1
|
Hughes AM, Kolb AD, Shupp AB, Shine KM, Bussard KM. Printing the Pathway Forward in Bone Metastatic Cancer Research: Applications of 3D Engineered Models and Bioprinted Scaffolds to Recapitulate the Bone-Tumor Niche. Cancers (Basel) 2021; 13:507. [PMID: 33572757 PMCID: PMC7865550 DOI: 10.3390/cancers13030507] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
Breast cancer commonly metastasizes to bone, resulting in osteolytic lesions and poor patient quality of life. The bone extracellular matrix (ECM) plays a critical role in cancer cell metastasis by means of the physical and biochemical cues it provides to support cellular crosstalk. Current two-dimensional in-vitro models lack the spatial and biochemical complexities of the native ECM and do not fully recapitulate crosstalk that occurs between the tumor and endogenous stromal cells. Engineered models such as bone-on-a-chip, extramedullary bone, and bioreactors are presently used to model cellular crosstalk and bone-tumor cell interactions, but fall short of providing a bone-biomimetic microenvironment. Three-dimensional bioprinting allows for the deposition of biocompatible materials and living cells in complex architectures, as well as provides a means to better replicate biological tissue niches in-vitro. In cancer research specifically, 3D constructs have been instrumental in seminal work modeling cancer cell dissemination to bone and bone-tumor cell crosstalk in the skeleton. Furthermore, the use of biocompatible materials, such as hydroxyapatite, allows for printing of bone-like microenvironments with the ability to be implanted and studied in in-vivo animal models. Moreover, the use of bioprinted models could drive the development of novel cancer therapies and drug delivery vehicles.
Collapse
Affiliation(s)
- Anne M. Hughes
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA;
| | - Alexus D. Kolb
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.D.K.); (A.B.S.)
| | - Alison B. Shupp
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.D.K.); (A.B.S.)
| | - Kristy M. Shine
- Health Design Lab, Jefferson Bioprinting Lab, Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Karen M. Bussard
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.D.K.); (A.B.S.)
| |
Collapse
|
2
|
Wang L, Fang D, Xu J, Luo R. Various pathways of zoledronic acid against osteoclasts and bone cancer metastasis: a brief review. BMC Cancer 2020; 20:1059. [PMID: 33143662 PMCID: PMC7607850 DOI: 10.1186/s12885-020-07568-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Zoledronic acid (ZA) is one of the most important and effective class of anti-resorptive drug available among bisphosphonate (BP), which could effectively reduce the risk of skeletal-related events, and lead to a treatment paradigm for patients with skeletal involvement from advanced cancers. However, the exact molecular mechanisms of its anticancer effects have only recently been identified. In this review, we elaborate the detail mechanisms of ZA through inhibiting osteoclasts and cancer cells, which include the inhibition of differentiation of osteoclasts via suppressing receptor activator of nuclear factor κB ligand (RANKL)/receptor activator of nuclear factor κB (RANK) pathway, non-canonical Wnt/Ca2+/calmodulin dependent protein kinase II (CaMKII) pathway, and preventing of macrophage differentiation into osteoclasts, in addition, induction of apoptosis of osteoclasts through inhibiting farnesyl pyrophosphate synthase (FPPS)-mediated mevalonate pathway, and activation of reactive oxygen species (ROS)-induced pathway. Furthermore, ZA also inhibits cancer cells proliferation, viability, motility, invasion and angiogenesis; induces cancer cell apoptosis; reverts chemoresistance and stimulates immune response; and acts in synergy with other anti-cancer drugs. In addition, some new ways for delivering ZA against cancer is introduced. We hope this review will provide more information in support of future studies of ZA in the treatment of cancers and bone cancer metastasis.
Collapse
Affiliation(s)
- Lianwei Wang
- Department of General Surgery, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Dengyang Fang
- Department of General Surgery, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Jinming Xu
- Department of General Surgery, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Runlan Luo
- Department of Ultrasound, Fuling Central Hospital of Chongqing City, Chongqing, 408300, China.
| |
Collapse
|
3
|
Zhang GL, Gutter-Kapon L, Ilan N, Batool T, Singh K, Digre A, Luo Z, Sandler S, Shaked Y, Sanderson RD, Wang XM, Li JP, Vlodavsky I. Significance of host heparanase in promoting tumor growth and metastasis. Matrix Biol 2020; 93:25-42. [PMID: 32534153 PMCID: PMC7704762 DOI: 10.1016/j.matbio.2020.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 02/08/2023]
Abstract
Heparanase, the sole heparan sulfate degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, angiogenesis and metastasis. Much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth and metastasis. We have utilized mice over-expressing (Hpa-tg) heparanase to reveal the role of host heparanase in tumor initiation, growth and metastasis. While in wild type mice tumor development in response to DMBA carcinogenesis was restricted to the mammary gland, Hpa-tg mice developed tumors also in their lungs and liver, associating with reduced survival of the tumor-bearing mice. Consistently, xenograft tumors (lymphoma, melanoma, lung carcinoma, pancreatic carcinoma) transplanted in Hpa-tg mice exhibited accelerated tumor growth and shorter survival of the tumor-bearing mice compared with wild type mice. Hpa-tg mice were also more prone to the development of metastases following intravenous or subcutaneous injection of tumor cells. In some models, the growth advantage was associated with infiltration of heparanase-high host cells into the tumors. However, in other models, heparanase-high host cells were not detected in the primary tumor, implying that the growth advantage in Hpa-tg mice is due to systemic factors. Indeed, we found that plasma from Hpa-tg mice enhanced tumor cell migration and invasion attributed to increased levels of pro-tumorigenic factors (i.e., RANKL, SPARC, MIP-2) in the plasma of Hpa-Tg vs. wild type mice. Furthermore, tumor aggressiveness and short survival time were demonstrated in wild type mice transplanted with bone marrow derived from Hpa-tg but not wild type mice. These results were attributed, among other factors, to upregulation of pro-tumorigenic (i.e., IL35+) and downregulation of anti-tumorigenic (i.e., IFN-γ+) T-cell subpopulations in the spleen, lymph nodes and blood of Hpa-tg vs. wild type mice and their increased infiltration into the primary tumor. Collectively, our results emphasize the significance of host heparanase in mediating the pro-tumorigenic and pro-metastatic interactions between the tumor cells and the host tumor microenvironment, immune cells and systemic factors.
Collapse
Affiliation(s)
- Gan-Lin Zhang
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden
| | - Lilach Gutter-Kapon
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, P. O. Box 9649, Technion, Haifa 31096, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, P. O. Box 9649, Technion, Haifa 31096, Israel
| | - Tahira Batool
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden
| | - Kailash Singh
- Department of Cell Biology, University of Uppsala, Uppsala, Sweden
| | - Andreas Digre
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden
| | - Zhengkang Luo
- Department of Cell Biology, University of Uppsala, Uppsala, Sweden
| | - Stellan Sandler
- Department of Cell Biology, University of Uppsala, Uppsala, Sweden
| | - Yuval Shaked
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, P. O. Box 9649, Technion, Haifa 31096, Israel
| | - Ralph D Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xiao-Min Wang
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Jin-Ping Li
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden.
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, P. O. Box 9649, Technion, Haifa 31096, Israel.
| |
Collapse
|
4
|
Nordin N, Yeap SK, Rahman HS, Zamberi NR, Mohamad NE, Abu N, Masarudin MJ, Abdullah R, Alitheen NB. Antitumor and Anti-Metastatic Effects of Citral-Loaded Nanostructured Lipid Carrier in 4T1-Induced Breast Cancer Mouse Model. Molecules 2020; 25:molecules25112670. [PMID: 32526880 PMCID: PMC7321383 DOI: 10.3390/molecules25112670] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/05/2020] [Accepted: 04/11/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer nano-therapy has been progressing rapidly with the introduction of many novel drug delivery systems. The previous study has reported on the in vitro cytotoxicity of citral-loaded nanostructured lipid carrier (NLC-Citral) on MDA-MB-231 cells and some preliminary in vivo antitumor effects on 4T1 breast cancer cells challenged mice. However, the in vivo apoptosis induction and anti-metastatic effects of NLC-Citral have yet to be reported. In this study, the in vitro cytotoxic, anti-migration, and anti-invasion effects of NLC-Citral were tested on 4T1 breast cancer cells. In addition, the in vivo antitumor effects of oral delivery of NLC-Citral was also evaluated on BALB/c mice induced with 4T1 cells. In vitro cytotoxicity results showed that NLC-Citral and citral gave similar IC50 values on 4T1 cells. However, wound healing, migration, and invasion assays reflected better in vitro anti-metastasis potential for NLC-Citral than citral alone. Results from the in vivo study indicated that both NLC-Citral and citral have anti-tumor and anti-metastasis effects, whereby the NLC-Citral showed better efficacy than citral in all experiments. Also, the delay of tumor progression was through the suppression of the c-myc gene expression and induction of apoptosis in the tumor. In addition, the inhibition of metastasis of 4T1 cells to lung and bone marrow by the NLC-Citral and citral treatments was correlated with the downregulation of metastasis-related genes expression including MMP-9, ICAM, iNOS, and NF-kB and the angiogenesis-related proteins including G-CSF alpha, Eotaxin, bFGF, VEGF, IL-1alpha, and M-CSF in the tumor. Moreover, NLC-Citral showed greater downregulation of MMP-9, iNOS, ICAM, Eotaxin, bFGF, VEGF, and M-CSF than citral treatment in the 4T1-challenged mice, which may contribute to the better anti-metastatic effect of the encapsulated citral. This study suggests that NLC is a potential and effective delivery system for citral to target triple-negative breast cancer.
Collapse
Affiliation(s)
- Noraini Nordin
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (N.N.); (H.S.R.); (N.R.Z.); (N.E.M.); (N.A.); (M.J.M.)
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Sepang 43900, Malaysia;
| | - Heshu Sulaiman Rahman
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (N.N.); (H.S.R.); (N.R.Z.); (N.E.M.); (N.A.); (M.J.M.)
- Department of Physiology, College of Medicine, University of Sulaimani, Sulaymaniyah 46001, Kurdistan Region, Iraq
| | - Nur Rizi Zamberi
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (N.N.); (H.S.R.); (N.R.Z.); (N.E.M.); (N.A.); (M.J.M.)
| | - Nurul Elyani Mohamad
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (N.N.); (H.S.R.); (N.R.Z.); (N.E.M.); (N.A.); (M.J.M.)
| | - Nadiah Abu
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (N.N.); (H.S.R.); (N.R.Z.); (N.E.M.); (N.A.); (M.J.M.)
- UKM Medical Centre, UKM Medical Molecular Biology Institute (UMBI), Cheras 56000, Kuala Lumpur, Malaysia
| | - Mas Jaffri Masarudin
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (N.N.); (H.S.R.); (N.R.Z.); (N.E.M.); (N.A.); (M.J.M.)
| | - Rasedee Abdullah
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor 43400, Malaysia;
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (N.N.); (H.S.R.); (N.R.Z.); (N.E.M.); (N.A.); (M.J.M.)
- Institute of Bioscience, Universiti Putra Malaysia, Selangor 43400, Malaysia
- Correspondence: ; Tel.: +60-389467471
| |
Collapse
|
5
|
Breast Cancer Derived Extracellular Vesicles in Bone Metastasis Induction and Their Clinical Implications as Biomarkers. Int J Mol Sci 2020; 21:ijms21103573. [PMID: 32443642 PMCID: PMC7278927 DOI: 10.3390/ijms21103573] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer incidence and mortality are rapidly growing worldwide. The main risk factors for cancer can be associated with aging as well as the growth of the population and socioeconomic condition. Breast cancer, a crucial public health problem, is the second cause of death among women. About 70% of patients with advanced breast cancer have bone metastases. In bone metastasis, cancer cells and osteoclasts form a vicious cycle: cancer cells promote osteoclast differentiation and activation that, in turn, induce cancer cell seeding and proliferation in the bone. Growing evidence shows that extracellular vesicles (EVs) play a key role in carcinogenesis, proliferation, pre-metastatic niche formation, angiogenesis, metastasis, and chemoresistance in several tumors, such as breast, lung, prostate, and liver cancer. Here, we discuss the role of EVs released by breast cancer cells, focusing on bone metastasis induction and their clinical implications as biomarkers.
Collapse
|
6
|
Yang Z, Yue Z, Ma X, Xu Z. Calcium Homeostasis: A Potential Vicious Cycle of Bone Metastasis in Breast Cancers. Front Oncol 2020; 10:293. [PMID: 32211326 PMCID: PMC7076168 DOI: 10.3389/fonc.2020.00293] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/19/2020] [Indexed: 12/12/2022] Open
Abstract
Cancers have been considered as one of the most severe health problems in the world. Efforts to elucidate the cancer progression reveal the importance of bone metastasis for tumor malignancy, one of the leading causes for high mortality rate. Multiple cancers develop bone metastasis, from which breast cancers exhibit the highest rate and have been well-recognized. Numerous cells and environmental factors have been believed to synergistically facilitate bone metastasis in breast cancers, from which breast cancer cells, osteoclasts, osteoblasts, and their produced cytokines have been well-recognized to form a vicious cycle that aggravates tumor malignancy. Except the cytokines or chemokines, calcium ions are another element largely released from bones during bone metastasis that leads to hypercalcemia, however, have not been well-characterized yet in modulation of bone metastasis. Calcium ions act as a type of unique second messenger that exhibits omnipotent functions in numerous cells, including tumor cells, osteoclasts, and osteoblasts. Calcium ions cannot be produced in the cells and are dynamically fluxed among extracellular calcium pools, intracellular calcium storages and cytosolic calcium signals, namely calcium homeostasis, raising a possibility that calcium ions released from bone during bone metastasis would further enhance bone metastasis and aggravate tumor progression via the vicious cycle due to abnormal calcium homeostasis in breast cancer cells, osteoclasts and osteoblasts. TRPs, VGCCs, SOCE, and P2Xs are four major calcium channels/routes mediating extracellular calcium entry and affect calcium homeostasis. Here we will summarize the overall functions of these four calcium channels in breast cancer cells, osteoclasts and osteoblasts, providing evidence of calcium homeostasis as a vicious cycle in modulation of bone metastasis in breast cancers.
Collapse
Affiliation(s)
- Zhengfeng Yang
- Shanghai Institute of Immunology Center for Microbiota & Immune Related Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiying Yue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinrun Ma
- Shanghai Institute of Immunology Center for Microbiota & Immune Related Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyao Xu
- Shanghai Institute of Immunology Center for Microbiota & Immune Related Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
The Effects of TGF-β Signaling on Cancer Cells and Cancer Stem Cells in the Bone Microenvironment. Int J Mol Sci 2019; 20:ijms20205117. [PMID: 31619018 PMCID: PMC6829436 DOI: 10.3390/ijms20205117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/02/2019] [Accepted: 10/14/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) plays a key role in bone metastasis formation; we hypothesized the possible involvement of TGF-β in the induction of cancer stem cells (CSCs) in the bone microenvironment (micro-E), which may be responsible for chemo-resistance. METHODS Mouse mammary tumor cells were implanted under the dorsal skin flap over the calvaria and into a subcutaneous (subQ) lesions in female mice, generating tumors in the bone and subQ micro-Es. After implantation of the tumor cells, mice were treated with a TGF-β R1 kinase inhibitor (R1-Ki). RESULTS Treatment with R1-Ki decreased tumor volume and cell proliferation in the bone micro-E, but not in the subQ micro-E. R1-Ki treatment did not affect the induction of necrosis or apoptosis in either bone or subQ micro-E. The number of cells positive for the CSC markers, SOX2, and CD166 in the bone micro-E, were significantly higher than those in the subQ micro-E. R1-Ki treatment significantly decreased the number of CSC marker positive cells in the bone micro-E but not in the subQ micro-E. TGF-β activation of the MAPK/ERK and AKT pathways was the underlying mechanism of cell proliferation in the bone micro-E. BMP signaling did not play a role in cell proliferation in either micro-E. CONCLUSION Our results indicated that the bone micro-E is a key niche for CSC generation, and TGF-β signaling has important roles in generating CSCs and tumor cell proliferation in the bone micro-E. Therefore, it is critically important to evaluate responses to chemotherapeutic agents on both cancer stem cells and proliferating tumor cells in different tumor microenvironments in vivo.
Collapse
|
8
|
Li G, Ni A, Yu M. Pretumor microenvironment of hepatocellular carcinoma: Cancerization or anticancerization? Gene 2019; 701:46-54. [PMID: 30902783 DOI: 10.1016/j.gene.2019.03.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/11/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tumor microenvironment (TM) has been deeply concerned. However, the pretumor microenvironment (PTM) was poorly understood. The purpose in this study was to explore the possible pathophysiological features of PTM before hepatocellular carcinoma (HCC) appearance. METHODS Mouse livers with no swelling but with tumors present elsewhere in the body after subcutaneous injection of H22 in the fore underarm were considered a PTM, HCC tumors presenting far away from the PTM were regarded as a TM, and the healthy livers of mice without injection of H22 were regarded as a physiological microenvironment (PM). The transcriptomes of samples were generated using RNA-seq and validated using RT-qPCR. RESULTS Overall, 4483 differentially expressed genes (DEGs) were found in the TM compared with the PTM (TM/PTM), but only 194 were altered in the PTM compared with the PM (PTM/PM). Among those 194 DEGs, 104 displayed upregulation and 90 downregulation. Some of these DEGs could promote the ability to resist cancerization or facilitate cancer metastasis, while others indicated liver impairment. The DEGs were involved in 16 relevant pathways. Additionally, the frequency of alternative splicing (AS) in the DEGs in various samples was positively related to the expression of those DEGs. CONCLUSIONS The PTM initiatively armed itself to combat cancerization when its indications appeared although the PTM did not manifest any tissue swelling. However, the cancer cells were negatively influencing immunity to prevent clearance and positively promoting transformation to construct a suitable environment. During transformation by cancer cells, some genes with acquired AS participated in the construction of the PTM. This alteration created an invadable space and an appropriate environment for cancer cells.
Collapse
Affiliation(s)
- Genliang Li
- Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China.
| | - Anni Ni
- Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Mengyao Yu
- Donghai county hospital, Lianyungang 222000, Jiangsu, China
| |
Collapse
|
9
|
CC chemokines are differentially expressed in Breast Cancer and are associated with disparity in overall survival. Sci Rep 2019; 9:4014. [PMID: 30850664 PMCID: PMC6408438 DOI: 10.1038/s41598-019-40514-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/18/2019] [Indexed: 12/21/2022] Open
Abstract
Despite recent advances, breast cancer (BrCa) still affects many women and the impact is disproportional in African Americans (AA) compared to European Americans (EA). Addressing socioeconomic and behavioral status has not been enough to reduce disparity, suggesting contribution of biological differences in BrCa disparity. Our laboratory was first to show involvement of CC chemokines in BrCa. In this study, using ONCOMINE, TCGA, bc-GenExMiner and KMplotter, we examined the association of CC chemokines in BrCa outcomes and disparity. We show over-expression of CCL5, -7, -11, -17, -20, -22 and -25 in BrCa tissues. High mRNA levels of CCL7, -8, -17, -20 and -25 predicted a decrease in overall survival (OS). CCL7 and CCL8 were associated with decreased relapse-free survival. Expression of CCL17 and CCL25 was associated with decreased OS in AA. In EA, CCL8 was associated with decreased OS. Expression of CCL5, -7, -8, -17, -20 and -25 was highest in TNBC. Expression of CCL11 and CCL22 was associated with HER2. CCL7, -8, -17, -20 and -25 were elevated in AAs. In conclusion, our analysis suggests significant association of CC-chemokines in BrCa progression, OS and disparate disease outcome in AA compared to EA patients.
Collapse
|
10
|
Kolb AD, Shupp AB, Mukhopadhyay D, Marini FC, Bussard KM. Osteoblasts are "educated" by crosstalk with metastatic breast cancer cells in the bone tumor microenvironment. Breast Cancer Res 2019; 21:31. [PMID: 30813947 PMCID: PMC6391840 DOI: 10.1186/s13058-019-1117-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/07/2019] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION In a cancer-free environment in the adult, the skeleton continuously undergoes remodeling. Bone-resorbing osteoclasts excavate erosion cavities, and bone-depositing osteoblasts synthesize osteoid matrix that forms new bone, with no net bone gain or loss. When metastatic breast cancer cells invade the bone, this balance is disrupted. Patients with bone metastatic breast cancer frequently suffer from osteolytic bone lesions that elicit severe bone pain and fractures. Bisphosphonate treatments are not curative. Under ideal circumstances, osteoblasts would synthesize new matrix to fill in erosion cavities caused by osteoclasts, but this is not what occurs. Our prior evidence demonstrated that osteoblasts are diverted from laying down bone matrix to producing cytokines that facilitate breast cancer cell maintenance in late-stage disease. Here, we have new evidence to suggest that there are subpopulations of osteoblasts in the tumor niche as evidenced by their protein marker expression that have distinct roles in tumor progression in the bone. METHODS Tumor-bearing tibia of mice was interrogated by immunofluorescent staining for the presence of osteoblasts and alterations in niche protein expression. De-identified tissue from patients with bone metastatic breast cancer was analyzed for osteoblast subpopulations via multi-plex immunofluorescent staining. Effects of breast cancer cells on osteoblasts were recapitulated in vitro by osteoblast exposure to breast cancer-conditioned medium. Triple-negative and estrogen receptor-positive breast cancer proliferation, cell cycle, and p21 expression were assessed upon contact with "educated" osteoblasts. RESULTS A subpopulation of osteoblasts was identified in the bone tumor microenvironment in vivo of both humans and mice with bone metastatic breast cancer that express RUNX2/OCN/OPN but is negative for IL-6 and alpha-smooth muscle actin. These tumor "educated" osteoblasts (EOs) have altered properties compared to "uneducated" osteoblasts and suppress both triple-negative and estrogen receptor-positive breast cancer cell proliferation and increase cancer cell p21 expression. EO effects on breast cancer proliferation were mediated by NOV and decorin. Importantly, the presence of EO cells in the tibia of mice bearing tumors led to increased amounts of alkaline phosphatase and suppressed the expression of inflammatory cytokines in vivo. CONCLUSIONS Our work reveals that there is a subpopulation of osteoblasts in the bone tumor microenvironment that demonstrate a functional role in retarding breast cancer cell growth.
Collapse
Affiliation(s)
- Alexus D. Kolb
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Alison B. Shupp
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Dimpi Mukhopadhyay
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Frank C. Marini
- Comprehensive Cancer Center Wake Forest University and Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC USA
| | - Karen M. Bussard
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| |
Collapse
|
11
|
Kar S, Molla MS, Katti DR, Katti KS. Tissue-engineered nanoclay-based 3D in vitro breast cancer model for studying breast cancer metastasis to bone. J Tissue Eng Regen Med 2019; 13:119-130. [PMID: 30466156 DOI: 10.1002/term.2773] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/07/2018] [Accepted: 11/19/2018] [Indexed: 11/06/2022]
Abstract
Breast cancer (BrCa) preferentially spreads to bone and colonises within the bone marrow to cause bone metastases. To improve the outcome of patients with BrCa bone metastasis, we need to understand better the mechanisms underlying bone metastasis. Researchers have relied heavily upon in vivo xenografts due to limited availability of human bone metastasis samples. A significant limitation of these is that they do not have a human bone microenvironment. To address this issue, we have developed a nanoclay-based 3D in vitro model of BrCa bone metastasis using human mesenchymal stem cells (MSCs) and human BrCa cells mimicking late stage of BrCa pathogenesis at the metastatic site. This 3D model can provide a microenvironment suitable for cell-cell and cell-matrix interactions whilst retaining the behaviour of BrCa cells with different metastasis potential (i.e., highly metastatic MDA-MB-231 and low metastatic MCF-7) as shown by the production of alkaline phosphatase and matrix metalloproteinase-9. The sequential culture of MSCs with MCF-7 exhibited 3D tumouroids formation and also occurrence of mesenchymal to epithelial transition of cancer metastasis as evidenced by gene expression and immunocytochemistry. The unique and distinct behaviour of highly metastatic MDA-MB-231 and the low metastatic MCF-7 was observed at the bone metastasis site. The changes to migratory capabilities and invasiveness in MDA-MB-231 in comparison with tumour growth with MCF-7 was observed. Together, a novel bone-mimetic 3D in vitro BrCa model has been developed that could be used to study mechanisms governing the later stage of cancer pathogenesis in bone.
Collapse
Affiliation(s)
- Sumanta Kar
- Department of Civil and Environmental Engineering, CIE 201, NDSU, Fargo, North Dakota, United States
| | - Md Shahjahan Molla
- Department of Civil and Environmental Engineering, CIE 201, NDSU, Fargo, North Dakota, United States
| | - Dinesh R Katti
- Department of Civil and Environmental Engineering, CIE 201, NDSU, Fargo, North Dakota, United States
| | - Kalpana S Katti
- Department of Civil and Environmental Engineering, CIE 201, NDSU, Fargo, North Dakota, United States
| |
Collapse
|
12
|
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: 89] [Impact Index Per Article: 12.7] [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
|
13
|
Futakuchi M, Nitanda T, Ando S, Matsumoto H, Yoshimoto E, Fukamachi K, Suzui M. Therapeutic and Preventive Effects of Osteoclastogenesis Inhibitory Factor on Osteolysis, Proliferation of Mammary Tumor Cell and Induction of Cancer Stem Cells in the Bone Microenvironment. Int J Mol Sci 2018; 19:ijms19030888. [PMID: 29547583 PMCID: PMC5877749 DOI: 10.3390/ijms19030888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/02/2018] [Accepted: 03/14/2018] [Indexed: 11/24/2022] Open
Abstract
Background: We examined the effects of recombinant human osteoclastogenesis inhibitory factor (hOCIF) on osteolysis, proliferation of mammary tumor cells, and induction of cancer stem cells (CSCs) in the tumor-bone and tumor-subcutaneous microenvironments (TB- and TS-microE). Methods: Mouse mammary tumor cells were transplanted onto the calvaria or into a subcutaneous lesion of female mice, creating a TB-microE and a TS-microE, and the mice were then treated with hOCIF. To investigate the preventive effects of hOCIF, mice were treated with hOCIF before tumor cell implantation onto the calvaria (Pre), after (Post), and both before and after (Whole). The number of CSCs and cytokine levels were evaluated by IHC and ELISA assay, respectively. Results: hOCIF suppressed osteolysis, and growth of mammary tumors in the TB-microE, but not in the TS-microE. In the Pre, Post, and Whole groups, hOCIF suppressed osteolysis, and cell proliferation. hOCIF increased mouse osteoprotegrin (mOPG) levels in vivo, which suppressed mammary tumor cell proliferation in vitro. These preventive effects were observed in the dose-dependent. hOCIF did not affect the induction of CSCs in either microenvironment. Conclusion: While receptor activator of NF-κB ligand (RANKL) targeting therapy may not affect the induction of CSCs, RANKL is a potential target for prevention as well as treatment of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Mitsuru Futakuchi
- Department of Pathology, Nagasaki University Hospital, Nagasaki 851-8501, Japan.
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.
- Department of Molecular Toxicology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Takao Nitanda
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.
| | - Saeko Ando
- Department of Molecular Toxicology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Harutoshi Matsumoto
- Department of Molecular Toxicology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Eri Yoshimoto
- Department of Molecular Toxicology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Katsumi Fukamachi
- Department of Molecular Toxicology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Masumi Suzui
- Department of Molecular Toxicology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| |
Collapse
|
14
|
Salamanna F, Borsari V, Brogini S, Giavaresi G, Parrilli A, Cepollaro S, Cadossi M, Martini L, Mazzotti A, Fini M. An in vitro 3D bone metastasis model by using a human bone tissue culture and human sex-related cancer cells. Oncotarget 2018; 7:76966-76983. [PMID: 27765913 PMCID: PMC5363563 DOI: 10.18632/oncotarget.12763] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 09/27/2016] [Indexed: 01/08/2023] Open
Abstract
One of the main limitations, when studying cancer-bone metastasis, is the complex nature of the native bone environment and the lack of reliable, simple, inexpensive models that closely mimic the biological processes occurring in patients and allowing the correct translation of results. To enhance the understanding of the mechanisms underlying human bone metastases and in order to find new therapies, we developed an in vitro three-dimensional (3D) cancer-bone metastasis model by culturing human breast or prostate cancer cells with human bone tissue isolated from female and male patients, respectively. Bone tissue discarded from total hip replacement surgery was cultured in a rolling apparatus system in a normoxic or hypoxic environment. Gene expression profile, protein levels, histological, immunohistochemical and four-dimensional (4D) micro-CT analyses showed a noticeable specificity of breast and prostate cancer cells for bone colonization and ingrowth, thus highlighting the species-specific and sex-specific osteotropism and the need to widen the current knowledge on cancer-bone metastasis spread in human bone tissues. The results of this study support the application of this model in preclinical studies on bone metastases and also follow the 3R principles, the guiding principles, aimed at replacing/reducing/refining (3R) animal use and their suffering for scientific purposes.
Collapse
Affiliation(s)
- Francesca Salamanna
- Laboratory of Biocompatibility, Technological Innovation and Advanced Therapy, Rizzoli RIT, Rizzoli Orthopedic Institute, Bologna, Italy.,Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Veronica Borsari
- Laboratory of Biocompatibility, Technological Innovation and Advanced Therapy, Rizzoli RIT, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Silvia Brogini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Gianluca Giavaresi
- Laboratory of Biocompatibility, Technological Innovation and Advanced Therapy, Rizzoli RIT, Rizzoli Orthopedic Institute, Bologna, Italy.,Laboratory of Tissue Engineering-Innovative Technology Platforms for Tissue Engineering, Rizzoli Orthopedic Institute, Palermo, Italy
| | - Annapaola Parrilli
- Laboratory of Biocompatibility, Technological Innovation and Advanced Therapy, Rizzoli RIT, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Simona Cepollaro
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Matteo Cadossi
- I Orthopaedics and Trauma Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy.,University of Bologna, Bologna, Italy
| | - Lucia Martini
- Laboratory of Biocompatibility, Technological Innovation and Advanced Therapy, Rizzoli RIT, Rizzoli Orthopedic Institute, Bologna, Italy.,Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Antonio Mazzotti
- I Orthopaedics and Trauma Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy.,University of Bologna, Bologna, Italy
| | - Milena Fini
- Laboratory of Biocompatibility, Technological Innovation and Advanced Therapy, Rizzoli RIT, Rizzoli Orthopedic Institute, Bologna, Italy.,Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|
15
|
Quantitative contrast-enhanced CT attenuation evaluation of osseous metastases following chemotherapy. Skeletal Radiol 2017; 46:1385-1395. [PMID: 28667362 DOI: 10.1007/s00256-017-2706-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/14/2017] [Accepted: 06/19/2017] [Indexed: 02/02/2023]
Abstract
PURPOSE Osseous metastases often undergo an osteoblastic healing response following chemotherapy. The purpose of our study was to demonstrate the quantitative CT changes in attenuation of osseous metastases before and after chemotherapy. MATERIALS AND METHODS Our study was IRB approved and HIPAA compliant. Our cohort consisted of 86 consecutive cancer patients with contrast-enhanced CTs before and 14 ± 2 (12-25) months after initiation of chemotherapy (60 ± 11 years, 36 males, 50 females). The average and maximum metastasis attenuations were measured in Hounsfield units (HU) by two readers. Treatment effects were assessed using paired t-tests and Fisher exact tests. Intraclass correlation coefficients (ICCs) were calculated. Patient records were reviewed to determine the patient's clinical status (worse, unchanged, or improved) at the time of follow-up CT. RESULTS The distribution of lesion types was as follows: lytic (30/86, 35%), blastic (43/86, 50%), and mixed lytic-blastic (13/86, 15%). There was a significant increase in average and maximum CT attenuation of metastases following chemotherapy for all patients, which remained statistically significant when stratified by lesion type, clinical status (worsening or improving/stable), cancer type (breast, lung), and radiation therapy (P < 0.05). In a subgroup of patients whose osseous metastases decreased in average attenuation (14/86, 16%), more patients had a worse clinical status (11/14, 79%) (P = 0.02). ICC was almost perfect for average attenuation and substantial for maximum attenuation. CONCLUSION Quantitative assessment of osseous metastatic disease using CT attenuation measurements demonstrated a statistically significant increase in attenuation more than 12 months after initiation of chemotherapy.
Collapse
|
16
|
Sun Y, Wu YX, Zhang P, Peng G, Yu SY. Anti-rheumatic drug iguratimod protects against cancer-induced bone pain and bone destruction in a rat model. Oncol Lett 2017; 13:4849-4856. [PMID: 28588731 PMCID: PMC5452928 DOI: 10.3892/ol.2017.6045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/26/2017] [Indexed: 01/14/2023] Open
Abstract
The bone is one of the most common sites of metastasis in patients with cancer. Current treatments for bone metastases include bisphosphonates, denosumab, non-steroidal anti-inflammatory drugs and analgesics, but each of them has certain limitations. Cytokines and mediators released from various cells in the bone microenvironment may drive a vicious cycle of osteolytic bone metastases. Iguratimod (T-614), a novel disease-modifying anti-rheumatic drug, has demonstrated therapeutic effects by suppressing the production of inflammatory cytokines in rats and patients with rheumatoid arthritis. Therefore, the current study evaluated the hypothesis that iguratimod may protect against cancer-induced bone pain and bone metastasis in a rat model. For this purpose, rats inoculated with Walker 256 cells were treated with iguratimod from days 11–17 post-surgery. Mechanical paw withdrawal thresholds and expression levels of phosphorylated extracellular signal-related kinase (pERK) and c-Fos in the spinal cord were investigated to detect changes in bone pain. Bone destruction levels were detected using X-rays, hematoxylin and eosin and tartrate-resistant acid phosphatase staining. The results revealed that mechanical paw withdrawal thresholds and the expression levels of pERK and c-Fos declined in a dose-dependent manner in rats treated with iguratimod, and bone destruction severity was also reduced. These findings may provide important new insights into the treatment of bone metastasis symptoms.
Collapse
Affiliation(s)
- Yue Sun
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ying-Xing Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Peng Zhang
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Guang Peng
- Department of Clinical Cancer Prevention, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Shi-Ying Yu
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
17
|
Jackson W, Sosnoski DM, Ohanessian SE, Chandler P, Mobley A, Meisel KD, Mastro AM. Role of Megakaryocytes in Breast Cancer Metastasis to Bone. Cancer Res 2017; 77:1942-1954. [PMID: 28202531 DOI: 10.1158/0008-5472.can-16-1084] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 12/08/2016] [Accepted: 12/26/2016] [Indexed: 11/16/2022]
Abstract
Little is known about how megakaryocytes may affect metastasis beyond serving as a source of platelets. In this study, we explored the functional implications of megakaryocyte accumulation in the femurs of mice after injection of metastatic or non-metastatic breast cancer cells in 4T1.2 BALB/cJ and MDA-MB-231 nude mouse models. At bone metastatic sites, but not primary growth sites, tumor growth was associated with increased megakaryopoiesis in both model systems. In the orthotopic BALB/cJ model, extramedullary hematopoiesis occurred in the spleen, resulting in a four-fold increase in megakaryocytes. In support of the hypothesis that reducing megakaryocytes may reduce metastasis, we found that thrombopoietin-deficient mice exhibited a 90% relative decrease in megakaryocytes, yet they developed more aggressive metastasis than wild-type hosts. In human clinical specimens, we observed an increase in megakaryocytes in the bone marrow of 6/8 patients with metastatic breast cancer compared with age- and gender-matched controls. Taken together, our results suggested that an increase in megakaryocytes occurring in response to metastatic cells entering the bone marrow confers some measure of protection against metastasis, challenging present views on the role of megakaryocytes in this setting. Cancer Res; 77(8); 1942-54. ©2017 AACR.
Collapse
Affiliation(s)
- Walter Jackson
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania
| | - Donna M Sosnoski
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania
| | - Sara E Ohanessian
- Pathology and Laboratory Medicine, Penn State Hershey Medical Center, Hershey, Pennsylvania
| | - Paige Chandler
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania
| | - Adam Mobley
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania
| | - Kacey D Meisel
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania
| | - Andrea M Mastro
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania.
| |
Collapse
|
18
|
Abubakar AA, Noordin MM, Azmi TI, Kaka U, Loqman MY. The use of rats and mice as animal models in ex vivo bone growth and development studies. Bone Joint Res 2016; 5:610-618. [PMID: 27965220 PMCID: PMC5227059 DOI: 10.1302/2046-3758.512.bjr-2016-0102.r2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 10/06/2016] [Indexed: 01/09/2023] Open
Abstract
In vivo animal experimentation has been one of the cornerstones of biological and biomedical research, particularly in the field of clinical medicine and pharmaceuticals. The conventional in vivo model system is invariably associated with high production costs and strict ethical considerations. These limitations led to the evolution of an ex vivo model system which partially or completely surmounted some of the constraints faced in an in vivo model system. The ex vivo rodent bone culture system has been used to elucidate the understanding of skeletal physiology and pathophysiology for more than 90 years. This review attempts to provide a brief summary of the historical evolution of the rodent bone culture system with emphasis on the strengths and limitations of the model. It encompasses the frequency of use of rats and mice for ex vivo bone studies, nutritional requirements in ex vivo bone growth and emerging developments and technologies. This compilation of information could assist researchers in the field of regenerative medicine and bone tissue engineering towards a better understanding of skeletal growth and development for application in general clinical medicine.Cite this article: A. A. Abubakar, M. M. Noordin, T. I. Azmi, U. Kaka, M. Y. Loqman. The use of rats and mice as animal models in ex vivo bone growth and development studies. Bone Joint Res 2016;5:610-618. DOI: 10.1302/2046-3758.512.BJR-2016-0102.R2.
Collapse
Affiliation(s)
- A A Abubakar
- Department of Pre-Clinical Veterinary Sciences, Universiti Putra Malaysia, Malaysia
| | - M M Noordin
- Department of Pre-Clinical Veterinary Sciences, Universiti Putra Malaysia, Malaysia
| | - T I Azmi
- Department of Pre-Clinical Veterinary Sciences, Universiti Putra Malaysia, Malaysia
| | - U Kaka
- Department of Pre-Clinical Veterinary Sciences, Universiti Putra Malaysia, Malaysia
| | - M Y Loqman
- Department of Pre-Clinical Veterinary Sciences, Universiti Putra Malaysia, Malaysia
| |
Collapse
|
19
|
Zhou X, Zhu W, Nowicki M, Miao S, Cui H, Holmes B, Glazer RI, Zhang LG. 3D Bioprinting a Cell-Laden Bone Matrix for Breast Cancer Metastasis Study. ACS APPLIED MATERIALS & INTERFACES 2016; 8:30017-30026. [PMID: 27766838 DOI: 10.1021/acsami.6b10673] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Metastasis is one of the deadliest consequences of breast cancer, with bone being one of the primary sites of occurrence. Insufficient 3D biomimetic models currently exist to replicate this process in vitro. In this study, we developed a biomimetic bone matrix using 3D bioprinting technology to investigate the interaction between breast cancer (BrCa) cells and bone stromal cells (fetal osteoblasts and human bone marrow mesenchymal stem cells (MSCs)). A tabletop stereolithography 3D bioprinter was employed to fabricate a series of bone matrices consisting of osteoblasts or MSCs encapsulated in gelatin methacrylate (GelMA) hydrogel with nanocrystalline hydroxyapatite (nHA). When BrCa cells were introduced into the stromal cell-laden bioprinted matrices, we found that the growth of BrCa cells was enhanced by the presence of osteoblasts or MSCs, whereas the proliferation of the osteoblasts or MSCs was inhibited by the BrCa cells. The BrCa cells co-cultured with MSCs or osteoblasts presented increased vascular endothelial growth factor (VEGF) secretion in comparison to that of monocultured BrCa cells. Additionally, the alkaline phosphatase activity of MSCs or osteoblasts was reduced after BrCa cell co-culture. These results demonstrate that the 3D bioprinted matrix, with BrCa cells and bone stromal cells, provides a suitable model with which to study the interactive effects of cells in the context of an artificial bone microenvironment and thus may serve as a valuable tool for the investigation of postmetastatic breast cancer progression in bone.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Robert I Glazer
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center , Washington, D.C. 20007, United States
| | | |
Collapse
|
20
|
Anti-rheumatic drug iguratimod (T-614) alleviates cancer-induced bone destruction via down-regulating interleukin-6 production in a nuclear factor-κB-dependent manner. ACTA ACUST UNITED AC 2016; 36:691-699. [PMID: 27752889 DOI: 10.1007/s11596-016-1646-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/25/2016] [Indexed: 12/31/2022]
Abstract
Cytokines are believed to be involved in a "vicious circle" of progressive interactions in bone metastasis. Iguratimod is a novel anti-rheumatic drug which is reported to have the capability of anti-cytokines. In this study, a rat model was constructed to investigate the effect of iguratimod on bone metastasis and it was found that iguratimod alleviated cancer-induced bone destruction. To further explore whether an anti-tumor activity of iguratimod contributes to the effect of bone resorption suppression, two human breast cancer cell lines MDA-MB-231 and MCF-7 were studied. The effect of iguratimod on tumor proliferation was detected by CCK-8 assay and flow cytometry. The effects of iguratimod on migration and invasion of cancer cells were determined by wound-healing and Transwell assays. Results showed that high dose (30 μg/mL) iguratimod slightly suppressed the proliferation of cancer cells but failed to inhibit their migration and invasion capacity. Interestingly, iguratimod decreased the transcription level of IL-6 in MDA-MB-231 cells in a concentration-dependent manner. Moreover, iguratimod partially impaired NF-κB signaling by suppressing the phosphorylation of NF-κB p65 subunit. Our findings indicated that iguratimod may alleviate bone destruction by partially decreasing the expression of IL-6 in an NF-κB-dependent manner, while it has little effect on the tumor proliferation and invasion.
Collapse
|
21
|
Zoi I, Karamouzis MV, Adamopoulos C, Papavassiliou AG. RANKL Signaling and ErbB Receptors in Breast Carcinogenesis. Trends Mol Med 2016; 22:839-850. [PMID: 27567286 DOI: 10.1016/j.molmed.2016.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/26/2016] [Accepted: 07/29/2016] [Indexed: 02/07/2023]
|
22
|
Virtej A, Papadakou P, Sasaki H, Bletsa A, Berggreen E. VEGFR-2 reduces while combined VEGFR-2 and -3 signaling increases inflammation in apical periodontitis. J Oral Microbiol 2016; 8:32433. [PMID: 27650043 PMCID: PMC5030260 DOI: 10.3402/jom.v8.32433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/15/2016] [Accepted: 08/16/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND In apical periodontitis, oral pathogens provoke an inflammatory response in the apical area that induces bone resorptive lesions. In inflammation, angio- and lymphangiogenesis take place. Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are key players in these processes and are expressed in immune cells and endothelial cells in the lesions. OBJECTIVE We aimed at testing the role of VEGFR-2 and -3 in periapical lesion development and investigated their role in lymphangiogenesis in the draining lymph nodes. DESIGN We induced lesions by pulp exposure in the lower first molars of C57BL/6 mice. The mice received IgG injections or blocking antibodies against VEGFR-2 (anti-R2), VEGFR-3 (anti-R3), or combined VEGFR-2 and -3, starting on day 0 until day 10 or 21 post-exposure. RESULTS Lesions developed faster in the anti-R2 and anti-R3 group than in the control and anti-R2/R3 groups. In the anti-R2 group, a strong inflammatory response was found expressed as increased number of neutrophils and osteoclasts. A decreased level of pro-inflammatory cytokines was found in the anti-R2/R3 group. Lymphangiogenesis in the draining lymph nodes was inhibited after blocking of VEGFR-2 and/or -3, while the largest lymph node size was seen after anti-R2 treatment. CONCLUSIONS We demonstrate an anti-inflammatory effect of VEGFR-2 signaling in periapical lesions which seems to involve neutrophil regulation and is independent of angiogenesis. Combined signaling of VEGFR-2 and -3 has a pro-inflammatory effect. Lymph node lymphangiogenesis is promoted through activation of VEGFR-2 and/or VEGFR-3.
Collapse
Affiliation(s)
- Anca Virtej
- Department of Biomedicine, University of Bergen, Bergen, Norway;
| | | | - Hajime Sasaki
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA
| | - Athanasia Bletsa
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Ellen Berggreen
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
23
|
Distinguishing Untreated Osteoblastic Metastases From Enostoses Using CT Attenuation Measurements. AJR Am J Roentgenol 2016; 207:362-8. [DOI: 10.2214/ajr.15.15559] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Heterogeneity of tumor cells in the bone microenvironment: Mechanisms and therapeutic targets for bone metastasis of prostate or breast cancer. Adv Drug Deliv Rev 2016; 99:206-211. [PMID: 26656603 DOI: 10.1016/j.addr.2015.11.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 11/19/2015] [Accepted: 11/25/2015] [Indexed: 01/08/2023]
Abstract
Bone is the most common target organ of metastasis of prostate and breast cancers. This produces considerable morbidity due to skeletal-related events, SREs, including bone pain, hypercalcemia, pathologic fracture, and compression of the spinal cord. The mechanism of bone metastasis is complex and involves cooperative reciprocal interaction among tumor cells, osteoblasts, osteoclasts, and the mineralized bone matrix. The interaction between the metastatic tumor and bone stromal cells has been commonly referred to as the "vicious cycle". Tumor cells stimulate osteoblasts, which in turn stimulate osteoclasts through the secretion of cytokines such as the TNF family member receptor activator of nuclear κB ligand (RANKL). Activated osteoclasts degrade the bone matrix by producing strong acid and proteinases. Bone degradation by osteoclasts releases TGFβ and other growth factors stored in the bone matrix, that further stimulate tumor cells. Bone modifying agents, targeting osteoclast activity, such as bisphosphonate and RANKL antibodies are considered as the standard of care for reducing SREs of patients with bone metastatic diseases. These agents decrease osteoclast activity and delay worsening of skeletal pain and aggravation of bone metastatic diseases. While the management of SREs by these agents may improve patients' lives, this treatment does not address the specific issues of the patients with bone metastasis such as tumor dormancy, drug resistance, or improvement of survival. Here, we review the mechanisms of bone metastasis formation, tumor heterogeneity in the bone microenvironment, and conventional therapy for bone metastatic diseases and discuss the potential development of new therapies targeting tumor heterogeneity in the bone microenvironment.
Collapse
|
25
|
Positive Feedback Loops Between Inflammatory, Bone and Cancer Cells During Metastatic Niche Construction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 936:137-148. [PMID: 27739046 DOI: 10.1007/978-3-319-42023-3_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone, which includes several cell populations and numerous cytokines and chemokines that provide cell-cell signaling, is a common destination for many cancer metastases. Bone metastasis skews this signaling to develop vicious cycles between immune, bone and cancer populations that lead to abnormal bone remodeling during cancer niche construction. Temporal models utilize positive feedback systems as an integrative tool providing insights into the rate-limiting processes that determine multiple stages of the bone metastasis. We develop a logical-transient-threshold framework by linking temporal responses of the cancer, bone and immune systems through macrophages during ecological niche construction of cancer in host bone.
Collapse
|
26
|
Templeton ZS, Bachmann MH, Alluri RV, Maloney WJ, Contag CH, King BL. Methods for culturing human femur tissue explants to study breast cancer cell colonization of the metastatic niche. J Vis Exp 2015:52656. [PMID: 25867136 PMCID: PMC4401351 DOI: 10.3791/52656] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Bone is the most common site of breast cancer metastasis. Although it is widely accepted that the microenvironment influences cancer cell behavior, little is known about breast cancer cell properties and behaviors within the native microenvironment of human bone tissue.We have developed approaches to track, quantify and modulate human breast cancer cells within the microenvironment of cultured human bone tissue fragments isolated from discarded femoral heads following total hip replacement surgeries. Using breast cancer cells engineered for luciferase and enhanced green fluorescent protein (EGFP) expression, we are able to reproducibly quantitate migration and proliferation patterns using bioluminescence imaging (BLI), track cell interactions within the bone fragments using fluorescence microscopy, and evaluate breast cells after colonization with flow cytometry. The key advantages of this model include: 1) a native, architecturally intact tissue microenvironment that includes relevant human cell types, and 2) direct access to the microenvironment, which facilitates rapid quantitative and qualitative monitoring and perturbation of breast and bone cell properties, behaviors and interactions. A primary limitation, at present, is the finite viability of the tissue fragments, which confines the window of study to short-term culture. Applications of the model system include studying the basic biology of breast cancer and other bone-seeking malignancies within the metastatic niche, and developing therapeutic strategies to effectively target breast cancer cells in bone tissues.
Collapse
Affiliation(s)
| | | | - Rajiv V Alluri
- Department of Pediatrics, Stanford University School of Medicine
| | - William J Maloney
- Department of Orthopaedic Surgery, Stanford University School of Medicine
| | | | - Bonnie L King
- Department of Pediatrics, Stanford University School of Medicine;
| |
Collapse
|
27
|
Dormancy and growth of metastatic breast cancer cells in a bone-like microenvironment. Clin Exp Metastasis 2015; 32:335-44. [PMID: 25749879 DOI: 10.1007/s10585-015-9710-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/28/2015] [Indexed: 12/19/2022]
Abstract
Breast cancer can reoccur, often as bone metastasis, many years if not decades after the primary tumor has been treated. The factors that stimulate dormant metastases to grow are not known, but bone metastases are often associated with skeletal trauma. We used a dormancy model of MDA-MB-231BRMS1, a metastasis-suppressed human breast cancer cell line, co-cultured with MC3T3-E1 osteoblasts in a long term, three dimensional culture system to test the hypothesis that bone remodeling cytokines could stimulate dormant cells to grow. The cancer cells attached to the matrix produced by MC3T3-E1 osteoblasts but grew slowly or not at all until the addition of bone remodeling cytokines, TNFα and IL-β. Stimulation of cell proliferation by these cytokines was suppressed with indomethacin, an inhibitor of cyclooxygenase and of prostaglandin production, or a prostaglandin E2 (PGE2) receptor antagonist. Addition of PGE2 directly to the cultures also stimulated cell proliferation. MCF-7, non-metastatic breast cancer cells, remained dormant when co-cultured with normal human osteoblast and fibroblast growth factor. Similar to the MDA-MB-231BRMS1 cells, MCF-7 proliferation increased in response to TNFα and IL-β. These findings suggest that changes in the bone microenvironment due to inflammatory cytokines associated with bone repair or excess turnover may trigger the occurrence of latent bone metastasis.
Collapse
|
28
|
Contag CH, Lie WR, Bammer MC, Hardy JW, Schmidt TL, Maloney WJ, King BL. Monitoring dynamic interactions between breast cancer cells and human bone tissue in a co-culture model. Mol Imaging Biol 2014; 16:158-66. [PMID: 24008275 DOI: 10.1007/s11307-013-0685-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Bone is a preferential site of breast cancer metastasis, and models are needed to study this process at the level of the microenvironment. We have used bioluminescence imaging (BLI) and multiplex biomarker immunoassays to monitor dynamic breast cancer cell behaviors in co-culture with human bone tissue. PROCEDURES Femur tissue fragments harvested from hip replacement surgeries were co-cultured with luciferase-positive MDA-MB-231-fLuc cells. BLI was performed to quantify breast cell proliferation and track migration relative to bone tissue. Breast cell colonization of bone tissues was assessed with immunohistochemistry. Biomarkers in co-culture supernatants were profiled with MILLIPLEX(®) immunoassays. RESULTS BLI demonstrated increased MDA-MB-231-fLuc cell proliferation (p < 0.001) in the presence vs. absence of bones and revealed breast cell migration toward bone. Immunohistochemistry illustrated MDA-MB-231-fLuc cell colonization of bone, and MILLIPLEX(®) profiles of culture supernatants suggested breast/bone crosstalk. CONCLUSIONS Breast cell behaviors that facilitate metastasis occur reproducibly in human bone tissue co-cultures and can be monitored and quantified using BLI and multiplex immunoassays.
Collapse
Affiliation(s)
- Christopher H Contag
- Department of Pediatrics, Stanford University School of Medicine, 150E Clark Center, 318 Campus Drive, Stanford, CA, 94305-5427, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Lu X, Yarbrough WG. Negative regulation of RelA phosphorylation: emerging players and their roles in cancer. Cytokine Growth Factor Rev 2014; 26:7-13. [PMID: 25438737 DOI: 10.1016/j.cytogfr.2014.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 09/03/2014] [Indexed: 01/25/2023]
Abstract
NF-κB signaling contributes to human disease processes, notably inflammatory diseases and cancer. Many advances have been made in understanding mechanisms responsible for abnormal NF-κB activation with RelA post-translational modification, particularly phosphorylation, proven to be critical for RelA function. While the majority of studies have focused on identifying kinases responsible for NF-κB phosphorylation and pathway activation, recently progress has also been made in understanding the negative regulators important for restraining RelA activity. Here we summarize negative regulators of RelA phosphorylation, their targeting sites in RelA and biological functions through negative regulation of RelA activation. Finally, we emphasize the tumor suppressor-like roles that these negative regulators can assume in human cancers.
Collapse
Affiliation(s)
- Xinyuan Lu
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA; Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Wendell G Yarbrough
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, CT, USA; Department of Pathology, Yale University, New Haven, CT, USA; Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
30
|
Singh T, Kaur V, Kumar M, Kaur P, Murthy RSR, Rawal RK. The critical role of bisphosphonates to target bone cancer metastasis: an overview. J Drug Target 2014; 23:1-15. [DOI: 10.3109/1061186x.2014.950668] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
31
|
Shapovalov Y, Zettel M, Spielman SC, Amico-Ruvio SA, Kelly EA, Sipe GO, Dickerson IM, Majewska AK, Brown EB. Fluoxetine modulates breast cancer metastasis to the brain in a murine model. BMC Cancer 2014; 14:598. [PMID: 25129445 PMCID: PMC4242485 DOI: 10.1186/1471-2407-14-598] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 08/06/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Despite advances in the treatment of primary breast tumors, the outcome of metastatic breast cancer remains dismal. Brain metastases present a particularly difficult therapeutic target due to the "sanctuary" status of the brain, with resulting inability of most chemotherapeutic agents to effectively eliminate cancer cells in the brain parenchyma. A large number of breast cancer patients receive various neuroactive drugs to combat complications of systemic anti-tumor therapies and to treat concomitant diseases. One of the most prescribed groups of neuroactive medications is anti-depressants, in particular selective serotonin reuptake inhibitors (SSRIs). Since SSRIs have profound effects on the brain, it is possible that their use in breast cancer patients could affect the development of brain metastases. This would provide important insight into the mechanisms underlying brain metastasis. Surprisingly, this possibility has been poorly explored. METHODS We studied the effect of fluoxetine, an SSRI, on the development of brain metastatic breast cancer using MDA-MB-231BR cells in a mouse model. RESULTS The data demonstrate that fluoxetine treatment increases the number of brain metastases, an effect accompanied by elevated permeability of the blood-brain barrier, pro-inflammatory changes in the brain, and glial activation. This suggests a possible role of brain-resident immune cells and glia in promoting increased development of brain metastases. CONCLUSION Our results offer experimental evidence that neuroactive substances may influence the pathogenesis of brain metastatic disease. This provides a starting point for further investigations into possible mechanisms of interaction between various neuroactive drugs, tumor cells, and the brain microenvironment, which may lead to the discovery of compounds that inhibit metastasis to the brain.
Collapse
Affiliation(s)
- Yuriy Shapovalov
- />Department of Neurobiology and Anatomy, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Box 603, Rochester, NY 14642 USA
- />Department of Biomedical Engineering, University of Rochester, Box 270168, Rochester, NY 14627 USA
| | - Martha Zettel
- />Department of Neurobiology and Anatomy, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Box 603, Rochester, NY 14642 USA
| | - Sara C Spielman
- />Department of Neurobiology and Anatomy, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Box 603, Rochester, NY 14642 USA
| | - Stacy A Amico-Ruvio
- />Department of Neurobiology and Anatomy, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Box 603, Rochester, NY 14642 USA
| | - Emily A Kelly
- />Department of Neurobiology and Anatomy, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Box 603, Rochester, NY 14642 USA
| | - Grayson O Sipe
- />Department of Neurobiology and Anatomy, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Box 603, Rochester, NY 14642 USA
| | - Ian M Dickerson
- />Department of Neurobiology and Anatomy, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Box 603, Rochester, NY 14642 USA
| | - Ania K Majewska
- />Department of Neurobiology and Anatomy, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Box 603, Rochester, NY 14642 USA
| | - Edward B Brown
- />Department of Biomedical Engineering, University of Rochester, Box 270168, Rochester, NY 14627 USA
| |
Collapse
|
32
|
Krishnan V, Vogler EA, Sosnoski DM, Mastro AM. In Vitro Mimics of Bone Remodeling and the Vicious Cycle of Cancer in Bone. J Cell Physiol 2013; 229:453-62. [DOI: 10.1002/jcp.24464] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 08/27/2013] [Indexed: 01/16/2023]
Affiliation(s)
- Venkatesh Krishnan
- Department of Biochemistry and Molecular Biology; The Pennsylvania State University; University Park Pennsylvania
- The Huck Institutes of Life Sciences; The Pennsylvania State University; University Park Pennsylvania
| | - Erwin A. Vogler
- Department of Materials Science and Engineering; The Pennsylvania State University; University Park Pennsylvania
- Department of Bioengineering; The Pennsylvania State University; University Park Pennsylvania
- Materials Research Institute; The Pennsylvania State University; University Park Pennsylvania
| | - Donna M. Sosnoski
- Department of Biochemistry and Molecular Biology; The Pennsylvania State University; University Park Pennsylvania
| | - Andrea M. Mastro
- Department of Biochemistry and Molecular Biology; The Pennsylvania State University; University Park Pennsylvania
| |
Collapse
|
33
|
Mandel K, Yang Y, Schambach A, Glage S, Otte A, Hass R. Mesenchymal stem cells directly interact with breast cancer cells and promote tumor cell growth in vitro and in vivo. Stem Cells Dev 2013; 22:3114-3127. [PMID: 23895436 DOI: 10.1089/scd.2013.0249] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cellular interactions were investigated between human mesenchymal stem cells (MSC) and human breast cancer cells. Co-culture of the two cell populations was associated with an MSC-mediated growth stimulation of MDA-MB-231 breast cancer cells. A continuous expansion of tumor cell colonies was progressively surrounded by MSC(GFP) displaying elongated cell bodies. Moreover, some MSC(GFP) and MDA-MB-231(cherry) cells spontaneously generated hybrid/chimeric cell populations, demonstrating a dual (green fluorescent protein+cherry) fluorescence. During a co-culture of 5-6 days, MSC also induced expression of the GPI-anchored CD90 molecule in breast cancer cells, which could not be observed in a transwell assay, suggesting the requirement of direct cellular interactions. Indeed, MSC-mediated CD90 induction in the breast cancer cells could be partially blocked by a gap junction inhibitor and by inhibition of the notch signaling pathway, respectively. Similar findings were observed in vivo by which a subcutaneous injection of a co-culture of primary MSC with MDA-MB-231(GFP) cells into NOD/scid mice exhibited an about 10-fold increased tumor size and enhanced metastatic capacity as compared with the MDA-MB-231(GFP) mono-culture. Flow cytometric evaluation of the co-culture tumors revealed more than 90% of breast cancer cells with about 3% of CD90-positive cells, also suggesting an MSC-mediated in vivo induction of CD90 in MDA-MB-231 cells. Furthermore, immunohistochemical analysis demonstrated an elevated neovascularization and viability in the MSC/MDA-MB-231(GFP)-derived tumors. Together, these data suggested an MSC-mediated growth stimulation of breast cancer cells in vitro and in vivo by which the altered MSC morphology and the appearance of hybrid/chimeric cells and breast cancer-expressing CD90(+) cells indicate mutual cellular alterations.
Collapse
Affiliation(s)
- Katharina Mandel
- 1 Biochemistry and Tumor Biology Lab, Gynecology Research Unit , Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
34
|
Changes in the peripheral blood and bone marrow from untreated advanced breast cancer patients that are associated with the establishment of bone metastases. Clin Exp Metastasis 2013; 31:213-32. [PMID: 24173696 DOI: 10.1007/s10585-013-9622-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 10/13/2013] [Indexed: 10/26/2022]
Abstract
Bone metastasis is an incurable complication of breast cancer affecting 70-80 % of advanced patients. It is a multistep process that includes tumour cell mobilisation, intravasation, survival in the circulation, extravasation, migration and proliferation in the bone marrow/bone. Although novel findings demonstrate the bone marrow microenvironment significance in bone metastatic progression, a majority of studies have focused on end-stage disease and little is known about how the pre-metastatic niche arises in the bone marrow/bone tissues. We demonstrated a significant increase in patients' peripheral blood plasma ability to induce transendothelial migration of MCF-7 cells compared with healthy volunteers. Moreover, high RANKL, MIF and OPG levels in patients' peripheral blood could play a role in the intravasation, angiogenesis, survival and epithelial-mesenchymal transition of circulating tumour cells. Also, we observed a significant increase in patients' bone marrow plasma capacity to induce transendothelial migration of MDA-MB231 and MCF-7 cells compared with healthy volunteers. Furthermore, patients' bone marrow mesenchymal stem cells could control the recruitment of tumour cells, modifying the MCF-7 and MDA-MB231 cell migration. In addition, we found a significantly higher MDA-MB231 cell proliferation when we used patients' bone marrow plasma compared with healthy volunteers. Interestingly, PDGF-AB, ICAM-1 and VCAM-1 levels in patients' bone marrow were significantly higher than the values of healthy volunteers, suggesting that they could be involved in the cancer cell extravasation, bone resorption and cancer cell proliferation. We believe that these results can reveal new information about what alterations happen in the bone marrow of advanced breast cancer patients before bone colonisation, changes that create optimal soil for the metastatic cascade progression.
Collapse
|
35
|
Lu X, An H, Jin R, Zou M, Guo Y, Su PF, Liu D, Shyr Y, Yarbrough WG. PPM1A is a RelA phosphatase with tumor suppressor-like activity. Oncogene 2013; 33:2918-27. [PMID: 23812431 DOI: 10.1038/onc.2013.246] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 05/06/2013] [Accepted: 05/20/2013] [Indexed: 12/28/2022]
Abstract
Nuclear factor-κB (NF-κB) signaling contributes to human disease processes, notably inflammatory diseases and cancer. NF-κB has a role in tumorigenesis and tumor growth, as well as promotion of metastases. Mechanisms responsible for abnormal NF-κB activation are not fully elucidated; however, RelA phosphorylation, particularly at serine residues S536 and S276, is critical for RelA function. Kinases that phosphorylate RelA promote oncogenic behaviors, suggesting that phosphatases targeting RelA could have tumor-inhibiting activities; however, few RelA phosphatases have been identified. Here, we identified tumor inhibitory and RelA phosphatase activities of the protein phosphatase 2C (PP2C) phosphatase family member, PPM1A. We show that PPM1A directly dephosphorylated RelA at residues S536 and S276 and selectively inhibited NF-κB transcriptional activity, resulting in decreased expression of monocyte chemotactic protein-1/chemokine (C-C motif) ligand 2 and interleukin-6, cytokines implicated in cancer metastasis. PPM1A depletion enhanced NF-κB-dependent cell invasion, whereas PPM1A expression inhibited invasion. Analyses of human expression data revealed that metastatic prostate cancer deposits had lower PPM1A expression compared with primary tumors without distant metastases. A hematogenous metastasis mouse model revealed that PPM1A expression inhibited bony metastases of prostate cancer cells after vascular injection. In summary, our findings suggest that PPM1A is a RelA phosphatase that regulates NF-κB activity and that PPM1A has tumor suppressor-like activity. Our analyses also suggest that PPM1A inhibits prostate cancer metastases and as neither gene deletions nor inactivating mutations of PPM1A have been described, increasing PPM1A activity in tumors represents a potential therapeutic strategy to inhibit NF-κB signaling or bony metastases in human cancer.
Collapse
Affiliation(s)
- X Lu
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - H An
- 1] Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA [2] Division of Surgical Sciences, Vanderbilt University, Nashville, TN, USA
| | - R Jin
- 1] Vanderbilt Prostate Cancer Center, Vanderbilt University, Nashville, TN, USA [2] Department of Urology, Vanderbilt University, Nashville, TN, USA
| | - M Zou
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, CT, USA
| | - Y Guo
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - P-F Su
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - D Liu
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, CT, USA
| | - Y Shyr
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - W G Yarbrough
- 1] Division of Otolaryngology, Department of Surgery, Yale University, New Haven, CT, USA [2] Department of Pathology, Yale University, New Haven, CT, USA [3] Yale Cancer Center, New Haven, CT, USA
| |
Collapse
|
36
|
Agarwal M, He C, Siddiqui J, Wei J, Macoska JA. CCL11 (eotaxin-1): a new diagnostic serum marker for prostate cancer. Prostate 2013; 73:573-81. [PMID: 23059958 PMCID: PMC3594486 DOI: 10.1002/pros.22597] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 09/10/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND The recent recommendation of the U.S. Preventive Services Task Force against PSA-based screening for prostate cancer was based, in part, on the lack of demonstrated diagnostic utility of serum PSA values in the low, but detectable range to successfully predict prostate cancer. Though controversial, this recommendation reinforced the critical need to develop, validate, and determine the utility of other serum and/or urine transcript and protein markers as diagnostic markers for PCa. The studies described here were intended to determine whether inflammatory cytokines might augment serum PSA as a diagnostic marker for prostate cancer. METHODS Multiplex ELISA assays were performed to quantify CCL1, CCL2, CCL5, CCL8, CCL11, CCL17, CXCL1, CXCL5, CXCL8, CXCL10, CXCL12, and IL-6 protein levels in the serum of 272 men demonstrating serum PSA values of <10 ng/ml and undergoing a 12 core diagnostic needle biopsy for detection of prostate cancer. Logistic regression was used to identify the associations between specific chemokines and prostate cancer status adjusted for prostate volume, and baseline PSA. RESULTS Serum levels for CCL1 (I-309) were significantly elevated among all men with enlarged prostates (P < 0.04). Serum levels for CCL11 (Eotaxin-1) were significantly elevated among men with prostate cancer regardless of prostate size (P < 0.01). The remaining 10 cytokines examined in this study did not exhibit significant correlations with either prostate volume or cancer status. CONCLUSIONS Serum CCL11 values may provide a useful diagnostic tool to help distinguish between prostatic enlargement and prostate cancer among men demonstrating low, but detectable, serum PSA values.
Collapse
Affiliation(s)
- Manisha Agarwal
- Department of Urology, The University of Michigan School of Medicine, Ann Arbor, MI
| | - Chang He
- Department of Urology, The University of Michigan School of Medicine, Ann Arbor, MI
| | - Javed Siddiqui
- Center for Translational Pathology, The University of Michigan School of Medicine, Ann Arbor, MI
| | - John Wei
- Department of Urology, The University of Michigan School of Medicine, Ann Arbor, MI
- Center for Translational Pathology, The University of Michigan School of Medicine, Ann Arbor, MI
| | - Jill A. Macoska
- Department of Urology, The University of Michigan School of Medicine, Ann Arbor, MI
- Address Correspondence To: Jill A. Macoska, Ph.D., Department of Urology, The University of Michigan, 6217 Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109-0944, (734) 647-8121 TEL, (734) 647-9271 FAX,
| |
Collapse
|
37
|
Das Roy L, Curry JM, Sahraei M, Besmer DM, Kidiyoor A, Gruber HE, Mukherjee P. Arthritis augments breast cancer metastasis: role of mast cells and SCF/c-Kit signaling. Breast Cancer Res 2013; 15:R32. [PMID: 23577751 PMCID: PMC3672823 DOI: 10.1186/bcr3412] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 04/11/2013] [Indexed: 12/16/2022] Open
Abstract
Introduction Breast cancer remains the second leading cause of cancer-related deaths for women in the United States. Metastasis is regulated not only by intrinsic genetic changes in malignant cells, but also by the microenvironment, especially those associated with chronic inflammation. We recently reported that mice with autoimmune arthritis have significantly increased incidence of bone and lung metastasis and decreased survival associated with breast cancer. In this study, we evaluated the mechanism underlying the increased metastasis. Methods We used two mouse models; one that develops spontaneous autoimmune arthritis (SKG mice) injected with metastatic breast cancer cells (4T1), and another that develops spontaneous breast cancer (MMTV-PyV MT mice) injected with type II collagen to induce autoimmune arthritis. Mast cell levels and metastasis were monitored. Results First, we confirmed that breast tumor-bearing arthritic mice have a significantly higher incidence of bone and lung metastasis than do their nonarthritic counterparts. Next, we showed increased recruitment of mast cells within the primary tumor of arthritic mice, which facilitates metastasis. Next, we report that arthritic mice without any tumors have higher numbers of mast cells in the bones and lungs, which may be the underlying cause for the enhanced lung and bone metastases observed in the arthritic mice. Next, we showed that once the tumor cells populate the metastatic niches (bones and lungs), they further increase the mast cell population within the niche and assist in enhancing metastasis. This may primarily be due to the interaction of c-Kit receptor present on mast cells and stem cell factor (SCF, the ligand for ckit) expressed on tumor cells. Finally, we showed that targeting the SCF/cKit interaction with an anti-ckit antibody reduces the differentiation of mast cells and consequently reduces metastasis. Conclusion This is the first report to show that mast cells may play a critical role in remodeling not only the tumor microenvironment but also the metastatic niche to facilitate efficient metastasis through SCF/cKit interaction in breast cancer with arthritis.
Collapse
|
38
|
Virtej A, Løes SS, Berggreen E, Bletsa A. Localization and signaling patterns of vascular endothelial growth factors and receptors in human periapical lesions. J Endod 2013; 39:605-11. [PMID: 23611377 DOI: 10.1016/j.joen.2012.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 11/28/2012] [Accepted: 12/10/2012] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are key players in vasculogenesis and are also involved in pathologic conditions with bone destruction. Vasculogenesis is critical for disease progression, and bone resorption is a hallmark of apical periodontitis. However, the localization of VEGFs and VEGFRs and their gene signaling pathways in human apical periodontitis have not been thoroughly investigated. The aim of this study was to localize VEGFs and VEGFRs and analyze their gene expression as well as signaling pathways in human periapical lesions. METHODS Tissue was collected after endodontic surgery from patients diagnosed with chronic apical periodontitis. Periodontal ligament samples from extracted healthy wisdom teeth was also collected and used as control tissue. In lesion cryosections, VEGFs/VEGFRs were identified by immunohistochemistry/double immunofluorescence by using specific antibodies. A human VEGF signaling polymerase chain reaction array system was used for gene expression analysis comparing lesions with periodontal ligament samples. RESULTS The histologic evaluation revealed heterogeneous morphology of the periapical lesions with various degrees of inflammatory infiltrates. In the lesions, all investigated factors and receptors were identified in blood vessels and various immune cells. No lymphatic vessels were detected. Gene expression analysis revealed up-regulation of VEGF-A and VEGFR-3, although not significant. Phosphatidylinositol-3-kinases, protein kinase C, mitogen-activated protein kinases, and phospholipases, all known to be involved in VEGF-mediated angiogenic activity, were significantly up-regulated. CONCLUSIONS The cellular and vascular expressions of VEGFs and VEGFRs in chronic apical periodontitis, along with significant alterations of genes mediating VEGF-induced angiogenic responses, suggest ongoing vascular remodeling in established chronic periapical lesions.
Collapse
Affiliation(s)
- Anca Virtej
- Institute of Biomedicine, Faculty of Medicine-Dentistry, University of Bergen, Bergen, Norway
| | | | | | | |
Collapse
|
39
|
Animal model for mammary tumor growth in the bone microenvironment. Breast Cancer 2013; 20:195-203. [DOI: 10.1007/s12282-013-0439-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 01/07/2013] [Indexed: 02/06/2023]
|