Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Lab Invest ; 100(5): 777-785, 2020 05.
Article in English | MEDLINE | ID: mdl-31896813

ABSTRACT

TRAIL-activating therapy is promising in treating various cancers, including pancreatic cancer, a highly malignant neoplasm with poor prognosis. However, many pancreatic cancer cells are resistant to TRAIL-induced apoptosis despite their expression of intact death receptors (DRs). Protein O-GlcNAcylation is a versatile posttranslational modification that regulates various biological processes. Elevated protein O-GlcNAcylation has been recently linked to cancer cell growth and survival. In this study, we evaluated the role of protein O-GlcNAcylation in pancreatic cancer TRAIL resistance, and identified higher levels of O-GlcNAcylation in TRAIL-resistant pancreatic cancer cells. With gain- and loss-of-function of the O-GlcNAc-adding enzyme, O-GlcNActransferase (OGT), we determined that increasing O-GlcNAcylation rendered TRAIL-sensitive cells more resistant to TRA-8-induced apoptosis, while inhibiting O-GlcNAcylation promoted TRA-8-induced apoptosis in TRAIL-resistance cells. Furthermore, we demonstrated that OGT knockdown sensitized TRAIL-resistant cells to TRA-8 therapy in a mouse model in vivo. Mechanistic studies revealed direct O-GlcNAc modifications of DR5, which regulated TRA-8-induced DR5 oligomerization. We further defined that DR5 O-GlcNAcylation was independent of FADD, the adapter protein for the downstream death-inducing signaling. These studies have demonstrated an important role of protein O-GlcNAcylation in regulating TRAIL resistance of pancreatic cancer cells; and uncovered the contribution of O-GlcNAcylation to DR5 oligomerization and thus mediating DR-inducing signaling.


Subject(s)
Drug Resistance, Neoplasm/genetics , N-Acetylglucosaminyltransferases , Pancreatic Neoplasms , TNF-Related Apoptosis-Inducing Ligand , Acetylglucosamine/metabolism , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Knockout , Mice, Nude , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
2.
J Biol Chem ; 292(25): 10390-10397, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28476883

ABSTRACT

Pancreatic cancer is a malignant neoplasm with a high mortality rate. Therapeutic agents that activate TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis have shown promising efficacy, but many pancreatic cancers are resistant to TRAIL therapy. Epigenetic regulation plays important roles in tumor pathogenesis and resistance, and a recent study indicated that the long non-coding RNA HOX transcript antisense RNA (HOTAIR) is overexpressed in pancreatic cancer. However, the role of HOTAIR in pancreatic cancer resistance to anticancer agents is unknown. The present study determined the role of HOTAIR in pancreatic cancer TRAIL resistance and investigated the underlying molecular mechanisms. We observed that TRAIL-resistant pancreatic cancer cells had higher levels of HOTAIR expression, whereas TRAIL-sensitive pancreatic cancer cells had lower HOTAIR levels. Overexpressing HOTAIR in TRAIL-sensitive cells attenuated TRAIL-induced apoptosis, and shRNA-mediated HOTAIR knockdown in TRAIL-resistant PANC-1 cells sensitized them to TRAIL-induced apoptosis. These results support a causative effect of HOTAIR on TRAIL sensitivity. Mechanistically, we found that increased HOTAIR expression inhibited the expression of the TRAIL receptor death receptor 5 (DR5), whereas HOTAIR knockdown increased DR5 expression. We further demonstrated that HOTAIR regulates DR5 expression via the epigenetic regulator enhancer of zeste homolog 2 (EZH2) and that EZH2 controls histone H3 lysine 27 trimethylation on the DR5 gene. Taken together, these results demonstrate that high HOTAIR levels increase the resistance of pancreatic cancer cells to TRAIL-induced apoptosis via epigenetic regulation of DR5 expression. Our study therefore supports the notion that targeting HOTAIR function may represent a strategy to overcome TRAIL resistance in pancreatic cancer.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm/drug effects , Pancreatic Neoplasms/metabolism , RNA, Long Noncoding/biosynthesis , RNA, Neoplasm/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histones/genetics , Histones/metabolism , Humans , Methylation/drug effects , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
3.
Oncotarget ; 6(28): 25308-19, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26320171

ABSTRACT

Pancreatic cancer is highly malignant with limited therapy and a poor prognosis. TRAIL-activating therapy has been promising, however, clinical trials have shown resistance and limited responses of pancreatic cancers. We investigated the effects of calmodulin(CaM) antagonists, trifluoperazine(TFP) and tamoxifen(TMX), on TRA-8-induced apoptosis and tumorigenesis of TRA-8-resistant pancreatic cancer cells, and underlying mechanisms. TFP or TMX alone did not induce apoptosis of resistant PANC-1 cells, while they dose-dependently enhanced TRA-8-induced apoptosis. TMX treatment enhanced efficacy of TRA-8 therapy on tumorigenesis in vivo. Analysis of TRA-8-induced death-inducing-signaling-complex (DISC) identified recruitment of survival signals, CaM/Src, into DR5-associated DISC, which was inhibited by TMX/TFP. In contrast, TMX/TFP increased TRA-8-induced DISC recruitment/activation of caspase-8. Consistently, caspase-8 inhibition blocked the effects of TFP/TMX on TRA-8-induced apoptosis. Moreover, TFP/TMX induced DR5 expression. With a series of deletion/point mutants, we identified CaM antagonist-responsive region in the putative Sp1-binding domain between -295 to -300 base pairs of DR5 gene. Altogether, we have demonstrated that CaM antagonists enhance TRA-8-induced apoptosis of TRA-8-resistant pancreatic cancer cells by increasing DR5 expression and enhancing recruitment of apoptotic signal while decreasing survival signals in DR5-associated DISC. Our studies support the use of these readily available CaM antagonists combined with TRAIL-activating agents for pancreatic cancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Calmodulin/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Pancreatic Neoplasms/drug therapy , Tamoxifen/pharmacology , Trifluoperazine/pharmacology , Animals , Apoptosis/drug effects , Binding Sites , Calmodulin/metabolism , Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Male , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Protein Binding , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , Sp1 Transcription Factor/metabolism , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
4.
Biochemistry ; 53(16): 2680-8, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24702583

ABSTRACT

The Fas death receptor-activated death-inducing signaling complex (DISC) regulates apoptosis in many normal and cancer cells. Qualitative biochemical experiments demonstrate that calmodulin (CaM) binds to the death domain of Fas. The interaction between CaM and Fas regulates Fas-mediated DISC formation. A quantitative understanding of the interaction between CaM and Fas is important for the optimal design of antagonists for CaM or Fas to regulate the CaM-Fas interaction, thus modulating Fas-mediated DISC formation and apoptosis. The V254N mutation of the Fas death domain (Fas DD) is analogous to an identified mutant allele of Fas in lpr-cg mice that have a deficiency in Fas-mediated apoptosis. In this study, the interactions of CaM with the Fas DD wild type (Fas DD WT) and with the Fas DD V254N mutant were characterized using isothermal titration calorimetry (ITC), circular dichroism spectroscopy (CD), and molecular dynamics (MD) simulations. ITC results reveal an endothermic binding characteristic and an entropy-driven interaction of CaM with Fas DD WT or with Fas DD V254N. The Fas DD V254N mutation decreased the association constant (Ka) for CaM-Fas DD binding from (1.79 ± 0.20) × 10(6) to (0.88 ± 0.14) × 10(6) M(-1) and slightly increased a standard state Gibbs free energy (ΔG°) for CaM-Fas DD binding from -8.87 ± 0.07 to -8.43 ± 0.10 kcal/mol. CD secondary structure analysis and MD simulation results did not show significant secondary structural changes of the Fas DD caused by the V254N mutation. The conformational and dynamical motion analyses, the analyses of hydrogen bond formation within the CaM binding region, the contact numbers of each residue, and the electrostatic potential for the CaM binding region based on MD simulations demonstrated changes caused by the Fas DD V254N mutation. These changes caused by the Fas DD V254N mutation could affect the van der Waals interactions and electrostatic interactions between CaM and Fas DD, thereby affecting CaM-Fas DD interactions. Results from this study characterize CaM-Fas DD interactions in a quantitative way, providing structural and thermodynamic evidence of the role of the Fas DD V254N mutation in the CaM-Fas DD interaction. Furthermore, the results could help to identify novel strategies for regulating CaM-Fas DD interactions and Fas DD conformation and thus to modulate Fas-mediated DISC formation and thus Fas-mediated apoptosis.


Subject(s)
Calmodulin/metabolism , Protein Interaction Domains and Motifs , fas Receptor/metabolism , Calmodulin/chemistry , Calorimetry/methods , Circular Dichroism , Molecular Dynamics Simulation , Mutation , Protein Structure, Secondary , Thermodynamics , fas Receptor/chemistry , fas Receptor/genetics
5.
Proteins ; 81(3): 377-85, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23042204

ABSTRACT

Fas binding to Fas-associated death domain (FADD) activates FADD-caspase-8 binding to form death-inducing signaling complex (DISC) that triggers apoptosis. The Fas-Fas association exists primarily as dimer in the Fas-FADD complex, and the Fas-FADD tetramer complexes have the tendency to form higher order oligomer. The importance of the oligomerized Fas-FADD complex in DISC formation has been confirmed. This study sought to provide structural insight for the roles of Fas death domain (Fas DD) binding to FADD and the oligomerization of Fas DD-FADD complex in activating FADD-procaspase-8 binding. Results show Fas DD binding to FADD stabilized the FADD conformation, including the increased stability of the critical residues in FADD death effector domain (FADD DED) for FADD-procaspase-8 binding. Fas DD binding to FADD resulted in the decreased degree of both correlated and anticorrelated motion of the residues in FADD and caused the reversed correlated motion between FADD DED and FADD death domain (FADD DD). The exposure of procaspase-8 binding residues in FADD that allows FADD to interact with procaspase-8 was observed with Fas DD binding to FADD. We also observed different degrees of conformational and motion changes of FADD in the Fas DD-FADD complex with different degrees of oligomerization. The increased conformational stability and the decreased degree of correlated motion of the residues in FADD in Fas DD-FADD tetramer complex were observed compared to those in Fas DD-FADD dimer complex. This study provides structural evidence for the roles of Fas DD binding to FADD and the oligomerization degree of Fas DD-FADD complex in DISC formation to signal apoptosis.


Subject(s)
Computational Biology/methods , Fas-Associated Death Domain Protein/chemistry , Multiprotein Complexes/chemistry , Protein Multimerization , Signal Transduction , fas Receptor/chemistry , Binding Sites , Caspase 8/chemistry , Molecular Dynamics Simulation , Principal Component Analysis , Protein Binding , Protein Interaction Mapping/methods , Protein Stability , Protein Structure, Secondary , Static Electricity
6.
Lab Invest ; 92(1): 82-90, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21912376

ABSTRACT

We have previously demonstrated that calmodulin (CaM) binds directly to c-FLIP(L) in a Ca(2+)-dependent manner. Deletion of the CaM-binding region (amino acid 197-213) results in reduced CaM binding, and increased Fas-mediated apoptosis and decreased tumorigenesis of cholangiocarcinoma cells. The present studies were designed to identify the precise amino acids between 197 and 213 that are responsible for CaM/FLIP binding, and their roles in mediating the anti-apoptotic function of c-FLIP(L). Sequence analysis of the CaM-binding region at 197-213 predicted three unique positively charged residues at 204, 207 and 209, which might be responsible for the CaM/FLIP binding. A point mutation at H204 of c-FLIP(L) was found to markedly reduce CaM binding, whereas point mutation at R207 or K209 did not affect c-FLIP(L) binding to CaM. Decreased CaM/FLIP binding was confirmed in cholangiocarcinoma cells overexpressing the H204 c-FLIP(L) mutant. Reduced CaM binding by the H204 mutant resulted in increased sensitivity to Fas-mediated apoptosis and inhibited tumor growth in mice compared with wild-type c-FLIP(L). Death-inducing signaling complex (DISC) analysis showed that the reduced CaM binding to H204 mutant resulted in less c-FLIP(L) recruited into the DISC. Concurrently, increased caspase 8 was recruited to the DISC, which resulted in increased cleavage and activation of caspase 8, activation of downstream caspase 3 and increased apoptosis. Therefore, these results demonstrate that the H204 residue is responsible for c-FLIP(L) binding to CaM, which mediates the anti-apoptotic function of c-FLIP(L), most likely through affecting recruitment of caspase 8 into the DISC and thus caspase 8 activation. These studies further characterized CaM/FLIP interaction and its function in regulating Fas-mediated apoptosis and tumorigenesis, which may provide new therapeutic targets for cancer therapy.


Subject(s)
Apoptosis , Bile Duct Neoplasms/prevention & control , Bile Ducts, Intrahepatic , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Calmodulin/metabolism , Cholangiocarcinoma/prevention & control , fas Receptor/physiology , Animals , Caspases/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Humans , Male , Mice , Point Mutation
7.
Proteins ; 79(8): 2543-56, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21656570

ABSTRACT

Death-inducing signaling complex (DISC) formation is a critical step in Fas-mediated signaling for apoptosis. Previous experiments have demonstrated that the calmodulin (CaM) antagonist, trifluoperazine (TFP) regulates CaM-Fas binding and affects Fas-mediated DISC formation. In this study, we investigated the anti-cooperative characteristics of TFP binding to CaM and the effect of TFP on the CaM-Fas interaction from both structural and thermodynamic perspectives using combined molecular dynamics simulations and binding free energy analyses. We studied the interactions of different numbers of TFP molecules with CaM and explored the effects of the resulting conformational changes in CaM on CaM-Fas binding. Results from these analyses showed that the number of TFP molecules bound to CaM directly influenced α-helix formation and hydrogen bond occupancy within the α-helices of CaM, contributing to the conformational and motion changes in CaM. These changes affected CaM binding to Fas, resulting in secondary structural changes in Fas and conformational and motion changes of Fas in CaM-Fas complexes, potentially perturbing the recruitment of Fas-associated death domain for DISC formation. The computational results from this study reveal the structural and molecular mechanisms that underlie the role of the CaM antagonist, TFP, in regulation of CaM-Fas binding and Fas-mediated DISC formation in a concentration-dependent manner.


Subject(s)
Calmodulin/metabolism , Computational Biology/methods , Protein Binding/drug effects , Trifluoperazine/pharmacology , fas Receptor/metabolism , Animals , Humans , Mice , Models, Molecular , Molecular Dynamics Simulation , Protein Structure, Secondary , Thermodynamics
8.
J Biol Chem ; 286(28): 24776-84, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21613217

ABSTRACT

Pancreatic cancer remains a devastating malignancy with a poor prognosis and is largely resistant to current therapies. To understand the resistance of pancreatic tumors to Fas death receptor-induced apoptosis, we investigated the molecular mechanisms of Fas-activated survival signaling in pancreatic cancer cells. We found that knockdown of the Fas-associated protein with death domain (FADD), the adaptor that mediates downstream signaling upon Fas activation, rendered Fas-sensitive MiaPaCa-2 and BxPC-3 pancreatic cells resistant to Fas-induced apoptosis. By contrast, Fas activation promoted the survival of the FADD knockdown MiaPaCa-2 and BxPC-3 cells in a concentration-dependent manner. The pharmacological inhibitor of ERK, PD98059, abrogated Fas-promoted cell survival in FADD knockdown MiaPaCa-2 and BxPC-3 cells. Furthermore, increased phosphorylation of Src was demonstrated to mediate Fas-induced ERK activation and cell survival. Immunoprecipitation of Fas in the FADD knockdown cells identified the presence of increased calmodulin, Src, and phosphorylated Src in the Fas-associated protein complex upon Fas activation. Trifluoperazine, a calmodulin antagonist, inhibited Fas-induced recruitment of calmodulin, Src, and phosphorylated Src. Consistently, trifluoperazine blocked Fas-promoted cell survival. A direct interaction of calmodulin and Src and their binding site were identified with recombinant proteins. These results support an essential role of calmodulin in mediating Fas-induced FADD-independent activation of Src-ERK signaling pathways, which promote survival signaling in pancreatic cancer cells. Understanding the molecular mechanisms responsible for the resistance of pancreatic cells to apoptosis induced by Fas-death receptor signaling may provide molecular insights into designing novel therapies to treat pancreatic tumors.


Subject(s)
Calmodulin/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fas-Associated Death Domain Protein/metabolism , MAP Kinase Signaling System , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , fas Receptor/metabolism , src-Family Kinases/metabolism , Calmodulin/genetics , Cell Line, Tumor , Cell Survival , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/genetics , Fas-Associated Death Domain Protein/genetics , Humans , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Phosphorylation , fas Receptor/genetics , src-Family Kinases/genetics
9.
Lab Invest ; 91(6): 896-904, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21464824

ABSTRACT

Cholangiocarcinoma is a highly malignant tumor with limited therapeutic options. We have previously reported that tamoxifen (TMX) induces apoptosis of cholangiocarcinoma cells and reduces cholangiocarcinoma tumorigenesis in mice. In the present studies, we determined the effect of combination therapy of TMX and gemcitabine (GMT), another chemotherapeutical reagent for many cancers, on cholangiocarcinoma tumorigenesis and investigated the responsible mechanisms. GMT inhibited cell growth and induced apoptosis of cholangiocarcinoma cells in a concentration-dependent manner. TMX enhanced GMT-induced apoptosis of cholangiocarcinoma cells. Consistently, GMT (15 mg/kg) inhibited cholangiocarcinoma tumorigenesis in nude mice by 50%. TMX (15 mg/kg) enhanced the inhibitory effect of GMT on tumorigenesis by 33%. The inhibition of tumor growth correlated with enhanced apoptosis in tumor tissues. To elucidate the mechanisms underlying the additive effects of TMX on GMT-induced apoptosis, we determined the activation of caspases in cholangiocarcinoma cells exposed to GMT, TMX, or both. Activation of caspases 9 and 3, as well as cytochrome c release to the cytosol, was demonstrated in cells exposed to both reagents. In contrast, TMX activated caspase 2, whereas GMT had no effect. Inhibition of caspase 2 activation decreased TMX-, but not GMT-, induced activation of caspase 3 and apoptosis of cholangiocarcinoma cells. Similarly, activation of caspase 2 was found in tumors from TMX-treated mice, but not GMT-treated mice. Therefore, the enhanced effect of TMX on GMT-induced cholangiocarcinoma cell death is partially mediated by activation of caspase 2. TMX and GMT both induce apoptosis and inhibit cholangiocarcinoma tumorigenesis, which may be attributed to the activation of distinct apoptosis signals by TMX and GMT. Our studies provide in vivo evidence and molecular insight to support the use of TMX and GMT in combination as an effective therapy for cholangiocarcinoma.


Subject(s)
Caspases/metabolism , Cholangiocarcinoma/drug therapy , Deoxycytidine/analogs & derivatives , Tamoxifen/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/physiopathology , Cytochromes c/metabolism , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Therapy, Combination , Enzyme Activation/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Nude , Tamoxifen/therapeutic use , Gemcitabine
10.
J Bone Miner Res ; 26(8): 1913-25, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21472777

ABSTRACT

The innate immune system and its components play an important role in the pathogenesis of inflammatory bone destruction. Blockade of inflammatory cytokines does not completely arrest bone erosion, suggesting that other mediators also may be involved in osteolysis. Previously we showed that nucleosides promote osteoclastogenesis and bone-resorption activity in the presence of receptor activator for nuclear factor κB ligand (RANKL) in vitro. The studies described here further demonstrate that selected nucleosides and nucleoside analogues accelerate bone destruction in mice immunized with collagen II alone (CII) but also further enhance bone erosion in mice immunized by collagen II plus complete Freund's adjuvant (CII + CFA). Abundant osteoclasts are accumulated in destructive joints. These data indicate that nucleosides act as innate immune activators distinct from CFA, synergistically accelerating osteoclast formation and inflammatory osteolysis. The potential roles of the surface triggering receptor expressed on myeloid cells (TREM) and the intracellular inflammasome in nucleoside-enhanced osteoclastogenesis have been studied. These observations provide new insight into the pathogenesis and underlying mechanism of bone destruction in inflammatory autoimmune osteoarthritis.


Subject(s)
Immunity, Innate/drug effects , Inflammation/complications , Inflammation/immunology , Nucleosides/pharmacology , Osteolysis/complications , Osteolysis/immunology , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Bone Resorption/complications , Bone Resorption/pathology , Carrier Proteins/metabolism , Femur/drug effects , Femur/pathology , Inflammasomes/metabolism , Inflammation/pathology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Osteoclasts/drug effects , Osteoclasts/pathology , Osteogenesis/drug effects , Osteolysis/pathology , Receptors, Immunologic/metabolism , Receptors, Purinergic/metabolism , Signal Transduction/drug effects
11.
Annu Rev Pathol ; 6: 121-45, 2011.
Article in English | MEDLINE | ID: mdl-20936937

ABSTRACT

The skeleton provides mechanical support for stature and locomotion, protects vital organs, and controls mineral homeostasis. A healthy skeleton must be maintained by constant bone modeling to carry out these crucial functions throughout life. Bone remodeling involves the removal of old or damaged bone by osteoclasts (bone resorption) and the subsequent replacement of new bone formed by osteoblasts (bone formation). Normal bone remodeling requires a tight coupling of bone resorption to bone formation to guarantee no alteration in bone mass or quality after each remodeling cycle. However, this important physiological process can be derailed by a variety of factors, including menopause-associated hormonal changes, age-related factors, changes in physical activity, drugs, and secondary diseases, which lead to the development of various bone disorders in both women and men. We review the major diseases of bone remodeling, emphasizing our current understanding of the underlying pathophysiological mechanisms.


Subject(s)
Bone Diseases/pathology , Bone Diseases/physiopathology , Bone Remodeling/physiology , Animals , Humans
12.
J Bone Miner Metab ; 29(1): 111-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20589403

ABSTRACT

Mechanical forces are essential to maintain skeletal integrity, and microgravity exposure leads to bone loss. The underlying molecular mechanisms leading to the changes in osteoblasts and osteoclast differentiation and function remain to be fully elucidated. Because of the infrequency of spaceflights and payload constraints, establishing in vitro and in vivo systems that mimic microgravity conditions becomes necessary. We have established a simulated microgravity (modeled microgravity, MMG) system to study the changes induced in osteoclast precursors. We observed that MMG, on its own, was unable to induce osteoclastogenesis of osteoclast precursors; however, 24 h of MMG activates osteoclastogenesis-related signaling molecules ERK, p38, PLCγ2, and NFATc1. Receptor activator of NFkB ligand (RANKL) (with or without M-CSF) stimulation for 3-4 days in gravity of cells that had been exposed to MMG for 24 h enhanced the formation of very large tartrate-resistant acid phosphatase (TRAP)-positive multinucleated (>30 nuclei) osteoclasts accompanied by an upregulation of the osteoclast marker genes TRAP and cathepsin K. To validate the in vitro system, we studied the hindlimb unloading (HLU) system using BALB/c mice and observed a decrease in BMD of femurs and a loss of 3D microstructure of both cortical and trabecular bone as determined by micro-CT. There was a marked stimulation of osteoclastogenesis as determined by the total number of TRAP-positive multinucleated osteoclasts formed and also an increase in RANKL-stimulated osteoclastogenesis from precursors removed from the tibias of mice after 28 days of HLU. In contrast to earlier reported findings, we did not observe any histomorphometric changes in the bone formation parameters. Thus, the foregoing observations indicate that microgravity sensitizes osteoclast precursors for increased differentiation. The in vitro model system described here is potentially a valid system for testing drugs for preventing microgravity-induced bone loss by targeting the molecular events occurring in microgravity-induced enhanced osteoclastogenesis.


Subject(s)
Osteoclasts/cytology , RANK Ligand/pharmacology , Weightlessness , Acid Phosphatase/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Bone Density/drug effects , Cell Line , Hindlimb Suspension/physiology , Isoenzymes/metabolism , Male , Mice , Mice, Inbred BALB C , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/drug effects , Receptor Activator of Nuclear Factor-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tartrate-Resistant Acid Phosphatase
13.
Ann N Y Acad Sci ; 1192: 358-64, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20392260

ABSTRACT

Calcium signaling plays a key role in bone turnover, regulating both osteoblasts and osteoclasts. Despite this the role of calmodulin, the primary intracellular calcium receptor regulatory protein, has received little attention. In this brief review, the function of Ca(2+)/calmodulin signaling in osteoclast development, function, and apoptosis is reviewed. Considerable evidence supports an important regulatory role for Ca(2+)/calmodulin signaling in each of these processes. The overall role of Ca(2+)/calmodulin in regulating bone turnover is also supported by animal and human studies showing that calmodulin antagonists preserve bone mass.


Subject(s)
Calmodulin/physiology , Osteoclasts/metabolism , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Bone Remodeling/genetics , Bone Remodeling/physiology , Calcium/pharmacology , Calmodulin/antagonists & inhibitors , Calmodulin/genetics , Calmodulin/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Female , Humans , Osteoclasts/drug effects , Osteoclasts/physiology , Ovariectomy , Rodentia , Signal Transduction/drug effects , Signal Transduction/genetics
14.
Lab Invest ; 89(11): 1221-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19752858

ABSTRACT

Obesity is a risk factor for breast cancer and is associated with increased plasma concentrations of free fatty acids (FFAs). We and others have demonstrated that FFA induces plasminogen activator inhibitor-1 (PAI-1) expression in a variety of cells. Emerging evidence supports elevation of PAI-1 as a prognostic marker for breast cancer. Therefore, we hypothesized that FFAs might increase expression of PAI-1 in breast cancer cells and facilitate breast cancer progression. Secreted PAI-1 was higher in invasive and metastatic MDA-MB-231 cells compared with less invasive and non-metastatic Hs578T cells. Utilizing FFAs with different saturation and chain lengths, we demonstrated that linoleic acid induced expression of PAI-1 in MDA-MB-231 cells. Linoleic acid also induced in vitro migration of MDA-MB-231. By contrast, other FFAs tested had little or no effect on PAI-1 expression or migration. Linoleic acid-induced breast cancer cell migration was completely inhibited by virally expressed antisense PAI-1 RNA. Furthermore, increased expression of PAI-1 by FFAs was not detected in the SMAD4-deficient MDA-MB-468 breast carcinoma cells. Electrophoretic mobility-shift assay confirmed that linoleic acid-induced expression of PAI-1 was mediated, at least in part, by SMAD4 in MDA-MB-231 cells. That linoleic acid induces PAI-1 expression in breast cancer cells through SMAD4 provides a novel insight into understanding the relationships between two migration-associated molecules, FFAs, and PAI-1.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement/drug effects , Fatty Acids, Nonesterified/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Smad4 Protein/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Neoplasm Invasiveness , Oligoribonucleotides, Antisense/pharmacology , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Smad4 Protein/genetics
15.
Clin Cancer Res ; 15(4): 1288-96, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19228732

ABSTRACT

PURPOSE: Cholangiocarcinoma is a fatal tumor with limited therapeutic options. We have reported that calmodulin antagonists tamoxifen and trifluoperazine induced apoptosis in cholangiocarcinoma cells. Here, we determined the effects of tamoxifen on tumorigenesis and the molecular mechanisms of tamoxifen-induced apoptosis. EXPERIMENTAL DESIGN: Nude mice xenograft model of cholangiocarcinoma was used and tamoxifen was given i.p. and intratumorally. Cholangiocarcinoma cells were used to characterize molecular mechanisms of tamoxifen-induced apoptosis in vitro. RESULTS: I.p. or intratumoral injection of tamoxifen decreased cholangiocarcinoma tumorigenesis by 40% to 80% in nude mice. In cells isolated from tumor xenografts, tamoxifen inhibited phosphorylation of AKT (pAKT) and cellular FLICE like inhibitory protein (c-FLIP). Immunohistochemical analysis further showed that pAKT was identified in all nontreated tumors but was absent in tamoxifen-treated tumors. In vitro, tamoxifen activated caspase-8 and caspase-10, and their respective inhibitors partially blocked tamoxifen-induced apoptosis. Overexpression of c-FLIP inhibited tamoxifen-induced apoptosis and enhanced tumorigenesis of cholangiocarcinoma cells in nude mice, whereas deletion of the calmodulin-binding domain on c-FLIP restored the sensitivity to tamoxifen and inhibited tumorigenesis. With two additional cholangiocarcinoma cell lines, we confirmed that the expression of FLIP is an important factor in mediating spontaneous and tamoxifen-induced apoptosis. CONCLUSIONS: Thus, tamoxifen inhibits cholangiocarcinoma tumorigenesis in nude mice. Tamoxifen-induced apoptosis is partially dependent on caspases, inhibition of pAKT, and FLIP expression. Further, calmodulin-FLIP binding seems to be important in FLIP-mediated resistance to tamoxifen. Therefore, the present studies support the concept that tamoxifen may be used as a therapy for cholangiocarcinoma and possibly other malignancies in which the calmodulin targets AKT and c-FLIP play important roles in the tumor pathogenesis.


Subject(s)
Bile Duct Neoplasms/drug therapy , Calmodulin/antagonists & inhibitors , Cholangiocarcinoma/drug therapy , Tamoxifen/pharmacology , Animals , Apoptosis/drug effects , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic , CASP8 and FADD-Like Apoptosis Regulating Protein/antagonists & inhibitors , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Calmodulin/physiology , Caspases/physiology , Cell Line, Tumor , Cholangiocarcinoma/pathology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Tamoxifen/therapeutic use , Xenograft Model Antitumor Assays
16.
J Cell Biochem ; 106(1): 25-32, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19021144

ABSTRACT

Notch signaling is associated with prostate osteoblastic bone metastases and calcium/calmodulin-dependent kinase II (CaMKII) is associated with osteoblastogenesis of human mesenchymal stem cells. Here we show that prostate cancer cell lines C4-2B and PC3, both derived from bone metastases and express Notch-1, have all four isoforms of CaMKII (alpha, beta, gamma, delta). In contrast, prostate cancer cell lines LNcaP and DU145, which are not derived from bone metastases and lack the Notch-1 receptor, both lack the alpha isoform of CaMKII. In addition, DU145 cells also lack the beta-isoform. In C4-2B cells, inhibition of CaMKII by KN93 or gamma-secretase by L-685,458 inhibited the formation of the cleaved form of Notch-1 thus inhibiting Notch signaling. KN93 inhibited down stream Notch-1 signaling including Hes-1 gene expression, Hes-1 promoter activity, and c-Myc expression. In addition, both KN93 and L-685,458 inhibited proliferation and Matrigel invasion by C4-2B cells. The activity of gamma-secretase was unaffected by KN93 but markedly inhibited by L-685,458. Inhibition of the expression of alpha, beta, or gamma-isoform by siRNA did not affect Hes-1 gene expression, however when expression of one isoform was inhibited by siRNA, there were compensatory changes in the expression of the other isoforms. Over-expression of CaMKII-alpha increased Hes-1 expression, consistent with Notch-1 signaling being at least partially dependent upon CaMKII. This unique crosstalk between CaMKII and Notch-1 pathways provides new insight into Notch signaling and potentially provides new targets for pharmacotherapeutics.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Prostatic Neoplasms/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Amyloid Precursor Protein Secretases/metabolism , Animals , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/enzymology , Sulfonamides/pharmacology
17.
Biophys J ; 95(12): 5913-21, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18820240

ABSTRACT

Previous studies have demonstrated a calcium-dependent interaction of calmodulin (CaM) and Fas that is regulated during Fas-induced apoptosis in several cell lines, including cholangiocarcinoma, Jurkat cells, and osteoclasts. The binding of CaM and Fas has been identified on residues 231-254 of Fas; the V254N point mutation decreases the CaM/Fas binding, and the C-terminal deletion mutation increases the CaM/Fas binding. Recent studies have shown that CaM is recruited into the Fas-mediated death-inducing signaling complex (DISC) in a calcium-dependent manner. However, the molecular mechanisms whereby Fas mutations and CaM/Fas binding might regulate Fas-mediated DISC formation are unknown. In this study we investigated the binding thermodynamics and conformation of the CaM/Fas complexes with combined explicit solvent molecular-dynamics simulations and implicit solvent binding free-energy calculations. The binding free-energy analysis demonstrated that the Fas V254N point mutation reduced its binding affinity with CaM. In contrast, the Fas mutant with the deletion of the 15 amino acid at the C-terminus increased its binding to CaM. These observations are consistent with previous findings from biochemical studies. Conformational analyses further showed that the Fas V254N mutation resulted in an unstable conformation, whereas the C-terminal deletion mutation stabilized the Fas conformation, and both mutations resulted in changes of the degree of correlation between the motions of the residues in Fas. Analysis of the CaM/Fas complex revealed that CaM/Fas binding stabilized the conformation of both CaM and Fas and changed the degree of correlated motion of the residues of CaM and Fas. The results presented here provide structural evidence for the roles of Fas mutations and CaM/Fas binding in Fas-induced DISC formation. Understanding the molecular mechanisms of CaM/Fas binding in Fas-mediated DISC formation should provide important insights into the function of Fas mutations and CaM in regulating Fas-mediated apoptosis.


Subject(s)
Calcium/metabolism , Calmodulin/chemistry , Calmodulin/metabolism , fas Receptor/chemistry , fas Receptor/metabolism , Models, Molecular , Movement , Protein Binding , Protein Conformation , Protein Stability , Protein Structure, Tertiary , Sequence Deletion , Signal Transduction , Solvents/metabolism , Thermodynamics , fas Receptor/genetics
18.
J Biol Chem ; 283(43): 29593-601, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18728019

ABSTRACT

Phospholipase Cgamma2 (PLCgamma2) is an important signaling effector of multiple receptors in the immune system. Here we show that PLCgamma2-deficient mice displayed impaired lymph node organogenesis but normal splenic structure and Peyer's patches. Receptor activator of NF-kappaB ligand (RANKL) is a tumor necrosis factor family cytokine and is essential for lymph node organogenesis. Importantly, PLCgamma2 deficiency severely impaired RANKL signaling, resulting in marked reduction of RANKL-induced activation of MAPKs, p38 and JNK, but not ERK. The lack of PLCgamma2 markedly diminished RANKL-induced activation of NF-kappaB, AP-1, and NFATc1. Moreover, PLCgamma2 deficiency impaired RANKL-mediated biological function, leading to failure of the PLCgamma2-deficient bone marrow macrophage precursors to differentiate into osteoclasts after RANKL stimulation. Re-introduction of PLCgamma2 but not PLCgamma1 restores RANKL-mediated osteoclast differentiation of PLCgamma2-deficient bone marrow-derived monocyte/macrophage. Taken together, PLCgamma2 is essential for RANK signaling, and its deficiency leads to defective lymph node organogenesis and osteoclast differentiation.


Subject(s)
Phospholipase C gamma/physiology , RANK Ligand/physiology , Animals , Cell Differentiation , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lymph Nodes/pathology , Mice , Mice, Transgenic , Models, Biological , Organogenesis , Osteoclasts/metabolism , Phospholipase C gamma/metabolism , Signal Transduction , Spleen/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Biol Chem ; 283(22): 15319-27, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18378684

ABSTRACT

Oxidative stress plays a critical role in the pathogenesis of atherosclerosis including the formation of lipid laden macrophages and the development of inflammation. However, oxidative stress-induced molecular signaling that regulates the development of vascular calcification has not been investigated in depth. Osteogenic differentiation of vascular smooth muscle cells (VSMC) is critical in the development of calcification in atherosclerotic lesions. An important contributor to oxidative stress in atherosclerotic lesions is the formation of hydrogen peroxide from diverse sources in vascular cells. In this study we defined molecular signaling that is operative in the H2O2-induced VSMC calcification. We found that H2O2 promotes a phenotypic switch of VSMC from contractile to osteogenic phenotype. This response was associated with an increased expression and transactivity of Runx2, a key transcription factor for osteogenic differentiation. The essential role of Runx2 in oxidative stress-induced VSMC calcification was further confirmed by Runx2 depletion and overexpression. Inhibition of Runx2 using short hairpin RNA blocked VSMC calcification, and adenovirus-mediated overexpression of Runx2 alone induced VSMC calcification. Inhibition of H2O2-activated AKT signaling blocked VSMC calcification and Runx2 induction concurrently. This blockage did not cause VSMC apoptosis. Taken together, our data demonstrate a critical role for AKT-mediated induction of Runx2 in oxidative stress-induced VSMC calcification.


Subject(s)
Calcinosis/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Hydrogen Peroxide/metabolism , Muscle, Smooth, Vascular/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adenoviridae , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Calcinosis/pathology , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Mice , Muscle, Smooth, Vascular/pathology , Transcriptional Activation
20.
AIDS Res Hum Retroviruses ; 24(4): 607-16, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18370588

ABSTRACT

The cytosolic domain of human immunodeficiency virus gp160 glycoprotein contains two calmodulin-binding regions. The role of these domains in modulating intracellular calmodulin signaling is of considerable interest in unraveling the mechanism whereby calmodulin regulates Fas-mediated apoptosis in HIV-infected cells. In this investigation we have employed 2D-NMR spectroscopy to determine the solution structure of the 30-residue calmodulin-binding domain corresponding to residues 826-855 of gp160. In solution, the gp160 (826-855) peptide exhibits a high degree of segmental flexibility. Within its conformational manifold, we have detected two separate flexible amphipathic helices involving residues 826-841 and 846-855 connected by a highly flexible type-II beta-turn at Pro-843 and Arg-844. The observed NOE pattern as well as the observation of long-range NOE contacts between the side chains of His-841 and Ile-846 are compatible with the presence of this turn in the conformational manifold of this peptide. This investigation focusing on the properties of the free peptide in solution paves the way for extending the investigations on the interaction of calmodulin with HIV-1 gp160.


Subject(s)
Calmodulin/metabolism , HIV Envelope Protein gp160/chemistry , HIV Envelope Protein gp160/metabolism , HIV-1/chemistry , Humans , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Solutions
SELECTION OF CITATIONS
SEARCH DETAIL
...