Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(3)2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35159039

ABSTRACT

Multiple myeloma (MM) remains incurable for most patients due to the emergence of drug resistant clones. Here we report a p53-independent mechanism responsible for Growth Factor Independence-1 (GFI1) support of MM cell survival by its modulation of sphingolipid metabolism to increase the sphingosine-1-phosphate (S1P) level regardless of the p53 status. We found that expression of enzymes that control S1P biosynthesis, SphK1, dephosphorylation, and SGPP1 were differentially correlated with GFI1 levels in MM cells. We detected GFI1 occupancy on the SGGP1 gene in MM cells in a predicted enhancer region at the 5' end of intron 1, which correlated with decreased SGGP1 expression and increased S1P levels in GFI1 overexpressing cells, regardless of their p53 status. The high S1P:Ceramide intracellular ratio in MM cells protected c-Myc protein stability in a PP2A-dependent manner. The decreased MM viability by SphK1 inhibition was dependent on the induction of autophagy in both p53WT and p53mut MM. An autophagic blockade prevented GFI1 support for viability only in p53mut MM, demonstrating that GFI1 increases MM cell survival via both p53WT inhibition and upregulation of S1P independently. Therefore, GFI1 may be a key therapeutic target for all types of MM that may significantly benefit patients that are highly resistant to current therapies.

2.
J Bone Miner Res ; 35(1): 181-195, 2020 01.
Article in English | MEDLINE | ID: mdl-31487061

ABSTRACT

Key osteoclast (OCL) regulatory gene promoters in bone marrow-derived monocytes harbor bivalent histone modifications that combine activating Histone 3 lysine 4 tri-methyl (H3K4me3) and repressive H3K27me3 marks, which upon RANKL stimulation resolve into repressive or activating architecture. Enhancer of zeste homologue 2 (EZH2) is the histone methyltransferase component of the polycomb repressive complex 2, which catalyzes H3K27me3 modifications. Immunofluorescence microscopy reveals that EZH2 localization during murine osteoclastogenesis is dynamically regulated. Using EZH2 knockdown and small molecule EZH2 inhibitor GSK126, we show that EZH2 plays a critical epigenetic role in OCL precursors (OCLp) during the first 24 hours of RANKL activation. RANKL triggers EZH2 translocation into the nucleus where it represses OCL-negative regulators MafB, Irf8, and Arg1. Consistent with its cytoplasmic localization in OCLp, EZH2 methyltransferase activity is required during early RANKL signaling for phosphorylation of AKT, resulting in downstream activation of the mTOR complex, which is essential for induction of OCL differentiation. Inhibition of RANKL-induced pmTOR-pS6RP signaling by GSK126 altered the translation ratio of the C/EBPß-LAP and C/EBPß-LIP isoforms and reduced nuclear translocation of the inhibitory C/EBPß-LIP, which is necessary for transcriptional repression of the OCL negative-regulatory transcription factor MafB. EZH2 in multinucleated OCL is primarily cytoplasmic and mature OCL cultured on bone segments in the presence of GSK126 exhibit defective cytoskeletal architecture and reduced resorptive activity. Here we present new evidence that EZH2 plays epigenetic and cytoplasmic roles during OCL differentiation by suppressing MafB transcription and regulating early phases of PI3K-AKT-mTOR-mediated RANKL signaling, respectively. Consistent with its cytoplasmic localization, EZH2 is required for cytoskeletal dynamics during resorption by mature OCL. Thus, EZH2 exhibits complex roles in supporting osteoclast differentiation and function. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption , Osteoclasts , Animals , Bone Resorption/genetics , Cell Differentiation , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Mice , Phosphatidylinositol 3-Kinases
3.
J Hematol Oncol ; 11(1): 123, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30286780

ABSTRACT

BACKGROUND: In spite of major advances in treatment, multiple myeloma (MM) is currently an incurable malignancy due to the emergence of drug-resistant clones. We previously showed that MM cells upregulate the transcriptional repressor, growth factor independence 1 (Gfi1), in bone marrow stromal cells (BMSCs) that induces prolonged inhibition of osteoblast differentiation. However, the role of Gfi1 in MM cells is unknown. METHODS: Human primary CD138+ and BMSC were purified from normal donors and MM patients' bone marrow aspirates. Gfi1 knockdown and overexpressing cells were generated by lentiviral-mediated shRNA. Proliferation/apoptosis studies were done by flow cytometry, and protein levels were determined by Western blot and/or immunohistochemistry. An experimental MM mouse model was generated to investigate the effects of MM cells overexpressing Gfi1 on tumor burden and osteolysis in vivo. RESULTS: We found that Gfi1 expression is increased in patient's MM cells and MM cell lines and was further increased by co-culture with BMSC, IL-6, and sphingosine-1-phosphate. Modulation of Gfi1 in MM cells had major effects on their survival and growth. Knockdown of Gfi1 induced apoptosis in p53-wt, p53-mutant, and p53-deficient MM cells, while Gfi1 overexpression enhanced MM cell growth and protected MM cells from bortezomib-induced cell death. Gfi1 enhanced cell survival of p53-wt MM cells by binding to p53, thereby blocking binding to the promoters of the pro-apoptotic BAX and NOXA genes. Further, Gfi1-p53 binding could be blocked by HDAC inhibitors. Importantly, inoculation of MM cells overexpressing Gfi1 in mice induced increased bone destruction, increased osteoclast number and size, and enhanced tumor growth. CONCLUSIONS: These results support that Gfi1 plays a key role in MM tumor growth, survival, and bone destruction and contributes to bortezomib resistance, suggesting that Gfi1 may be a novel therapeutic target for MM.


Subject(s)
DNA-Binding Proteins/biosynthesis , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Osteogenesis/physiology , Transcription Factors/biosynthesis , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Female , Humans , Mice
4.
Article in English | MEDLINE | ID: mdl-30008697

ABSTRACT

Multiple myeloma bone disease (MMBD) is characterized by non-healing lytic bone lesions that persist even after a patient has achieved a hematologic remission. We previously reported that p62 (sequestosome-1) in bone marrow stromal cells (BMSC) is critical for the formation of MM-induced signaling complexes that mediate OB suppression. Importantly, XRK3F2, an inhibitor of the p62-ZZ domain, blunted MM-induced Runx2 suppression in vitro, and induced new bone formation and remodeling in the presence of tumor in vivo. Additionally, we reported that MM cells induce the formation of repressive chromatin on the Runx2 gene in BMSC via direct binding of the transcriptional repressor GFI1, which recruits the histone modifiers, histone deacetylase 1 (HDAC1) and Enhancer of zeste homolog 2 (EZH2). In this study we investigated the mechanism by which blocking p62-ZZ domain-dependent signaling prevents MM-induced suppression of Runx2 in BMSC. XRK3F2 prevented MM-induced upregulation of Gfi1 and repression of the Runx2 gene when present in MM-preOB co-cultures. We also show that p62-ZZ-domain blocking by XRK3F2 also prevented MM conditioned media and TNF plus IL7-mediated Gfi1 mRNA upregulation and the concomitant Runx2 repression, indicating that XRK3F2's prevention of p62-ZZ domain signaling within preOB is involved in the response. Chromatin immunoprecipitation (ChIP) analyses revealed that XRK3F2 decreased MM-induced GFI1 occupancy at the Runx2-P1 promoter and prevented recruitment of HDAC1, thus preserving the transcriptionally permissive chromatin mark H3K9ac on Runx2 and allowing osteogenic differentiation. Furthermore, treatment of MM-exposed preOB with XRK3F2 after MM removal decreased GFI1 enrichment at Runx2-P1 and rescued MM-induced suppression of Runx2 mRNA and its downstream osteogenic gene targets together with increased osteogenic differentiation. Further, primary BMSC (hBMSC) from MM patients (MM-hBMSC) had little ability to increase H3K9ac on the Runx2 promoter in osteogenic conditions when compared to hBMSC from healthy donors (HD). XRK3F2 treatment enriched Runx2 gene H3K9ac levels in MM-hBMSC to the level observed in HD-hBMSC, but did not alter HD-hBMSC H3K9ac. Importantly, XRK3F2 treatment of long-term MM-hBMSC cultures rescued osteogenic differentiation and mineralization. Our data show that blocking p62-ZZ domain-dependent signaling with XRK3F2 can reverse epigenetic-based mechanisms of MM-induced Runx2 suppression and promote osteogenic differentiation.

5.
ACS Appl Mater Interfaces ; 9(39): 33624-33631, 2017 Oct 04.
Article in English | MEDLINE | ID: mdl-28915002

ABSTRACT

The use of cancer-relevant microRNA molecules as endogenous drug release stimuli is promising for personalized cancer treatment yet remains a great challenge because of their low abundance. Herein, we report a new type of microRNA-catalyzed drug release system based on DNA-programmed gold nanoparticle (GNP)-quantum dot (QD) complex. We show that a trace amount of miRNA-21 molecules could specifically catalyze the disassembly of doxorubicin (Dox)-loaded GNP-QDs complex through entropy driven process, during which the Dox-intercalating sites are destructed for drug release. This catalytic reaction could proceed both in fixed cells and live cells with miRNA-21 overexpression. Dox molecules could be efficiently released in the cells and translocate to cell nuclei. QD photoluminescence is simultaneously activated during catalytic disassembly process, thus providing a reliable feedback for microRNA-triggered drug release. The GNP-QDs-Dox complex exhibits much higher drug potency than free Dox molecules, and therefore represents a promising platform for accurate and effective cancer cell treatment.


Subject(s)
Nanoparticles , Biocatalysis , Cell Line, Tumor , DNA , Doxorubicin , Gold , Humans , MicroRNAs , Neoplasms
6.
Mol Cancer Res ; 15(4): 405-417, 2017 04.
Article in English | MEDLINE | ID: mdl-28119431

ABSTRACT

In multiple myeloma, osteolytic lesions rarely heal because of persistent suppressed osteoblast differentiation resulting in a high fracture risk. Herein, chromatin immunoprecipitation analyses reveal that multiple myeloma cells induce repressive epigenetic histone changes at the Runx2 locus that prevent osteoblast differentiation. The most pronounced multiple myeloma-induced changes were at the Runx2-P1 promoter, converting it from a poised bivalent state to a repressed state. Previously, it was observed that multiple myeloma induces the transcription repressor GFI1 in osteoblast precursors, which correlates with decreased Runx2 expression, thus prompting detailed characterization of the multiple myeloma and TNFα-dependent GFI1 response element within the Runx2-P1 promoter. Further analyses reveal that multiple myeloma-induced GFI1 binding to Runx2 in osteoblast precursors and recruitment of the histone modifiers HDAC1, LSD1, and EZH2 is required to establish and maintain Runx2 repression in osteogenic conditions. These GFI1-mediated repressive chromatin changes persist even after removal of multiple myeloma. Ectopic GFI1 is sufficient to bind to Runx2, recruit HDAC1 and EZH2, increase H3K27me3 on the gene, and prevent osteogenic induction of endogenous Runx2 expression. Gfi1 knockdown in MC4 cells blocked multiple myeloma-induced recruitment of HDAC1 and EZH2 to Runx2, acquisition of repressive chromatin architecture, and suppression of osteoblast differentiation. Importantly, inhibition of EZH2 or HDAC1 activity in pre-osteoblasts after multiple myeloma exposure in vitro or in osteoblast precursors from patients with multiple myeloma reversed the repressive chromatin architecture at Runx2 and rescued osteoblast differentiation.Implications: This study suggests that therapeutically targeting EZH2 or HDAC1 activity may reverse the profound multiple myeloma-induced osteoblast suppression and allow repair of the lytic lesions. Mol Cancer Res; 15(4); 405-17. ©2017 AACR.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Histone Deacetylase 1/metabolism , Multiple Myeloma/genetics , Osteoblasts/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Coculture Techniques , Core Binding Factor Alpha 1 Subunit/metabolism , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Epigenesis, Genetic , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , Indoles/administration & dosage , Indoles/pharmacology , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Osteoblasts/drug effects , Promoter Regions, Genetic , Pyridones/administration & dosage , Pyridones/pharmacology , Transcription Factors/genetics
7.
J Bone Miner Res ; 29(1): 90-102, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23794264

ABSTRACT

Paget's disease of bone (PDB) is characterized by abnormal osteoclasts with unique characteristics that include increased sensitivity of osteoclast progenitors to 1,25(OH)2 D3 , receptor activator of NF-κB ligand (RANKL), and TNF-α; increased osteoclast numbers; and increased expression of IL-6 and several transcription factors. We recently reported that measles virus nucleocapsid protein (MVNP) plays a key role in the development of these abnormal osteoclasts. MVNP can induce the pagetic osteoclast phenotype in vitro and in vivo in TRAP-MVNP transgenic mice. However, the molecular mechanisms by which MVNP generates pagetic osteoclasts have not been determined. TANK-binding kinase 1 (TBK1) and IκB kinase-ϵ (IKKϵ) are IKK family members that complex with MVNP and activate both IRF3 and NF-κB pathways. MVNP increases the amount of TBK1 protein in bone marrow monocytes (BMM). Interestingly, we found that RANKL increased TBK1 and IKKϵ early in osteoclast differentiation, suggesting a possible role in normal osteoclastogenesis. However, only TBK1 is further increased in osteoclasts formed by TRAP-MVNP BMM owing to increased TBK1 protein stability. TBK1 overexpression induced IL6 promoter reporter activity, and elevated endogenous IL6 mRNA and p65 NF-κB, TAF12, and ATF7 proteins in several cell lines. Overexpression of TBK1 was insufficient to induce pagetic osteoclasts from WT BMM but synergized with MVNP to increase pagetic osteoclast formation from TRAP-MVNP BMM. BX795 inhibition of TBK1 impaired MVNP-induced IL-6 expression in both NIH3T3 cells and BMM, and shRNA knockdown of Tbk1 in NIH3T3 cells impaired IL-6 secretion induced by MVNP and decreased TAF12 and ATF7, factors involved in 1,25(OH)2 D3 hypersensitivity of pagetic osteoclasts. Similarly, Tbk1 knockdown in BMM from TRAP-MVNP and WT mice specifically impaired development of the MVNP-induced osteoclast pagetic phenotype. These results demonstrate that TBK1 plays a critical role in mediating the effects of MVNP on osteoclast differentiation and on the expression of IL-6, a key contributor to the pagetic osteoclast phenotype.


Subject(s)
I-kappa B Kinase/metabolism , Nucleocapsid Proteins/metabolism , Protein Serine-Threonine Kinases/physiology , Acid Phosphatase , Animals , HEK293 Cells , Humans , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Isoenzymes , Mice , NIH 3T3 Cells , Osteitis Deformans/genetics , Osteitis Deformans/prevention & control , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , RANK Ligand/pharmacology , Tartrate-Resistant Acid Phosphatase , Tumor Necrosis Factor-alpha/pharmacology
8.
Bone ; 53(1): 269-76, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23262029

ABSTRACT

Measles virus plays an important role as an environmental factor in the pathogenesis of Paget's disease (PD). Previous studies have shown that IL-6 is increased in the bone marrow of Paget's patients and that measles virus nucleocapsid protein (MVNP) induces IL-6 secretion by pagetic osteoclasts. Further, IL-6 plays a critical role in the development of pagetic osteoclasts and bone lesions induced by PD, but the mechanisms regulating IL-6 production by MVNP remain unclear. Our current studies revealed that MVNP expression in osteoclast precursors down-regulated Sirt1 mRNA and protein, a negative regulator of NF-κB activity, which is a key factor for IL-6 expression. MVNP expression in NIH3T3 cells also elevated Il-6 transcription and impaired the expression of Sirt1 mRNA both under basal conditions and upon activation of the Sirt1 upstream regulator FoxO3 by LY294002 (a PI3K/AKT inhibitor). Luciferase activity assays showed that constitutively active FoxO3 abolished the repressive effect of MVNP on reporters driven by either FoxO3 response elements or the Sirt1 promoter. Further, protein stability assays revealed that FoxO3 was degraded more rapidly in MVNP-expressing cells than in control cells following the addition of cycloheximide. Similarly, co-transfection of MVNP and FoxO3 into HEK293 cells demonstrated that MVNP decreased the protein levels of over-expressed FoxO3 in a dose-dependent manner. Treatment with the proteasome inhibitor, MG132, blocked the MVNP-triggered decrease of FoxO3, and the treatment with the serine/threonine phosphatase inhibitor, calyculin A, revealed that MVNP increased phosphorylation of FoxO3. Further, over-expression of Sirt1 or treatment with the Sirt1 activator resveratrol blocked the increase in Il-6 transcription by MVNP. Finally, resveratrol reduced the numbers of TRAP positive multi-nuclear cells in bone marrow cultures from TRAP-MVNP transgenic mice to wild type levels. These results indicate that MVNP decreases FoxO3/Sirt1 signaling to enhance the levels of IL-6, which in part mediate MVNP's contribution to the development of Paget's disease.


Subject(s)
Down-Regulation , Forkhead Transcription Factors/metabolism , Interleukin-6/metabolism , Measles virus/physiology , Nucleocapsid Proteins/physiology , Osteitis Deformans/metabolism , Signal Transduction , Sirtuin 1/metabolism , Animals , Base Sequence , Blotting, Western , DNA Primers , Forkhead Box Protein O3 , Forkhead Transcription Factors/physiology , Mice , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1/genetics , Transcription, Genetic/physiology
9.
PLoS One ; 7(8): e43158, 2012.
Article in English | MEDLINE | ID: mdl-22912816

ABSTRACT

Constitutive activation of pro-survival kinases has become a promising target of small molecules with an increasing interest in developing multi-targeted agents. The mechanisms underlying the responsiveness to most agents targeting cancer specific survival pathways are still poorly understood but critical for their clinical application. In this study, we found that sunitinib, a small molecule inhibitor of multiple tyrosine kinases including VEGFRs and PDGFRs induces apoptosis and inhibits cell growth in colon cancer cells in cell culture and xenograft models via the BH3-only protein PUMA. Sunitinib treatment induced PUMA transcription via the AKT/FoxO3a axis. PUMA, BH3 mimetics, or 5-Flurourical sensitized colon cancer cells to sunitinib-induced apoptosis. Furthermore, PUMA was induced by sunitinib treatment in xenograft tumors, and deficiency in PUMA significantly suppressed the anti-tumor effects of sunitinib. Our study suggests that PUMA-mediated apoptosis is important for the therapeutic responses to sunitinib, and activation of the mitochondrial pathway by BH3 mimetics or PUMA manipulation may be useful for improving the antitumor activity of sunitinib. Modulation of PUMA and selective Bcl-2 family members might be potential biomarkers for predicting sunitinib responses.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Indoles/pharmacology , Proto-Oncogene Proteins/metabolism , Pyrroles/pharmacology , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Colonic Neoplasms/metabolism , DNA Primers/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Histological Techniques , Humans , In Situ Nick-End Labeling , Luciferases , Oncogene Protein v-akt/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sunitinib
10.
Blood ; 118(26): 6871-80, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22042697

ABSTRACT

Protracted inhibition of osteoblast (OB) differentiation characterizes multiple myeloma (MM) bone disease and persists even when patients are in long-term remission. However, the underlying pathophysiology for this prolonged OB suppression is unknown. Therefore, we developed a mouse MM model in which the bone marrow stromal cells (BMSCs) remained unresponsive to OB differentiation signals after removal of MM cells. We found that BMSCs from both MM-bearing mice and MM patients had increased levels of the transcriptional repressor Gfi1 compared with controls and that Gfi1 was a novel transcriptional repressor of the critical OB transcription factor Runx2. Trichostatin-A blocked the effects of Gfi1, suggesting that it induces epigenetic changes in the Runx2 promoter. MM-BMSC cell-cell contact was not required for MM cells to increase Gfi1 and repress Runx2 levels in MC-4 before OBs or naive primary BMSCs, and Gfi1 induction was blocked by anti-TNF-α and anti-IL-7 antibodies. Importantly, BMSCs isolated from Gfi1(-/-) mice were significantly resistant to MM-induced OB suppression. Strikingly, siRNA knockdown of Gfi1 in BMSCs from MM patients significantly restored expression of Runx2 and OB differentiation markers. Thus, Gfi1 may have an important role in prolonged MM-induced OB suppression and provide a new therapeutic target for MM bone disease.


Subject(s)
Bone Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Multiple Myeloma/metabolism , Osteoblasts/metabolism , Stromal Cells/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Blotting, Western , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , DNA-Binding Proteins/genetics , Female , Gene Expression , Humans , Interleukin-7/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Osteoblasts/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/pathology , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
Cancer Res ; 71(10): 3625-34, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21444671

ABSTRACT

Autophagy is an evolutionarily conserved stress response mechanism that often occurs in apoptosis-defective cancer cells and can protect against cell death. In this study, we investigated how apoptosis and autophagy affect each other in cancer cells in response to chemotherapeutic treatment. We found that specific ablation of the proapoptotic function of cytochrome c, a key regulator of mitochondria-mediated apoptosis, enhanced autophagy following chemotherapeutic treatment. Induction of autophagy required Beclin 1 and was associated with blockage of Beclin 1 cleavage by caspase 8 at two sites. To investigate the role of Beclin 1 cleavage in the suppression of autophagy and cell survival, a caspase-resistant mutant of Beclin 1 was knocked into HCT116 colon cancer cells. Beclin 1 mutant knockin resulted in markedly increased autophagy and improved long-term cell survival after chemotherapeutic treatment but without affecting apoptosis and caspase activation. Furthermore, Beclin 1 mutant tumors were significantly less responsive to chemotherapeutic treatment than were wild-type tumors. These results show that chemotherapy-induced apoptosis inhibits autophagy at the execution stage subsequent to cytochrome c release through caspase 8-mediated cleavage of Beclin 1. If apoptosis fails to execute, autophagy is unleashed due to lack of Beclin 1 cleavage by caspases and can contribute to cancer cell survival and therapeutic resistance. Therefore, Beclin 1 may be a useful target for inhibiting autophagy to sensitize chemotherapy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy , Caspase 8/metabolism , Cytochromes c/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Mutation , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Beclin-1 , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/genetics
12.
Clin Cancer Res ; 17(8): 2361-72, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21242120

ABSTRACT

PURPOSE: Overexpression of inhibitors of apoptosis proteins (IAP) contributes to therapeutic resistance. Second mitochondria-derived activator of caspase (Smac) promotes caspase activation by binding to IAPs upon release from the mitochondria. IAP antagonists, also called SMAC mimetics, are promising anticancer agents modeled after this mechanism. We investigated the role and mechanisms of Smac- and Smac mimetic-mediated chemosensitization in head and neck squamous cell carcinoma (HNSCC) cells. EXPERIMENTAL DESIGN: The effects of SMAC knockdown, SMAC overexpression, and a small molecule Smac mimetic on the chemosensitivities of HNSCC cells were determined. The mechanisms of Smac- and Smac mimetic-mediated chemosensitization were investigated by analyzing growth suppression, the mitochondrial apoptotic pathway, caspase activation, and IAP proteins. The therapeutic responses of HNSCC cells with different levels of Smac were compared in xenograft models. RESULTS: We found that Smac mediates apoptosis induced by several classes of therapeutic agents through the mitochondrial pathway. SMAC knockdown led to impaired caspase activation, mitochondrial membrane depolarization, and release of cytochrome c. A small molecule Smac mimetic, at nanomolar concentrations, significantly sensitized HNSCC cells to gemcitabine-induced apoptosis and restored gemcitabine sensitivity in SMAC knockdown cells, through caspase activation, X-linked IAP dissociation, and mitochondria-associated events, but not the TNF-α pathway. Furthermore, Smac levels modulated the therapeutic response of HNSCC cells to gemcitabine in xenograft models. CONCLUSIONS: Our results establish a critical role of Smac in mediating therapeutic responses of HNSCC cells and provide a strong rationale for combining Smac mimetics with other anticancer agents to treat HNSCC.


Subject(s)
Apoptosis/drug effects , Deoxycytidine/analogs & derivatives , Head and Neck Neoplasms/drug therapy , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis Regulatory Proteins , Biomimetic Materials/pharmacology , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Deoxycytidine/pharmacology , Drug Synergism , Enzyme Activation/drug effects , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/genetics , RNA Interference , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
13.
Mol Cancer Ther ; 9(11): 2893-902, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20978166

ABSTRACT

Most targeted anticancer drugs are inhibitors of kinases that are aberrantly activated in cancer cells. However, the mechanisms by which kinase inhibitors suppress tumor growth remain unclear. In this study, we found that UCN-01, a staurosporine analogue and broad-range kinase inhibitor used in clinical trials, inhibits colon cancer cell growth by inducing apoptosis via PUMA, a BH3-only Bcl-2 family member and a p53 target. PUMA expression was markedly elevated in a p53-independent fashion following UCN-01 treatment. The induction of PUMA by UCN-01 was mediated by direct binding of FoxO3a to the PUMA promoter following inhibition of AKT signaling. Deficiency in PUMA abrogated UCN-01-induced apoptosis, caspase activation, and mitochondrial dysfunction, and rendered UCN-01 resistance in a clonogenic assay, whereas elevated PUMA expression or a BH3 mimetic sensitized UCN-01 induced apoptosis. Chemosensitization by UCN-01 seemed to involve simultaneous PUMA induction through both p53-dependent and p53-independent mechanisms. Furthermore, deficiency in PUMA suppressed the antitumor effects of UCN-01 in a xenograft model, concurrent with reduced apoptosis and caspase activation in vivo. These results suggest that PUMA-mediated apoptosis is pivotal for the anticancer activities of UCN-01, and possibly other clinically used kinase inhibitor drugs, and that PUMA manipulation may be useful for improving their anticancer activities.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Drug Resistance, Neoplasm/genetics , Forkhead Transcription Factors/physiology , Proto-Oncogene Proteins/genetics , Staurosporine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/physiology , Drug Resistance, Neoplasm/drug effects , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Nude , Models, Biological , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Staurosporine/pharmacology , Tumor Cells, Cultured , Up-Regulation/drug effects , Up-Regulation/physiology , Xenograft Model Antitumor Assays
14.
Cancer Res ; 69(15): 6339-46, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19584278

ABSTRACT

The high mortality rate of lung cancer is largely due to the spread of disease to other organs. However, the molecular changes driving lung cancer invasion and metastasis remain unclear. In this study, we identified fibulin-5, a vascular ligand for integrin receptors, as a suppressor of lung cancer invasion and metastasis. Fibulin-5 was silenced by promoter hypermethylation in a majority of lung cancer cell lines and primary tumors. It inhibited lung cancer cell invasion and down-regulated matrix metalloproteinase-7 (MMP-7), which promoted lung cancer cell invasion. Knockdown of fibulin-5 was sufficient to stimulate cell invasion and MMP-7 expression. The expression levels of fibulin-5 and MMP-7 were inversely correlated in lung tumors. Suppression of MMP-7 expression by fibulin-5 was mediated by an integrin-binding RGD motif via the extracellular signal-regulated kinase (ERK) pathway. Furthermore, overexpression of fibulin-5 in H460 lung cancer cells inhibited metastasis in mice. Collectively, these results suggest that epigenetic silencing of fibulin-5 promotes lung cancer invasion and metastasis by activating MMP-7 expression through the ERK pathway.


Subject(s)
Extracellular Matrix Proteins/metabolism , Lung Neoplasms/metabolism , Matrix Metalloproteinase 7/biosynthesis , Matrix Metalloproteinase Inhibitors , Animals , Cell Line, Tumor , DNA Methylation , Down-Regulation , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Silencing , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Matrix Metalloproteinase 7/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Oligopeptides/metabolism , Promoter Regions, Genetic , Transfection , Xenograft Model Antitumor Assays
15.
Hum Reprod ; 23(7): 1581-93, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18296448

ABSTRACT

BACKGROUND: In order to study the novel genes related to rat embryonic implantation, a novel implantation-associated gene, Iag-1, was identified and characterized from rat uterus of early pregnancy. Iag-1 was initially derived from suppressive subtracted hybridization of a cDNA library of rat uterus, which was used to analyse differentially expressed genes between the preimplantation and implantation period. METHODS: The full-length cDNA sequence of Iag-1 was cloned from rat uterus on D5.5 of pregnancy by 5'- and 3'-RACE. The expression of Iag-1 in the uterus of early pregnancy, pseudopregnancy, artificial decidualization and activation of delayed implantation was detected by northern blotting, in situ hybridization, western blotting and immunofluorescence. Endometrial stromal cells (ESCs) were isolated from rat uterus. The effect of Iag-1 on ESCs proliferation and apoptosis were determined by MTT assay, TUNEL and Hoechst staining. Apoptosis-related proteins in ESCs were detected by western blotting. RESULTS: Differential patterns of Iag-1 expression were detected in rat embryo and in the uterus during the peri-implantation period. Iag-1 was specifically localized in glandular epithelium and luminal epithelium. In contrast, the expression of Iag-1 was not significantly altered in uterus of pseudopregnancy and artificial decidualization, but was significantly increased in the uterus after activation of delayed implantation. Stable expression of introduced Iag-1 inhibited the proliferation of in vitro-cultured ESCs. Significant apoptosis was also detected in the ESCs overexpressing Iag-1, along with the enhancement of p53 and Bax protein expression. CONCLUSIONS: Overexpression of Iag-1 can inhibit ESCs proliferation and induce ESCs apoptosis, and p53 and Bax may play an important role in the process of Iag-1-induced apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Embryo Implantation/physiology , Uterus/physiology , Amino Acid Sequence , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Base Sequence , Cell Proliferation , Cloning, Molecular , Decidua/physiology , Embryo Implantation, Delayed/physiology , Female , In Situ Nick-End Labeling , Molecular Sequence Data , Pregnancy , Pseudopregnancy/physiopathology , RNA, Messenger/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Stromal Cells/cytology , Stromal Cells/physiology , Tissue Distribution , Tumor Suppressor Protein p53/physiology , Uterus/cytology , bcl-2-Associated X Protein/physiology
16.
Carcinogenesis ; 29(1): 84-92, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18048388

ABSTRACT

Although the oncogenic role of the Wnt/beta-catenin pathway is well defined, it remains unclear how this pathway is aberrantly activated in lung cancer. We found that Dickkopf (Dkk)-3, a member of Dkk family of Wnt antagonists, is frequently inactivated in lung cancer and plays a role in suppressing lung cancer cell growth through inhibition of beta-catenin/T-cell factor (TCF)-4 signaling. Dkk3 is the only Dkk family member abundantly expressed in normal lung, but silenced by promoter hypermethylation in a large fraction of lung cancer cell lines and lung tumors. Downregulation of Dkk3 was correlated with tumor progression and expression of nuclear beta-catenin in lung tumors. Ectopic expression of Dkk3 in lung cancer cells with Dkk3 hypermethylation induced apoptosis and inhibited TCF-4 activity as well as nuclear accumulation of beta-catenin and expression of TCF-4 targets c-Myc and cyclin D1. Furthermore, small interference RNA knock down of Dkk3 in cells lacking Dkk3 hypermethylation was sufficient to promote cell proliferation, beta-catenin nuclear translocation and expression of c-Myc. These observations suggested that epigenetic inactivation of Dkk3 activates the Wnt/beta-catenin pathway, thereby promoting the growth of lung cancer cells.


Subject(s)
Down-Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Signal Transduction , TCF Transcription Factors/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Chemokines , Humans , Transcription Factor 7-Like 2 Protein
17.
Mol Cancer Ther ; 6(12 Pt 1): 3180-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18089712

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) ranks the eighth most common cancer worldwide. The patients often present with advanced disease, which responds poorly to chemoradiation therapy. PUMA is a BH3-only Bcl-2 family protein and a p53 target that is required for apoptosis induced by p53 and various chemotherapeutic agents. In this study, we found that PUMA induction by chemotherapeutic agents is abrogated in most HNSCC cell lines. Adenoviral gene delivery of PUMA induced apoptosis and chemosensitization more potently than did adenoviral delivery of p53 in HNSCC cells. Finally, we showed that PUMA suppressed the growth of HNSCC xenograft tumors and sensitized them to cisplatin through induction of apoptosis. Our data suggest that absence of PUMA activation in HNSCC cells contributes to chemoresistance and that gene therapy with PUMA might be an efficient substitute for p53 to enhance the responses of HNSCC cells to chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/physiology , Carcinoma, Squamous Cell/drug therapy , Cisplatin/therapeutic use , Head and Neck Neoplasms/drug therapy , Proto-Oncogene Proteins/physiology , Animals , Apoptosis/physiology , Cell Division/physiology , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Transplantation, Heterologous
18.
Clin Cancer Res ; 13(15 Pt 1): 4336-44, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671114

ABSTRACT

PURPOSE: The goal of this study is to identify novel genes frequently silenced by promoter hypermethylation in lung cancer. EXPERIMENTAL DESIGNS: Bioinformatic analysis was done to identify candidate genes significantly down-regulated in lung cancer. The effects of DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine on the expression of the candidate genes were determined. Methylated CpG sites in the promoters of the candidate genes were identified using bisulfite DNA sequencing. Methylation-specific PCR was developed and used to analyze DNA methylation in cell lines and clinical specimen. Pathologic and functional analyses were done to study the role of one candidate gene, receptor activity-modifying protein 2 (RAMP2), in suppressing lung cancer cell growth. RESULTS: Among 54 candidate genes down-regulated in lung cancer, 31 were found to contain CpG islands in their promoters. Six of these 31 genes could be reactivated by 5-aza-2'-deoxycytidine in at least four of six lung cancer cell lines analyzed. Promoter hypermethylation of RAMP2, epidermal growth factor-containing fibulin-like extracellular matrix protein 1, and deleted in U Twenty Twenty cells was detected in 36% to 77% of 22 lung cancer cell lines and in 38% to 50% of 32 primary lung tumors, whereas hypermethylathion of these genes was rarely found in the matched normal samples. The methylation frequencies of these genes in lung cancer were similar to those of commonly used methylation markers, such as RAS association domain family protein 1A, p16, and methylguanine-DNA methyltransferase. Immunohistochemistry showed that RAMP2 was down-regulated in a majority of lung tumors, and RAMP2 down-regulation was correlated with high tumor grade. Ectopic expression of RAMP2 inhibited lung cancer cell growth and caused apoptotic cell death. Knockdown of RAMP2 by RNA interference stimulated cell proliferation. CONCLUSIONS: Studying the newly identified genes may provide new insight into lung tumorigenesis. These genes might be useful as molecular markers of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation , Extracellular Matrix Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, Immunologic/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , CpG Islands , DNA Mutational Analysis , DNA, Neoplasm/genetics , Down-Regulation , Epigenesis, Genetic , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Immunoenzyme Techniques , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/genetics , Receptor Activity-Modifying Protein 2 , Receptor Activity-Modifying Proteins , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Tumor Cells, Cultured , Roundabout Proteins
19.
J Interferon Cytokine Res ; 27(7): 567-78, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17651018

ABSTRACT

Growth and development of placentas in all pregnancy periods and that of fetuses in late pregnancy were inhibited after administration of interferon-gamma (IFN-gamma). Apoptosis can be detected by TUNEL at the maternal-fetal interface during normal rat pregnancy. Apoptosis locations at the maternal-fetal interface changed according to the period of pregnancy. The results of immunohistochemistry and the DNA ladder assay showed that IFN-gamma could promote the apoptosis levels during the entire pregnancy, but it did not change the apoptosis locations. IFN regulatory factor-1 (IRF-1), FasL, and p53 expressions were modulated by IFN-gamma during the entire pregnancy. In vitro cell proliferation assay indicated that IFN-gamma could inhibit proliferation of human cytotrophoblast cells, and flow assay showed that this effect was mainly due to apoptosis induction. TUNEL and Hoechst staining also showed that IFN-gamma could induce apoptosis of human cytotrophoblast cells. Expression of IRF-1 was induced and expression of active caspase-3 was promoted by IFN-gamma treatment, but IFN-gamma did not affect the expression of IFNGR and p53.


Subject(s)
Apoptosis , Fetus/metabolism , Interferon-gamma/pharmacology , Placenta/cytology , Trophoblasts/cytology , Uterus/cytology , Animals , Cell Proliferation , Female , Fetus/drug effects , Gene Expression Regulation , Humans , In Situ Nick-End Labeling , Interferon-gamma/metabolism , Placenta/drug effects , Placenta/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Trophoblasts/drug effects , Trophoblasts/metabolism , Uterus/drug effects , Uterus/metabolism
20.
Reprod Fertil Dev ; 19(3): 510-9, 2007.
Article in English | MEDLINE | ID: mdl-17394800

ABSTRACT

In the present study, the possible mechanisms by which interferon (IFN)-gamma affects pregnancy were investigated using the cytokine network model. The IFN-gamma-induced expression of interleukin (IL)-1beta was examined using western blotting, immunohistochemistry and immunofluorescence. The results showed that IFN-gamma treatment significantly decreased the expression of uterine IL-1beta protein during the preimplantation, post-implantation and mid-gestation periods. The expression of IL-1beta protein was increased after IFN-gamma treatment compared with the control group in late pregnancy. In the placenta, IL-1beta protein levels were significantly increased after IFN-gamma treatment in early and mid-pregnancy. In late pregnancy, IFN-gamma treatment significantly decreased placental IL-1beta protein levels. IL-1beta was mainly expressed in the myometrium, uterine arteries, decidua basalis, trophospongium of the junctional layer and trophoblastic epithelium of the labyrinthine layers. IL-1beta was mainly located in the cytoplasm of in vitro cultured endometrial stromal cells (ESCs). IFN-gamma treatment did not affect the distribution of IL-1beta, only the expression of IL-1beta. The effects of IFN-gamma on the proliferation of ESCs were determined using an MTS (a novel tetrazolium compound) assay. IFN-gamma treatment inhibited the proliferation of ESCs and decreased the weight of the fetus and placenta. These results indicate that exogenous IFN-gamma affects the expression of IL-1beta and inhibits ESC proliferation.


Subject(s)
Fetus/drug effects , Interferon-gamma/pharmacology , Interleukin-1beta/metabolism , Placenta/drug effects , Animals , Cell Proliferation/drug effects , Endometrium/cytology , Endometrium/drug effects , Female , Fetus/immunology , Interleukin-1beta/analysis , Placenta/chemistry , Placenta/immunology , Pregnancy , Rats , Rats, Sprague-Dawley , Stromal Cells/drug effects , Uterus/drug effects , Uterus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...