Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Protoc ; 12(4): 639-663, 2017 04.
Article in English | MEDLINE | ID: mdl-28253234

ABSTRACT

Current in vivo models for investigating human primary bone tumors and cancer metastasis to the bone rely on the injection of human cancer cells into the mouse skeleton. This approach does not mimic species-specific mechanisms occurring in human diseases and may preclude successful clinical translation. We have developed a protocol to engineer humanized bone within immunodeficient hosts, which can be adapted to study the interactions between human cancer cells and a humanized bone microenvironment in vivo. A researcher trained in the principles of tissue engineering will be able to execute the protocol and yield study results within 4-6 months. Additive biomanufactured scaffolds seeded and cultured with human bone-forming cells are implanted ectopically in combination with osteogenic factors into mice to generate a physiological bone 'organ', which is partially humanized. The model comprises human bone cells and secreted extracellular matrix (ECM); however, other components of the engineered tissue, such as the vasculature, are of murine origin. The model can be further humanized through the engraftment of human hematopoietic stem cells (HSCs) that can lead to human hematopoiesis within the murine host. The humanized organ bone model has been well characterized and validated and allows dissection of some of the mechanisms of the bone metastatic processes in prostate and breast cancer.


Subject(s)
Bone Neoplasms/secondary , Bone and Bones/pathology , Tissue Engineering/methods , Adenocarcinoma , Animals , Bone Morphogenetic Protein 7/pharmacology , Bone Neoplasms/pathology , Bone and Bones/drug effects , Breast Neoplasms/pathology , Disease Models, Animal , Electricity , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Hematopoietic Stem Cell Transplantation , Humans , Male , Mice , Prostatic Neoplasms/pathology , Tissue Engineering/instrumentation
2.
Biomed Tech (Berl) ; 61(3): 253-66, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-25781662

ABSTRACT

We set out to compare the osteogenicity of human mesenchymal stem (hMSCs) and osteoblasts (hOBs). Upon osteogenic induction in monolayer, hMSCs showed superior matrix mineralization expressing characteristic bone-related genes. For scaffold cultures, both cell types presented spindle-shaped, osteoblast-like morphologies forming a dense, interconnected network of high viability. On the scaffolds, hOBs proliferated faster. A general upregulation of parathyroid hormone-related protein (PTHrP), osteoprotegrin (OPG), receptor activator of NF-κB ligand (RANKL), sclerostin (SOST), and dentin matrix protein 1 (DMP1) was observed for both cell types. Simultaneously, PTHrP, RANKL and DMP-1 expression decreased under osteogenic stimulation, while OPG and SOST increased significantly. Following transplantation into NOD/SCID mice, µCT and histology showed increased bone deposition with hOBs. The bone was vascularized, and amounts further increased for both cell types after recombinant human bone morphogenic protein 7 (rhBMP-7) addition also stimulating osteoclastogenesis. Complete bone organogenesis was evidenced by the presence of osteocytes and hematopoietic precursors. Our study results support the asking to develop 3D cellular models closely mimicking the functions of living tissues suitable for in vivo translation.


Subject(s)
Bone Resorption/physiopathology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis/physiology , Parathyroid Hormone-Related Protein/chemistry , Animals , Bone Resorption/metabolism , Humans , Mice , Mice, SCID , Osteoblasts/chemistry , Parathyroid Hormone-Related Protein/metabolism , Parathyroid Hormone-Related Protein/physiology
3.
Cancer Metastasis Rev ; 33(2-3): 721-35, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24771149

ABSTRACT

Bone metastasis is a complication that occurs in 80 % of women with advanced breast cancer. Despite the prevalence of bone metastatic disease, the avenues for its clinical management are still restricted to palliative treatment options. In fact, the underlying mechanisms of breast cancer osteotropism have not yet been fully elucidated due to a lack of suitable in vivo models that are able to recapitulate the human disease. In this work, we review the current transplantation-based models to investigate breast cancer-induced bone metastasis and delineate the strengths and limitations of the use of different grafting techniques, tissue sources, and hosts. We further show that humanized xenograft models incorporating human cells or tissue grafts at the primary tumor site or the metastatic site mimic more closely the human disease. Tissue-engineered constructs are emerging as a reproducible alternative to recapitulate functional humanized tissues in these murine models. The development of advanced humanized animal models may provide better platforms to investigate the mutual interactions between human cancer cells and their microenvironment and ultimately improve the translation of preclinical drug trials to the clinic.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Animals , Disease Models, Animal , Female , Heterografts , Humans , Isografts , Neoplasm Transplantation
4.
Dis Model Mech ; 7(2): 299-309, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24713276

ABSTRACT

The skeleton is a preferred homing site for breast cancer metastasis. To date, treatment options for patients with bone metastases are mostly palliative and the disease is still incurable. Indeed, key mechanisms involved in breast cancer osteotropism are still only partially understood due to the lack of suitable animal models to mimic metastasis of human tumor cells to a human bone microenvironment. In the presented study, we investigate the use of a human tissue-engineered bone construct to develop a humanized xenograft model of breast cancer-induced bone metastasis in a murine host. Primary human osteoblastic cell-seeded melt electrospun scaffolds in combination with recombinant human bone morphogenetic protein 7 were implanted subcutaneously in non-obese diabetic/severe combined immunodeficient mice. The tissue-engineered constructs led to the formation of a morphologically intact 'organ' bone incorporating a high amount of mineralized tissue, live osteocytes and bone marrow spaces. The newly formed bone was largely humanized, as indicated by the incorporation of human bone cells and human-derived matrix proteins. After intracardiac injection, the dissemination of luciferase-expressing human breast cancer cell lines to the humanized bone ossicles was detected by bioluminescent imaging. Histological analysis revealed the presence of metastases with clear osteolysis in the newly formed bone. Thus, human tissue-engineered bone constructs can be applied efficiently as a target tissue for human breast cancer cells injected into the blood circulation and replicate the osteolytic phenotype associated with breast cancer-induced bone lesions. In conclusion, we have developed an appropriate model for investigation of species-specific mechanisms of human breast cancer-related bone metastasis in vivo.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Heterografts/pathology , Models, Biological , Tissue Engineering/methods , Animals , Female , Humans , Mice , Mice, SCID , Reproducibility of Results , Tumor Microenvironment
5.
Stem Cell Res Ther ; 4(5): 105, 2013.
Article in English | MEDLINE | ID: mdl-24004723

ABSTRACT

INTRODUCTION: To stimulate healing of large bone defects research has concentrated on the application of mesenchymal stem cells (MSCs). METHODS: In the present study, we induced the overexpression of the growth factors bone morphogenetic protein 2 (BMP-2) and/or Indian hedgehog (IHH) in human MSCs by adenoviral transduction to increase their osteogenic potential. GFP and nontransduced MSCs served as controls. The influence of the respective genetic modification on cell metabolic activity, proliferation, alkaline phosphatase (ALP) activity, mineralization in cell culture, and osteogenic marker gene expression was investigated. RESULTS: Transduction had no negative influence on cell metabolic activity or proliferation. ALP activity showed a typical rise-and-fall pattern with a maximal activity at day 14 and 21 after osteogenic induction. Enzyme activity was significantly higher in groups cultured with osteogenic media. The overexpression of BMP-2 and especially IHH + BMP-2 resulted in a significantly higher mineralization after 28 days. This was in line with obtained quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) analyses, which showed a significant increase in osteopontin and osteocalcin expression for osteogenically induced BMP-2 and IHH + BMP-2 transduced cells when compared with the other groups. Moreover, an increase in runx2 expression was observed in all osteogenic groups toward day 21. It was again more pronounced for BMP-2 and IHH + BMP-2 transduced cells cultured in osteogenic media. CONCLUSIONS: In summary, viral transduction did not negatively influence cell metabolic activity and proliferation. The overexpression of BMP-2 in combination with or without IHH resulted in an increased deposition of mineralized extracellular matrix, and expression of osteogenic marker genes. Viral transduction therefore represents a promising means to increase the osteogenic potential of MSCs and the combination of different transgenes may result in synergistic effects.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Hedgehog Proteins/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis , Adenoviridae/genetics , Bone Marrow Cells/cytology , Bone Morphogenetic Protein 2/genetics , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Extracellular Matrix/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Hedgehog Proteins/genetics , Humans , Mesenchymal Stem Cells/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Time Factors , Transfection
6.
J Bone Miner Res ; 28(6): 1399-411, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23362043

ABSTRACT

The mechanisms leading to colonization of metastatic breast cancer cells (BCa) in the skeleton are still not fully understood. Here, we demonstrate that mineralized extracellular matrices secreted by primary human osteoblasts (hOBM) modulate cellular processes associated with BCa colonization of bone. A panel of four BCa cell lines of different bone-metastatic potential (T47D, SUM1315, MDA-MB-231, and the bone-seeking subline MDA-MB-231BO) was cultured on hOBM. After 3 days, the metastatic BCa cells had undergone morphological changes on hOBM and were aligned along the hOBM's collagen type I fibrils that were decorated with bone-specific proteins. In contrast, nonmetastatic BCa cells showed a random orientation on hOBM. Atomic force microscopy-based single-cell force spectroscopy revealed that the metastatic cell lines adhered more strongly to hOBM compared with nonmetastatic cells. Function-blocking experiments indicated that ß1 -integrins mediated cell adhesion to hOBM. In addition, metastatic BCa cells migrated directionally and invaded hOBM, which was accompanied by enhanced MMP-2 and -9 secretion. Furthermore, we observed gene expression changes associated with osteomimickry in BCa cultured on hOBM. As such, osteopontin mRNA levels were significantly increased in SUM1315 and MDA-MB-231BO cells in a ß1 -integrin-dependent manner after growing for 3 days on hOBM compared with tissue culture plastic. In conclusion, our results show that extracellular matrices derived from human osteoblasts represent a powerful experimental platform to dissect mechanisms underlying critical steps in the development of bone metastases.


Subject(s)
Bone Substitutes/chemistry , Breast Neoplasms/metabolism , Cell Movement , Tumor Microenvironment , Animals , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Female , Humans , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Rats , Tissue Engineering
7.
Gynecol Oncol ; 127(3): 569-78, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22964375

ABSTRACT

OBJECTIVE: Chemoresistance is a critical feature of advanced ovarian cancer with only 30% of patients surviving longer than 5 years. We have previously shown that four kallikrein-related (KLK) peptidases, KLK4, KLK5, KLK6 and KLK7 (KLK4-7), are implicated in peritoneal invasion and tumour growth, but underlying mechanisms were not identified. We also reported that KLK7 overexpression confers chemoresistance to paclitaxel, and cell survival via integrins. In this study, we further explored the functional consequenses of overexpression of all four KLKs (KLK4-7) simultaneously in the ovarian cancer cell line, OV-MZ-6, and its impact on integrin expression and signalling, cell adhesion and survival as contributors to chemoresistance and metastatic progression. METHODS: Quantitative gene and protein expression analyses, confocal microscopy, cell adhesion and chemosensitivity assays were performed. RESULTS: Expression of α5ß1/αvß3 integrins was downregulated upon combined stable KLK4-7 overexpression in OV-MZ-6 cells. Accordingly, the adhesion of these cells to vitronectin and fibronectin, the extracellular matrix binding proteins of α5ß1/αvß3 integrins and two predominant proteins of the peritoneal matrix, was decreased. KLK4-7-transfected cells were more resistant to paclitaxel (10-100 nmol/L: 38-54%), but not to carboplatin, which was associated with decreased apoptotic stimuli. However, the KLK4-7-induced paclitaxel resistance was not blocked by the MEK1/2 inhibitor, U0126. CONCLUSIONS: This study demonstrates that combined KLK4-7 expression by ovarian cancer cells promotes reduced integrin expression with consequently less cell-matrix attachment, and insensitivity to paclitaxel mediated by complex integrin and MAPK independent interactions, indicative of a malignant phenotype and disease progression suggesting a role for these KLKs in this process.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Kallikreins/physiology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Adhesion/drug effects , Cell Line, Tumor , Female , Humans , Integrins/metabolism , Kallikreins/genetics , MAP Kinase Signaling System , Ovarian Neoplasms/metabolism , Paclitaxel/therapeutic use
8.
Biomaterials ; 31(31): 7928-36, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20688384

ABSTRACT

Prostate cancer metastasis is reliant on the reciprocal interactions between cancer cells and the bone niche/micro-environment. The production of suitable matrices to study metastasis, carcinogenesis and in particular prostate cancer/bone micro-environment interaction has been limited to specific protein matrices or matrix secreted by immortalised cell lines that may have undergone transformation processes altering signaling pathways and modifying gene or receptor expression. We hypothesize that matrices produced by primary human osteoblasts are a suitable means to develop an in vitro model system for bone metastasis research mimicking in vivo conditions. We have used a decellularized matrix secreted from primary human osteoblasts as a model for prostate cancer function in the bone micro-environment. We show that this collagen I rich matrix is of fibrillar appearance, highly mineralized, and contains proteins, such as osteocalcin, osteonectin and osteopontin, and growth factors characteristic of bone extracellular matrix (ECM). LNCaP and PC3 cells grown on this matrix, adhere strongly, proliferate, and express markers consistent with a loss of epithelial phenotype. Moreover, growth of these cells on the matrix is accompanied by the induction of genes associated with attachment, migration, increased invasive potential, Ca(2+) signaling and osteolysis. In summary, we show that growth of prostate cancer cells on matrices produced by primary human osteoblasts mimics key features of prostate cancer bone metastases and thus is a suitable model system to study the tumor/bone micro-environment interaction in this disease.


Subject(s)
Bone Matrix/metabolism , Calcification, Physiologic , Cell Communication , Models, Biological , Osteoblasts/metabolism , Prostatic Neoplasms/pathology , Tumor Microenvironment , Bone and Bones/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Cell Shape , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Keratin-8/genetics , Keratin-8/metabolism , Male , Matrix Metalloproteinases/metabolism , Osteoblasts/cytology , Osteoblasts/ultrastructure , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics
9.
J Tissue Eng Regen Med ; 4(7): 565-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20568083

ABSTRACT

Recently, research has focused on bone marrow derived multipotent mesenchymal precursor cells (MPC) and osteoblasts (OB) for clinical use in bone engineering. Prior to clinical application, cell based treatment concepts need to be evaluated in preclinical, large animal models. Sheep in particular are considered a valid model for orthopaedic and trauma related research. However, only sheep aged > 6 years show secondary osteon formation characteristic of human bone. Osteogenic cells isolated from animals of this age group remain poorly characterized. In the present study, ex vivo expanded MPC isolated from ovine bone marrow proliferated at a higher rate than OB derived from tibial compact bone as assessed in standard 2D cultures. MPC expressed the respective phenotypic profile typical for different mesenchymal cell populations (CD14(-)/CD31(-)/CD45(-)/CD29(+)/CD44(+)/CD166(+)) and showed a multilineage differentiation potential. When compared to OB, MPC had a higher mineralization potential under standard osteogenic culture conditions and expressed typical bone related markers such as osteocalcin, osteonectin and type I collagen at the mRNA and protein level. After 4 weeks in 3D culture, MPC constructs demonstrated higher cell density and mineralization, whilst cell viability on the scaffolds was assessed > 90%. Cells displayed a spindle-like morphology and formed interconnected networks. In contrast, when implanted subcutaneously into NOD/SCID mice, MPC presented a lower osteogenic potential than OB. In summary, this study provides a detailed characterisation of ovine MPC and OB from a bone engineering perspective and suggests that MPC and OB provide promising means for future bone disease related treatment applications.


Subject(s)
Bone and Bones , Mesenchymal Stem Cells , Multipotent Stem Cells , Osteoblasts , Tissue Engineering/methods , Tissue Scaffolds , Animals , Antigens, Differentiation/biosynthesis , Bone Diseases/therapy , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Calcification, Physiologic , Cell Differentiation , Cell Survival , Cells, Cultured , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Sheep , Transplantation, Heterologous , Transplantation, Homologous
10.
J Cell Mol Med ; 14(4): 1003-13, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20082656

ABSTRACT

Cell proliferation is a critical and frequently studied feature of molecular biology in cancer research. Therefore, various assays are available using different strategies to measure cell proliferation. Metabolic assays such as AlamarBlue, water-soluble tetrazolium salt and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide, which were originally developed to determine cell toxicity, are used to assess cell numbers. Additionally, proliferative activity can be determined by quantification of DNA content using fluorophores such as CyQuant and PicoGreen. Referring to data published in high ranking cancer journals, these assays were applied in 945 publications over the past 14 years to examine the proliferative behaviour of diverse cell types. In these studies, however, mainly metabolic assays were used to quantify changes in cell growth yet these assays may not accurately reflect cellular proliferation rates due to a miscorrelation of metabolic activity and cell number. Testing this hypothesis, we compared the metabolic activity of different cell types, human cancer cells and primary cells, over a time period of 4 days using AlamarBlue and the fluorometric assays CyQuant and PicoGreen to determine their DNA content. Our results show certain discrepancies in terms of over-estimation of cell proliferation with respect to the metabolic assay in comparison to DNA binding fluorophores.


Subject(s)
Biomedical Research/methods , DNA, Neoplasm/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Aged , Biological Assay , Cell Line, Tumor , Cell Proliferation , Female , Humans , Oxazines/metabolism , Publications , Time Factors , Xanthenes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...