. Cell-cycle regulators cdk2ap1 and bicalutamide suppress malignant biological interactions between prostate cancer and bone cells. Prostate 2011; 71:353-67. [PMID: 20812223 DOI: 10.1002/pros.21249] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 07/22/2010] [Indexed: 11/06/2022]
Abstract
INTRODUCTION We examined whether the novel cell-cycle regulator cdk2-associated protein 1 (p12(cdk2ap1) or cdk2ap1), recently shown to regulate prostate cancer cell cycle and apoptosis, could have the capacity to reduce invasiveness and/or reduce malignant biological interactions between prostate cancer and bone cells. We also examined whether combining two cell-cycle arrest stimuli, cdk2ap1 plus bicalutamide (or casodex, CDX), could help enhance inhibition of prostate cancer cell phenotypes. METHODS We stably expressed cdk2ap1 in prostate cancer cell lines using lentiviral vectors, as well as several different co-culture assays to quantify cellular invasion, migration, and the effect of the treatments on interaction with the bone microenvironment. RESULTS We have determined that cdk2ap1 can further augment the effects of CDX on cell-cycle arrest, growth inhibition, and cellular invasion. Using a coculture model, we observed that either cdk2ap1 or cdk2ap1/CDX combination were able to reduce chemotaxis towards osteoblasts, and also reduce the osteoblastic proliferative response to prostate cancer. Also modified by cdk2ap1 and CDX were several signaling pathways associated with prostate cancer/bone crosstalk mechanisms involved in prostate cancer progression. CONCLUSIONS These results suggest that either cdk2ap1 or the cdk2ap1/CDX combination hold promise in regulating prostate cancer growth and malignant phenotypes, and potentially also in reducing procarcinogenic interactions with a bone microenvironment model, restoring malignant phenotypes and signaling to a more benign state.
Collapse
Affiliation(s)
- Olga Zolochevska
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | | |
Collapse
|
28
|
Singh A, Morris RJ. The Yin and Yang of bone morphogenetic proteins in cancer. Cytokine Growth Factor Rev 2010; 21:299-313. [PMID: 20688557 DOI: 10.1016/j.cytogfr.2010.06.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 06/21/2010] [Accepted: 06/28/2010] [Indexed: 01/22/2023]
Abstract
Bone morphogenetic proteins (BMPs) were first studied as growth factors or morphogens of the transforming growth factor-beta superfamily. These growth molecules, originally associated with bone and cartilage development, are now known to play an important role in morphogenesis and homeostasis in many other tissues. More recently, significant contributions from BMPs, their receptors, and interacting molecules have been linked to carcinogenesis and tumor progression. On the other hand, BMPs can sometimes function as a tumor suppressor. Our report highlights these new roles in the pathogenesis of cancer that may suggest novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ashok Singh
- Laboratory of Stem Cells and Cancer, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | | |
Collapse
|