Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 17319, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33057033

ABSTRACT

Multiple Myeloma (MM) induces bone destruction, decreases bone formation, and increases marrow angiogenesis in patients. We reported that osteocytes (Ocys) directly interact with MM cells to increase tumor growth and expression of Ocy-derived factors that promote bone resorption and suppress bone formation. However, the contribution of Ocys to enhanced marrow vascularization in MM is unclear. Since the MM microenvironment is hypoxic, we assessed if hypoxia and/or interactions with MM cells increases pro-angiogenic signaling in Ocys. Hypoxia and/or co-culture with MM cells significantly increased Vegf-a expression in MLOA5-Ocys, and conditioned media (CM) from MLOA5s or MM-MLOA5 co-cultured in hypoxia, significantly increased endothelial tube length compared to normoxic CM. Further, Vegf-a knockdown in MLOA5s or primary Ocys co-cultured with MM cells or neutralizing Vegf-a in MM-Ocy co-culture CM completely blocked the increased endothelial activity. Importantly, Vegf-a-expressing Ocy numbers were significantly increased in MM-injected mouse bones, positively correlating with tumor vessel area. Finally, we demonstrate that direct contact with MM cells increases Ocy Fgf23, which enhanced Vegf-a expression in Ocys. Fgf23 deletion in Ocys blocked these changes. These results suggest hypoxia and MM cells induce a pro-angiogenic phenotype in Ocys via Fgf23 and Vegf-a signaling, which can promote MM-induced marrow vascularization.


Subject(s)
Bone Marrow/blood supply , Fibroblast Growth Factors/physiology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neovascularization, Pathologic/genetics , Osteocytes/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone Resorption/etiology , Cell Line , Female , Fibroblast Growth Factor-23 , Gene Expression/genetics , Humans , Mice , Mice, Inbred C57BL , Osteocytes/metabolism , Osteogenesis , Tumor Microenvironment
2.
Leuk Res ; 41: 85-95, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26740054

ABSTRACT

Bortezomib, a clinical drug for multiple myeloma (MM) and mantle cell lymphoma, exhibits complex mechanisms of action, which vary depending on the cancer type and the critical genetic alterations of each cancer. Here we investigated the signaling mechanisms of bortezomib in mouse B lymphoma and human MM cells deficient in a new tumor suppressor gene, TRAF3. We found that bortezomib consistently induced up-regulation of the cell cycle inhibitor p21(WAF1) and the pro-apoptotic protein Noxa as well as cleavage of the anti-apoptotic protein Mcl-1. Interestingly, bortezomib induced the activation of NF-κB1 and the accumulation of the oncoprotein c-Myc, but inhibited the activation of NF-κB2. Furthermore, we demonstrated that oridonin (an inhibitor of NF-κB1 and NF-κB2) or AD 198 (a drug targeting c-Myc) drastically potentiated the anti-cancer effects of bortezomib in TRAF3-deficient malignant B cells. Taken together, our findings increase the understanding of the mechanisms of action of bortezomib, which would aid the design of novel bortezomib-based combination therapies. Our results also provide a rationale for clinical evaluation of the combinations of bortezomib and oridonin (or other inhibitors of NF-κB1/2) or AD 198 (or other drugs targeting c-Myc) in the treatment of lymphoma and MM, especially in patients containing TRAF3 deletions or relevant mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Lymphoma, B-Cell/genetics , Multiple Myeloma/genetics , Signal Transduction/drug effects , TNF Receptor-Associated Factor 3/deficiency , Animals , Disease Models, Animal , Humans , Immunoblotting , Immunoprecipitation , Mice , Proteasome Inhibitors/pharmacology , TNF Receptor-Associated Factor 3/genetics , Transduction, Genetic
3.
J Hematol Oncol ; 7: 56, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25200342

ABSTRACT

BACKGROUND: Identification of novel genetic risk factors is imperative for a better understanding of B lymphomagenesis and for the development of novel therapeutic strategies. TRAF3, a critical regulator of B cell survival, was recently recognized as a tumor suppressor gene in B lymphocytes. The present study aimed to identify novel oncogenes involved in malignant transformation of TRAF3-deficient B cells. METHODS: We used microarray analysis to identify genes differentially expressed in TRAF3-/- mouse splenic B lymphomas. We employed lentiviral vector-mediated knockdown or overexpression to manipulate gene expression in human multiple myeloma (MM) cell lines. We analyzed cell apoptosis and proliferation using flow cytometry, and performed biochemical studies to investigate signaling mechanisms. To delineate protein-protein interactions, we applied affinity purification followed by mass spectrometry-based sequencing. RESULTS: We identified mutated in colorectal cancer (MCC) as a gene strikingly up-regulated in TRAF3-deficient mouse B lymphomas and human MM cell lines. Aberrant up-regulation of MCC also occurs in a variety of primary human B cell malignancies, including non-Hodgkin lymphoma (NHL) and MM. In contrast, MCC expression was not detected in normal or premalignant TRAF3-/- B cells even after treatment with B cell stimuli, suggesting that aberrant up-regulation of MCC is specifically associated with malignant transformation of B cells. In elucidating the functional roles of MCC in malignant B cells, we found that lentiviral shRNA vector-mediated knockdown of MCC induced apoptosis and inhibited proliferation in human MM cells. Experiments of knockdown and overexpression of MCC allowed us to identify several downstream targets of MCC in human MM cells, including phospho-ERK, c-Myc, p27, cyclin B1, Mcl-1, caspases 8 and 3. Furthermore, we identified 365 proteins (including 326 novel MCC-interactors) in the MCC interactome, among which PARP1 and PHB2 were two hubs of MCC signaling pathways in human MM cells. CONCLUSIONS: Our results indicate that in sharp contrast to its tumor suppressive role in colorectal cancer, MCC functions as an oncogene in B cells. Our findings suggest that MCC may serve as a diagnostic marker and therapeutic target in B cell malignancies, including NHL and MM.


Subject(s)
B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , Genes, MCC/genetics , Oncogenes/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Lymphoma, Non-Hodgkin/genetics , Mice , Mice, Knockout , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , Prohibitins , Reverse Transcriptase Polymerase Chain Reaction , TNF Receptor-Associated Factor 3/deficiency , Tandem Mass Spectrometry
4.
Leuk Res ; 38(3): 393-401, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24418753

ABSTRACT

Using a mouse model with the tumor suppressor TRAF3 deleted from B cells, we identified Sox5 as a gene strikingly up-regulated in B lymphomas. Sox5 proteins were not detected in normal or premalignant TRAF3(-/-) B cells even after treatment with B cell stimuli. The Sox5 expressed in TRAF3(-/-) B lymphomas represents a novel isoform of Sox5, and was localized in the nucleus of malignant B cells. Overexpression of Sox5 inhibited cell cycle progression, and up-regulated the protein levels of p27 and ß-catenin in human multiple myeloma cells. Together, our findings indicate that Sox5 regulates the proliferation of malignant B cells.


Subject(s)
B-Lymphocytes/metabolism , Cell Nucleus/genetics , Gene Expression Regulation, Neoplastic , SOXD Transcription Factors/genetics , TNF Receptor-Associated Factor 3/genetics , Animals , Antibodies/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Cell Cycle/genetics , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Proliferation , Humans , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , SOXD Transcription Factors/metabolism , Signal Transduction , TNF Receptor-Associated Factor 3/deficiency , beta Catenin/genetics , beta Catenin/metabolism
5.
BMC Cancer ; 13: 481, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24131623

ABSTRACT

BACKGROUND: TRAF3, a new tumor suppressor identified in human non-Hodgkin lymphoma (NHL) and multiple myeloma (MM), induces PKCδ nuclear translocation in B cells. The present study aimed to evaluate the therapeutic potential of two PKCδ activators, N-Benzyladriamycin-14-valerate (AD 198) and ingenol-3-angelate (PEP005), on NHL and MM. METHODS: In vitro anti-tumor activities of AD 198 and PEP005 were determined using TRAF3-/- mouse B lymphoma and human patient-derived MM cell lines as model systems. In vivo therapeutic effects of AD 198 were assessed using NOD SCID mice transplanted with TRAF3-/- mouse B lymphoma cells. Biochemical studies were performed to investigate signaling mechanisms induced by AD 198 or PEP005, including subcellular translocation of PKCδ. RESULTS: We found that AD 198 exhibited potent in vitro and in vivo anti-tumor activity on TRAF3-/- tumor B cells, while PEP005 displayed contradictory anti- or pro-tumor activities on different cell lines. Detailed mechanistic investigation revealed that AD 198 did not affect PKCδ nuclear translocation, but strikingly suppressed c-Myc expression and inhibited the phosphorylation of ERK, p38 and JNK in TRAF3-/- tumor B cells. In contrast, PEP005 activated multiple signaling pathways in these cells, including PKCδ, PKCα, PKCε, NF-κB1, ERK, JNK, and Akt. Additionally, AD198 also potently inhibited the proliferation/survival and suppressed c-Myc expression in TRAF3-sufficient mouse and human B lymphoma cell lines. Furthermore, we found that reconstitution of c-Myc expression conferred partial resistance to the anti-proliferative/apoptosis-inducing effects of AD198 in human MM cells. CONCLUSIONS: AD 198 and PEP005 have differential effects on malignant B cells through distinct biochemical mechanisms. Our findings uncovered a novel, PKCδ-independent mechanism of the anti-tumor effects of AD 198, and suggest that AD 198 has therapeutic potential for the treatment of NHL and MM involving TRAF3 inactivation or c-Myc up-regulation.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/analogs & derivatives , Lymphoma, B-Cell/genetics , Multiple Myeloma/genetics , TNF Receptor-Associated Factor 3/deficiency , Animals , Antibiotics, Antineoplastic/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Diterpenes/administration & dosage , Diterpenes/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Genetic Vectors/genetics , Humans , Isografts , Lentivirus/genetics , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/mortality , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Protein Kinase C-delta/metabolism , Protein Transport , Proteolysis/drug effects , Proto-Oncogene Proteins c-myc/genetics , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...