Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 26(1): 169-81, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20683884

ABSTRACT

ADAM8 expression is increased in the interface tissue around a loosened hip prosthesis and in the pannus and synovium of patients with rheumatoid arthritis, but its potential role in these processes is unclear. ADAM8 stimulates osteoclast (OCL) formation, but the effects of overexpression or loss of expression of ADAM8 in vivo and the mechanisms responsible for the effects of ADAM8 on osteoclastogenesis are unknown. Therefore, to determine the effects of modulating ADAM expression, we generated tartrate-resistant acid phosphatase (TRAP)-ADAM8 transgenic mice that overexpress ADAM8 in the OCL lineage and ADAM8 knockout (ADAM8 KO) mice. TRAP-ADAM8 mice developed osteopenia and had increased numbers of OCL precursors that formed hypermultinucleated OCLs with an increased bone-resorbing capacity per OCL. They also had an enhanced differentiation capacity, increased TRAF6 expression, and increased NF-κB, Erk, and Akt signaling compared with wild-type (WT) littermates. This increased bone-resorbing capacity per OCL was associated with increased levels of p-Pyk2 and p-Src activation. In contrast, ADAM8 KO mice did not display a bone phenotype in vivo, but unlike WT littermates, they did not increase RANKL production, OCL formation, or calvarial fibrosis in response to tumor necrosis factor α (TNF-α) in vivo. Since loss of ADAM8 does not inhibit basal bone remodeling but only blocks the enhanced OCL formation in response to TNF-α, these results suggest that ADAM8 may be an attractive therapeutic target for preventing bone destruction associated with inflammatory disease.


Subject(s)
ADAM Proteins/metabolism , Antigens, CD/metabolism , Membrane Proteins/metabolism , Osteoclasts/cytology , Osteoclasts/enzymology , Stem Cells/cytology , Stem Cells/enzymology , Acid Phosphatase/metabolism , Animals , Biomarkers/metabolism , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Count , Cell Differentiation/drug effects , Cell Fusion , Enzyme Activation/drug effects , Isoenzymes/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Organ Size/drug effects , Osteoclasts/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/pharmacology , Signal Transduction/drug effects , Stem Cells/drug effects , Tartrate-Resistant Acid Phosphatase , Tumor Necrosis Factor-alpha/pharmacology , src-Family Kinases/metabolism
2.
J Cell Biochem ; 105(2): 370-80, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18636554

ABSTRACT

One of the most life-threatening complications of prostate cancer is skeletal metastasis. In order to develop treatment for metastasis, it is important to understand its molecular mechanisms. Our work in this field has drawn parallels between hematopoietic stem cell and prostate cancer homing to the marrow. Our recent work demonstrated that annexin II expressed by osteoblasts and endothelial cells plays a critical role in niche selection. In this study, we demonstrate that annexin II and its receptor play a crucial role in establishing metastasis of prostate cancer. Prostate cancer cell lines migrate toward annexin II and the adhesion of prostate cancer to osteoblasts and endothelial cells was inhibited by annexin II. By blocking annexin II or its receptor in animal models, short-term and long-term localization of prostate cancers are limited. Annexin II may also facilitate the growth of prostate cancer in vitro and in vivo by the MAPK pathway. These data strongly suggest that annexin II and its receptor axis plays a central role in prostate cancer metastasis, and that prostate cancer utilize the hematopoietic stem cell homing mechanisms to gain access to the niche.


Subject(s)
Annexin A2/physiology , Cell Physiological Phenomena , Neoplasm Metastasis/pathology , Prostatic Neoplasms/pathology , Receptors, Peptide/physiology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Coculture Techniques , Endothelial Cells , Humans , Male , Osteoblasts
3.
Cancer Res ; 67(8): 3777-84, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440091

ABSTRACT

To better understand the molecular changes that occur in Waldenstrom macroglobulinemia (WM), we employed antibody-based protein microarrays to compare patterns of protein expression between untreated WM and normal bone marrow controls. Protein expression was defined as a >2-fold or 1.3-fold change in at least 67% of the tumor samples. Proteins up-regulated by >2-fold included Ras family proteins, such as Rab-4 and p62DOK, and Rho family proteins, such as CDC42GAP and ROKalpha. Other proteins up-regulated by >1.3-fold included cyclin-dependent kinases, apoptosis regulators, and histone deacetylases (HDAC). We then compared the samples of patients with symptomatic and asymptomatic WM and showed similar protein expression signatures, indicating that the dysregulation of signaling pathways occurs early in the disease course. Three proteins were different by >2-fold in symptomatic versus asymptomatic, including the heat shock protein HSP90. Elevated protein expression was confirmed by immunohistochemistry and immunoblotting. Functional significance was validated by the induction of apoptosis and inhibition of proliferation using specific HDAC and HSP90 inhibitors. This study, therefore, identifies, for the first time, multiple novel proteins that are dysregulated in WM, which both enhance our understanding of disease pathogenesis and represent targets of novel therapeutics.


Subject(s)
Waldenstrom Macroglobulinemia/metabolism , Aged , Apoptosis/physiology , Benzoquinones/pharmacology , Bone Marrow Cells/metabolism , Cell Growth Processes/physiology , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Lactams, Macrocyclic/pharmacology , Lymphocytes/metabolism , Male , Middle Aged , Plasma Cells/metabolism , Proteomics , Reproducibility of Results , Waldenstrom Macroglobulinemia/pathology
4.
Cell Signal ; 19(4): 867-79, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17196367

ABSTRACT

Protein kinase D localizes in the Golgi and regulates protein transport from the Golgi to the plasma membrane. In the present study, we found that PKD3, a novel member of the PKD family, and its fluorescent protein fusions localized in the Golgi and in the vesicular structures that are in part marked by endosome markers. Fluorescent recovery after photobleaching (FRAP) showed that the PKD3-associated vesicular structures were constantly forming and dissolving, reflecting active subcellular structures. FRAP on plasma membrane-located PKD3 indicated a slower recovery of PKD3 fluorescent signal compared to those of PKC isoforms, implying a different targeting mechanism at the plasma membrane. VAMP2, the vesicle-localized v-SNARE, was later identified as a novel binding partner of PKD3 through yeast two-hybrid screening. PKD3 directly interacted with VAMP2 in vitro and in vivo, and colocalized in part with VAMP2 vesicles in cells. PKD3 did not phosphorylate VAMP-GFP and the purified GST-VAMP2 protein in in vitro phosphorylation assays. Rather, PKD3 was found to promote the recruitment of VAMP2 vesicles to the plasma membrane in response to PMA, while the kinase dead PKD3 abolished this effect. Thus, the kinase activity of PKD3 was required for PMA-induced plasma membrane trafficking of VAMP2. In summary, our findings suggest that PKD3 localizes to vesicular structures that are part of the endocytic compartment. The vesicular distribution may be attributed in part to the direct interaction between PKD3 and vesicle-associated membrane protein VAMP2, through which PKD3 may regulate VAMP2 vesicle trafficking by facilitating its recruitment to the target membrane.


Subject(s)
Cytoplasmic Vesicles/enzymology , Protein Kinase C/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Animals , CHO Cells , Cell Membrane/enzymology , Cricetinae , Cricetulus , Cytosol/enzymology , Endosomes/enzymology , Golgi Apparatus/enzymology , Humans , Phosphorylation , Protein Binding , Protein Transport , Recombinant Fusion Proteins/metabolism
5.
Blood ; 109(7): 2708-17, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17119115

ABSTRACT

The mechanisms by which multiple myeloma (MM) cells migrate and home to the bone marrow are not well understood. In this study, we sought to determine the effect of the chemokine SDF-1 (CXCL12) and its receptor CXCR4 on the migration and homing of MM cells. We demonstrated that CXCR4 is differentially expressed at high levels in the peripheral blood and is down-regulated in the bone marrow in response to high levels of SDF-1. SDF-1 induced motility, internalization, and cytoskeletal rearrangement in MM cells evidenced by confocal microscopy. The specific CXCR4 inhibitor AMD3100 and the anti-CXCR4 antibody MAB171 inhibited the migration of MM cells in vitro. CXCR4 knockdown experiments demonstrated that SDF-1-dependent migration was regulated by the P13K and ERK/ MAPK pathways but not by p38 MAPK. In addition, we demonstrated that AMD3100 inhibited the homing of MM cells to the bone marrow niches using in vivo flow cytometry, in vivo confocal microscopy, and whole body bioluminescence imaging. This study, therefore, demonstrates that SDF-1/CXCR4 is a critical regulator of MM homing and that it provides the framework for inhibitors of this pathway to be used in future clinical trials to abrogate MM trafficking.


Subject(s)
Chemokines, CXC/physiology , Multiple Myeloma/immunology , Receptors, CXCR4/physiology , Animals , Antibodies, Monoclonal/pharmacology , Benzylamines , Bone Marrow/immunology , Bone Marrow/pathology , Case-Control Studies , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CXCL12 , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/blood , Chemotaxis/drug effects , Chemotaxis/physiology , Cyclams , Cytoskeleton/physiology , Heterocyclic Compounds/pharmacology , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/pathology , Multiple Myeloma/physiopathology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/blood , Receptors, CXCR4/genetics
6.
Clin Cancer Res ; 12(22): 6826-35, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17121904

ABSTRACT

PURPOSE: The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (mTOR) pathway and the heat shock protein family are up-regulated in multiple myeloma and are both regulators of the cyclin D/retinoblastoma pathway, a critical pathway in multiple myeloma. Inhibitors of mTOR and HSP90 protein have showed in vitro and in vivo single-agent activity in multiple myeloma. Our objective was to determine the effects of the mTOR inhibitor rapamycin and the HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) on multiple myeloma cells. EXPERIMENTAL DESIGN: Multiple myeloma cell lines were incubated with rapamycin (0.1-100 nmol/L) and 17-AAG (100-600 nmol/L) alone and in combination. RESULTS: In this study, we showed that the combination of rapamycin and 17-AAG synergistically inhibited proliferation, induced apoptosis and cell cycle arrest, induced cleavage of poly(ADP-ribose) polymerase and caspase-8/caspase-9, and dysregulated signaling in the phosphatidylinositol 3-kinase/AKT/mTOR and cyclin D1/retinoblastoma pathways. In addition, we showed that both 17-AAG and rapamycin inhibited angiogenesis and osteoclast formation, indicating that these agents target not only multiple myeloma cells but also the bone marrow microenvironment. CONCLUSIONS: These studies provide the basis for potential clinical evaluation of this combination for multiple myeloma patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzoquinones/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/therapeutic use , Multiple Myeloma/drug therapy , Protein Kinases/metabolism , Sirolimus/therapeutic use , Apoptosis/drug effects , Benzoquinones/administration & dosage , Benzoquinones/pharmacology , Bone Marrow Cells/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Drug Synergism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lactams, Macrocyclic/administration & dosage , Lactams, Macrocyclic/pharmacology , Models, Biological , Neovascularization, Physiologic/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
7.
J Bone Miner Res ; 21(10): 1657-65, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16995821

ABSTRACT

UNLABELLED: We identified a previously unknown integrin, alpha(9)beta(1), on OCLs and their precursors. Antibody to alpha(9) inhibited OCL formation in human marrow cultures, and OCLs from alpha(9) knockout mice had a defect in actin ring reorganization and an impaired bone resorption capacity. INTRODUCTION: Integrins play important roles in osteoclast (OCL) formation and function. Mature OCLs mainly express alpha(v)beta(3) integrin, a heterodimer adhesion receptor that has been implicated in osteoclastic bone resorption. We identified ADAM8, a disintegrin and metalloproteinase, as a novel stimulator of OCL differentiation and showed that the disintegrin domain of ADAM8 mediated its effects on OCL formation. Because the disintegrin domain of ADAM8 does not bind Arg-Gly-Asp (RGD) sequences, we determined which integrin bound ADAM8 and characterized its role in OCL formation and activity. MATERIALS AND METHODS: Chinese hamster ovary cells (CHO) expressing different integrin subunits were tested for their capacity to bind the disintegrin domain of ADAM8. Mouse or human bone marrow cells and purified OCL precursors were tested for alpha(9)beta(1) integrin expression by Western blot, immunocytochemistry, and real-time RT-PCR. A monoclonal antibody to human alpha(9) was used to block alpha(9)beta(1) on OCL precursors stimulated by 1alpha,25-dihydroxyvitamin D(3) [1alpha,25(OH)(2)D(3)] or RANKL. Vertebrae of 7-day-old alpha(9)(-/-) mice and wildtype (WT) littermates were compared using bone histomorphometry and 3D microCT analysis. RESULTS: Alpha(9) integrin was expressed by mouse and human bone marrow-derived OCLs and their precursors. Importantly, the anti-alpha(9) antibody inhibited human OCL formation stimulated by 1alpha,25(OH)(2)D(3) or RANKL dose-dependently. Furthermore, analysis of OCLs formed in marrow cultures from alpha(9)(-/-) mice showed that the OCLs formed were more contracted and formed significantly less bone resorption pits on dentin slices. Histologic analysis of alpha(9)(-/-) vertebrae showed thickened trabecular regions and retained cartilage within vertebral bodies of alpha(9)(-/-) mice. 3D microCT analysis of alpha(9)(-/-) vertebrae also showed a significant increase in trabecular bone volume/total tissue volume and a tendency for decreased trabecular separation compared with WT mice. CONCLUSIONS: These results support a previously unknown role for alpha(9)beta(1) integrin in OCL formation and function.


Subject(s)
Bone Resorption , Integrins/metabolism , Osteoclasts/physiology , ADAM Proteins/metabolism , Animals , Antigens, CD/metabolism , CHO Cells , Cell Differentiation , Cricetinae , Cytoskeleton/metabolism , Humans , Immunohistochemistry , Integrin alphaVbeta3/metabolism , Integrins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
8.
J Biol Chem ; 281(41): 30542-50, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16895901

ABSTRACT

Annexin II is a heterotetramer, consisting of two 11-kDa (p11) and two 36-kDa (p36) subunits, that is produced by osteoclasts and stimulates osteoclast formation. However, its receptor is unknown. We showed that annexin II binds to normal primary human marrow stromal cells and the Paget's marrow-derived PSV10 stromal cell line to induce osteoclast formation. 125I-Labeled annexin II binding assays with PSV10 cells demonstrated that there was a single class of annexin II receptors with a Kd of 5.79 nm and Bmax of 2.13 x 10(5) receptors/cell. Annexin III or annexin V did not bind this receptor. Using 125I-labeled annexin II binding to screen NIH3T3 transfected with a human marrow cDNA expression library, we identified a putative annexin II receptor clone, which encoded a novel 26-kDa type I membrane receptor protein when expressed in HEK 293 cells. HEK 293 cells transformed with the cloned annexin II receptor cDNA showed a similar binding affinity to annexin II as that observed in PSV10 cells. Chemical cross-linking experiments with biotinylated annexin II and intact PSV10 cells identified a 55-kDa band on Western blot analysis that reacted with both an anti-p11 antibody and streptavidin but not anti-p36 antibody. A rabbit polyclonal antibody raised against the putative recombinant annexin II receptor also recognized the same 26-kDa protein band detected in PSV10 cells. Importantly, the annexin II receptor antibody dose-dependently blocked the stimulatory effects of annexin II on human osteoclast formation, demonstrating that the receptor mediates the effects of annexin II on osteoclast formation.


Subject(s)
Annexin A2/chemistry , Bone Marrow Cells/metabolism , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Stromal Cells/metabolism , Animals , Cloning, Molecular , Cross-Linking Reagents/pharmacology , Humans , Kinetics , Mice , Molecular Sequence Data , NIH 3T3 Cells , Osteoclasts/metabolism
9.
J Bone Miner Res ; 20(7): 1161-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15940368

ABSTRACT

UNLABELLED: We report that AX-II, in addition to inducing GM-CSF expression, also increases membrane-bound RANKL synthesis by marrow stromal cells and does so through a previously unreported MAPK-dependent pathway. Thus, both GM-CSF and RANKL are required for AX-II stimulation of OCL formation. INTRODUCTION: Annexin II (AX-II) is an autocrine/paracrine factor secreted by osteoclasts (OCLs) that stimulates human OCL formation and bone resorption in vitro by inducing bone marrow stromal cells and activated CD4+ T cells to produce granulocyte-macrophage colony-stimulating factor (GM-CSF). GM-CSF in turn increases OCL precursor proliferation and further enhances OCL formation. However, the induction of GM-CSF by AX-II cannot fully explain its effects on OCL formation. In this study, we tested the capacity of AX-II to induce the expression of RANKL and the corresponding signaling pathways AX-II employs in human marrow stromal cells to induce RANKL. We also showed that both GM-CSF and RANKL are required for OCL formation induced by AX-II. MATERIALS AND METHODS: Real-time RT-PCR and Western blot analysis were used to detect RANKL and osteoprotegerin (OPG) mRNA and protein expression in unfractionated human bone marrow mononuclear cells stimulated with AX-II. Soluble RANKL in the conditioned medium was analyzed by ELISA. Activation of the MAPK pathway by AX-II was tested by Western blot. The effects of OPG and anti-GM-CSF on AX-II-induced OCL formation were also examined. RESULTS AND CONCLUSION: In addition to upregulating GM-CSF mRNA, AX-II increased RANKL mRNA expression dose-dependently in unfractionated human bone marrow mononuclear cells and modestly increased soluble RANKL in unfractionated human bone marrow mononuclear cell conditioned medium. However, AX-II markedly increased membrane-bound RANKL on human bone marrow stromal cells. Treatment of marrow stromal cells with AX-II activated MAP-kinase (ERKs) and PD 98059 abolished the effect but did not block the increase in GM-CSF. Interestingly, OPG, a natural decoy receptor for RANKL, or anti-GM-CSF partially inhibited OCL formation by AX-II in human bone marrow cells, and the combination of OPG and anti-GM-CSF completely blocked AX-II-induced OCL formation. These data show that AX-II stimulates both the proliferation and differentiation of OCL precursors through production of GM-CSF and RANKL respectively.


Subject(s)
Annexin A2/physiology , Bone Marrow Cells/metabolism , Carrier Proteins/biosynthesis , Membrane Glycoproteins/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/physiology , Annexin A2/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Carrier Proteins/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Flavonoids/pharmacology , Glycoproteins/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Membrane Glycoproteins/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Monocytes/drug effects , Monocytes/metabolism , Osteoprotegerin , Protein Kinase Inhibitors/pharmacology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor , Stromal Cells/drug effects , Stromal Cells/metabolism , Up-Regulation
10.
Mol Pharmacol ; 67(1): 152-62, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15496505

ABSTRACT

Protein kinase C (PKC) and protein kinase D (PKD) coordinate and regulate many fundamental cellular processes. In this study, we evaluate the role of classic and novel PKC (c/nPKC) and PKD in glucose transport in L6 myotubes. c/nPKC is either activated by short-term phorbol 12-myristate 13-acetate (PMA) treatment or down-regulated by prolonged PMA treatment at a high dose in L6 myotubes. Our results indicate that PMA treatments have little impact on basal and insulin-stimulated glucose uptake and insulin-induced Akt activation. In contrast, the PKC inhibitors Go6976 [12-(2-cyanoethyl)-6,7,12,13-tetrahydro-13-methyl-5-oxo-5H-indolo[2,3-a]pyrrolo[3,4-c] carbazole], Go6983 [2-[1-(3-dimethylaminopropyl)-5-methoxyindol-3-yl]-3-(1H-indol-3-yl)maleimide], GF 109203X [bisindolylmaleimide I; 2-[1-(3-dimethylaminopropyl)indol-3-yl]-3-(1H-indol-3-yl)maleimide], and Ro 31-8220 [bisindolylmaleimide IX; 2-{1-[3-(amidinothio)propyl]-1H-indol3-yl}-3-(1-methylindol-3-yl)maleimide] block basal and insulin-stimulated glucose uptake, and their inhibitory effects persist upon down-regulation of c/nPKC by PMA, implying the presence of PKC-independent effectors in mediating their inhibition of glucose uptake. Go6976, the potent cPKC inhibitor that also effectively inhibits PKD, dose-dependently blocks basal glucose uptake in L6 myotubes, whereas Go6983, the nonselective PKC inhibitor that is ineffective for PKD, has little effect on basal glucose uptake, implying the involvement of PKD in this process. Most prominently, adenoviral gene expression of a dominant-negative PKD isoform, PKD3, primarily inhibits basal glucose uptake and, to a lesser extent, insulin-stimulated glucose uptake, whereas overexpression of wild-type PKD3 significantly enhances basal glucose uptake. Moreover, expression of a PKD3-targeted siRNA significantly inhibits basal glucose uptake. Taken together, our results indicate that PKD, specifically PKD3, directly contributes to insulin-independent basal glucose uptake in L6 skeletal muscle cells.


Subject(s)
Glucose/metabolism , Muscle Fibers, Skeletal/metabolism , Protein Kinase C/metabolism , Animals , Biological Transport , Cell Line , Deoxyglucose/pharmacokinetics , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Insulin/pharmacology , Kinetics , Monosaccharide Transport Proteins/metabolism , Muscle Proteins/metabolism , Rats , Tetradecanoylphorbol Acetate/pharmacology
11.
J Leukoc Biol ; 77(3): 408-13, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15604121

ABSTRACT

Dendritic cells (DC) and natural killer (NK) cells are essential components of the innate immune system, which rapidly sense and eliminate invading pathogens and transformed cells, mediate inflammation, and initiate adaptive immune responses. During the early immune events, DC and NK cells interact and regulate each other. The cellular "cross talk" and its molecular mediators are believed to be critical to the quality and magnitude of innate and adaptive immune responses. The goal of the present manuscript is to identify and initially assess major molecular mediators of DC-NK cell interaction. We have previously shown that DC and NK cells constitutively express several tumor necrosis factor family ligands (TNFfLs) and corresponding TNF family receptors (TNFfRs). Therefore, DC and NK cells might be able to interact via cognate interplays of TNFfLs and TNFfRs. Here, we provide initial experimental evidence supporting this possibility. We found that combined but not individual ligation of several TNFfRs induced substantial increases in secretion of interleukin-12 and interferon-gamma by DC and NK cells, respectively. In contrast, specific, individual disruptions of the engagements of the corresponding TNfL-TNFfR pairs greatly impaired DC and NK cell abilities to reciprocally mediate the increases in cytokine secretion. These findings indicate that multiple TNFfLs mediate DC-NK cell interaction.


Subject(s)
Dendritic Cells/immunology , Killer Cells, Natural/immunology , Receptors, Tumor Necrosis Factor/immunology , Animals , Cell Communication/immunology , Female , Ligands , Mice , Mice, Inbred C57BL , Models, Biological
12.
Mol Cancer Res ; 2(2): 129-40, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14985469

ABSTRACT

The catalytic domain of overexpressed protein kinase C (PKC)-delta mediates phorbol 12-myristate 13-acetate (PMA)-induced differentiation or apoptosis in appropriate model cell lines. To define the portions of the catalytic domain that are critical for these isozyme-specific functions, we constructed reciprocal chimeras, PKC-delta/epsilonV5 and -epsilon/deltaV5, by swapping the V5 domains of PKC-delta and -epsilon. PKC-delta/epsilonV5 failed to mediate PMA-induced differentiation of 32D cells, showing the essential nature of the V5 domain for PKC-delta's functionality. The other chimera, PKC-epsilon/deltaV5, endowed inactive PKC-epsilon with nearly all PKC-delta's apoptotic ability, confirming the importance of PKC-delta in this function. Green fluorescent protein (GFP)-tagged PKC-deltaV5 and -epsilon/deltaV5 in A7r5 cells showed substantial basal nuclear localization, while GFP-tagged PKC-epsilon and -delta/epsilonV5 showed significantly less, indicating that the V5 region of PKC-delta contains determinants critical to its nuclear distribution. PKC-epsilon/deltaV5-GFP showed much slower kinetics of translocation to membranes in response to PMA than parental PKC-epsilon, implicating the PKC-epsilonV5 domain in membrane targeting. Thus, the V5 domain is critical in several of the isozyme-specific functions of PKC-delta and -epsilon.


Subject(s)
Protein Kinase C/chemistry , Protein Kinase C/metabolism , Animals , Apoptosis/drug effects , Catalytic Domain , Cell Differentiation/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Mice , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase C/genetics , Protein Kinase C-delta , Protein Kinase C-epsilon , Protein Structure, Tertiary , Protein Transport/drug effects , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology
13.
J Immunol ; 168(4): 1823-30, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11823515

ABSTRACT

Dendritic cells (DCs) mediate cross-priming of tumor-specific T cells by acquiring tumor Ags from dead cancer cells. The process of cross-priming would be most economical and efficient if DCs also induce death of cancer cells. In this study, we demonstrate that normal human in vitro generated immature DCs consistently and efficiently induce apoptosis in cancer cell lines, freshly isolated noncultured cancer cells, and normal proliferating endothelial cells, but not in most normal cells. In addition, in vivo generated noncultured peripheral blood immature DCs mediate similar tumoricidal activity as their in vitro counterpart, indicating that this DC activity might be biologically relevant. In contrast to immature DCs, freshly isolated monocytes (myeloid DC precursors) and in vitro generated mature DCs are not cytotoxic or are less cytotoxic, respectively, suggesting that DC-mediated killing of cancer cells is developmentally regulated. Comparable cytotoxic activity is mediated by untreated DCs, paraformaldehyde-fixed DCs, and soluble products of DCs, and is destructible by proteases, indicating that both cell membrane-bound and secreted proteins mediate this DC function. Overall, our data demonstrate that human immature DCs are capable of inducing apoptosis in cancer cells and thus to both directly mediate anticancer activity and initiate processing of cellular tumor Ags.


Subject(s)
Apoptosis , Dendritic Cells/immunology , Neoplasms/immunology , Caspases/physiology , Cell Line , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytotoxicity Tests, Immunologic , Dendritic Cells/classification , Humans , Immunophenotyping , Kinetics , Membrane Proteins/physiology , Monocytes/immunology , Neoplasms/pathology , Signal Transduction , Stem Cells/immunology , Tumor Cells, Cultured
14.
J Immunol ; 168(4): 1831-9, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11823516

ABSTRACT

Our recent studies have demonstrated that human immature dendritic cells (DCs) are able to directly and effectively mediate apoptotic killing against a wide array of cultured and freshly-isolated cancer cells without harming normal cells. In the present study, we demonstrate that this tumoricidal activity is mediated by multiple cytotoxic TNF family ligands. We determine that human immature DCs express on their cell surface four different cytotoxic TNF family ligands: TNF, lymphotoxin-alpha(1)beta(2), Fas ligand, and TNF-related apoptosis inducing ligand; while cancer cells express the corresponding death receptors. Disruptions of interactions between the four ligands expressed on DCs and corresponding death-signaling receptors expressed on cancer cells using specific Abs or R:Fc fusion proteins block the cytotoxic activity of DCs directed against cancer cells. The novel findings suggest that DC killing of cancer cells is mediated by the concerted engagement of four TNF family ligands of DCs with corresponding death receptors of cancer cells. Overall, our data demonstrate that DCs are fully equipped for an efficient direct apoptotic killing of cancer cells and suggest that this mechanism may play a critical role in both afferent and efferent anti-tumor immunity.


Subject(s)
Apoptosis , Dendritic Cells/immunology , Neoplasms/immunology , Apoptosis Regulatory Proteins , Cell Line , Cells, Cultured , Cytotoxicity Tests, Immunologic , Fas Ligand Protein , Humans , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/physiology , Lymphotoxin-beta , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Membrane Proteins/genetics , Membrane Proteins/physiology , Microscopy, Confocal , Monocytes/immunology , Neoplasms/pathology , RNA, Messenger/biosynthesis , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Stem Cells/immunology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...