Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 14189, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578352

ABSTRACT

Interactions between multiple myeloma (MM) and bone marrow (BM) are well documented to support tumour growth, yet the cellular mechanisms underlying pain in MM are poorly understood. We have used in vivo murine models of MM to show significant induction of nerve growth factor (NGF) by the tumour-bearing bone microenvironment, alongside other known pain-related characteristics such as spinal glial cell activation and reduced locomotion. NGF was not expressed by MM cells, yet bone stromal cells such as osteoblasts expressed and upregulated NGF when cultured with MM cells, or MM-related factors such as TNF-α. Adiponectin is a known MM-suppressive BM-derived factor, and we show that TNF-α-mediated NGF induction is suppressed by adiponectin-directed therapeutics such as AdipoRON and L-4F, as well as NF-κB signalling inhibitor BMS-345541. Our study reveals a further mechanism by which cellular interactions within the tumour-bone microenvironment contribute to disease, by promoting pain-related properties, and suggests a novel direction for analgesic development.


Subject(s)
Adiponectin/genetics , Multiple Myeloma/drug therapy , Nerve Growth Factor/genetics , Pain/drug therapy , Tumor Necrosis Factor-alpha/genetics , Adiponectin/antagonists & inhibitors , Animals , Bone Marrow/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Mice , Multiple Myeloma/complications , Multiple Myeloma/genetics , Multiple Myeloma/pathology , NF-kappa B/antagonists & inhibitors , Neuroglia/metabolism , Neuroglia/pathology , Osteoblasts/drug effects , Pain/complications , Pain/genetics , Pain/pathology , Peptides/pharmacology , Piperidines/pharmacology , Quinoxalines/pharmacology , Stromal Cells/drug effects , Tumor Microenvironment/drug effects
2.
Bonekey Rep ; 5: 772, 2016.
Article in English | MEDLINE | ID: mdl-26909147

ABSTRACT

Multiple myeloma is an incurable plasma-cell malignancy characterized by osteolytic bone disease and immunosuppression. Murine models of multiple myeloma and myeloma bone disease are critical tools for an improved understanding of the pathogenesis of the disease and the development of novel therapeutic strategies. This review will cover commonly used immunocompetent and xenograft models of myeloma, describing the advantages and disadvantages of each model system. In addition, this review provides detailed protocols for establishing systemic and local models of myeloma using both murine and human myeloma cell lines.

3.
J Bone Miner Res ; 28(4): 960-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23172686

ABSTRACT

Silent information regulator T1 (SirT1) is linked to longevity and negatively controls NF-κB signaling, a crucial mediator of survival and regulator of both osteoclasts and osteoblasts. Here we show that NF-κB repression by SirT1 in both osteoclasts and osteoblasts is necessary for proper bone remodeling and may contribute to the mechanisms linking aging and bone loss. Osteoclast- or osteoblast-specific SirT1 deletion using the Sirt(flox/flox) mice crossed to lysozyme M-cre and the 2.3 kb col1a1-cre transgenic mice, respectively, resulted in decreased bone mass caused by increased resorption and reduced bone formation. In osteoclasts, lack of SirT1 promoted osteoclastogenesis in vitro and activated NF-κB by increasing acetylation of Lysine 310. Importantly, this increase in osteoclastogenesis was blocked by pharmacological inhibition of NF-κB. In osteoblasts, decreased SirT1 reduced osteoblast differentiation, which could also be rescued by inhibition of NF-κB. In further support of the critical role of NF-κB signaling in bone remodeling, elevated NF-κB activity in IκBα(+/-) mice uncoupled bone resorption and formation, leading to reduced bone mass. These findings support the notion that SirT1 is a genetic determinant of bone mass, acting in a cell-autonomous manner in both osteoblasts and osteoclasts, through control of NF-κB and bone cell differentiation.


Subject(s)
Bone Remodeling , NF-kappa B/metabolism , Signal Transduction , Sirtuin 1/metabolism , Acetylation , Aging/metabolism , Animals , Bone and Bones/anatomy & histology , Bone and Bones/metabolism , Gene Deletion , Gene Knockdown Techniques , I-kappa B Proteins/metabolism , Mice , NF-KappaB Inhibitor alpha , Organ Size , Organ Specificity , Osteoblasts/metabolism , Osteoclasts/metabolism
4.
PLoS One ; 7(11): e48871, 2012.
Article in English | MEDLINE | ID: mdl-23173040

ABSTRACT

Osteoclasts play a key role in the development of cancer-associated osteolytic lesions. The number and activity of osteoclasts are often enhanced by tumors. However, the origin of osteoclasts is unknown. Myeloid-derived suppressor cells (MDSCs) are one of the pre-metastatic niche components that are induced to expand by tumor cells. Here we show that the MDSCs can differentiate into mature and functional osteoclasts in vitro and in vivo. Inoculation of 5TGM1-GFP myeloma cells into C57BL6/KaLwRij mice led to a significant expansion of MDSCs in blood, spleen, and bone marrow over time. When grown in osteoclastogenic media in vitro, MDSCs from tumor-challenged mice displayed 14 times greater potential to differentiate into mature and functional osteoclasts than those from non-tumor controls. Importantly, MDSCs from tumor-challenged LacZ transgenic mice differentiated into LacZ+osteoclasts in vivo. Furthermore, a significant increase in tumor burden and bone loss accompanied by increased number of osteoclasts was observed in mice co-inoculated with tumor-challenged MDSCs and 5TGM1 cells compared to the control animals received 5TGM1 cells alone. Finally, treatment of MDSCs from myeloma-challenged mice with Zoledronic acid (ZA), a potent inhibitor of bone resorption, inhibited the number of osteoclasts formed in MDSC cultures and the expansion of MDSCs and bone lesions in mice. Collectively, these data provide in vitro and in vivo evidence that tumor-induced MDSCs exacerbate cancer-associated bone destruction by directly serving as osteoclast precursors.


Subject(s)
CD11b Antigen/metabolism , Multiple Myeloma/pathology , Myeloid Cells/pathology , Osteoclasts/pathology , Tumor Microenvironment , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Diphosphonates/pharmacology , Female , Humans , Imidazoles/pharmacology , Mice , Multiple Myeloma/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Prenylation/drug effects , Tumor Microenvironment/drug effects , Zoledronic Acid
5.
Cancer Res ; 72(9): 2183-9, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22374979

ABSTRACT

The rapid progression of multiple myeloma is dependent upon cellular interactions within the bone marrow microenvironment. In vitro studies suggest that bone marrow stromal cells (BMSC) can promote myeloma growth and survival and osteolytic bone disease. However, it is not possible to recreate all cellular aspects of the bone marrow microenvironment in an in vitro system, and the contributions of BMSCs to myeloma pathogenesis in an intact, immune competent, in vivo system are unknown. To investigate this, we used a murine myeloma model that replicates many features of the human disease. Coinoculation of myeloma cells and a BMSC line, isolated from myeloma-permissive mice, into otherwise nonpermissive mice resulted in myeloma development, associated with tumor growth within bone marrow and osteolytic bone disease. In contrast, inoculation of myeloma cells alone did not result in myeloma. BMSCs inoculated alone induced osteoblast suppression, associated with an increase in serum concentrations of the Wnt signaling inhibitor, Dkk1. Dkk1 was highly expressed in BMSCs and in myeloma-permissive bone marrow. Knockdown of Dkk1 expression in BMSCs decreased their ability to promote myeloma and the associated bone disease in mice. Collectively, our results show novel roles of BMSCs and BMSC-derived Dkk1 in the pathogenesis of multiple myeloma in vivo.


Subject(s)
Bone Marrow Cells/pathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Multiple Myeloma/pathology , Stromal Cells/pathology , Animals , Bone Marrow Cells/metabolism , Female , Gene Knockdown Techniques , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Stromal Cells/metabolism , Tumor Microenvironment , Wnt Proteins/antagonists & inhibitors
6.
Blood ; 118(22): 5872-82, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21908434

ABSTRACT

The contributions of the host microenvironment to the pathogenesis of multiple myeloma, including progression from the non-malignant disorder monoclonal gammopathy of undetermined significance, are poorly understood. In the present study, microarray analysis of a murine model requiring a unique host microenvironment for myeloma development identified decreased host-derived adiponectin compared with normal mice. In support, clinical analysis revealed decreased serum adiponectin concentrations in monoclonal gammopathy of undetermined significance patients who subsequently progressed to myeloma. We investigated the role of adiponectin in myeloma pathogenesis and as a treatment approach, using both mice deficient in adiponectin and pharmacologic enhancement of circulating adiponectin. Increased tumor burden and bone disease were observed in myeloma-bearing adiponectin-deficient mice, and adiponectin was found to induce myeloma cell apoptosis. The apolipoprotein peptide mimetic L-4F was used for pharmacologic enhancement of adiponectin. L-4F reduced tumor burden, increased survival of myeloma-bearing mice, and prevented myeloma bone disease. Collectively, our studies have identified a novel mechanism whereby decreased host-derived adiponectin promotes myeloma tumor growth and osteolysis. Furthermore, we have established the potential therapeutic benefit of increasing adiponectin for the treatment of myeloma and the associated bone disease.


Subject(s)
Bone Neoplasms/therapy , Molecular Targeted Therapy , Multiple Myeloma/therapy , Adiponectin/genetics , Adiponectin/physiology , Animals , Bone Diseases/etiology , Bone Diseases/genetics , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Cell Line, Tumor , Female , Genetic Predisposition to Disease , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Transplantation , Peptides/administration & dosage , Peptides/therapeutic use , Tumor Cells, Cultured , Tumor Microenvironment , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Xenograft Model Antitumor Assays
7.
J Bone Miner Res ; 25(11): 2419-26, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20499365

ABSTRACT

Transforming growth factor ß (TGF-ß) is an abundant bone matrix protein that influences osteoblast and osteoclast interactions to control bone remodeling. As such, TGF-ß represents an obvious pharmacologic target with the potential to regulate both bone formation and resorption to improve bone volume and strength. To investigate the skeletal effect of TGF-ß inhibition in vivo, we used an antibody (1D11) specifically directed at all three isoforms of TGF-ß. Normal mice were treated with 1D11 or control antibody (4 weeks), and cortical and trabecular bone was assessed by micro-computed tomographic (µCT) scanning. Bone volume and cellular distribution were determined by histomorphometric analysis of vertebrae and long bones. Also, whole-bone strength was assessed biomechanically by three-point bend testing, and tissue-level modulus and composition were analyzed by nanoindentation and Raman microspectroscopy, respectively. TGF-ß blockade by 1D11 increased bone mineral density (BMD), trabecular thickness, and bone volume by up to 54%, accompanied by elevated osteoblast numbers and decreased osteoclasts. Biomechanical properties of bone also were enhanced significantly by 1D11 treatment, with increased bending strength and tissue-level modulus. In addition, Raman microspectroscopy demonstrated that 1D11-mediated TGF-ß inhibition in the bone environment led to an 11% increase in the mineral-to-collagen ratio of trabecular bone. Together these studies demonstrate that neutralizing TGF-ß with 1D11 increases osteoblast numbers while simultaneously decreasing active osteoclasts in the marrow, resulting in a profound increase in bone volume and quality, similar to that seen in parathyroid hormone (PTH)-treated rodent studies.


Subject(s)
Antibodies/pharmacology , Bone and Bones/drug effects , Bone and Bones/pathology , Signal Transduction/drug effects , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Animals , Biomechanical Phenomena/drug effects , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects
8.
Dis Model Mech ; 2(11-12): 604-11, 2009.
Article in English | MEDLINE | ID: mdl-19779066

ABSTRACT

Multiple myeloma, and the associated osteolytic bone disease, is highly dependent upon cellular interactions within the bone marrow microenvironment. A major limitation of existing myeloma models is the requirement for a specific host strain of mouse, preventing molecular examination of the bone marrow microenvironment. The aim of the current study was to develop a model of myeloma in which the host microenvironment could be modified genetically. The Radl 5T murine model of myeloma is well characterized and closely mimics human myeloma. In the current study, we demonstrate 5T myeloma establishment in recombination activating gene 2 (RAG-2)-deficient mice, which have improper B- and T-cell development. Importantly, these mice can be easily bred with genetically modified mice to generate double knockout mice, allowing manipulation of the host microenvironment at a molecular level. Inoculation of 5TGM1 myeloma cells into RAG-2(-/-) mice resulted in myeloma development, which was associated with tumor growth within bone and an osteolytic bone disease, as assessed by microcomputed tomography (microCT), histology and histomorphometry. Myeloma-bearing RAG-2(-/-) mice displayed many features that were similar to both human myeloma and the original Radl 5T model. To demonstrate the use of this model, we have examined the effect of host-derived matrix metalloproteinase 9 (MMP-9) in the development of myeloma in vivo. Inoculation of 5TGM1 myeloma cells into mice that are deficient in RAG-2 and MMP-9 resulted in a reduction in both tumor burden and osteolytic bone disease when compared with RAG-2-deficient wild-type myeloma-bearing mice. The establishment of myeloma in RAG-2(-/-) mice permits molecular examination of the host contribution to myeloma pathogenesis in vivo.


Subject(s)
Bone Diseases/genetics , Disease Models, Animal , Multiple Myeloma/genetics , Multiple Myeloma/physiopathology , Animals , Bone Diseases/physiopathology , DNA-Binding Proteins/genetics , Female , Femur/pathology , Genetic Techniques , Immunohistochemistry/methods , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tibia/pathology , Tomography, X-Ray Computed/methods
9.
Am J Hematol ; 84(5): 268-72, 2009 May.
Article in English | MEDLINE | ID: mdl-19296472

ABSTRACT

The proteasome inhibitor bortezomib has a striking clinical benefit in patients with multiple myeloma. It is unknown whether the bone marrow microenvironment directly contributes to the dramatic response of myeloma cells to proteasome inhibition in vivo. We have used the well-characterized 5TGM1 murine model of myeloma to investigate myeloma growth within bone and response to the proteasome inhibitor bortezomib in vivo. Myeloma cells freshly isolated from the bone marrow of myeloma-bearing mice were found to have an increase in proteasome activity and an enhanced response to in vitro proteasome inhibition, as compared with pre-inoculation myeloma cells. Treatment of myeloma-bearing mice with bortezomib resulted in a greater reduction in tumor burden when the myeloma cells were located within the bone marrow when compared with extra-osseous sites. Our results demonstrate that myeloma cells exhibit an increase in proteasome activity and an enhanced response to bortezomib treatment when located within the bone marrow microenvironment in vivo.


Subject(s)
Boronic Acids/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Animals , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bortezomib , Cell Communication/drug effects , Cell Line, Tumor , Enzyme Activation , Immunoglobulin G/blood , Mice , Multiple Myeloma/pathology , Neoplasm Transplantation , Proteasome Endopeptidase Complex/metabolism , Tumor Burden/drug effects
10.
Blood ; 111(5): 2833-42, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18094333

ABSTRACT

There is increasing evidence to suggest that the Wnt signaling pathway plays a critical role in the pathogenesis of myeloma bone disease. In the present study, we determined whether increasing Wnt signaling within the bone marrow microenvironment in myeloma counteracts development of osteolytic bone disease. C57BL/KaLwRij mice were inoculated intravenously with murine 5TGM1 myeloma cells, resulting in tumor growth in bone and development of myeloma bone disease. Lithium chloride (LiCl) treatment activated Wnt signaling in osteoblasts, inhibited myeloma bone disease, and decreased tumor burden in bone, but increased tumor growth when 5TGM1 cells were inoculated subcutaneously. Abrogation of beta-catenin activity and disruption of Wnt signaling in 5TGM1 cells by stable overexpression of a dominant-negative TCF4 prevented the LiCl-induced increase in subcutaneous growth but had no effect on LiCl-induced reduction in tumor burden within bone or on osteolysis in myeloma-bearing mice. Together, these data highlight the importance of the local microenvironment in the effect of Wnt signaling on the development of myeloma bone disease and demonstrate that, despite a direct effect to increase tumor growth at extraosseous sites, increasing Wnt signaling in the bone marrow microenvironment can prevent the development of myeloma bone disease and inhibit myeloma growth within bone in vivo.


Subject(s)
Bone Diseases/pathology , Bone Marrow/metabolism , Bone and Bones/pathology , Multiple Myeloma/pathology , Signal Transduction , Tumor Burden , Wnt Proteins/metabolism , Animals , Bone Diseases/complications , Bone Marrow/drug effects , Bone and Bones/drug effects , Cell Line, Tumor , Female , Humans , Lithium Chloride/pharmacology , Mice , Mice, Inbred C57BL , Multiple Myeloma/complications , Neoplasm Transplantation , Plasmacytoma/pathology , Signal Transduction/drug effects , Tumor Burden/drug effects , beta Catenin/metabolism
11.
J Mol Cell Cardiol ; 44(2): 405-10, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18048055

ABSTRACT

Transgenic expression of enhanced green fluorescent protein (eGFP) in myocardium can result in cardiac dysfunction and cardiomyopathy, presumably through toxic effects that disrupt normal cellular signaling. The multifunctional Ca(2+)- and calmodulin-dependent protein kinase II (CaMKII) is widely expressed in myocardium and CaMKII activity is increased in human and animal models of cardiomyopathy, so we hypothesized that increased CaMKII activity is important for cardiomyopathy due to transgenic expression of eGFP. Here we report that cardiomyocyte-delimited eGFP over-expression causes increased CaMKII activity that predicts left ventricular dilation and dysfunction. On the other hand, transgenic co-expression of a CaMKII inhibitory peptide with eGFP prevents eGFP-mediated left ventricular dilation and dysfunction. These findings suggest that increased CaMKII activity is a critical pathological signal in transgenic cardiomyopathy due to eGFP over-expression.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cardiomyopathies/enzymology , Green Fluorescent Proteins/metabolism , Animals , Cardiomyopathies/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hypertrophy , Mice , Mice, Transgenic , Organ Size/drug effects , Peptides/pharmacology , Ventricular Function, Left/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...