13.11.2014 Views

Haematologica 2003 - Supplements

Haematologica 2003 - Supplements

Haematologica 2003 - Supplements

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

integrin in IGF-1-triggered MM cell adhesion and migration. A<br />

functional blocking antibody against β1 integrin and a RGD<br />

peptide, as well as cytochalasin D (cyt D: a cytoskeleton<br />

inhibitory agent), significantly reduced IGF-1-induced cell<br />

adhesion. Immunoprecipitation studies demonstrated that IGF-1<br />

rapidly and transiently induces association of IGF-1R and 1<br />

integrin, which is associated with phosphorylation of IGF-1R,<br />

IRS-1, and p85/PI3K. IGF-1 also triggers phosphorylation of<br />

AKT and ERK. We next demonstrated that IGF-1R and 1<br />

integrin comigrate to cholesterol-rich microdomains on plasma<br />

membrane (membrane rafts) following IGF-1 stimulation, using<br />

western blotting for the Triton-insoluble and Triton-soluble<br />

fraction of the cells and dual immunofluorescence staining. Using<br />

methyl-β-cyclodextrin to disrupt membrane raft structure, we<br />

further show that β1 integrin-mediated IGF-1-stimulated<br />

adhesion requires intact lipid rafts. In addition, IGF-1 (100 ng/ml<br />

and 40 ng/ml for MM.1S and OPM6, respectively) triggers<br />

polymerization of F-actin, indicating focal adhesion formation,<br />

and immunoprecipitation experiments confirm that IGF-1 induces<br />

interaction of β1 integrin with cytoskeletal and signaling proteins<br />

localized at focal adhesions, including focal adhesion kinase<br />

(p125FAK), α-actinin, and paxillin. Pretreatment of cyt D<br />

abrogates activation of p125FAK and paxillin induced by IGF-1.<br />

Importantly, IGF-1-induced MM cell adhesion to FN is achieved<br />

only when 1 integrin and PI3K/AKT are activated. In a 96-well<br />

transmigration assay, IGF-1 induces a 2-3 (MM.1S) and 3-4-fold<br />

(OPM6) increase in migration. Conversely, IR3 and a blocking<br />

anti-β1 integrin mAb, as well as Cyt D, abrogate MM cell<br />

transmigration. Interestingly, IGF-1 induces MM cell migration<br />

independent of de no vo protein synthesis. Finally, primary<br />

CD138+ MM cells isolated from 3 patients were also responsive<br />

to IGF-1-induced adhesion on FN-coated plates and BM stromal<br />

cells. Together, these studies demonstrate that IGF-1 induces MM<br />

cell adhesion and migration, suggesting a role of IGF-1 in the<br />

trafficking and localization of these cells in the BM<br />

microenvironment. Morover, these results identify a functional<br />

cooperation between IGF-1R and β1 integrin in MM homing,<br />

supporting blockade of IGF-1/IGF1R system as a novel treatment<br />

strategy of MM.<br />

113<br />

HHV-8 viral IL-6 homologue is as active as human IL-6<br />

on human myeloma cells<br />

Andreas Günther1, Frank Bakker1, Wolfgang Baum1,<br />

Renate Burger1, Frank Neipel2 and Martin Gramatzki1<br />

1Department of Medicine III / Div. of Hematology/Oncology and<br />

2Institute for Clinical and Molecular Virology, University of<br />

Erlangen, Germany<br />

Kaposi-Sarcoma-associated Virus/ Human-Herpesvirus-8<br />

(KSHV/HHV-8) is associated with at least three human<br />

lymphoproliferative disorders: primary effusion lymphomas<br />

(PEL), Castleman’s disease (CD) and plasmablastic lymphoma.<br />

However, the role of HHV-8 in multiple myeloma (MM) is still<br />

controversial. Some groups detected HHV-8-DNA in bone<br />

marrow samples of MM patients whereas other groups failed to<br />

confirm these data. An important issue is the potential functional<br />

role of HHV-8 which encodes for several viral cytokines. Most<br />

interest was given to the viral homologue of IL-6 (vIL-6) since<br />

human IL-6 (huIL-6) is not only an essential growth and survival<br />

factor for malignant plasma cells but plays an important role in<br />

PEL, CD and plasmablastic lymphoma. We could show that<br />

prokaryotically expressed vIL-6 (pro-v-IL-6) has biological<br />

activity on human myeloma cells in vitro (Burger et al., Blood<br />

1998). However, the vIL-6 concentrations needed for a maximum<br />

stimulatory growth effect on the strictly IL-6 dependent human<br />

myeloma cell line INA-6 were dramatically higher (4000 ng/ml)<br />

than those needed for recombinant huIL-6 (1.25 ng/ml) This<br />

requirement for very high amounts of vIL-6 argued against a<br />

causative role of vIL-6 in human diseases. However, this study<br />

was performed with prokaryotically expressed recombinant vIL-6<br />

which could be less active than vIL-6 expressed in human cells.<br />

Now, we could overcome these limitations. First we used<br />

conditioned medium (CM) of the HHV-8 infected PEL cell line<br />

BCBL-1, containing only small amounts of hu IL-6 (45 pg/ml).<br />

The addition of 50% CM was able to induce a proliferative<br />

response of the INA-6 plasma cell line which would have<br />

required 1 ng/ml of recombinant huIL-6. Anti-gp130 antibody<br />

(Ab) or the combination of anti-IL-6 receptor and anti-gp130<br />

Abs, but not anti-IL-6R Ab alone, were effective in inhibiting<br />

BCBL-1 CM activity on INA-6. These results are similar to those<br />

obtained with vIL-6 and indicate that the growth induction of<br />

BCBL-1 CM is mainly caused by vIL-6. In a second approach we<br />

used a baculovirus vector expression system in eukaryotic cells.<br />

Eukaryotically expressed vIL-6 (eu-v-IL-6) had a significantly<br />

higher activity than pro-v-IL-6 and induced a maximum<br />

stimulatory growth response in INA-6 cells at the same<br />

concentration as huIL-6 (1.25 ng/ml). Similar to the results<br />

published previously for pro-vIL-6 and obtained for BCBL-1<br />

CM. The growth effects induced by eu-v-IL-6 could be blocked<br />

by anti-gp 130 Ab or a combination of anti-IL-6R and anti-gp130<br />

Abs, but not anti-IL-6R Ab alone. Thus, our data indicate that<br />

the HHV-8 viral homologue of IL-6 is as active on human cells as<br />

hu IL-6. HHV-8 vIL-6 may therefore act as a substitute for the<br />

human cytokine in multiple myeloma and HHV-8 associated<br />

diseases and could play a causative role in their development.<br />

114<br />

Macrophage inflammatory protein 1-alpha (MIP-1α)<br />

triggers migration and signaling cascades mediating<br />

survival and proliferation in multiple myeloma cells<br />

Suzanne Lentzsch, Margarete Gries, Martin Janz, Ralf<br />

Bargou, Bernd Dorken, and Markus Y. Mapara.<br />

Humboldt University of Berlin, Charite Campus Buch, Robert-<br />

Roessle-Klinik, 13125 Berlin, Germany, Tel.: +49-30-94171370,<br />

FAX:+49-30-94171209, e-mail: lentzsch@rrk-berlin.de<br />

Recently, several studies have suggested that MIP-1 contributes<br />

to the pathophysiology of multiple myeloma (MM). Thus, it has<br />

been demonstrated that MIP-1α is an osteoclast stimulatory<br />

factor and that MIP-1α neutralizing antibody (ab) or antisense<br />

inhibit bone destruction and reduce tumor load in a SCID-mice<br />

model of MM. Furthermore, MM patients have significantly<br />

higher bone marrow plasma levels of MIP-1α than healthy<br />

controls.<br />

The current study was designed to determine the direct effects of<br />

MIP-1α on MM cells. We found expression of very high levels of<br />

MIP-1α and its receptor CCR5 in MM cell lines and patientderived<br />

primary cells. Furthermore, we were able to demonstrate<br />

that MIP-1α significantly increased proliferation of different MM<br />

cell lines (MM1.S, H929, OPM-2, INA-6) and patient-derived<br />

primary MM cells. MIP-1α specific migration could be observed<br />

in a dose-dependent fashion up to 21 fold compared to control. In<br />

MM cell line and patient MM cells, MIP-1α induces<br />

phosphorylation of p44/42 mitogen-activated protein kinase<br />

(MAPK) as well as AKT and its downstream target FKHR.<br />

STAT3 was not activated by MIP-1α. In addition, inhibition of<br />

AKT activation by the PI3-Kinase (PI3-K) inhibitors wortmannin<br />

S138

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!