1
|
Cramer EEA, Hermsen KCJ, Kock LM, Ito K, Hofmann S. Culture system for longitudinal monitoring of bone dynamics ex vivo. Biotechnol Bioeng 2024. [PMID: 39295202 DOI: 10.1002/bit.28848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 07/19/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024]
Abstract
To quantify and visualize both bone formation and resorption within osteochondral explants cultured ex vivo is challenging with the current analysis techniques. An approach that enables monitoring of bone remodeling dynamics is longitudinal microcomputed tomography (µCT), a non-destructive technique that relies on repeated µCT scanning and subsequent registration of consecutive scans. In this study, a two-compartment culture system suitable for osteochondral explants that allowed for µCT scanning during ex vivo culture was established. Explants were scanned repeatedly in a fixed orientation, which allowed assessment of bone remodeling due to adequate image registration. Using this method, bone formation was found to be restricted to the outer surfaces when cultured statically. To demonstrate that the culture system could capture differences in bone remodeling, explants were cultured statically and under dynamic compression as loading promotes osteogenesis. No quantitative differences between static and dynamic culture were revealed. Still, only in dynamic conditions, bone formation was visualized on trabecular surfaces located within the inner cores, suggesting enhanced bone formation towards the center of the explants upon mechanical loading. Taken together, the ex vivo culture system in combination with longitudinal µCT scanning and subsequent registration of images demonstrated potential for evaluating bone remodeling within explants.
Collapse
Affiliation(s)
- Esther E A Cramer
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Kim C J Hermsen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Linda M Kock
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- LifeTec Group BV, Eindhoven, The Netherlands
| | - Keita Ito
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Sandra Hofmann
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
2
|
Nuckhir M, Withey D, Cabral S, Harrison H, Clarke RB. State of the Art Modelling of the Breast Cancer Metastatic Microenvironment: Where Are We? J Mammary Gland Biol Neoplasia 2024; 29:14. [PMID: 39012440 PMCID: PMC11252219 DOI: 10.1007/s10911-024-09567-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/09/2024] [Indexed: 07/17/2024] Open
Abstract
Metastatic spread of tumour cells to tissues and organs around the body is the most frequent cause of death from breast cancer. This has been modelled mainly using mouse models such as syngeneic mammary cancer or human in mouse xenograft models. These have limitations for modelling human disease progression and cannot easily be used for investigation of drug resistance and novel therapy screening. To complement these approaches, advances are being made in ex vivo and 3D in vitro models, which are becoming progressively better at reliably replicating the tumour microenvironment and will in the future facilitate drug development and screening. These approaches include microfluidics, organ-on-a-chip and use of advanced biomaterials. The relevant tissues to be modelled include those that are frequent and clinically important sites of metastasis such as bone, lung, brain, liver for invasive ductal carcinomas and a distinct set of common metastatic sites for lobular breast cancer. These sites all have challenges to model due to their unique cellular compositions, structure and complexity. The models, particularly in vivo, provide key information on the intricate interactions between cancer cells and the native tissue, and will guide us in producing specific therapies that are helpful in different context of metastasis.
Collapse
Affiliation(s)
- Mia Nuckhir
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - David Withey
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - Sara Cabral
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - Hannah Harrison
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK.
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK.
| |
Collapse
|
3
|
GAO HUAN, ZHANG JIE, KLEIJN TONYG, WU ZHAOYONG, LIU BING, MA YUJIN, DING BAOYUE, YIN DONGFENG. Dual ligand-targeted Pluronic P123 polymeric micelles enhance the therapeutic effect of breast cancer with bone metastases. Oncol Res 2024; 32:769-784. [PMID: 38560569 PMCID: PMC10972726 DOI: 10.32604/or.2023.044276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/13/2023] [Indexed: 04/04/2024] Open
Abstract
Bone metastasis secondary to breast cancer negatively impacts patient quality of life and survival. The treatment of bone metastases is challenging since many anticancer drugs are not effectively delivered to the bone to exert a therapeutic effect. To improve the treatment efficacy, we developed Pluronic P123 (P123)-based polymeric micelles dually decorated with alendronate (ALN) and cancer-specific phage protein DMPGTVLP (DP-8) for targeted drug delivery to breast cancer bone metastases. Doxorubicin (DOX) was selected as the anticancer drug and was encapsulated into the hydrophobic core of the micelles with a high drug loading capacity (3.44%). The DOX-loaded polymeric micelles were spherical, 123 nm in diameter on average, and exhibited a narrow size distribution. The in vitro experiments demonstrated that a pH decrease from 7.4 to 5.0 markedly accelerated DOX release. The micelles were well internalized by cultured breast cancer cells and the cell death rate of micelle-treated breast cancer cells was increased compared to that of free DOX-treated cells. Rapid binding of the micelles to hydroxyapatite (HA) microparticles indicated their high affinity for bone. P123-ALN/DP-8@DOX inhibited tumor growth and reduced bone resorption in a 3D cancer bone metastasis model. In vivo experiments using a breast cancer bone metastasis nude model demonstrated increased accumulation of the micelles in the tumor region and considerable antitumor activity with no organ-specific histological damage and minimal systemic toxicity. In conclusion, our study provided strong evidence that these pH-sensitive dual ligand-targeted polymeric micelles may be a successful treatment strategy for breast cancer bone metastasis.
Collapse
Affiliation(s)
- HUAN GAO
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
- Department of Pharmacy, The General Hospital of Xinjiang Military Region, Urumqi, 830000, China
| | - JIE ZHANG
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - TONY G. KLEIJN
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands
- Department of Pathology, Laboratory of Experimental Oncology, Erasmus MC, Rotterdam, 3015 GD, The Netherlands
| | - ZHAOYONG WU
- Department of Pharmacy, Jiaxing Maternal and Child Health Care Hospital, Affiliated Hospital of Jiaxing University, Jiaxing, 314001, China
| | - BING LIU
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
- Qinghai Enlu Biotechnology Co., Ltd., Haidong, 810700, China
| | - YUJIN MA
- Qinghai Enlu Biotechnology Co., Ltd., Haidong, 810700, China
| | - BAOYUE DING
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - DONGFENG YIN
- Department of Pharmacy, The General Hospital of Xinjiang Military Region, Urumqi, 830000, China
| |
Collapse
|
4
|
Cramer EEA, de Wildt BWM, Hendriks JGE, Ito K, Hofmann S. Integration of osteoclastogenesis through addition of PBMCs in human osteochondral explants cultured ex vivo. Bone 2024; 178:116935. [PMID: 37852425 DOI: 10.1016/j.bone.2023.116935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023]
Abstract
The preservation of tissue specific cells in their native 3D extracellular matrix in bone explants provides a unique platform to study remodeling. Thus far, studies involving bone explant cultures showed a clear focus on achieving bone formation and neglected osteoclast activity and resorption. To simulate the homeostatic bone environment ex vivo, both key elements of bone remodeling need to be represented. This study aimed to assess and include osteoclastogenesis in human osteochondral explants through medium supplementation with RANKL and M-CSF and addition of peripheral blood mononuclear cells (PBMCs), providing osteoclast precursors. Osteochondral explants were freshly harvested from human femoral heads obtained from hip surgeries and cultured for 20 days in a two-compartment culture system. Osteochondral explants preserved viability and cellular abundance over the culture period, but histology demonstrated that resident osteoclasts were no longer present after 4 days of culture. Quantitative extracellular tartrate resistant acid phosphatase (TRAP) analysis confirmed depletion of osteoclast activity on day 4 even when stimulated with RANKL and M-CSF. Upon addition of PBMCs, a significant upregulation of TRAP activity was measured from day 10 onwards. Evaluation of bone loss trough μCT registration and measurement of extracellular cathepsin K activity revealed indications of enhanced resorption upon addition of PBMCs. Based on the results we suggest that an external source of osteoclast precursors, such as PBMCs, needs to be added in long-term bone explant cultures to maintain osteoclastic activity, and bone remodeling.
Collapse
Affiliation(s)
- Esther E A Cramer
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Bregje W M de Wildt
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Johannes G E Hendriks
- Department of Orthopedic Surgery & Trauma, Máxima Medical Center Eindhoven/Veldhoven, 5631 BM Eindhoven, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands.
| |
Collapse
|
5
|
Liu B, Yang Q, Cheng Y, Liu M, Ji Q, Zhang B, Yang Z, Zhou S, Liu D. Calcium phosphate hybrid micelles inhibit orthotopic bone metastasis from triple negative breast cancer by simultaneously killing cancer cells and reprogramming the microenvironment of bone resorption and immunosuppression. Acta Biomater 2023; 166:640-654. [PMID: 37236576 DOI: 10.1016/j.actbio.2023.05.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023]
Abstract
Triple negative breast cancer (TNBC) is prone to develop drug resistance and metastasis. Bone is the most common distant metastasis site of breast cancer cell. Patients with bone metastasis from TNBC suffer from unbearable pain due to the growth of bone metastasis and bone destruction. Simultaneously blocking the growth of bone metastasis and reprogramming the microenvironment of bone resorption and immunosuppression is a promising strategy to treat bone metastasis from TNBC. Herein, we prepared a pH and redox responsive drug delivery system, named DZ@CPH, by encapsulating docetaxel (DTX) with hyaluronic acid-polylactic acid micelle then reinforcing with calcium phosphate and zoledronate for targeting to bone metastasis from TNBC. DZ@CPH reduced the activation of osteoclast and inhibited bone resorption by decreasing the expression of nuclear factor κB receptor ligand and increasing the expression of osteoprotegerin in drug-resistant bone metastasis tissue. At the same time, DZ@CPH inhibited the invasion of bone metastatic TNBC cells by regulating the apoptosis-related and invasion-related protein expression. It also increased the sensitivity of orthotopic drug-resistant bone metastasis to DTX by inhibiting the expression of P-glycoprotein, Bcl-2 and transforming growth factor-β in tissue of drug-resistant bone metastasis. Moreover, the ratio between M1 type macrophage to M2 type macrophage in bone metastasis tissue was increased by DZ@CPH. In a word, DZ@CPH blocked the growth of bone metastasis from drug-resistant TNBC through inducing the apoptosis of drug-resistant TNBC cells and reprogramming the microenvironment of bone resorption and immunosuppression. DZ@CPH has a great potential in clinical application for the treatment of bone metastasis from drug-resistant TNBC. STATEMENT OF SIGNIFICANCE: Triple negative breast cancer (TNBC) is prone to develop bone metastasis. Now bone metastasis is still an intractable disease. In this study, docetaxel and zoledronate co-loaded calcium phosphate hybrid micelles (DZ@CPH) were prepared. DZ@CPH reduced the activation of osteoclasts and inhibited bone resorption. At the same time, DZ@CPH inhibited the invasion of bone metastatic TNBC cells by regulating the expression of apoptosis and invasion related protein in bone metastasis tissue. Moreover, the ratio between M1 type macrophages to M2 type macrophages in bone metastases tissue was increased by DZ@CPH. In a word, DZ@CPH blocked vicious cycle between the growth of bone metastasis and bone resorption, which greatly improved the therapeutic effect on bone metastasis from drug-resistant TNBC.
Collapse
Affiliation(s)
- Bao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Qian Yang
- Department of pharmacy, School of Medicine, Shaanxi Energy Institute, Xianyang, 712000, China
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Bangle Zhang
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Zhifu Yang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an 710032, China.
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China.
| | - Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
6
|
Dozzo A, Chullipalliyalil K, McAuliffe M, O’Driscoll CM, Ryan KB. Nano-Hydroxyapatite/PLGA Mixed Scaffolds as a Tool for Drug Development and to Study Metastatic Prostate Cancer in the Bone. Pharmaceutics 2023; 15:pharmaceutics15010242. [PMID: 36678871 PMCID: PMC9864166 DOI: 10.3390/pharmaceutics15010242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
(1) Background: Three-dimensional (3D) in vitro, biorelevant culture models that recapitulate cancer progression can help elucidate physio-pathological disease cues and enhance the screening of more effective therapies. Insufficient research has been conducted to generate in vitro 3D models to replicate the spread of prostate cancer to the bone, a key metastatic site of the disease, and to understand the interplay between the key cell players. In this study, we aim to investigate PLGA and nano-hydroxyapatite (nHA)/PLGA mixed scaffolds as a predictive preclinical tool to study metastatic prostate cancer (mPC) in the bone and reduce the gap that exists with traditional 2D cultures. (2) Methods: nHA/PLGA mixed scaffolds were produced by electrospraying, compacting, and foaming PLGA polymer microparticles, +/- nano-hydroxyapatite (nHA), and a salt porogen to produce 3D, porous scaffolds. Physicochemical scaffold characterisation together with an evaluation of osteoblastic (hFOB 1.19) and mPC (PC-3) cell behaviour (RT-qPCR, viability, and differentiation) in mono- and co-culture, was undertaken. (3) Results: The results show that the addition of nHA, particularly at the higher-level impacted scaffolds in terms of mechanical and degradation behaviour. The nHA 4 mg resulted in weaker scaffolds, but cell viability increased. Qualitatively, fluorescent imaging of cultures showed an increase in PC-3 cells compared to osteoblasts despite lower initial PC-3 seeding densities. Osteoblast monocultures, in general, caused an upregulation (or at least equivalent to controls) in gene production, which was highest in plain scaffolds and decreased with increases in nHA. Additionally, the genes were downregulated in PC3 and co-cultures. Further, drug toxicity tests demonstrated a significant effect in 2D and 3D co-cultures. (4) Conclusions: The results demonstrate that culture conditions and environment (2D versus 3D, monoculture versus co-culture) and scaffold composition all impact cell behaviour and model development.
Collapse
Affiliation(s)
- Annachiara Dozzo
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, T12 K8AF Cork, Ireland
| | | | - Michael McAuliffe
- Centre for Advanced Photonics & Process Analysis, Munster Technological University Cork, T12 P928 Cork, Ireland
| | - Caitriona M. O’Driscoll
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, T12 K8AF Cork, Ireland
| | - Katie B. Ryan
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, T12 K8AF Cork, Ireland
- Correspondence:
| |
Collapse
|
7
|
Tian Q, Lu Y, Yan B, Wu C. Integrative Bioinformatics Analysis Reveals That miR-524-5p/MEF2C Regulates Bone Metastasis in Prostate Cancer and Breast Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5211329. [PMID: 36128051 PMCID: PMC9482681 DOI: 10.1155/2022/5211329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/28/2022] [Indexed: 11/30/2022]
Abstract
Bone metastases are highly prevalent in patients with advanced prostate cancer and breast cancer and have a serious impact on the survival time and quality of life of these patients. It has been reported that microRNAs (miRNAs) are expressed abnormally in different types of cancer and metastases. However, it remains unknown whether the underlying miRNAs are associated with prostate and breast cancer bone metastasis. Differentially expressed miRNAs (DE-miRNAs) and their potential targets in the metastatic process were identified by bioinformatics analysis. Additionally, qPCR confirmed that the miR-524-5p expression was downregulated in prostate and breast cancer cells. The overexpression of miR-524-5p restrained cell proliferation, invasion, and metastasis in prostate and breast cancer cells. Meanwhile, miR-524-5p could target and inhibit the expression of MEF2C, which was verified by a luciferase assay. In conclusion, our data strongly suggest that downregulation of miR-524-5p appears to be a precocious event in prostate and breast cancer, and the miR-524-5p/MEF2C axis plays a novel role in bone metastases from prostate and breast cancers.
Collapse
Affiliation(s)
- QingHua Tian
- Department of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - YingYing Lu
- Department of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - BiCong Yan
- Department of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - ChunGen Wu
- Department of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
8
|
Costard LS, Hosn RR, Ramanayake H, O'Brien FJ, Curtin CM. Influences of the 3D microenvironment on cancer cell behaviour and treatment responsiveness: A recent update on lung, breast and prostate cancer models. Acta Biomater 2021; 132:360-378. [PMID: 33484910 DOI: 10.1016/j.actbio.2021.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
The majority of in vitro studies assessing cancer treatments are performed in two-dimensional (2D) monolayers and are subsequently validated in in vivo animal models. However, 2D models fail to accurately model the tumour microenvironment. Furthermore, animal models are not directly applicable to mimic the human scenario. Three-dimensional (3D) culture models may help to address the discrepancies of 2D and animal models. When cancer cells escape the primary tumour, they can invade at distant organs building secondary tumours, called metastasis. The development of metastasis leads to a dramatic decrease in the life expectancy of patients. Therefore, 3D systems to model the microenvironment of metastasis have also been developed. Several studies have demonstrated changes in cell behaviour and gene expression when cells are cultured in 3D compared to 2D and concluded a better comparability to cells in vivo. Of special importance is the effect seen in response to anti-cancer treatments as models are built primarily to serve as drug-testing platforms. This review highlights these changes between cancer cells grown in 2D and 3D models for some of the most common cancers including lung, breast and prostate tumours. In addition to models aiming to mimic the primary tumour site, the effects of 3D cell culturing in bone metastasis models are also described. STATEMENT OF SIGNIFICANCE: Most in vitro studies in cancer research are performed in 2D and are subsequently validated in in vivo animal models. However, both models possess numerous limitations: 2D models fail to accurately model the tumour microenvironment while animal models are expensive, time-consuming and can differ considerably from humans. It is accepted that the cancer microenvironment plays a critical role in the disease, thus, 3D models have been proposed as a potential solution to address the discrepancies of 2D and animal models. This review highlights changes in cell behaviour, including proliferation, gene expression and chemosensitivity, between cancer cells grown in 2D and 3D models for some of the most common cancers including lung, breast and prostate cancer as well as bone metastasis.
Collapse
|
9
|
Bahmad HF, Jalloul M, Azar J, Moubarak MM, Samad TA, Mukherji D, Al-Sayegh M, Abou-Kheir W. Tumor Microenvironment in Prostate Cancer: Toward Identification of Novel Molecular Biomarkers for Diagnosis, Prognosis, and Therapy Development. Front Genet 2021; 12:652747. [PMID: 33841508 PMCID: PMC8033163 DOI: 10.3389/fgene.2021.652747] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is by far the most commonly diagnosed cancer in men worldwide. Despite sensitivity to androgen deprivation, patients with advanced disease eventually develop resistance to therapy and may die of metastatic castration-resistant prostate cancer (mCRPC). A key challenge in the management of PCa is the clinical heterogeneity that is hard to predict using existing biomarkers. Defining molecular biomarkers for PCa that can reliably aid in diagnosis and distinguishing patients who require aggressive therapy from those who should avoid overtreatment is a significant unmet need. Mechanisms underlying the development of PCa are not confined to cancer epithelial cells, but also involve the tumor microenvironment. The crosstalk between epithelial cells and stroma in PCa has been shown to play an integral role in disease progression and metastasis. A number of key markers of reactive stroma has been identified including stem/progenitor cell markers, stromal-derived mediators of inflammation, regulators of angiogenesis, connective tissue growth factors, wingless homologs (Wnts), and integrins. Here, we provide a synopsis of the stromal-epithelial crosstalk in PCa focusing on the relevant molecular biomarkers pertaining to the tumor microenvironment and their role in diagnosis, prognosis, and therapy development.
Collapse
Affiliation(s)
- Hisham F Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL, United States
| | - Mohammad Jalloul
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Joseph Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Maya M Moubarak
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Tamara Abdul Samad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Deborah Mukherji
- Department of Internal Medicine, Division of Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Novel therapies for damaged and diseased bone are being developed in a preclinical testing process consisting of in vitro cell experiments followed by in vivo animal studies. The in vitro results are often not representative of the results observed in vivo. This could be caused by the complexity of the natural bone environment that is missing in vitro. Ex vivo bone explant cultures provide a model in which cells are preserved in their native three-dimensional environment. Herein, it is aimed to review the current status of bone explant culture models in relation to their potential in complementing the preclinical evaluation process with specific attention paid to the incorporation of mechanical loading within ex vivo culture systems. RECENT FINDINGS Bone explant cultures are often performed with physiologically less relevant bone, immature bone, and explants derived from rodents, which complicates translatability into clinical practice. Mature bone explants encounter difficulties with maintaining viability, especially in static culture. The integration of mechanical stimuli was able to extend the lifespan of explants and to induce new bone formation. Bone explant cultures provide unique platforms for bone research and mechanical loading was demonstrated to be an important component in achieving osteogenesis ex vivo. However, more research is needed to establish a representative, reliable, and reproducible bone explant culture system that includes both components of bone remodeling, i.e., formation and resorption, in order to bridge the gap between in vitro and in vivo research in preclinical testing.
Collapse
Affiliation(s)
- E E A Cramer
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - K Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - S Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands.
| |
Collapse
|
11
|
Abstract
The biological signals that coordinate the three-dimensional outgrowth and patterning of the vertebrate limb bud have been well delineated. These include a number of vital embryonic signaling pathways, including the fibroblast growth factor, WNT, transforming growth factor, and hedgehog. Collectively these signals converge on multiple progenitor populations to drive the formation of a variety of tissues that make up the limb musculoskeletal system, such as muscle, tendon, cartilage, stroma, and bone. The basic mechanisms regulating the commitment and differentiation of diverse limb progenitor populations has been successfully modeled in vitro using high density primary limb mesenchymal or micromass cultures. However, this approach is limited in its ability to more faithfully recapitulate the assembly of progenitors into organized tissues that span the entire musculoskeletal system. Other biological systems have benefitted from the development and availability of three-dimensional organoid cultures which have transformed our understanding of tissue development, homeostasis and regeneration. Such a system does not exist that effectively models the complexity of limb development. However, limb bud organ cultures while still necessitating the use of collected embryonic tissue have proved to be a powerful model system to elucidate the molecular underpinning of musculoskeletal development. In this methods article, the derivation and use of limb bud organ cultures from murine limb buds will be described, along with strategies to manipulate signaling pathways, examine gene expression and for longitudinal lineage tracking.
Collapse
Affiliation(s)
- Martin Arostegui
- Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - T Michael Underhill
- Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada.
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
12
|
Investigating the Multitarget Mechanism of Traditional Chinese Medicine Prescription for Cancer-Related Pain by Using Network Pharmacology and Molecular Docking Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7617261. [PMID: 33224254 PMCID: PMC7673937 DOI: 10.1155/2020/7617261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/30/2020] [Accepted: 10/24/2020] [Indexed: 01/04/2023]
Abstract
Gu-tong formula (GTF) has achieved good curative effects in the treatment of cancer-related pain. However, its potential mechanisms have not been explored. We used network pharmacology and molecular docking to investigate the molecular mechanism and the effective compounds of the prescription. Through the analysis and research in this paper, we obtained 74 effective compounds and 125 drug-disease intersection targets to construct a network, indicating that quercetin, kaempferol, and β-sitosterol were possibly the most important compounds in GTF. The key targets of GTF for cancer-related pain were Jun proto-oncogene (JUN), mitogen-activated protein kinase 1 (MAPK1), and RELA proto-oncogene (RELA). 2204 GO entries and 148 pathways were obtained by GO and KEGG enrichment, respectively, which proved that chemokine, MAPK, and transient receptor potential (TRP) channels can be regulated by GTF. The results of molecular docking showed that stigmasterol had strong binding activity with arginine vasopressin receptor 2 (AVPR2) and C-X3-C motif chemokine ligand 1 (CX3CL1) and cholesterol was more stable with p38 MAPK, prostaglandin-endoperoxide synthase 2 (PTGS2), and transient receptor potential vanilloid-1 (TRPV1). In conclusion, the therapeutic effect of GTF on cancer-related pain is based on the comprehensive pharmacological effect of multicomponent, multitarget, and multichannel pathways. This study provides a theoretical basis for further experimental research in the future.
Collapse
|
13
|
Zheng RJ, Song JL, Wu XH, Watts DC. Evaluation of bone formation in neonatal mouse calvariae using micro-CT and histomorphometry: an in vitro study. Acta Histochem 2020; 122:151614. [PMID: 33066836 DOI: 10.1016/j.acthis.2020.151614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/27/2020] [Accepted: 08/10/2020] [Indexed: 11/17/2022]
Abstract
Neonatal calvarial bone has been widely used for investigating the biological behaviour of intramembranous bones. This work evaluated the bone formation of neonatal calvarial bone by microcomputed tomography (micro-CT) and histomorphometry. Moreover, the viability of neonatal calvarial bone and the effect of micro-CT radiation exposure on neonatal calvarial bone viability were investigated. The calvarial bones of 4-day-old CD-1 mice were cultured in Dulbecco's modified Eagle's medium (DMEM) or osteogenic medium (OM) for 23 days. Micro-CT scanning and histological analysis were performed on days 2, 9, 16 and 23. An "OM-control" group was scanned only on days 2 and 23 to evaluate the effect of a single micro-CT radiation dose on calvarial bones. Histomorphometric measurements revealed that the number of osteoblasts per unit bone surface area (N. Ob/BS, /mm2) (days 9, 16 and 23) and the number of osteoclasts per unit bone surface area (N. Oc/BS, /mm2) (days 9 and 16) were higher and lower, respectively, in the OM group than in the DMEM group. Moreover, the calvarial bone survived for at least 16 days in vitro, as indicated by tartrate-resistant acid phosphatase (TRAP)-positive staining. Micro-CT assessment revealed that the bone surface (BS), bone volume (BV), bone surface density (BS/TV(Tissue volume)) and percent bone volume (BV/TV) were greater in the OM group than in the DMEM group except at baseline on day 2. All bone parameters of calvariae cultured in OM and OM-control conditions were not significantly different on days 2 and 23. Thus, the radiation dose from micro-CT in our study design had no perceptible effect on the formation of mouse calvarial bone in vitro.
Collapse
Affiliation(s)
- Ren-Jian Zheng
- Department of Prosthodontics, Stomatological Hospital of Chongqing Medical University, No. 426 Songshibei Road, Yubei, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Jin-Lin Song
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China; College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Xiao-Hong Wu
- Department of Prosthodontics, Stomatological Hospital of Chongqing Medical University, No. 426 Songshibei Road, Yubei, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - David C Watts
- School of Medical Sciences and Photon Science Institute, University of Manchester, Manchester M13 9PL, UK; Institute of Material Science and Technology, Friedrich-Schiller-University, Jena, Löbdergraben 32, 07743, Germany.
| |
Collapse
|
14
|
Use of in vitro bone models to screen for altered bone metabolism, osteopathies, and fracture healing: challenges of complex models. Arch Toxicol 2020; 94:3937-3958. [PMID: 32910238 PMCID: PMC7655582 DOI: 10.1007/s00204-020-02906-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Approx. every third hospitalized patient in Europe suffers from musculoskeletal injuries or diseases. Up to 20% of these patients need costly surgical revisions after delayed or impaired fracture healing. Reasons for this are the severity of the trauma, individual factors, e.g, the patients’ age, individual lifestyle, chronic diseases, medication, and, over 70 diseases that negatively affect the bone quality. To investigate the various disease constellations and/or develop new treatment strategies, many in vivo, ex vivo, and in vitro models can be applied. Analyzing these various models more closely, it is obvious that many of them have limits and/or restrictions. Undoubtedly, in vivo models most completely represent the biological situation. Besides possible species-specific differences, ethical concerns may question the use of in vivo models especially for large screening approaches. Challenging whether ex vivo or in vitro bone models can be used as an adequate replacement for such screenings, we here summarize the advantages and challenges of frequently used ex vivo and in vitro bone models to study disturbed bone metabolism and fracture healing. Using own examples, we discuss the common challenge of cell-specific normalization of data obtained from more complex in vitro models as one example of the analytical limits which lower the full potential of these complex model systems.
Collapse
|
15
|
Laranga R, Duchi S, Ibrahim T, Guerrieri AN, Donati DM, Lucarelli E. Trends in Bone Metastasis Modeling. Cancers (Basel) 2020; 12:E2315. [PMID: 32824479 PMCID: PMC7464021 DOI: 10.3390/cancers12082315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Bone is one of the most common sites for cancer metastasis. Bone tissue is composed by different kinds of cells that coexist in a coordinated balance. Due to the complexity of bone, it is impossible to capture the intricate interactions between cells under either physiological or pathological conditions. Hence, a variety of in vivo and in vitro approaches have been developed. Various models of tumor-bone diseases are routinely used to provide valuable information on the relationship between metastatic cancer cells and the bone tissue. Ideally, when modeling the metastasis of human cancers to bone, models would replicate the intra-tumor heterogeneity, as well as the genetic and phenotypic changes that occur with human cancers; such models would be scalable and reproducible to allow high-throughput investigation. Despite the continuous progress, there is still a lack of solid, amenable, and affordable models that are able to fully recapitulate the biological processes happening in vivo, permitting a correct interpretation of results. In the last decades, researchers have demonstrated that three-dimensional (3D) methods could be an innovative approach that lies between bi-dimensional (2D) models and animal models. Scientific evidence supports that the tumor microenvironment can be better reproduced in a 3D system than a 2D cell culture, and the 3D systems can be scaled up for drug screening in the same way as the 2D systems thanks to the current technologies developed. However, 3D models cannot completely recapitulate the inter- and intra-tumor heterogeneity found in patients. In contrast, ex vivo cultures of fragments of bone preserve key cell-cell and cell-matrix interactions and allow the study of bone cells in their natural 3D environment. Moreover, ex vivo bone organ cultures could be a better model to resemble the human pathogenic metastasis condition and useful tools to predict in vivo response to therapies. The aim of our review is to provide an overview of the current trends in bone metastasis modeling. By showing the existing in vitro and ex vivo systems, we aspire to contribute to broaden the knowledge on bone metastasis models and make these tools more appealing for further translational studies.
Collapse
Affiliation(s)
- Roberta Laranga
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| | - Serena Duchi
- BioFab3D@ACMD, St Vincent’s Hospital, Melbourne, VIC 3065, Australia;
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Ania Naila Guerrieri
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| | - Davide Maria Donati
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
- Rizzoli Laboratory Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Via di Barbiano 1/10, 40136 Bologna, Italy
- 3rd Orthopaedic and Traumatologic Clinic Prevalently Oncologic, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy
| | - Enrico Lucarelli
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| |
Collapse
|
16
|
Cui H, Esworthy T, Zhou X, Hann SY, Glazer RI, Li R, Zhang LG. Engineering a Novel 3D Printed Vascularized Tissue Model for Investigating Breast Cancer Metastasis to Bone. Adv Healthc Mater 2020; 9:e1900924. [PMID: 31846231 PMCID: PMC7297662 DOI: 10.1002/adhm.201900924] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Cancer metastases are a challenge for cancer treatment due to their organ specificity and pathophysiological complexity. Engineering 3D in vitro models capable of replicating native cancer dissemination can significantly improve the understanding of cancer biology and can help to guide the development of more effective treatments. In order to better mimic the behavior of native cancer, a triculture metastatic model is created using a stereolithography printing technique with optimized inks for investigating the invasion of breast cancer (BrCa) cells into vascularized bone tissue. The printed system allows to study transendothelial migration and the colony-forming behavior of metastatic BrCa cells. The key steps of BrCa cell progression including expansion, migration, and colonization are continuously monitored and the interactions between cancer cells, vascular cells, and bone cells are systematically investigated. The study results demonstrate that the 3D printed tissue construct by incorporating multiple cells and various favorable ink matrices provides a suitable model to study the interaction between these cells in a complex vascular microenvironment. As such, the 3D printed tricultured model may serve as a valuable tool for studying metastatic breast cancer progression in bone.
Collapse
Affiliation(s)
- Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, 3590 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, 3590 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, 3590 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Sung Yun Hann
- Department of Mechanical and Aerospace Engineering, The George Washington University, 3590 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Robert I Glazer
- Department of Oncology, and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Rong Li
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, 20052, USA
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, 3590 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
- Department of Electrical and Computer Engineering, The George Washington University, Washington, DC, 20052, USA
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA
- Department of Medicine, The George Washington University, Washington, DC, 20052, USA
| |
Collapse
|
17
|
Rao SR, Edwards CM, Edwards JR. Modeling the Human Bone-Tumor Niche: Reducing and Replacing the Need for Animal Data. JBMR Plus 2020; 4:e10356. [PMID: 32258970 PMCID: PMC7117847 DOI: 10.1002/jbm4.10356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/17/2022] Open
Abstract
Bone is the most common site for cancer metastasis. Understanding the interactions within the complex, heterogeneous bone-tumor microenvironment is essential for the development of new therapeutics. Various animal models of tumor-induced bone disease are routinely used to provide valuable information on the relationship between cancer cells and the skeleton. However, new model systems exist that offer an alternative approach to the use of animals and might more accurately reveal the cellular interactions occurring within the human bone-tumor niche. This review highlights replacement models that mimic the bone microenvironment and where cancer metastases and tumor growth might be assessed alongside bone turnover. Such culture models include the use of calcified regions of animal tissue and scaffolds made from bone mineral hydroxyapatite, synthetic polymers that can be manipulated during manufacture to create structures resembling trabecular bone surfaces, gel composites that can be modified for stiffness and porosity to resemble conditions in the tumor-bone microenvironment. Possibly the most accurate model system involves the use of fresh human bone samples, which can be cultured ex vivo in the presence of human tumor cells and demonstrate similar cancer cell-bone cell interactions as described in vivo. In addition, the use of mathematical modeling and computational biology approaches provide an alternative to preliminary animal testing. The use of such models offers the capacity to mimic significant elements of the human bone-tumor environment, and complement, refine, or replace the use of preclinical models. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Srinivasa R Rao
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences University of Oxford Oxford UK.,Nuffield Department of Surgical Sciences University of Oxford Oxford UK
| | - Claire M Edwards
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences University of Oxford Oxford UK.,Nuffield Department of Surgical Sciences University of Oxford Oxford UK
| | - James R Edwards
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences University of Oxford Oxford UK
| |
Collapse
|
18
|
Bellido T, Delgado-Calle J. Ex Vivo Organ Cultures as Models to Study Bone Biology. JBMR Plus 2020; 4:JBM410345. [PMID: 32161838 PMCID: PMC7059827 DOI: 10.1002/jbm4.10345] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/16/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
The integrity of the skeleton is maintained by the coordinated and balanced activities of the bone cells. Osteoclasts resorb bone, osteoblasts form bone, and osteocytes orchestrate the activities of osteoclasts and osteoblasts. A variety of in vitro approaches has been used in an attempt to reproduce the complex in vivo interactions among bone cells under physiological as well as pathological conditions and to test new therapies. Most cell culture systems lack the proper extracellular matrix, cellular diversity, and native spatial distribution of the components of the bone microenvironment. In contrast, ex vivo cultures of fragments of intact bone preserve key cell-cell and cell-matrix interactions and allow the study of bone cells in their natural 3D environment. Further, bone organ cultures predict the in vivo responses to genetic and pharmacologic interventions saving precious time and resources. Moreover, organ cultures using human bone reproduce human conditions and are a useful tool to test patient responses to therapeutic agents. Thus, these ex vivo approaches provide a platform to perform research in bone physiology and pathophysiology. In this review, we describe protocols optimized in our laboratories to establish ex vivo bone organ cultures and provide technical hints and suggestions. In addition, we present examples on how this technical approach can be employed to study osteocyte biology, drug responses in bone, cancer-induced bone disease, and cross-talk between bone and other organs © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Teresita Bellido
- Department of Anatomy, Cell Biology & Physiology Indiana University School of Medicine Indianapolis IN USA.,Division of Endocrinology, Department of Medicine Indiana University School of Medicine Indianapolis IN USA.,Indiana Center for Musculoskeletal Health Indiana University School of Medicine Indianapolis IN USA.,Richard L. Roudebush Veterans Affairs Medical Center Indianapolis IN USA
| | - Jesus Delgado-Calle
- Department of Anatomy, Cell Biology & Physiology Indiana University School of Medicine Indianapolis IN USA.,Indiana Center for Musculoskeletal Health Indiana University School of Medicine Indianapolis IN USA.,Richard L. Roudebush Veterans Affairs Medical Center Indianapolis IN USA.,Division of Hematology/Oncology, Department of Medicine Indiana University School of Medicine Indianapolis IN USA
| |
Collapse
|
19
|
Molla MDS, Katti DR, Iswara J, Venkatesan R, Paulmurugan R, Katti KS. Prostate Cancer Phenotype Influences Bone Mineralization at Metastasis: A Study Using an In Vitro Prostate Cancer Metastasis Testbed. JBMR Plus 2020; 4:e10256. [PMID: 32083238 PMCID: PMC7017885 DOI: 10.1002/jbm4.10256] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/01/2019] [Accepted: 11/13/2019] [Indexed: 12/18/2022] Open
Abstract
In this study, two types of prostate cancer cell lines, highly metastatic PC-3 and low metastatic MDA PCa 2b (PCa) were cultured on bone mimetic scaffolds to recapitulate metastasis to bone. A unique in vitro 3D tumor model that uses a sequential culture (SC) of human mesenchymal stem cells followed by seeding with cancer cells after bone formation was initiated to study the phenotype-specific interaction between prostate cancer cells and bone microenvironment. The PCa cells were observed to be less prolific and less metastatic, and to form multicellular tumoroids in the bone microenvironment, whereas PC-3 cells were more prolific and were highly metastatic, and did not form multicellular tumoroids in the bone microenvironment. The metastatic process exhibited by these two prostate cancer cell lines showed a significant and different effect on bone mineralization and extracellular matrix formation. Excessive bone formation in the presence of PC-3 and significant osteolysis in the presence of PCa were observed, which was also indicated by osteocalcin and MMP-9 expression as measured by ELISA and qRT-PCR. The field emission scanning electron microscopy images revealed that the structure of mineralized collagen in the presence of PC-3 is different than the one observed in healthy bone. All experimental results indicated that both osteolytic and osteoblastic bone lesions can be recapitulated in our tumor testbed model and that different cancer phenotypes have a very different influence on bone at metastasis. The 3D in vitro model presented in this study provides an improved, reproducible, and controllable system that is a useful tool to elucidate osteotropism of prostate cancer cells. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- MD Shahjahan Molla
- Center for Engineered Cancer TestbedsNorth Dakota State UniversityFargoNDUSA
- Department of Civil and Environmental EngineeringNorth Dakota State UniversityFargoNDUSA
- Scintillon InstituteSan DiegoUSA
| | - Dinesh R Katti
- Center for Engineered Cancer TestbedsNorth Dakota State UniversityFargoNDUSA
- Department of Civil and Environmental EngineeringNorth Dakota State UniversityFargoNDUSA
| | - Jairam Iswara
- Department of Urology, Saint Elizabeth's Medical CenterTufts UniversityBostonMAUSA
| | - Renugopalkrishnan Venkatesan
- Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMAUSA
- Center for Life SciencesBoston Children's Hospital, Harvard Medical School, BostonMassachusettsUSA
| | - Ramasamy Paulmurugan
- Department of RadiologyCellular Pathway Imaging Laboratory (CPIL), Stanford University School of MedicinePalo AltoCAUSA
| | - Kalpana S Katti
- Center for Engineered Cancer TestbedsNorth Dakota State UniversityFargoNDUSA
| |
Collapse
|
20
|
Sieberath A, Della Bella E, Ferreira AM, Gentile P, Eglin D, Dalgarno K. A Comparison of Osteoblast and Osteoclast In Vitro Co-Culture Models and Their Translation for Preclinical Drug Testing Applications. Int J Mol Sci 2020; 21:E912. [PMID: 32019244 PMCID: PMC7037207 DOI: 10.3390/ijms21030912] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
As the population of western societies on average ages, the number of people affected by bone remodeling-associated diseases such as osteoporosis continues to increase. The development of new therapeutics is hampered by the high failure rates of drug candidates during clinical testing, which is in part due to the poor predictive character of animal models during preclinical drug testing. Co-culture models of osteoblasts and osteoclasts offer an alternative to animal testing and are considered to have the potential to improve drug development processes in the future. However, a robust, scalable, and reproducible 3D model combining osteoblasts and osteoclasts for preclinical drug testing purposes has not been developed to date. Here we review various types of osteoblast-osteoclast co-culture models and outline the remaining obstacles that must be overcome for their successful translation.
Collapse
Affiliation(s)
- Alexander Sieberath
- School of Engineering, Newcastle University, Newcastle-Upon-Tyne NE1 7RU, UK; (A.S.); (A.M.F.); (P.G.)
| | - Elena Della Bella
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland; (E.D.B.); (D.E.)
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle-Upon-Tyne NE1 7RU, UK; (A.S.); (A.M.F.); (P.G.)
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Newcastle-Upon-Tyne NE1 7RU, UK; (A.S.); (A.M.F.); (P.G.)
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland; (E.D.B.); (D.E.)
| | - Kenny Dalgarno
- School of Engineering, Newcastle University, Newcastle-Upon-Tyne NE1 7RU, UK; (A.S.); (A.M.F.); (P.G.)
| |
Collapse
|
21
|
Shokoohmand A, Ren J, Baldwin J, Atack A, Shafiee A, Theodoropoulos C, Wille ML, Tran PA, Bray LJ, Smith D, Chetty N, Pollock PM, Hutmacher DW, Clements JA, Williams ED, Bock N. Microenvironment engineering of osteoblastic bone metastases reveals osteomimicry of patient-derived prostate cancer xenografts. Biomaterials 2019; 220:119402. [PMID: 31400612 DOI: 10.1016/j.biomaterials.2019.119402] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/16/2019] [Accepted: 07/30/2019] [Indexed: 01/01/2023]
Abstract
Representative in vitro models that mimic the native bone tumor microenvironment are warranted to support the development of more successful treatments for bone metastases. Here, we have developed a primary cell 3D model consisting of a human osteoblast-derived tissue-engineered construct (hOTEC) indirectly co-cultured with patient-derived prostate cancer xenografts (PDXs), in order to study molecular interactions in a patient-derived microenvironment context. The engineered biomimetic microenvironment had high mineralization and embedded osteocytes, and supported a high degree of cancer cell osteomimicry at the gene, protein and mineralization levels when co-cultured with prostate cancer PDXs from a lymph node metastasis (LuCaP35) and bone metastasis (BM18) from patients with primary prostate cancer. This fully patient-derived model is a promising tool for the assessment of new molecular mechanisms and as a personalized pre-clinical platform for therapy testing for patients with prostate cancer bone metastases.
Collapse
Affiliation(s)
- Ali Shokoohmand
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Jiongyu Ren
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Jeremy Baldwin
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Anthony Atack
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Abbas Shafiee
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Christina Theodoropoulos
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Marie-Luise Wille
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Phong A Tran
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Laura J Bray
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Deborah Smith
- Cancer Pathology Research Group, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia; Department of Anatomical Pathology, Mater Hospital Brisbane, QLD, Australia
| | - Naven Chetty
- Cancer Pathology Research Group, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia; Department of Anatomical Pathology, Mater Hospital Brisbane, QLD, Australia
| | - Pamela M Pollock
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Judith A Clements
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Elizabeth D Williams
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Nathalie Bock
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia.
| |
Collapse
|
22
|
Engineered bone for probing organotypic growth and therapy response of prostate cancer tumoroids in vitro. Biomaterials 2019; 197:296-304. [PMID: 30682644 DOI: 10.1016/j.biomaterials.2019.01.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/24/2018] [Accepted: 01/20/2019] [Indexed: 01/23/2023]
Abstract
Mechanistic analysis of metastatic prostate cancer (PCa) biology and therapy response critically depends upon clinically relevant three-dimensional (3D) bone-like, organotypic culture. We here combine an engineered bone-mimetic environment (BME) with longitudinal microscopy to test the growth and therapy response of 3D PCa tumoroids. Besides promoting both tumor-cell autonomous and microenvironment-dependent growth in PCa cell lines and patient-derived xenograft cells, the BME enables in vivo-like tumor cell response to therapy, and reveals bone stroma dependent resistance to chemotherapy and BME-targeted localization and induction of cytoxicity by Radium-223. The BME platform will allow the propagation, compound screening and mechanistic dissection of patient-derived bone tumor isolates and applications toward personalized medicine.
Collapse
|
23
|
Abstract
This chapter elaborates on the state-of-the-art experimental procedures utilized in ex-vivo model systems of cancer-bone cell interactions under "static and dynamic" culture conditions and their potential use to understand cellular and molecular mechanisms as well as drug testing and discovery. An additional focus of this chapter is to provide details of how to incorporate varying oxygen tension, viz., hypoxic, normoxic, and hyperoxic, in such studies and regulate the bone biology toward dissociation of the bone remodeling stages to achieve only "bone resorption" or "bone formation" individually.
Collapse
Affiliation(s)
- Erdjan Salih
- Department of Periodontology, Henry M. Goldman School of Dental Medicine, Boston University Medical Center, Boston, MA, USA.
| |
Collapse
|
24
|
Choudhary S, Ramasundaram P, Dziopa E, Mannion C, Kissin Y, Tricoli L, Albanese C, Lee W, Zilberberg J. Human ex vivo 3D bone model recapitulates osteocyte response to metastatic prostate cancer. Sci Rep 2018; 8:17975. [PMID: 30568232 PMCID: PMC6299475 DOI: 10.1038/s41598-018-36424-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/20/2018] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer deaths among American men. Unfortunately, there is no cure once the tumor is established within the bone niche. Although osteocytes are master regulators of bone homeostasis and remodeling, their role in supporting PCa metastases remains poorly defined. This is largely due to a lack of suitable ex vivo models capable of recapitulating the physiological behavior of primary osteocytes. To address this need, we integrated an engineered bone tissue model formed by 3D-networked primary human osteocytes, with conditionally reprogrammed (CR) primary human PCa cells. CR PCa cells induced a significant increase in the expression of fibroblast growth factor 23 (FGF23) by osteocytes. The expression of the Wnt inhibitors sclerostin and dickkopf-1 (Dkk-1), exhibited contrasting trends, where sclerostin decreased while Dkk-1 increased. Furthermore, alkaline phosphatase (ALP) was induced with a concomitant increase in mineralization, consistent with the predominantly osteoblastic PCa-bone metastasis niche seen in patients. Lastly, we confirmed that traditional 2D culture failed to reproduce these key responses, making the use of our ex vivo engineered human 3D bone tissue an ideal platform for modeling PCa-bone interactions.
Collapse
Affiliation(s)
- Saba Choudhary
- Department of Biomedical Engineering, Chemistry and Biological Sciences, Stevens Institute of Technology, Hoboken, NJ, USA
| | - Poornema Ramasundaram
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ, USA
| | - Eugenia Dziopa
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ, USA
| | - Ciaran Mannion
- Department of Pathology, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Yair Kissin
- Insall Scott Kelly Institute for Orthopedics and Sports Medicine, New York, NY, USA.,Hackensack University Medical Center, Hackensack, NJ, USA.,Lenox Hill Hospital, New York, NY, USA
| | - Lucas Tricoli
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Christopher Albanese
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Woo Lee
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken, NJ, USA
| | - Jenny Zilberberg
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ, USA.
| |
Collapse
|
25
|
Fitzgerald W, Gomez-Lopez N, Erez O, Romero R, Margolis L. Extracellular vesicles generated by placental tissues ex vivo: A transport system for immune mediators and growth factors. Am J Reprod Immunol 2018; 80:e12860. [PMID: 29726582 PMCID: PMC6021205 DOI: 10.1111/aji.12860] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 03/27/2018] [Indexed: 12/12/2022] Open
Abstract
PROBLEM To study the mechanisms of placenta function and the role of extracellular vesicles (EVs) in pregnancy, it is necessary to develop an ex vivo system that retains placental cytoarchitecture and the primary metabolic aspects, in particular the release of EVs and soluble factors. Here, we developed such a system and investigated the pattern of secretion of cytokines, growth factors, and extracellular vesicles by placental villous and amnion tissues ex vivo. METHODS OF STUDY Placental villous and amnion explants were cultured for 2 weeks at the air/liquid interface and their morphology and the released cytokines and EVs were analyzed. Cytokines were analyzed with multiplexed bead assays, and individual EVs were analyzed with recently developed techniques that involved EV capture with magnetic nanoparticles coupled to anti-EV antibodies and flow cytometry. RESULTS Ex vivo tissues (i) remained viable and preserved their cytoarchitecture; (ii) maintained secretion of cytokines and growth factors; (iii) released EVs of syncytiotrophoblast and amnion epithelial cell origins that contain cytokines and growth factors. CONCLUSION A system of ex vivo placental villous and amnion tissues can be used as an adequate model to study placenta metabolic activity in normal and complicated pregnancies, in particular to characterize EVs by their surface markers and by encapsulated proteins. Establishment and benchmarking the placenta ex vivo system may provide new insight in the functional status of this organ in various placental disorders, particularly regarding the release of EVs and cytokines. Such EVs may have a prognostic value for pregnancy complications.
Collapse
Affiliation(s)
- Wendy Fitzgerald
- Section of Intercellular Interactions, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Leonid Margolis
- Section of Intercellular Interactions, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| |
Collapse
|
26
|
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: 3.5] [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
|
27
|
Harrison RA, Nam JY, Weathers SP, DeMonte F. Intracranial dural, calvarial, and skull base metastases. HANDBOOK OF CLINICAL NEUROLOGY 2018; 149:205-225. [PMID: 29307354 DOI: 10.1016/b978-0-12-811161-1.00014-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metastatic disease to the intracranial dura, the calvarium, and the skull base is relatively uncommon but presents unique diagnostic and management challenges in the patient with cancer. Modern imaging techniques have facilitated the detection of intracranial tumor deposits, leading to increased incidence. While dural and calvarial metastases often present with nonspecific symptoms, skull base metastases present with distinct clinical syndromes dependent on the local neurovascular structures affected. Intracranial dural metastases can often be confused with meningioma and pose a diagnostic challenge, as well as significant neurologic morbidity, especially in the setting of hemorrhage. Surgical intervention may be helpful in selected patients for symptomatic relief as well as survival benefit. Management paradigms need to take into account the relative risks, benefits, and likely outcomes for each possible modality of treatment. Surgical excision is useful in many patients and in combination with radiation therapy can provide significant palliation. While medical therapy is rarely an initial therapy in these entities, it may be of added benefit dependent on the underlying tumor histology and prior treatment history. Occasionally treatment with curative intent is justified.
Collapse
Affiliation(s)
- Rebecca A Harrison
- Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Joo Yeon Nam
- Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Shiao-Pei Weathers
- Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Franco DeMonte
- Department of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
28
|
Lovitt CJ, Shelper TB, Avery VM. Cancer drug discovery: recent innovative approaches to tumor modeling. Expert Opin Drug Discov 2017; 11:885-94. [PMID: 27454169 DOI: 10.1080/17460441.2016.1214562] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Cell culture models have been at the heart of anti-cancer drug discovery programs for over half a century. Advancements in cell culture techniques have seen the rapid evolution of more complex in vitro cell culture models investigated for use in drug discovery. Three-dimensional (3D) cell culture research has become a strong focal point, as this technique permits the recapitulation of the tumor microenvironment. Biologically relevant 3D cellular models have demonstrated significant promise in advancing cancer drug discovery, and will continue to play an increasing role in the future. AREAS COVERED In this review, recent advances in 3D cell culture techniques and their application in tumor modeling and anti-cancer drug discovery programs are discussed. The topics include selection of cancer cells, 3D cell culture assays (associated endpoint measurements and analysis), 3D microfluidic systems and 3D bio-printing. EXPERT OPINION Although advanced cancer cell culture models and techniques are becoming commonplace in many research groups, the use of these approaches has yet to be fully embraced in anti-cancer drug applications. Furthermore, limitations associated with analyzing information-rich biological data remain unaddressed.
Collapse
Affiliation(s)
- Carrie J Lovitt
- a Discovery Biology, Eskitis Institute for Drug Discovery , Griffith University , Nathan , Australia
| | - Todd B Shelper
- a Discovery Biology, Eskitis Institute for Drug Discovery , Griffith University , Nathan , Australia
| | - Vicky M Avery
- a Discovery Biology, Eskitis Institute for Drug Discovery , Griffith University , Nathan , Australia
| |
Collapse
|
29
|
Zhao YP, Ye WL, Liu DZ, Cui H, Cheng Y, Liu M, Zhang BL, Mei QB, Zhou SY. Redox and pH dual sensitive bone targeting nanoparticles to treat breast cancer bone metastases and inhibit bone resorption. NANOSCALE 2017; 9:6264-6277. [PMID: 28470315 DOI: 10.1039/c7nr00962c] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Bone is an especially prone metastatic site for breast cancer, and to block the vicious cycle between bone resorption and tumor growth is an important strategy for the treatment of breast cancer bone metastasis. In this paper, pH- and redox-sensitive as well as breast cancer bone metastasis-targeting nanoparticles (DOX@ALN-(HA-PASP)CL) were prepared, and also their anti-tumor activity and anti-bone resorption effect were investigated in detail. The in vitro experimental results indicated that DOX released from DOX@ALN-(HA-PASP)CL exhibited a GSH-, DTT- and pH-dependent manner. Moreover, in an in vitro 3D breast cancer bone metastasis model, DOX@ALN-(HA-PASP)CL decreased bone resorption through inhibiting the proliferation of human breast cancer cells (MDA-MB-231 cells) and reducing the activity of osteoclasts. The in vivo experimental results indicated that a large amount of DOX was delivered to a breast cancer bone metastasis site after tumor-bearing mice were treated with DOX@ALN-(HA-PASP)CL; meanwhile, DOX@ALN-(HA-PASP)CL significantly decreased the tumor volume and bone resorption in tumor-bearing mice without causing obvious systemic toxicity. In conclusion, the in vitro and in vivo experimental results indicate that DOX@ALN-(HA-PASP)CL has great potential in the treatment of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Yi-Pu Zhao
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Narkhede AA, Shevde LA, Rao SS. Biomimetic strategies to recapitulate organ specific microenvironments for studying breast cancer metastasis. Int J Cancer 2017; 141:1091-1109. [PMID: 28439901 DOI: 10.1002/ijc.30748] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/22/2017] [Accepted: 04/11/2017] [Indexed: 12/14/2022]
Abstract
The progression of breast cancer from the primary tumor setting to the metastatic setting is the critical event defining Stage IV disease, no longer considered curable. The microenvironment at specific organ sites is known to play a key role in influencing the ultimate fate of metastatic cells; yet microenvironmental mediated-molecular mechanisms underlying organ specific metastasis in breast cancer are not well understood. This review discusses biomimetic strategies employed to recapitulate metastatic organ microenvironments, particularly, bone, liver, lung and brain to elucidate the mechanisms dictating metastatic breast cancer cell homing and colonization. These biomimetic strategies include in vitro techniques such as biomaterial-based co-culturing techniques, microfluidics, organ-mimetic chips, bioreactor technologies, and decellularized matrices as well as cutting edge in vivo techniques to better understand the interactions between metastatic breast cancer cells and the stroma at the metastatic site. The advantages and disadvantages of these systems are discussed. In addition, how creation of biomimetic models will impact breast cancer metastasis research and their broad utility is explored.
Collapse
Affiliation(s)
- Akshay A Narkhede
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL
| | - Lalita A Shevde
- Department of Pathology and Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL
| |
Collapse
|
31
|
Krzeszinski JY, Schwaid AG, Cheng WY, Jin Z, Gallegos ZR, Saghatelian A, Wan Y. Lipid Osteoclastokines Regulate Breast Cancer Bone Metastasis. Endocrinology 2017; 158:477-489. [PMID: 27967239 PMCID: PMC5460780 DOI: 10.1210/en.2016-1570] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/13/2016] [Indexed: 11/19/2022]
Abstract
Bone metastasis is a deadly consequence of cancers, in which osteoclast forms a vicious cycle with tumor cells. Bone metastasis attenuation by clinical usage of osteoclast inhibitors and in our osteopetrotic mouse genetic models with β-catenin constitutive activation or peroxisome proliferator-activated receptor γ deficiency fully support the important role of osteoclast in driving the bone metastatic niche. However, the mechanisms for this "partnership in crime" are underexplored. Here we show that osteoclasts reprogram their lipid secretion to support cancer cells. Metabolomic profiling reveals elevated prometastatic arachidonic acid (AA) but reduced antimetastatic lysophosphatidylcholines (LPCs). This shift in lipid osteoclastokines synergistically stimulates tumor cell proliferation, migration, survival, and expression of prometastatic genes. Pharmacologically, combined treatment with LPCs and BW-755C, an inhibitor of AA signaling via blocking lipoxygenase and cyclooxygenase, impedes breast cancer bone metastasis. Our findings elucidate key paracrine mechanisms for the osteoclast-cancer vicious cycle and uncover important therapeutic targets for bone metastasis.
Collapse
Affiliation(s)
| | - Adam G. Schwaid
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138; and
| | | | | | - Zachary R. Gallegos
- Department of Pharmacology and
- Simmons Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390;
| | - Alan Saghatelian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138; and
- Clayton Foundation Laboratories of Peptide Biology and Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Yihong Wan
- Department of Pharmacology and
- Simmons Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390;
| |
Collapse
|
32
|
|
33
|
Coughlin TR, Romero-Moreno R, Mason DE, Nystrom L, Boerckel JD, Niebur GL, Littlepage LE. Bone: A Fertile Soil for Cancer Metastasis. Curr Drug Targets 2017; 18:1281-1295. [PMID: 28025941 PMCID: PMC7932754 DOI: 10.2174/1389450117666161226121650] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/06/2016] [Accepted: 10/26/2016] [Indexed: 02/08/2023]
Abstract
Bone is one of the most common and most dangerous sites for metastatic growth across cancer types, and bone metastasis remains incurable. Unfortunately, the processes by which cancers preferentially metastasize to bone are still not well understood. In this review, we summarize the morphological features, physical properties, and cell signaling events that make bone a unique site for metastasis and bone remodeling. The signaling crosstalk between the tumor cells and bone cells begins a vicious cycle - a self-sustaining feedback loop between the tumor cells and the bone microenvironment composed of osteoclasts, osteoblasts, other bone marrow cells, bone matrix, and vasculature to support both tumor growth and bone destruction. Through this crosstalk, bone provides a fertile microenvironment that can harbor dormant tumor cells, sometimes for long periods, and support their growth by releasing cytokines as the bone matrix is destroyed, similar to providing nutrients for a seed to germinate in soil. However, few models exist to study the late stages of bone colonization by metastatic tumor cells. We describe some of the current methodologies used to study bone metastasis, highlighting the limitations of these methods and alternative future strategies to be used to study bone metastasis. While <i>in vivo</i> animal and patient studies may provide the gold standard for studying metastasis, <i>ex vivo</i> models can be used as an alternative to enable more controlled experiments designed to study the late stages of bone metastasis.
Collapse
Affiliation(s)
- Thomas R. Coughlin
- Harper Cancer Research Institute, South Bend, IN
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN
| | - Ricardo Romero-Moreno
- Harper Cancer Research Institute, South Bend, IN
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN
| | - Devon E. Mason
- Harper Cancer Research Institute, South Bend, IN
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN
| | - Lukas Nystrom
- Department of Orthopaedic Surgery and Rehabilitation, Loyola University Chicago, Stritch School of Medicine, Maywood, IL
| | - Joel D. Boerckel
- Harper Cancer Research Institute, South Bend, IN
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN
| | - Glen L. Niebur
- Harper Cancer Research Institute, South Bend, IN
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN
| | - Laurie E. Littlepage
- Harper Cancer Research Institute, South Bend, IN
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN
| |
Collapse
|
34
|
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: 2.8] [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
|
35
|
Xu L, Mohammad KS, Wu H, Crean C, Poteat B, Cheng Y, Cardoso AA, Machal C, Hanenberg H, Abonour R, Kacena MA, Chirgwin J, Suvannasankha A, Srour EF. Cell Adhesion Molecule CD166 Drives Malignant Progression and Osteolytic Disease in Multiple Myeloma. Cancer Res 2016; 76:6901-6910. [PMID: 27634757 DOI: 10.1158/0008-5472.can-16-0517] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 08/04/2016] [Accepted: 08/19/2016] [Indexed: 12/31/2022]
Abstract
Multiple myeloma is incurable once osteolytic lesions have seeded at skeletal sites, but factors mediating this deadly pathogenic advance remain poorly understood. Here, we report evidence of a major role for the cell adhesion molecule CD166, which we discovered to be highly expressed in multiple myeloma cell lines and primary bone marrow cells from patients. CD166+ multiple myeloma cells homed more efficiently than CD166- cells to the bone marrow of engrafted immunodeficient NSG mice. CD166 silencing in multiple myeloma cells enabled longer survival, a smaller tumor burden, and less osteolytic lesions, as compared with mice bearing control cells. CD166 deficiency in multiple myeloma cell lines or CD138+ bone marrow cells from multiple myeloma patients compromised their ability to induce bone resorption in an ex vivo organ culture system. Furthermore, CD166 deficiency in multiple myeloma cells also reduced the formation of osteolytic disease in vivo after intratibial engraftment. Mechanistic investigation revealed that CD166 expression in multiple myeloma cells inhibited osteoblastogenesis of bone marrow-derived osteoblast progenitors by suppressing Runx2 gene expression. Conversely, CD166 expression in multiple myeloma cells promoted osteoclastogenesis by activating TRAF6-dependent signaling pathways in osteoclast progenitors. Overall, our results define CD166 as a pivotal director in multiple myeloma cell homing to the bone marrow and multiple myeloma progression, rationalizing its further study as a candidate therapeutic target for multiple myeloma treatment. Cancer Res; 76(23); 6901-10. ©2016 AACR.
Collapse
Affiliation(s)
- Linlin Xu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Khalid S Mohammad
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hao Wu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Colin Crean
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Bradley Poteat
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yinghua Cheng
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Angelo A Cardoso
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Helmut Hanenberg
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Dusseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Rafat Abonour
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - John Chirgwin
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.,Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, Indiana
| | - Attaya Suvannasankha
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.,Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, Indiana
| | - Edward F Srour
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana. .,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
36
|
Marino S, Staines KA, Brown G, Howard-Jones RA, Adamczyk M. Models of ex vivo explant cultures: applications in bone research. BONEKEY REPORTS 2016; 5:818. [PMID: 27408711 PMCID: PMC4926536 DOI: 10.1038/bonekey.2016.49] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/04/2016] [Indexed: 01/09/2023]
Abstract
Ex vivo explant culture models are powerful tools in bone research. They allow investigation of bone and cartilage responses to specific stimuli in a controlled manner that closely mimics the in vivo processes. Because of limitations in obtaining healthy human bone samples the explant growth of animal tissue serves as a platform to study the complex physico-chemical properties of the bone. Moreover, these models enable preserving important cell-cell and cell-matrix interactions in order to better understand the behaviour of cells in their natural three-dimensional environment. Thus, the use of bone ex vivo explant cultures can frequently be of more physiological relevance than the use of two-dimensional primary cells grown in vitro. Here, we describe isolation and ex vivo growth of different animal bone explant models including metatarsals, femoral heads, calvaria, mandibular slices and trabecular cores. We also describe how these explants are utilised to study bone development, cartilage and bone metabolism, cancer-induced bone diseases, stem cell-driven bone repair and mechanoadaptation. These techniques can be directly used to understand mechanisms linked with bone physiology or bone-associated diseases.
Collapse
Affiliation(s)
- Silvia Marino
- Academic Unit of Bone Biology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, Medical School, The University of Sheffield, Sheffield, UK
| | | | - Genevieve Brown
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Rachel Anne Howard-Jones
- Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Magdalena Adamczyk
- Academic Unit of Bone Biology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, Medical School, The University of Sheffield, Sheffield, UK
- Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
37
|
Suvannasankha A, Tompkins DR, Edwards DF, Petyaykina KV, Crean CD, Fournier PG, Parker JM, Sandusky GE, Ichikawa S, Imel EA, Chirgwin JM. FGF23 is elevated in multiple myeloma and increases heparanase expression by tumor cells. Oncotarget 2016; 6:19647-60. [PMID: 25944690 PMCID: PMC4637311 DOI: 10.18632/oncotarget.3794] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/10/2015] [Indexed: 01/09/2023] Open
Abstract
Multiply myeloma (MM) grows in and destroys bone, where osteocytes secrete FGF23, a hormone which affects phosphate homeostasis and aging. We report that multiple myeloma (MM) cells express receptors for and respond to FGF23. FGF23 increased mRNA for EGR1 and its target heparanase, a pro-osteolytic factor in MM. FGF23 signals through a complex of klotho and a classical FGF receptor (FGFR); both were expressed by MM cell lines and patient samples. Bone marrow plasma cells from 42 MM patients stained positively for klotho, while plasma cells from 8 patients with monoclonal gammopathy of undetermined significance (MGUS) and 6 controls were negative. Intact, active FGF23 was increased 2.9X in sera of MM patients compared to controls. FGF23 was not expressed by human MM cells, but co-culture with mouse bone increased its mRNA. The FGFR inhibitor NVP-BGJ398 blocked the heparanase response to FGF23. NVP-BGJ398 did not inhibit 8226 growth in vitro but significantly suppressed growth in bone and induction of the osteoclast regulator RANK ligand, while decreasing heparanase mRNA. The bone microenvironment provides resistance to some anti-tumor drugs but increased the activity of NVP-BGJ398 against 8226 cells. The FGF23/klotho/heparanase signaling axis may offer targets for treatment of MM in bone.
Collapse
Affiliation(s)
- Attaya Suvannasankha
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Douglas R Tompkins
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daniel F Edwards
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Katarina V Petyaykina
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Colin D Crean
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pierrick G Fournier
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jamie M Parker
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - George E Sandusky
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shoji Ichikawa
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Erik A Imel
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John M Chirgwin
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
38
|
Bone microenvironment-mediated resistance of cancer cells to bisphosphonates and impact on bone osteocytes/stem cells. Clin Exp Metastasis 2016; 33:563-88. [PMID: 27155840 DOI: 10.1007/s10585-016-9798-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 04/29/2016] [Indexed: 12/28/2022]
Abstract
Anti-resorptive bisphosphonates (BPs) have been clinically used to prevent cancer-bone metastasis and cancer-induced bone pathologies despite the fact that the phenotypic response of the cancer-bone interactions to BP exposure is "uncharted territory". This study offers unique insights into the interplay between cancer stem cells and osteocytes/osteoblasts and mesenchymal stem cells using a three-dimensional (3D) live cancer-bone interactive model. We provide extraordinary cryptic details of the biological events that occur as a result of alendronate (ALN) treatment using 3D live cancer-bone model systems under specific bone remodeling stages. While cancer cells are susceptible to BP treatment in the absence of bone, they are totally unaffected in the presence of bone. Cancer cells colonize live bone irrespective of whether the bone is committed to bone resorption or formation and hence, cancer-bone metastasis/interactions are though to be "independent of bone remodeling stages". In our 3D live bone model systems, ALN inhibited bone resorption at the osteoclast differentiation level through effects of mineral-bound ALN on osteocytes and osteoblasts. The mineral-bound ALN rendered bone incapable of osteoblast differentiation, while cancer cells colonize the bone with striking morphological adaptations which led to a conclusion that a direct anti-cancer effect of BPs in a "live or in vivo" bone microenvironment is implausible. The above studies were complemented with mass spectrometric analysis of the media from cancer-bone organ cultures in the absence and presence of ALN. The mineral-bound ALN impacts the bone organs by limiting transformation of mesenchymal stem cells to osteoblasts and leads to diminished endosteal cell population and degenerated osteocytes within the mineralized bone matrix.
Collapse
|
39
|
Salamanna F, Contartese D, Maglio M, Fini M. A systematic review on in vitro 3D bone metastases models: A new horizon to recapitulate the native clinical scenario? Oncotarget 2016; 7:44803-44820. [PMID: 27027241 PMCID: PMC5190136 DOI: 10.18632/oncotarget.8394] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/18/2016] [Indexed: 11/25/2022] Open
Affiliation(s)
- Francesca Salamanna
- Laboratory of Biocompatibility, Technological Innovation and Advanced Therapy, Rizzoli RIT, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Deyanira Contartese
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Melania Maglio
- Laboratory of Biocompatibility, Technological Innovation and Advanced Therapy, Rizzoli RIT, Rizzoli Orthopedic Institute, 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 Orthopedic Institute, Bologna, Italy
| |
Collapse
|
40
|
Mouse models for studying prostate cancer bone metastasis. BONEKEY REPORTS 2016; 5:777. [PMID: 26916039 DOI: 10.1038/bonekey.2016.4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/19/2015] [Indexed: 01/05/2023]
Abstract
Once tumor cells metastasize to the bone, the prognosis for prostate cancer patients is generally very poor. The mechanisms involved in bone metastasis, however, remain elusive, because of lack of relevant animal models. In this manuscript, we describe step-by-step protocols for the xenograft mouse models that are currently used for studying prostate cancer bone metastasis. The different routes of tumor inoculation (intraosseous, intracardiac, intravenous and orthotopic) presented are useful for exploring the biology of bone metastasis.
Collapse
|
41
|
Pagani S, Fini M, Giavaresi G, Salamanna F, Borsari V. The active role of osteoporosis in the interaction between osteoblasts and bone metastases. Bone 2015; 79:176-82. [PMID: 26057367 DOI: 10.1016/j.bone.2015.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 04/22/2015] [Accepted: 06/03/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION To minimize the severity of bone metastases and to delay their onset, it is important to analyze the underlying biological mechanisms. The present study focused on the link between OP and metastatic cells, with particular attention to osteoblast behavior. METHODS Osteoblasts (OB) were isolated from the trabecular bone of iliac crest of healthy (SHAM) and ovariectomized (OVX) adult female rats and co-cultured with MRMT-1 rat breast carcinoma cells as conditioned medium (CM) or alone (CTR) for 24h, 7 and 14 days and tested for cell viability, morphology and synthetic activity, i.e. C-terminal procollagen type I, alkaline phosphatase, osteoprotegerin, receptor activator for nuclear factor KB ligand and interleukin-8. RESULTS Osteoblast morphology showed a reduced organization in the OVX group, in particular in the CM condition. Conversely, the analysis of cell viability revealed significantly higher values in the OVXCM group with respect to the SHAMCM group at all experimental times, whereas the OVXCTR group had significantly lower values at 7 and 14 days in comparison to those of the SHAM group. ALP release was significantly lower in the CM condition than that of CTR at all timepoints, and so was procollagen type I at 7 and 14 days. The RANKL/OPG ratio showed significantly higher values in OVX osteoblasts in comparison with those of the SHAM group, both in CTR and in CM conditions at each experimental time. Finally, OVXCM showed significantly higher values of IL-8 than those of SHAMCM at 7 and 14 days. CONCLUSIONS The results clearly indicate an influence of the metastatic cells on the osteoblastic physiology at different levels: morphology, viability, release of typical proteins, and also IL-8 as a proinflammatory cytokine, especially marked by osteoporosis. Further investigations might highlight the relationship between osteoblasts and breast cancer cells, which might be useful to improve common drugs used against osteoporosis and bone metastases, by enhancing the bone deposition/tumor progression ratio.
Collapse
Affiliation(s)
- Stefania Pagani
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy; Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Department RIT Rizzoli, Rizzoli Orthopaedic Institute, Bologna, Italy.
| | - Milena Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy; Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Department RIT Rizzoli, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Gianluca Giavaresi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy; Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Department RIT Rizzoli, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Francesca Salamanna
- Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Department RIT Rizzoli, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Veronica Borsari
- Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Department RIT Rizzoli, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|
42
|
Holen I, Nutter F, Wilkinson JM, Evans CA, Avgoustou P, Ottewell PD. Human breast cancer bone metastasis in vitro and in vivo: a novel 3D model system for studies of tumour cell-bone cell interactions. Clin Exp Metastasis 2015; 32:689-702. [PMID: 26231669 DOI: 10.1007/s10585-015-9737-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/28/2015] [Indexed: 01/09/2023]
Abstract
Bone is established as the preferred site of breast cancer metastasis. However, the precise mechanisms responsible for this preference remain unidentified. In order to improve outcome for patients with advanced breast cancer and skeletal involvement, we need to better understand how this process is initiated and regulated. As bone metastasis cannot be easily studied in patients, researchers have to date mainly relied on in vivo xenograft models. A major limitation of these is that they do not contain a human bone microenvironment, increasingly considered to be an important component of metastases. In order to address this shortcoming, we have developed a novel humanised bone model, where 1 × 10(5) luciferase-expressing MDA-MB-231 or T47D human breast tumour cells are seeded on viable human subchaodral bone discs in vitro. These discs contain functional osteoclasts 2-weeks after in vitro culture and positive staining for calcine 1-week after culture demonstrating active bone resorption/formation. In vitro inoculation of MDA-MB-231 or T47D cells colonised human bone cores and remained viable for <4 weeks, however, use of matrigel to enhance adhesion or a moving platform to increase diffusion of nutrients provided no additional advantage. Following colonisation by the tumour cells, bone discs pre-seeded with MDA-MB-231 cells were implanted subcutaneously into NOD SCID mice, and tumour growth monitored using in vivo imaging for up to 6 weeks. Tumour growth progressed in human bone discs in 80 % of the animals mimicking the later stages of human bone metastasis. Immunohistochemical and PCR analysis revealed that growing MDA-MB-231 cells in human bone resulted in these cells acquiring a molecular phenotype previously associated with breast cancer bone metastases. MDA-MB-231 cells grown in human bone discs showed increased expression of IL-1B, HRAS and MMP9 and decreased expression of S100A4, whereas, DKK2 and FN1 were unaltered compared with the same cells grown in mammary fat pads of mice not implanted with human bone discs.
Collapse
Affiliation(s)
- I Holen
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - F Nutter
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - J M Wilkinson
- Department of Human Metabolism, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - C A Evans
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - P Avgoustou
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - Penelope D Ottewell
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK.
| |
Collapse
|
43
|
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: 1.7] [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
|
44
|
Zhu W, Wang M, Fu Y, Castro NJ, Fu SW, Zhang LG. Engineering a biomimetic three-dimensional nanostructured bone model for breast cancer bone metastasis study. Acta Biomater 2015; 14:164-74. [PMID: 25528534 DOI: 10.1016/j.actbio.2014.12.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/06/2014] [Accepted: 12/10/2014] [Indexed: 01/08/2023]
Abstract
Traditional breast cancer (BrCa) bone metastasis models contain many limitations with regards to controllability, reproducibility and flexibility of design. In this study, a novel biomimetic bone microenvironment was created by integrating hydroxyapatite (HA) and native bioactive factors deposited by osteogenic induction of human bone marrow mesenchymal stem cells (MSCs) within a cytocompatible chitosan hydrogel. It was found that a 10% nanocrystalline HA (nHA) chitosan scaffold exhibited the highest BrCa adhesion and proliferation when compared to chitosan scaffolds with 20% nHA, 10% and 20% microcrystalline HA as well as amorphous HA. This 3-D tunable bone scaffold can provide a biologically relevant environment, increase cell-cell and cell-matrix interactions as found in native bone, and retain the behavior of BrCa cells with different metastasis potential (i.e. highly metastatic MDA-MB-231, less metastatic MCF-7 and transfected MDA-MB-231). The co-culture of MSCs and MDA-MB-231 in this bone model illustrated that MSCs have the capacity to upregulate the expression of the well-known metastasis-associated gene metadherin within BrCa cells. In summary, this study illustrates the ability of our 3-D bone model to create a biomimetic environment conducive to recapitulating the behavior of metastatic BrCa cells, making it a promising tool for in vitro BrCa cell bone metastasis study and for the discovery of potential therapeutics.
Collapse
|
45
|
Murugan N, Kavitha L, Shinyjoy E, Rajeswari D, Vimala K, Kannan S, Gopi D. Smart rose flower like bioceramic/metal oxide dual layer coating with enhanced anti-bacterial, anti-cancer, anti-corrosive and biocompatible properties for improved orthopedic applications. RSC Adv 2015. [DOI: 10.1039/c5ra17747b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The flower like Se,Mn-HAP/ZrO2 dual layer coating on AZ91 magnesium alloy satisfies the requirements in bone cancer treatment and signifies progress in the field of implant materials.
Collapse
Affiliation(s)
- N. Murugan
- Department of Chemistry
- Periyar University
- Salem 636011
- India
| | - L. Kavitha
- Department of Physics
- School of Basic and Applied Sciences
- Central University of Tamilnadu
- Thiruvarur 610 101
- India
| | - E. Shinyjoy
- Department of Chemistry
- Periyar University
- Salem 636011
- India
| | - D. Rajeswari
- Department of Physics
- Periyar University
- Salem 636 011
- India
| | - K. Vimala
- Proteomics and Molecular Cell Physiology Laboratory
- Department of Zoology
- Periyar University
- Salem 636011
- India
| | - S. Kannan
- Proteomics and Molecular Cell Physiology Laboratory
- Department of Zoology
- Periyar University
- Salem 636011
- India
| | - D. Gopi
- Department of Chemistry
- Periyar University
- Salem 636011
- India
- Centre for Nanoscience and Nanotechnology
| |
Collapse
|
46
|
Engineered microenvironments provide new insights into ovarian and prostate cancer progression and drug responses. Adv Drug Deliv Rev 2014; 79-80:193-213. [PMID: 24969478 DOI: 10.1016/j.addr.2014.06.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 05/30/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Tissue engineering technologies, which have originally been designed to reconstitute damaged tissue structure and function, can mimic not only tissue regeneration processes but also cancer development and progression. Bioengineered approaches allow cell biologists to develop sophisticated experimentally and physiologically relevant cancer models to recapitulate the complexity of the disease seen in patients. Tissue engineering tools enable three-dimensionality based on the design of biomaterials and scaffolds that re-create the geometry, chemistry, function and signalling milieu of the native tumour microenvironment. Three-dimensional (3D) microenvironments, including cell-derived matrices, biomaterial-based cell culture models and integrated co-cultures with engineered stromal components, are powerful tools to study dynamic processes like proteolytic functions associated with cancer progression, metastasis and resistance to therapeutics. In this review, we discuss how biomimetic strategies can reproduce a humanised niche for human cancer cells, such as peritoneal or bone-like microenvironments, addressing specific aspects of ovarian and prostate cancer progression and therapy response.
Collapse
|
47
|
Taubenberger AV. In vitro microenvironments to study breast cancer bone colonisation. Adv Drug Deliv Rev 2014; 79-80:135-44. [PMID: 25453260 DOI: 10.1016/j.addr.2014.10.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 09/13/2014] [Accepted: 10/15/2014] [Indexed: 12/15/2022]
Abstract
Bone metastasis occurs frequently in patients with advanced breast cancer and is a major cause of morbidity and mortality in these patients. In order to advance current therapies, the mechanisms leading to the formation of bone metastases and their pathophysiology have to be better understood. Several in vitro models have been developed for systematic studies of interactions between breast cancer cells and the bone microenvironment. Such models can provide insights into the molecular basis of bone metastatic colonisation and also may provide a useful platform to design more physiologically relevant drug testing assays. This review describes different in vitro approaches and discusses their advantages and disadvantages.
Collapse
Affiliation(s)
- Anna V Taubenberger
- Group of Cellular Machines, Biotec TU Dresden, Tatzberg 47-51, 01307 Dresden, Germany; Institute of Health and Biomedical Innovation, Queensland University of Technology, Musk Avenue 60, Kelvin Grove, QLD, Australia.
| |
Collapse
|
48
|
Siclari VA, Mohammad KS, Tompkins DR, Davis H, McKenna CR, Peng X, Wessner LL, Niewolna M, Guise TA, Suvannasankha A, Chirgwin JM. Tumor-expressed adrenomedullin accelerates breast cancer bone metastasis. Breast Cancer Res 2014; 16:458. [PMID: 25439669 PMCID: PMC4303191 DOI: 10.1186/s13058-014-0458-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 10/09/2014] [Indexed: 01/23/2023] Open
Abstract
Introduction Adrenomedullin (AM) is secreted by breast cancer cells and increased by hypoxia. It is a multifunctional peptide that stimulates angiogenesis and proliferation. The peptide is also a potent paracrine stimulator of osteoblasts and bone formation, suggesting a role in skeletal metastases—a major site of treatment-refractory tumor growth in patients with advanced disease. Methods The role of adrenomedullin in bone metastases was tested by stable overexpression in MDA-MB-231 breast cancer cells, which cause osteolytic bone metastases in a standard animal model. Cells with fivefold increased expression of AM were characterized in vitro, inoculated into immunodeficient mice and compared for their ability to form bone metastases versus control subclones. Bone destruction was monitored by X-ray, and tumor burden and osteoclast numbers were determined by quantitative histomorphometry. The effects of AM overexpression on tumor growth and angiogenesis in the mammary fat pad were determined. The effects of AM peptide on osteoclast-like multinucleated cell formation were tested in vitro. A small-molecule AM antagonist was tested for its effects on AM-stimulated ex vivo bone cell cultures and co-cultures with tumor cells, where responses of tumor and bone were distinguished by species-specific real-time PCR. Results Overexpression of AM mRNA did not alter cell proliferation in vitro, expression of tumor-secreted factors or cell cycle progression. AM-overexpressing cells caused osteolytic bone metastases to develop more rapidly, which was accompanied by decreased survival. In the mammary fat pad, tumors grew more rapidly with unchanged blood vessel formation. Tumor growth in the bone was also more rapid, and osteoclasts were increased. AM peptide potently stimulated bone cultures ex vivo; responses that were blocked by small-molecule adrenomedullin antagonists in the absence of cellular toxicity. Antagonist treatment dramatically suppressed tumor growth in bone and decreased markers of osteoclast activity. Conclusions The results identify AM as a target for therapeutic intervention against bone metastases. Adrenomedullin potentiates osteolytic responses in bone to metastatic breast cancer cells. Small-molecule antagonists can effectively block bone-mediated responses to tumor-secreted adrenomedullin, and such agents warrant development for testing in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0458-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Valerie A Siclari
- Department of Biochemistry and Molecular Genetics, University of Virginia, PO Box 800733, Charlottesville, VA, 22908, USA.
| | - Khalid S Mohammad
- Division of Endocrinology and Metabolism, Department of Medicine, 450 Ray C Hunt Dr, University of Virginia, PO Box 801406, Charlottesville, VA, 22908, USA. .,Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 980 Walnut, St, C321-C, Indianapolis, IN, 46202, USA.
| | - Douglas R Tompkins
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 980 Walnut, St, C321-C, Indianapolis, IN, 46202, USA. .,Richard L. Roudebush VA Medical Center, 1481 W 10th St, Indianapolis, IN, 46202, USA.
| | - Holly Davis
- Division of Endocrinology and Metabolism, Department of Medicine, 450 Ray C Hunt Dr, University of Virginia, PO Box 801406, Charlottesville, VA, 22908, USA.
| | - C Ryan McKenna
- Division of Endocrinology and Metabolism, Department of Medicine, 450 Ray C Hunt Dr, University of Virginia, PO Box 801406, Charlottesville, VA, 22908, USA.
| | - Xianghong Peng
- Division of Endocrinology and Metabolism, Department of Medicine, 450 Ray C Hunt Dr, University of Virginia, PO Box 801406, Charlottesville, VA, 22908, USA. .,Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 980 Walnut, St, C321-C, Indianapolis, IN, 46202, USA.
| | - Lisa L Wessner
- Division of Endocrinology and Metabolism, Department of Medicine, 450 Ray C Hunt Dr, University of Virginia, PO Box 801406, Charlottesville, VA, 22908, USA.
| | - Maria Niewolna
- Division of Endocrinology and Metabolism, Department of Medicine, 450 Ray C Hunt Dr, University of Virginia, PO Box 801406, Charlottesville, VA, 22908, USA. .,Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 980 Walnut, St, C321-C, Indianapolis, IN, 46202, USA.
| | - Theresa A Guise
- Division of Endocrinology and Metabolism, Department of Medicine, 450 Ray C Hunt Dr, University of Virginia, PO Box 801406, Charlottesville, VA, 22908, USA. .,Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 980 Walnut, St, C321-C, Indianapolis, IN, 46202, USA.
| | - Attaya Suvannasankha
- Richard L. Roudebush VA Medical Center, 1481 W 10th St, Indianapolis, IN, 46202, USA. .,Division of Hematology/Oncology, Department of Medicine, 980 Walnut St, C321-H, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - John M Chirgwin
- Department of Biochemistry and Molecular Genetics, University of Virginia, PO Box 800733, Charlottesville, VA, 22908, USA. .,Division of Endocrinology and Metabolism, Department of Medicine, 450 Ray C Hunt Dr, University of Virginia, PO Box 801406, Charlottesville, VA, 22908, USA. .,Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 980 Walnut, St, C321-C, Indianapolis, IN, 46202, USA. .,Richard L. Roudebush VA Medical Center, 1481 W 10th St, Indianapolis, IN, 46202, USA.
| |
Collapse
|
49
|
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: 1.1] [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
|
50
|
Wu X, Wang L, Deng F, Watts DC. Mouse calvarial defect Model: An approach for the micro-tomographic evaluation of polymer scaffolds. Microsc Res Tech 2014; 77:1037-43. [PMID: 25233951 DOI: 10.1002/jemt.22433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 08/20/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Xiaohong Wu
- Department of Prosthodontics, the Affiliated Hospital of Stomatology; Chongqing Medical University; Yubei district Chongqing 401147 China
- Chongqing key Laboratory for Oral Diseases and Biomedical Sciences; Chongqing 401147 China
| | - Lu Wang
- Department of Prosthodontics, the Affiliated Hospital of Stomatology; Chongqing Medical University; Yubei district Chongqing 401147 China
- Chongqing key Laboratory for Oral Diseases and Biomedical Sciences; Chongqing 401147 China
| | - Feng Deng
- Chongqing key Laboratory for Oral Diseases and Biomedical Sciences; Chongqing 401147 China
- Department of Orthodontics, the Affiliated Hospital of Stomatology; Chongqing Medical University; Yubei district Chongqing 401147 China
| | - David C. Watts
- School of Dentistry and Photon Science Institute; The University of Manchester; Higher Cambridge Street Manchester M15 6FH United Kingdom
- Institute of Material Science and Technology; Friedrich-Schiller-University; Jena Löbdergraben 32 07743 Germany
| |
Collapse
|