Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 57
1.
Biochem Pharmacol ; 188: 114579, 2021 06.
Article En | MEDLINE | ID: mdl-33895161

Osteolytic diseases, including breast cancer-induced osteolysis and postmenopausal osteoporosis, are attributed to excessive bone resorption by osteoclasts. Spleen tyrosine kinase (SYK) is involved in osteoclastogenesis and bone resorption, whose role in breast cancer though remains controversial. Effects of PRT062607 (PRT), a highly specific inhibitor of SYK, on the osteoclast and breast cancer functionalities are yet to be clarified. This study demonstrated the in vitro inhibitory actions of PRT on the osteoclast-specific gene expression, bone resorption, and osteoclastogenesis caused by receptor activator of nuclear factor kappa B ligand (RANKL), as well as its in vitro suppressive effects on the growth, migration and invasion of breast carcinoma cell line MDA-MB-231, which were achieved through PLCγ2 and PI3K-AKT-mTOR pathways. Further, we proved that PRT could prevent post-ovariectomy (OVX) loss of bone and breast cancer-induced bone destruction in vivo, which agreed with the in vitro outcomes. In conclusion, our findings suggest the potential value of PRT in managing osteolytic diseases mediated by osteoclasts.


Breast Neoplasms/enzymology , Cyclohexylamines/therapeutic use , Osteolysis/enzymology , Ovariectomy/adverse effects , Pyrimidines/therapeutic use , Syk Kinase/antagonists & inhibitors , Syk Kinase/metabolism , Animals , Bone Resorption/enzymology , Bone Resorption/pathology , Bone Resorption/prevention & control , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cell Line, Tumor , Cyclohexylamines/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Osteolysis/pathology , Osteolysis/prevention & control , Pyrimidines/pharmacology
2.
Mol Med Rep ; 23(1)2021 01.
Article En | MEDLINE | ID: mdl-33236155

Matrix metalloproteinase 2 (MMP2) is a well­characterized protein that is indispensable for extracellular matrix remodeling and other pathological processes, such as tumor progression and skeletal dysplasia. Excessive activation of MMP2 promotes osteolytic metastasis and bone destruction in late­stage cancers, while its loss­of­function mutations result in the decreased bone mineralization and generalized osteolysis occurring progressively in skeletal developmental disorders, particularly in multicentric osteolysis, nodulosis and arthropathy (MONA). Either upregulation or downregulation of MMP2 activity can result in the same osteolytic effects. Thus, different functions of MMP2 have been recently identified that could explain this observation. While MMP2 can degrade bone matrix, facilitate osteoclastogenesis and amplify various signaling pathways that enhance osteolysis in bone metastasis, its role in maintaining the number of bone cells, supporting osteocytic canalicular network formation and suppressing leptin­mediated inhibition of bone formation has been implicated in osteolytic disorders caused by MMP2 deficiency. Furthermore, the proangiogenic activity of MMP2 is one of the potential mechanisms that are associated with both pathological situations. In the present article, the latest research on MMP2 in bone homeostasis is reviewed and the mechanisms underlying the role of this protein in skeletal metastasis and developmental osteolysis are discussed.


Bone Neoplasms , Bone and Bones , Matrix Metalloproteinase 2 , Neoplasm Proteins , Osteochondrodysplasias , Osteolysis , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Bone and Bones/enzymology , Bone and Bones/pathology , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Osteochondrodysplasias/enzymology , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Osteolysis/enzymology , Osteolysis/genetics , Osteolysis/pathology
3.
Biosci Rep ; 40(6)2020 06 26.
Article En | MEDLINE | ID: mdl-32478376

Bone metastasis of colorectal cancer (CRC) cells leads to osteolysis. Aberrant activation of osteoclasts is responsible for bone resorption in tumor. In general, bone marrow-derived monocytes (BMMs) differentiate into osteoclasts, however, how CRC cells interact with BMMs and how to regulate the differentiation is elusive. We here report that CRC cells promote bone resorption in bone metastasis. Transcriptomic profiling revealed CCL3 up-regulated in MC-38 conditional medium treated BMMs. Further investigation demonstrated that CCL3 produced by BMMs facilitated cell infusion and thus promoted the osteoclastogenesis. In addition, CRC cells derived EGF stimulated the production of CCL3 in BMMs through activation of ERK/CREB pathway. Blockage of EGF or CCL3 can efficiently attenuate the osteolysis in bone metastasis of CRC.


Bone Neoplasms/enzymology , Chemokine CCL3/metabolism , Colorectal Neoplasms/enzymology , Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Osteoclasts/enzymology , Osteogenesis , Osteolysis/enzymology , Tibia/enzymology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Cell Communication , Cell Line, Tumor , Chemokine CCL3/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Mice, Inbred C57BL , Osteoclasts/pathology , Osteolysis/genetics , Osteolysis/pathology , Signal Transduction , Tibia/pathology
4.
Cell Death Dis ; 9(5): 498, 2018 05 01.
Article En | MEDLINE | ID: mdl-29703893

Osteolysis is an osteolytic lesion featured by enhanced osteoclast formation and potent bone erosion. Lacking of effective regimen for treatment of the pathological process highlights the importance of identifying agents that can suppress the differentiation and function of osteoclast. Artemether is a natural compound derived from Artemisia annua L. and it is popularized for the treatment of malaria. In present study, we demonstrated that artemether could suppress RANKL-induced osteoclastogenesis and expression of osteoclast marker genes such as tartrate-resistant acid phosphatase, cathepsin K, matrix metalloproteinase 9, nuclear factor of activated T-cell cytoplasmic 1, and dendritic cell-specific transmembrane protein. It inhibited the osteoclastic bone resorption in a dose-dependent manner in vitro. Furthermore, artemether attenuated RANKL-induced MAPKs (ERK, JNK, p-38) activity. In addition, we have showed that artemether was able to mitigate bone erosion in a murine model of LPS-induced inflammatory bone loss. Taken together, these findings suggest that artemether reduces inflammatory bone loss via inhibition of MAPKs activation during osteoclast differentiation, and it might be a potential candidate for the treatment of osteoclast-related disorders.


Artemether/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteolysis/prevention & control , Animals , Disease Models, Animal , Gene Expression Regulation , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Osteoclasts/enzymology , Osteoclasts/pathology , Osteogenesis/genetics , Osteolysis/chemically induced , Osteolysis/enzymology , Osteolysis/pathology , RANK Ligand/pharmacology , RAW 264.7 Cells
5.
J Cell Physiol ; 233(3): 2398-2408, 2018 Mar.
Article En | MEDLINE | ID: mdl-28731198

Currently, there are no medications available to treat aseptic loosening of orthopedic implants. Using osteoprotegerin fusion protein (OPG-Fc), we previously blocked instability-induced osteoclast differentiation and peri-prosthetic osteolysis. Wnt/ß-catenin signaling, which regulates OPG secretion from osteoblasts, also modulates the bone tissue response to mechanical loading. We hypothesized that activating Wnt/ß-catenin signaling by inhibiting glycogen synthase kinase-3ß (GSK-3ß) would reduce instability-induced bone loss through regulation of both osteoblast and osteoclast differentiation. We examined effects of GSK-3ß inhibition on regulation of RANKL and OPG in a rat model of mechanical instability-induced peri-implant osteolysis. The rats were treated daily with a GSK-3ß inhibitor, AR28 (20 mg/kg bw), for up to 5 days. Bone tissue and blood serum were assessed by qRT-PCR, immunohistochemistry, and ELISA on days 3 and 5, and by micro-CT on day 5. After 3 days of treatment with AR28, mRNA levels of ß-catenin, Runx2, Osterix, Col1α1, and ALP were increased leading to higher osteoblast numbers compared to vehicle-treated animals. BMP-2 and Wnt16 mRNA levels were downregulated by mechanical instability and this was rescued by GSK-3ß inhibition. Osteoclast numbers were decreased significantly after 3 days of GSK-3ß inhibition, which correlated with enhanced OPG mRNA expression. This was accompanied by decreased serum levels of TRAP5b on days 3 and 5. Treatment with AR28 upregulated osteoblast differentiation, while osteoclastogenesis was blunted, leading to increased bone mass by day 5. These data suggest that GSK-3ß inactivation suppresses osteolysis through regulating both osteoblast and osteoclast differentiation in a rat model of instability-induced osteolysis.


Cell Differentiation/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteolysis/prevention & control , Prosthesis Failure , Protein Kinase Inhibitors/pharmacology , Tibia/drug effects , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Plates , Cell Proliferation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Gene Expression Regulation , Glycogen Synthase Kinase 3 beta/metabolism , Male , Osteoblasts/enzymology , Osteoblasts/pathology , Osteoclasts/enzymology , Osteoclasts/pathology , Osteolysis/enzymology , Osteolysis/genetics , Osteolysis/pathology , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Prosthesis Implantation/instrumentation , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase/blood , Tibia/enzymology , Tibia/pathology , Tibia/surgery , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/genetics , beta Catenin/metabolism
6.
J Cell Physiol ; 232(3): 617-624, 2017 Mar.
Article En | MEDLINE | ID: mdl-27312515

Bone homeostasis is maintained by a balance between resorption of the bone matrix and its replacement by new bone. Osteoclasts play a crucially important role in bone metabolism. They are responsible for bone resorption under pathophysiological conditions. Differentiation of these cells, which are derived from bone marrow cells, depends on receptor activator of NF-κB ligand (RANKL). RANKL-induced osteoclastogenesis is regulated by the phosphoinositide (PI) signaling pathway, in which diacylglycerol (DG) serves as a second messenger in signal transduction. In this study, we examined the functional implications of DG kinase (DGK), an enzyme family responsible for DG metabolism, for osteoclast differentiation and activity. Of DGKs, DGKζ is most abundantly expressed in osteoclast precursors such as bone marrow-derived monocytes/macrophages. During osteoclast differentiation from precursor cells, DGKζ is downregulated at the protein level. In this regard, we found that DGKζ deletion enhances osteoclast differentiation and bone resorption activity under inflammatory conditions in an animal model of osteolysis. Furthermore, DGKζ deficiency upregulates RANKL expression in response to TNFα stimulation. Collectively, results suggest that DGKζ is silent under normal conditions, but it serves as a negative regulator in osteoclast function under inflammatory conditions. Downregulation of DGKζ might be one factor predisposing a person to osteolytic bone destruction in pathological conditions. J. Cell. Physiol. 232: 617-624, 2017. © 2016 Wiley Periodicals, Inc.


Bone Resorption/enzymology , Bone Resorption/pathology , Cell Differentiation , Diacylglycerol Kinase/metabolism , Down-Regulation , Inflammation/pathology , Osteoclasts/pathology , Animals , Biomarkers/metabolism , Bone Density/drug effects , Bone Resorption/complications , Cell Differentiation/drug effects , Disease Models, Animal , Fibroblasts/metabolism , Inflammation/complications , Inflammation/enzymology , Isoenzymes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice, Knockout , Osteoclasts/drug effects , Osteolysis/complications , Osteolysis/enzymology , Osteolysis/pathology , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Tomography, X-Ray Computed , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
7.
Sci Transl Med ; 8(353): 353ra113, 2016 08 24.
Article En | MEDLINE | ID: mdl-27559096

Myelomatous bone disease is characterized by the development of lytic bone lesions and a concomitant reduction in bone formation, leading to chronic bone pain and fractures. To understand the underlying mechanism, we investigated the contribution of myeloma-expressed thymidine phosphorylase (TP) to bone lesions. In osteoblast progenitors, TP up-regulated the methylation of RUNX2 and osterix, leading to decreased bone formation. In osteoclast progenitors, TP up-regulated the methylation of IRF8 and thereby enhanced expression of NFATc1 (nuclear factor of activated T cells, cytoplasmic 1 protein), leading to increased bone resorption. TP reversibly catalyzes thymidine into thymine and 2-deoxy-d-ribose (2DDR). Myeloma-secreted 2DDR bound to integrin αVß3/α5ß1 in the progenitors, activated PI3K (phosphoinositide 3-kinase)/Akt signaling, and increased DNMT3A (DNA methyltransferase 3A) expression, resulting in hypermethylation of RUNX2, osterix, and IRF8 This study elucidates an important mechanism for myeloma-induced bone lesions, suggesting that targeting TP may be a viable approach to healing resorbed bone in patients. Because TP overexpression is common in bone-metastatic tumors, our findings could have additional mechanistic implications.


Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Bone Resorption/enzymology , Bone Resorption/pathology , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Osteogenesis/physiology , Thymidine Phosphorylase/metabolism , Bone Neoplasms/physiopathology , Bone Resorption/physiopathology , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/genetics , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , DNA Methyltransferase 3A , Down-Regulation , Humans , Interferon Regulatory Factors/genetics , Multiple Myeloma/physiopathology , Osteoblasts/pathology , Osteoblasts/physiology , Osteoclasts/pathology , Osteoclasts/physiology , Osteolysis/enzymology , Osteolysis/pathology , Osteolysis/prevention & control , RANK Ligand/metabolism , Sp7 Transcription Factor/genetics , Thymidine Phosphorylase/antagonists & inhibitors , Up-Regulation
8.
J Clin Invest ; 126(5): 1759-72, 2016 05 02.
Article En | MEDLINE | ID: mdl-27043283

Multiple myeloma (MM) cells secrete osteoclastogenic factors that promote osteolytic lesions; however, the identity of these factors is largely unknown. Here, we performed a screen of human myeloma cells to identify pro-osteoclastogenic agents that could potentially serve as therapeutic targets for ameliorating MM-associated bone disease. We found that myeloma cells express high levels of the matrix metalloproteinase MMP-13 and determined that MMP-13 directly enhances osteoclast multinucleation and bone-resorptive activity by triggering upregulation of the cell fusogen DC-STAMP. Moreover, this effect was independent of the proteolytic activity of the enzyme. Further, in mouse xenograft models, silencing MMP-13 expression in myeloma cells inhibited the development of osteolytic lesions. In patient cohorts, MMP-13 expression was localized to BM-associated myeloma cells, while elevated MMP-13 serum levels were able to correctly predict the presence of active bone disease. Together, these data demonstrate that MMP-13 is critical for the development of osteolytic lesions in MM and that targeting the MMP-13 protein - rather than its catalytic activity - constitutes a potential approach to mitigating bone disease in affected patients.


Matrix Metalloproteinase 13/metabolism , Multiple Myeloma/enzymology , Neoplasm Proteins/metabolism , Osteoclasts/enzymology , Osteolysis/enzymology , Animals , Cell Fusion , Female , Heterografts , Humans , Male , Matrix Metalloproteinase 13/genetics , Mice , Mice, Knockout , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Neoplasm Proteins/genetics , Neoplasm Transplantation , Osteoclasts/pathology , Osteolysis/genetics , Osteolysis/pathology , Osteolysis/therapy
9.
Am J Med Genet A ; 170A(2): 410-417, 2016 Feb.
Article En | MEDLINE | ID: mdl-26601801

​Multicentric osteolysis nodulosis and arthropathy (MONA) is an infrequently described autosomal recessive skeletal dysplasia characterized by progressive osteolysis and arthropathy. Inactivating mutations in MMP2, encoding matrix metalloproteinase-2, are known to cause this disorder. Fifteen families with mutations in MMP2 have been reported in literature. In this study we screened thirteen individuals from eleven families for MMP2 mutations and identified eight mutations (five novel and three known variants). We characterize the clinical, radiographic and molecular findings in all individuals with molecularly proven MONA from the present cohort and previous reports, and provide a comprehensive review of the MMP2 related disorders.


Matrix Metalloproteinase 2/genetics , Mutation/genetics , Osteolysis/genetics , Adolescent , Amino Acid Sequence , Child , Child, Preschool , Cohort Studies , Female , Homozygote , Humans , Infant , Infant, Newborn , Male , Molecular Sequence Data , Osteolysis/enzymology , Osteolysis/pathology , Prognosis , Sequence Homology, Amino Acid
10.
Biomaterials ; 69: 12-21, 2015 Nov.
Article En | MEDLINE | ID: mdl-26275858

Aseptic loosening is associated with the development of wear debris-induced peri-implant osteolytic bone disease caused by an increased osteoclastic bone resorption and decreased osteoblastic bone formation. However, no effective measures for the prevention and treatment of peri-implant osteolysis currently exist. The aim of this study was to determine whether lithium chloride (LiCl), a selective inhibitor of glycogen synthetase kinase 3 beta (GSK-3ß), mitigates wear debris-induced osteolysis in a murine calvarial model of osteolysis. GSK-3ß is activated by titanium (Ti) particles, and implantation of Ti particles on the calvarial surface in C57BL/6 mice resulted in osteolysis caused by an increase in the number of osteoclasts and a decrease in the number of osteoblasts. Mice implanted with Ti particles were gavage-fed LiCl (50 or 200 mg kg(-1)d(-1)), 6 days per week for 2 weeks. The LiCl treatment significantly inhibited GSK-3ß activity and increased ß-catenin and axin-2 expression in a dose-dependent manner, dramatically mitigating the Ti particle-induced suppression of osteoblast numbers and the expression of bone formation markers. Finally, we demonstrated that inhibition of GSK-3ß suppresses osteoclast differentiation and reduces the severity of Ti particle-induced osteolysis. The results of this study indicate that Ti particle-induced osteolysis is partly dependent on GSK-3ß and, therefore, the canonical Wnt signaling pathway. This suggests that selective inhibitors of GSK-3ß such as LiCl may help prevent and treat wear debris-induced osteolysis.


Glycogen Synthase Kinase 3/antagonists & inhibitors , Lithium Chloride/therapeutic use , Osteolysis/drug therapy , Osteolysis/enzymology , Protein Kinase Inhibitors/therapeutic use , Animals , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Mice , Mice, Inbred C57BL , Osteolysis/etiology , Osteolysis/pathology , Prostheses and Implants/adverse effects , Skull/drug effects , Skull/enzymology , Skull/pathology , Titanium/adverse effects
11.
Int J Mol Sci ; 15(12): 21913-34, 2014 Nov 28.
Article En | MEDLINE | ID: mdl-25464380

The aim of this study was to assess the effect of naringenin on osteoclastogenesis and titanium particle-induced osteolysis. Osteolysis from wear-induced particles and aseptic loosening are the most frequent late complications of total joint arthroplasty leading to revision of the prosthesis. Osteolysis during aseptic loosening is most likely due to increased bone resorption by osteoclasts. Through in vitro studies, we demonstrated that naringenin, a naturally occurring flavanone in grapefruit and tomatoes, exerts potent inhibitory effects on the ligand of the receptor activator of nuclear factor-κB (RANKL)-induced osteoclastogenesis and revealed that the mechanism of action of naringenin, which inhibited osteoclastogenesis by suppression of the p38 signaling pathway. Through in vivo studies, we proved that naringenin attenuated titanium particle-induced osteolysis in a mouse calvarial model. In general, we demonstrated that naringenin inhibited osteoclastogenesis via suppression of p38 signaling in vitro and attenuated titanium particle-induced osteolysis in vivo. This study also suggested that naringenin has significant potential for the treatment of osteolysis-related diseases caused by excessive osteoclast formation and activity.


Flavanones/pharmacology , MAP Kinase Signaling System/drug effects , Osteoclasts/enzymology , Osteogenesis/drug effects , Osteolysis/enzymology , RANK Ligand/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Actins/metabolism , Animals , Anisomycin/pharmacology , Bone Resorption/complications , Bone Resorption/pathology , Cell Death/drug effects , Cell Differentiation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavanones/chemistry , Gene Expression Regulation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Models, Biological , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteoclasts/pathology , Osteolysis/complications , Osteolysis/pathology , Titanium
12.
J Biol Chem ; 288(47): 33634-33641, 2013 Nov 22.
Article En | MEDLINE | ID: mdl-24081142

Phospholipase C γ2 (PLCγ2) is a critical regulator of innate immune cells and osteoclasts (OCs) during inflammatory arthritis. Both the catalytic domain and the adaptor motifs of PLCγ2 are required for OC formation and function. Due to the high homology between the catalytic domains of PLCγ2 and the ubiquitously expressed PLCγ1, molecules encompassing the adaptor motifs of PLCγ2 were designed to test the hypothesis that uncoupling the adaptor and catalytic functions of PLCγ2 could specifically inhibit osteoclastogenesis and bone erosion. Wild-type (WT) bone marrow macrophages (BMM) that overexpress the tandem Src homology 2 (SH2) domains of PLCγ2 (SH2(N+C)) failed to form mature OCs and resorb bone in vitro. Activation of the receptor activator of NF-κB (RANK) signaling pathway, which is critical for OC development, was impaired in cells expressing SH2(N+C). Arrest in OC differentiation was evidenced by a reduction of p38 and Iκ-Bα phosphorylation as well as decreased NFATc1 and c-Fos/c-Jun levels. Consistent with our hypothesis, SH2(N+C) abrogated formation of the RANK-Gab2 complex, which mediates NF-κB and AP-1 activation following RANK ligand (RANKL) stimulation. Furthermore, the ability of SH2(N+C) to prevent inflammatory osteolysis was examined in vivo following RANKL or LPS injections over the calvaria. Both models induced osteolysis in the control group, whereas the SH2(N+C)-treated cohort was largely protected from bone erosion. Collectively, these data indicate that inflammatory osteolysis can be abrogated by treatment with a molecule composed of the tandem SH2 domains of PLCγ2.


Bone Marrow Cells/enzymology , Osteoclasts/enzymology , Osteolysis/enzymology , Phospholipase C gamma/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Cells/pathology , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Lipopolysaccharides/toxicity , Mice , NF-KappaB Inhibitor alpha , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/pathology , Osteolysis/drug therapy , Osteolysis/genetics , Osteolysis/pathology , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , src Homology Domains
13.
J Biol Chem ; 288(42): 30399-30410, 2013 Oct 18.
Article En | MEDLINE | ID: mdl-24005670

Abnormal osteoclast formation and osteolysis are the hallmarks of multiple myeloma (MM) bone disease, yet the underlying molecular mechanisms are incompletely understood. Here, we show that the AKT pathway was up-regulated in primary bone marrow monocytes (BMM) from patients with MM, which resulted in sustained high expression of the receptor activator of NF-κB (RANK) in osteoclast precursors. The up-regulation of RANK expression and osteoclast formation in the MM BMM cultures was blocked by AKT inhibition. Conditioned media from MM cell cultures activated AKT and increased RANK expression and osteoclast formation in BMM cultures. Inhibiting AKT in cultured MM cells decreased their growth and ability to promote osteoclast formation. Of clinical significance, systemic administration of the AKT inhibitor LY294002 blocked the formation of tumor tissues in the bone marrow cavity and essentially abolished the MM-induced osteoclast formation and osteolysis in SCID mice. The level of activating transcription factor 4 (ATF4) protein was up-regulated in the BMM cultures from multiple myeloma patients. Adenoviral overexpression of ATF4 activated RANK expression in osteoclast precursors. These results demonstrate a new role of AKT in the MM promotion of osteoclast formation and bone osteolysis through, at least in part, the ATF4-dependent up-regulation of RANK expression in osteoclast precursors.


Gene Expression Regulation, Neoplastic , Multiple Myeloma/enzymology , Osteoclasts/enzymology , Osteolysis/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation , Activating Transcription Factor 4/metabolism , Animals , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Female , Heterografts , Humans , Male , Mice , Mice, SCID , Morpholines/pharmacology , Multiple Myeloma/pathology , Neoplasm Transplantation , Osteoclasts/pathology , Osteolysis/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/metabolism , Tumor Cells, Cultured
14.
Biomed Res Int ; 2013: 230805, 2013.
Article En | MEDLINE | ID: mdl-23862137

The leading complication of total joint replacement is periprosthetic osteolysis, which often results in aseptic loosening of the implant, leading to revision surgery. Extracellular matrix degradation and connective tissue remodeling around implants have been considered as major biological events in the periprosthetic loosening. Critical mediators of wear particle-induced inflammatory osteolysis released by periprosthetic synovial cells (mainly macrophages) are inflammatory cytokines, chemokines, and proteolytic enzymes, mainly matrix metalloproteinases (MMPs). Numerous studies reveal a strong interdependence of MMP expression and activity with the molecular mechanisms that control the composition and turnover of periprosthetic matrices. MMPs can either actively modulate or be modulated by the molecular mechanisms that determine the debris-induced remodeling of the periprosthetic microenvironment. In the present study, the molecular mechanisms that control the composition, turnover, and activity of matrix macromolecules within the periprosthetic microenvironment exposed to wear debris are summarized and presented. Special emphasis is given to MMPs and their endogenous tissue inhibitors (TIMPs), as well as to the proteasome pathway, which appears to be an elegant molecular regulator of specific matrix macromolecules (including specific MMPs and TIMPs). Furthermore, strong rationale for potential clinical applications of the described molecular mechanisms to the treatment of periprosthetic loosening and osteolysis is provided.


Extracellular Matrix/metabolism , Matrix Metalloproteinases/metabolism , Osteolysis/enzymology , Osteolysis/etiology , Prosthesis Failure/adverse effects , Proteasome Endopeptidase Complex/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Humans , Osteolysis/therapy
15.
Exp Hematol ; 41(6): 547-557.e2, 2013 Jun.
Article En | MEDLINE | ID: mdl-23435312

Multiple myeloma (MM) cells typically grow in focal lesions, stimulating osteoclasts that destroy bone and support MM. Osteoclasts and MM cells are hypermetabolic. The coenzyme nicotinamide adenine dinucleotide (NAD(+)) is not only essential for cellular metabolism; it also affects activity of NAD-dependent enzymes, such as PARP-1 and SIRT-1. Nicotinamide phosphoribosyltransferase (NAMPT/PBEF/visfatin, encoded by PBEF1) is a rate-limiting enzyme in NAD(+) biosynthesis from nicotinamide. Coculture of primary MM cells with osteoclasts induced PBEF1 upregulation in both cell types. PBEF1 expression was higher in experimental myelomatous bones than in nonmyelomatous bone and higher in MM patients' plasma cells than in healthy donors' counterparts. APO866 is a specific PBEF1 inhibitor known to deplete cellular NAD(+). APO866 at low nanomolar concentrations inhibited growth of primary MM cells or MM cell lines cultured alone or cocultured with osteoclasts and induced apoptosis in these cells. PBEF1 activity and NAD(+) content were reduced in MM cells by APO866, resulting in lower activity of PARP-1 and SIRT-1. The inhibitory effect of APO866 on MM cell growth was abrogated by supplementation of extracellular NAD(+) or NAM. APO866 inhibited NF-κB activity in osteoclast precursors and suppressed osteoclast formation and activity. PBEF1 knockdown similarly inhibited MM cell growth and osteoclast formation. In the SCID-rab model, APO866 inhibited growth of primary MM and H929 cells and prevented bone disease. These findings indicate that MM cells and osteoclasts are highly sensitive to NAD(+) depletion and that PBEF1 inhibition represents a novel approach to target cellular metabolism and inhibit PARP-1 and bone disease in MM.


Cytokines/physiology , Multiple Myeloma/enzymology , Neoplasm Proteins/physiology , Nicotinamide Phosphoribosyltransferase/physiology , Osteoclasts/enzymology , Osteolysis/enzymology , Acrylamides/pharmacology , Animals , Bone and Bones/pathology , Cell Differentiation/drug effects , Coculture Techniques , Cytokines/antagonists & inhibitors , Enzyme Induction , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Mice, SCID , Multiple Myeloma/complications , Multiple Myeloma/pathology , NAD/metabolism , NF-kappa B/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Niacinamide/metabolism , Nicotinamide Mononucleotide/analogs & derivatives , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Osteoclasts/physiology , Osteolysis/etiology , Osteolysis/pathology , Piperidines/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Rabbits , Sirtuin 1/metabolism , Tumor Cells, Cultured/metabolism , Up-Regulation
16.
Leukemia ; 26(9): 2114-23, 2012 Sep.
Article En | MEDLINE | ID: mdl-22425892

Bone destruction is a hallmark of multiple myeloma and affects more than 80% of patients. However, current therapy is unable to completely cure and/or prevent bone lesions. Although it is accepted that myeloma cells mediate bone destruction by inhibition of osteoblasts and activation of osteoclasts, the underlying mechanism is still poorly understood. This study demonstrates that constitutive activation of p38 mitogen-activated protein kinase in myeloma cells is responsible for myeloma-induced osteolysis. Our results show that p38 is constitutively activated in most myeloma cell lines and primary myeloma cells from patients. Myeloma cells with high/detectable p38 activity, but not those with low/undetectable p38 activity, injected into severe combined immunodeficient (SCID) or SCID-hu mice caused bone destruction. Inhibition or knockdown of p38 in human myeloma reduced or prevented myeloma-induced osteolytic bone lesions without affecting tumor growth, survival, or homing to bone. Mechanistic studies showed that myeloma cell p38 activity inhibited osteoblastogenesis and bone formation and activated osteoclastogenesis and bone resorption in myeloma-bearing SCID mice. This study elucidates a novel molecular mechanism-activation of p38 signaling in myeloma cells-by which myeloma cells induce osteolytic bone lesions, and indicates that targeting myeloma cell p38 may be a viable approach to treating or preventing myeloma bone disease.


Bone Diseases/etiology , Multiple Myeloma/complications , Multiple Myeloma/enzymology , Osteolysis/etiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Blotting, Western , Bone Diseases/enzymology , Bone Diseases/pathology , Case-Control Studies , Cell Communication , Cell Proliferation , Humans , Immunoenzyme Techniques , Mice , Mice, SCID , Multiple Myeloma/pathology , Osteolysis/enzymology , Osteolysis/pathology , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
17.
Acta Neurochir (Wien) ; 154(4): 611-20; discussion 620, 2012 Apr.
Article En | MEDLINE | ID: mdl-22327326

OBJECT: Although bone invasion and hyperostosis are common phenomena in patients with intracranial meningiomas, the basic pathomechanism is not fully understood. Based on an immunohistochemical study of surgically resected samples with hyperostosis, we postulate a possible mechanism of hyperostosis in patients with intracranial meningiomas. MATERIALS AND METHODS: Forty-six meningiomas were evaluated in this study. Twenty-six meningiomas associated with hyperostosis specimens served as the study group, and 20 meningiomas without any bony changes served as controls. An immunohistochemical staining technique was used to detect the expression of matrix metalloproteinase (MMP)-2, -9, and -13, membrane type (MT)1-MMP, estrogen receptor (ER), and progesterone receptor (PR) in the main tumor and hyperostotic portions of the studied samples. RESULTS: In the non-hyperostosis group, expression of MMP-13, MT1-MMP, and ER was significantly less than in the main tumor portion of hyperostotic meningiomas, while there was no difference in the expression of MMP-2 and -9 and PR in the main tumor between the two groups. In the hyperostosis group, the immunoreactivity of MMP-2 in the hyperostotic portion revealed a higher pattern of expression than the main tumor (p < 0.002). The expression of MMP-9, MT1-MMP, ER, and PR had relatively positive immunoreactivity in the main tumor portion (P < 0.05). CONCLUSIONS: Increased expression of MMP-13 and MT1-MMP in the tumor portion of hyperostosis of meningiomas might contribute to the initiation of osteolysis. Activated MMP-2 in hyperostotic lesions may change the physiological metabolism of the skull bone, thus playing an important role in hyperostosis formation.


Hyperostosis/enzymology , Matrix Metalloproteinases/physiology , Meningeal Neoplasms/enzymology , Meningioma/enzymology , Skull/enzymology , Biomarkers, Tumor/physiology , Female , Humans , Hyperostosis/pathology , Hyperostosis/physiopathology , Male , Matrix Metalloproteinase 13/physiology , Matrix Metalloproteinase 14/physiology , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinase 9/physiology , Meningeal Neoplasms/pathology , Meningeal Neoplasms/physiopathology , Meningioma/pathology , Meningioma/physiopathology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Osteolysis/enzymology , Osteolysis/pathology , Osteolysis/physiopathology , Skull/pathology , Skull/physiopathology
18.
PLoS One ; 7(1): e29615, 2012.
Article En | MEDLINE | ID: mdl-22253746

We investigated the effects of the matrix metalloproteinase 13 (MMP13)-selective inhibitor, 5-(4-{4-[4-(4-fluorophenyl)-1,3-oxazol-2-yl]phenoxy}phenoxy)-5-(2-methoxyethyl) pyrimidine-2,4,6(1H,3H,5H)-trione (Cmpd-1), on the primary tumor growth and breast cancer-associated bone remodeling using xenograft and syngeneic mouse models. We used human breast cancer MDA-MB-231 cells inoculated into the mammary fat pad and left ventricle of BALB/c Nu/Nu mice, respectively, and spontaneously metastasizing 4T1.2-Luc mouse mammary cells inoculated into mammary fat pad of BALB/c mice. In a prevention setting, treatment with Cmpd-1 markedly delayed the growth of primary tumors in both models, and reduced the onset and severity of osteolytic lesions in the MDA-MB-231 intracardiac model. Intervention treatment with Cmpd-1 on established MDA-MB-231 primary tumors also significantly inhibited subsequent growth. In contrast, no effects of Cmpd-1 were observed on soft organ metastatic burden following intracardiac or mammary fat pad inoculations of MDA-MB-231 and 4T1.2-Luc cells respectively. MMP13 immunostaining of clinical primary breast tumors and experimental mice tumors revealed intra-tumoral and stromal expression in most tumors, and vasculature expression in all. MMP13 was also detected in osteoblasts in clinical samples of breast-to-bone metastases. The data suggest that MMP13-selective inhibitors, which lack musculoskeletal side effects, may have therapeutic potential both in primary breast cancer and cancer-induced bone osteolysis.


Breast Neoplasms/complications , Breast Neoplasms/pathology , Matrix Metalloproteinase Inhibitors , Osteolysis/etiology , Osteolysis/pathology , Protease Inhibitors/pharmacology , Animals , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/enzymology , Breast Neoplasms/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Creatine Kinase/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Fluorescent Antibody Technique , Humans , Matrix Metalloproteinase 13/metabolism , Mice , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoblasts/pathology , Osteolysis/enzymology , Protease Inhibitors/blood , Xenograft Model Antitumor Assays
19.
Curr Protein Pept Sci ; 13(2): 164-79, 2012 Mar.
Article En | MEDLINE | ID: mdl-22044155

Over the last three decades, V-ATPases have emerged from the obscurity of poorly understood membrane proton transport phenomena to being recognized as ubiquitous proton pumps that underlie vital cellular processes in all eukaryotic and many prokaryotic cells. These exquisitely complex molecular motors also engage in diverse specialized roles contributing to development, tissue function and pH homeostasis within complex organisms. Increasingly, mutations and misappropriation of V-ATPase function have been linked to diseases, ranging from sclerosing bone pathologies and renal tubular acidosis to bone-loss disorders and cancer metastasis. Much remains to be learned about the details of V-ATPase cell and molecular biology; nevertheless, interest in V-ATPases as potential therapeutic targets has burgeoned in recent years. In this review, we present a history of our involvement and contributions to the understanding of V-ATPase structure and function and our nascent and ongoing contributions to translating the knowledge gained from basic research on the nature of V-ATPases into tools for drug discovery. We focus here primarily on the treatment of bone-loss pathologies, like osteoporosis, and present proof-of-concept for a drug screening strategy based on targeting a3-B2 subunit interactions within the V-ATPase complex.


Drug Discovery/methods , Osteoporosis/drug therapy , Osteoporosis/enzymology , Protein Interaction Maps/drug effects , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Humans , Models, Molecular , Osteolysis/drug therapy , Osteolysis/enzymology , Protein Subunits/chemistry , Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/chemistry
20.
Cancer Res ; 70(21): 8329-38, 2010 Nov 01.
Article En | MEDLINE | ID: mdl-20978204

Excessive bone destruction is a major cause of morbidity in myeloma patients. However, the biological mechanisms involved in the pathogenesis of myeloma-induced bone disease are not fully understood. Heparanase, an enzyme that cleaves the heparan sulfate chains of proteoglycans, is upregulated in a variety of human tumors, including multiple myeloma. We previously showed that heparanase promotes robust myeloma tumor growth and supports spontaneous metastasis of tumor cells to bone. In the present study, we show, for the first time, that the expression of heparanase by myeloma tumor cells remarkably enhances bone destruction locally within the tumor microenvironment. In addition, enhanced heparanase expression in the primary tumor also stimulated systemic osteoclastogenesis and osteolysis, thus mimicking the systemic osteoporosis often seen in myeloma patients. These effects occur, at least in part, as the result of a significant elevation in the expression and secretion of receptor activator of NF-κB ligand (RANKL) by heparanase-expressing myeloma cells. Moreover, analysis of bone marrow biopsies from myeloma patients reveals a positive correlation between the level of expression of heparanase and RANKL. Together, these discoveries reveal a novel and key role for heparanase in promoting tumor osteolysis and show that RANKL is central to the mechanism of heparanase-mediated osteolysis in myeloma.


Glucuronidase/physiology , Multiple Myeloma/enzymology , Osteolysis/enzymology , RANK Ligand/metabolism , Animals , Blotting, Western , Bone Resorption , Cell Differentiation , Cell Proliferation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Femur/metabolism , Femur/pathology , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Male , Mice , Mice, SCID , Multiple Myeloma/pathology , Osteoclasts/cytology , Osteoclasts/metabolism , Osteolysis/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tibia/metabolism , Tibia/pathology , Up-Regulation
...