Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Oncotarget ; 6(25): 21589-602, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26009993

ABSTRACT

Treatment of multiple myeloma with bortezomib can result in severe adverse effects, necessitating the development of targeted inhibitors of the proteasome. We show that stable expression of a dominant-negative F-box deleted (∆F) mutant of the E3 ubiquitin ligase, SCFß-TrCP/FWD1, in murine 5TGM1 myeloma cells dramatically attenuated their skeletal engraftment and survival when inoculated into immunocompetent C57BL/KaLwRij mice. Similar results were obtained in immunodeficient bg-nu-xid mice, suggesting that the observed effects were independent of host recipient immune status. Bone marrow stroma offered no protection for 5TGM1-∆F cells in cocultures treated with tumor necrosis factor (TNF), indicating a cell-autonomous anti-myeloma effect. Levels of p100, IκBα, Mcl-1, ATF4, total and cleaved caspase-3, and phospho-ß-catenin were elevated in 5TGM1-∆F cells whereas cIAP was down-regulated. TNF also activated caspase-3 and downregulated Bcl-2, correlating with the enhanced susceptibility of 5TGM1-∆F cells to apoptosis. Treatment of 5TGM1 tumor-bearing mice with a ß-TrCP1/FWD1 inhibitor, pyrrolidine dithiocarbamate (PDTC), significantly reduced tumor burden in bone. PDTC also increased levels of cleaved Mcl-1 and caspase-3 in U266 human myeloma cells, correlating with our murine data and validating the development of specific ß-TrCP inhibitors as an alternative therapy to nonspecific proteasome inhibitors for myeloma patients.


Subject(s)
Multiple Myeloma/metabolism , Mutation , Ubiquitin-Protein Ligases/genetics , beta-Transducin Repeat-Containing Proteins/genetics , Animals , Apoptosis , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Bortezomib/chemistry , Caspase 3/metabolism , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Female , Genes, Dominant , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Proteasome Endopeptidase Complex/chemistry , Pyrrolidines/chemistry , Stromal Cells/cytology , Thiocarbamates/chemistry , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/metabolism , beta Catenin/chemistry , beta-Transducin Repeat-Containing Proteins/metabolism
2.
PLoS Biol ; 10(7): e1001363, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22815651

ABSTRACT

Bone and lung metastases are responsible for the majority of deaths in patients with breast cancer. Following treatment of the primary cancer, emotional and psychosocial factors within this population precipitate time to recurrence and death, however the underlying mechanism(s) remain unclear. Using a mouse model of bone metastasis, we provide experimental evidence that activation of the sympathetic nervous system, which is one of many pathophysiological consequences of severe stress and depression, promotes MDA-231 breast cancer cell colonization of bone via a neurohormonal effect on the host bone marrow stroma. We demonstrate that induction of RANKL expression in bone marrow osteoblasts, following ß2AR stimulation, increases the migration of metastatic MDA-231 cells in vitro, independently of SDF1-CXCR4 signaling. We also show that the stimulatory effect of endogenous (chronic stress) or pharmacologic sympathetic activation on breast cancer bone metastasis in vivo can be blocked with the ß-blocker propranolol, and by knockdown of RANK expression in MDA-231 cells. These findings indicate that RANKL promotes breast cancer cell metastasis to bone via its pro-migratory effect on breast cancer cells, independently of its effect on bone turnover. The emerging clinical implication, supported by recent epidemiological studies, is that ßAR-blockers and drugs interfering with RANKL signaling, such as Denosumab, could increase patient survival if used as adjuvant therapy to inhibit both the early colonization of bone by metastatic breast cancer cells and the initiation of the "vicious cycle" of bone destruction induced by these cells.


Subject(s)
Bone Marrow Cells/cytology , Bone Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Stromal Cells/cytology , Sympathetic Nervous System/physiology , Adrenergic beta-Antagonists/pharmacology , Animals , Cell Movement , Female , Mice , Osteoblasts/metabolism , Propranolol/pharmacology , Receptors, Adrenergic, beta-2/drug effects , Signal Transduction , Sympathetic Nervous System/drug effects
3.
Bone ; 48(1): 141-51, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20685406

ABSTRACT

The majority of breast cancer and prostate cancer patients with metastatic disease will go on to develop bone metastases, which contribute largely to the patient's morbidity and mortality. Numerous small animal models of cancer metastasis to bone have been developed to study tumor-induced bone destruction, but the advancement of imaging modalities utilized for these models has lagged significantly behind clinical imaging. Therefore, there is a significant need for improvements to live small animal imaging, particularly when obtaining high-resolution images for longitudinal quantitative analyses. Recently, live animal micro-computed tomography (µCT) has gained popularity due to its ability to obtain high-resolution 3-dimensional images. However, the utility of µCT in bone metastasis models has been limited to end-point analyses due to off-target radiation effects on tumor cells. We hypothesized that live animal in vivo µCT can be utilized to perform reproducible and quantitative longitudinal analyses of bone volume in tumor-bearing mice, particularly in a drug treatment model of breast cancer metastasis to bone. To test this hypothesis, we utilized the MDA-MB-231 osteolytic breast cancer model in which the tumor cells are inoculated directly into the tibia of athymic nude mice and imaged mice weekly by Faxitron (radiography), Imtek µCT (in vivo), and Maestro (GFP-imaging). Exvivo µCT and histology were performed at end point for validation. After establishing a high-resolution scanning protocol for the Imtek CT, we determined whether clear, measurable differences in bone volume were detectable in mice undergoing bisphosphonate drug treatments. We found that in vivo µCT could be used to obtain quantifiable and longitudinal images of the progression of bone destruction over time without altering tumor cell growth. In addition, we found that we could detect lesions as early as week 1 and that this approach could be used to monitor the effect of drug treatment on bone. Taken together, these data indicate that in vivo µCT is an effective and reproducible method for longitudinal monitoring of tumor-associated bone destruction in mouse models of tumor-induced bone disease.


Subject(s)
Bone Neoplasms/diagnostic imaging , X-Ray Microtomography/methods , Animals , Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Cell Line, Tumor , Diphosphonates/therapeutic use , Disease Models, Animal , Disease Progression , Female , Humans , Longitudinal Studies , Male , Mice , Mice, Nude , Neoplasm Transplantation , Reproducibility of Results , Tibia/diagnostic imaging , Tibia/pathology , Tomography, X-Ray Computed/methods
4.
J Biomech ; 42(7): 891-7, 2009 May 11.
Article in English | MEDLINE | ID: mdl-19281987

ABSTRACT

Although fracture healing is frequently studied in pre-clinical models of long bone fractures using rodents, there is a dearth of objective quantitative techniques to assess successful healing. Biomechanical testing is possibly the most quantitative and relevant to a successful clinical outcome, but it is a destructive technique providing little insight into the cellular mechanisms associated with healing. The advent of X-ray computed tomography (CT) has provided the opportunity to quantitatively and non-destructively assess bone structure and density, but it is unknown how measurements derived using this technology relate to successful healing. To examine possible relationships, we used a pre-clinical model to test for statistically significant correlations between quantitative characteristics of the callus by micro-CT (microCT) and the bending strength, stiffness, and energy-to-failure of the callus as assessed by three-point bending of excised bones. A closed, transverse fracture was generated in the mid-shaft of rat femurs by impact loading. Shortly thereafter, the rats received a one-time, local injection of either the vehicle or one of four doses of lovastatin. Following sacrifice after 4 weeks of healing, fractured femurs were extracted for microCT analysis and then three-point bending. Setting the region of interest to be 3.2 mm above and below the fracture line, we acquired standard and new microCT-derived measurements. The mineralized callus volume and the mineral density of the callus correlated positively with callus strength (rxy = -0.315, p = 0.016 and rxy = 0.444, p<0.0005, respectively) and stiffness (rxy = -0.271, p = 0.040 and rxy = 0.325, p = 0.013, respectively), but the fraction of the callus that mineralized and the moment of inertia of the callus did not. This fraction did correlate with energy-to-failure (rxy = -0.343, p = 0.0085). Of the microCT-derived measurements, quantifying defects within the outer bridging cortices of the callus produced the strongest correlation with both callus strength (rxy = 0.557, p<0.0001) and stiffness (rxy = 0.468, p = 0.0002). By both reducing structural defects and increasing mineralization, lovastatin appears to increase the callus strength.


Subject(s)
Femoral Fractures/diagnostic imaging , Femoral Fractures/physiopathology , Animals , Femoral Fractures/drug therapy , Fracture Healing/drug effects , Lovastatin/therapeutic use , Male , Rats , Rats, Sprague-Dawley , X-Ray Microtomography
5.
Br J Haematol ; 139(3): 434-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17910634

ABSTRACT

Impaired bone formation contributes to the lack of bone healing in multiple myeloma and there is a need for agents with bone anabolic properties to reverse the bone deficit in patients. Bortezomib, a proteasome inhibitor with antitumour efficacy in myeloma patients, enhanced new bone formation in mouse calvarial cultures; this effect was blocked by dickkopf 1(Dkk1), an antagonist of Wnt signalling implicated in myeloma bone disease. Bortezomib inhibited Dkk1 expression in calvariae and bone marrow-derived stromal cells, suggesting a novel mechanism by which bortezomib exerts its effects in bone. Clinical trials in patients with myeloma bone disease are needed to validate these results.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Osteogenesis/drug effects , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Animals , Antineoplastic Agents/antagonists & inhibitors , Boronic Acids/antagonists & inhibitors , Bortezomib , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Inbred ICR , Organ Culture Techniques , Osteoblasts/cytology , Osteoblasts/drug effects , Pyrazines/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction/methods , Skull/drug effects , Skull/physiology
6.
Mol Cancer Ther ; 6(6): 1701-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17541032

ABSTRACT

Development of new therapies for myeloma has been hindered by the lack of suitable preclinical animal models of the disease in which widespread tumor foci in the skeleton can be detected reliably. Traditional means of detecting skeletal tumor infiltration such as histopathology are cumbersome and labor-intensive and do not allow temporal monitoring of tumor progression or regression in response to therapy. To resolve this problem, we modified the Radl 5TGM1 model of myeloma bone disease such that fluorescent myeloma tumors can be optically imaged in situ. Here, we show that murine myeloma 5TGM1 tumor cells, engineered to express enhanced green fluorescent protein (eGFP; 5TGM1-eGFP cells), can be imaged in a temporal fashion using a fluorescence illuminator and a charge-coupled device camera in skeletons of live C57BL/KaLwRij mice. High-resolution, whole-body images of tumor-bearing mice revealed that myeloma cells homed almost exclusively to the skeleton, with multiple focal tumor foci in the axial skeleton, consistent with myeloma tumor distribution in humans. Finally, the tested antitumor treatment effect of Velcade (bortezomib), a proteasome inhibitor used clinically in myeloma, was readily detected by GFP imaging, suggesting the power of the technique in combination with the Radl 5TGM1-eGFP model for rapid preclinical assessment and sensitive monitoring of novel and potential therapeutics. Whole-body GFP imaging is practical, convenient, inexpensive, and rapid, and these advantages should enable a high throughput when evaluating in vivo efficacy of new potential antimyeloma therapeutics and assessing response to treatment.


Subject(s)
Multiple Myeloma/diagnosis , Animals , Fluorescence , Green Fluorescent Proteins/genetics , Mice
7.
Cancer Res ; 66(15): 7548-53, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885353

ABSTRACT

Parathyroid hormone-related peptide (PTHrP) is a major factor involved in tumor-induced osteolysis caused by breast cancers that have metastasized to bone. However, the molecular mechanisms that mediate PTHrP production by breast cancer cells are not entirely clear. We hypothesized that Gli2, a downstream transcriptional effector of the Hedgehog (Hh) signaling pathway, regulates PTHrP expression in metastatic breast cancer because the Hh pathway regulates physiologic PTHrP expression in the developing growth plate. Here, we show that Gli2 is expressed in several human cancer cell lines that cause osteolytic lesions in vivo and produce PTHrP (MDA-MB-231, RWGT2, and PC-3) but is not expressed in nonosteolytic cancer cell lines that do not secrete PTHrP (MCF-7, ZR-75, and T47D). Transient expression of Gli2 in MDA-MB-231 and MCF-7 breast cancer cells increased PTHrP promoter-luciferase activity dose dependently. Stable expression of Gli2 in MDA-MB-231 cells resulted in an increase in PTHrP protein in the conditioned medium. Alternatively, MDA-MB-231 cells stably transfected with Gli2-EnR, a repressor of Gli2 activity, exhibited a 72% to 93% decrease in PTHrP mRNA by quantitative real-time PCR when compared with control cells. To examine the effects of Gli2 on breast cancer-mediated osteolysis in vivo, athymic nude mice were inoculated with MDA-MB-231 cells stably expressing Gli2 or the empty vector. Following tumor cell inoculation via the left cardiac ventricle, Gli2-expressing tumors caused significantly more osteolysis. Together, these data suggest that PTHrP expression and osteolysis in vivo in human breast cancer cells is driven at least in part by Gli2.


Subject(s)
Breast Neoplasms/metabolism , Kruppel-Like Transcription Factors/metabolism , Nuclear Proteins/metabolism , Osteolysis/metabolism , Parathyroid Hormone-Related Protein/biosynthesis , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Culture Media, Conditioned , Humans , Kruppel-Like Transcription Factors/antagonists & inhibitors , Mice , Mice, Nude , Neoplasm Metastasis , Nuclear Proteins/antagonists & inhibitors , Osteolysis/pathology , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Promoter Regions, Genetic , Radiography , Transfection , Zinc Finger Protein Gli2
8.
Blood ; 102(1): 311-9, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12649140

ABSTRACT

Recent data have implicated macrophage inflammatory protein-1alpha (MIP-1alpha) in multiple myeloma (MM)-associated osteolysis. However, it is unclear whether the chemokine's effects are direct, to enhance osteolysis, or indirect and mediated through a reduction in tumor burden, or both. It is also unclear whether MIP-1alpha requires other factors such as receptor activator of nuclear factor-kappaB ligand (RANKL) for its effects on bone. In murine 5TGM1 (Radl) myeloma-bearing mice, administration of neutralizing anti-MIP-1alpha antibodies reduced tumor load assessed by monoclonal paraprotein titers, prevented splenomegaly, limited development of osteolytic lesions, and concomitantly reduced tumor growth in bone. To determine the effects of MIP-1alpha on bone in vivo, Chinese hamster ovary (CHO) cells secreting human MIP-1alpha (CHO/MIP-1alpha) were inoculated into athymic mice. Mice bearing intramuscular CHO/MIP-1alpha tumors developed lytic lesions at distant skeletal sites, which occurred earlier and were larger than those in mice with CHO/empty vector (EV) tumors. When experimental metastases were induced via intracardiac inoculation, mice bearing CHO/MIP-1alpha tumors developed hypercalcemia and significantly more osteolytic lesions than mice bearing CHO/EV tumors, with intramedullary CHO/MIP-1alpha tumors associated with significantly more tartrate-resistant acid phosphatase-positive (TRAP+) osteoclasts. Injection of recombinant MIP-1alpha over calvariae of normal mice evoked a striking increase in osteoclast formation, an effect dependent on RANK/RANKL signaling because MIP-1alpha had no effect in RANK-/- mice. Together, these results establish that MIP-1alpha is sufficient to induce MM-like destructive lesions in bone in vivo. Because, in the 5TGM1 model, blockade of osteoclastic resorption in other situations does not decrease tumor burden, we conclude that MIP-1alpha exerts a dual effect in myeloma, on osteoclasts, and tumor cells.


Subject(s)
Macrophage Inflammatory Proteins/pharmacology , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Osteolysis/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Bone Resorption/chemically induced , Chemokine CCL3 , Chemokine CCL4 , Disease Models, Animal , Hypercalcemia , Injections, Intralesional , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/immunology , Mice , Multiple Myeloma/pathology , Osteoclasts/cytology , Osteoclasts/drug effects , Osteolysis/etiology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Skull/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...