Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Leukemia ; 31(12): 2678-2685, 2017 12.
Article in English | MEDLINE | ID: mdl-28490813

ABSTRACT

A hallmark of bone marrow changes with aging is the increase in adipocyte composition, but how this impacts development of multiple myeloma (MM) is unknown. Here, we report the role of the adipokine leptin as master regulator of anti-myeloma tumor immunity by modulating the invariant natural killer T (iNKT) cell function. A marked increase in serum leptin levels and leptin receptor (LR) expression on iNKT cells in MM patients and the 5T33 murine MM model was observed. MM cells and leptin synergistically counteracted anti-tumor functionality of both murine and human iNKT cells. In vivo blockade of LR signaling combined with iNKT stimulation resulted in superior anti-tumor protection. This was linked to persistent IFN-γ secretion upon repeated iNKT cell stimulation and a restoration of the dynamic antigen-induced motility arrest as observed by intravital microscopy, thereby showing alleviation of iNKT cell anergy. Overall our data reveal the LR axis as novel therapeutic target for checkpoint inhibition to treat MM.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/metabolism , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/metabolism , Receptors, Leptin/antagonists & inhibitors , Animals , Antibodies, Monoclonal , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/biosynthesis , Disease Models, Animal , Galactosylceramides/pharmacology , Humans , Leptin/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Natural Killer T-Cells/immunology , Xenograft Model Antitumor Assays
2.
Leukemia ; 31(8): 1706-1714, 2017 08.
Article in English | MEDLINE | ID: mdl-27909306

ABSTRACT

Chromosomal region 1p22 is deleted in ⩾20% of multiple myeloma (MM) patients, suggesting the presence of an unidentified tumor suppressor. Using high-resolution genomic profiling, we delimit a 58 kb minimal deleted region (MDR) on 1p22.1 encompassing two genes: ectopic viral integration site 5 (EVI5) and ribosomal protein L5 (RPL5). Low mRNA expression of EVI5 and RPL5 was associated with worse survival in diagnostic cases. Patients with 1p22 deletion had lower mRNA expression of EVI5 and RPL5, however, 1p22 deletion status is a bad predictor of RPL5 expression in some cases, suggesting that other mechanisms downregulate RPL5 expression. Interestingly, RPL5 but not EVI5 mRNA levels were significantly lower in relapsed patients responding to bortezomib and; both in newly diagnosed and relapsed patients, bortezomib treatment could overcome their bad prognosis by raising their progression-free survival to equal that of patients with high RPL5 expression. In conclusion, our genetic data restrict the MDR on 1p22 to EVI5 and RPL5 and although the role of these genes in promoting MM progression remains to be determined, we identify RPL5 mRNA expression as a biomarker for initial response to bortezomib in relapsed patients and subsequent survival benefit after long-term treatment in newly diagnosed and relapsed patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Chromosome Deletion , Chromosomes, Human, Pair 1 , Multiple Myeloma/genetics , Ribosomal Proteins/genetics , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Humans , MDS1 and EVI1 Complex Locus Protein , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Mutation , Proto-Oncogenes/genetics , RNA, Messenger/analysis , Transcription Factors/genetics
3.
Leukemia ; 30(3): 640-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26487273

ABSTRACT

Bortezomib (bort) has improved overall survival in patients with multiple myeloma (MM), but the majority of them develop drug resistance. In this study, we demonstrate that bone marrow (BM) fibroblasts (cancer-associated fibroblasts; CAFs) from bort-resistant patients are insensitive to bort and protect the RPMI8226 and patients' plasma cells against bort-induced apoptosis. Bort triggers CAFs to produce high levels of interleukin (IL)-6, IL-8, insulin-like growth factor (IGF)-1 and transforming growth factor (TGF) ß. Proteomic studies on CAFs demonstrate that bort resistance parallels activation of oxidative stress and pro-survival autophagy. Indeed, bort induces reactive oxygen species in bort-resistant CAFs and activates autophagy by increasing light chain 3 protein (LC3)-II and inhibiting p62 and phospho-mammalian target of rapamycin. The small-interfering RNA knockdown of Atg7, and treatment with 3-methyladenine, restores bort sensitivity in bort-resistant CAFs and produces cytotoxicity in plasma cells co-cultured with CAFs. In the syngeneic 5T33 MM model, bort-treatment induces the expansion of LC3-II(+) CAFs. TGFß mediates bort-induced autophagy, and its blockade by LY2109761, a selective TßRI/II inhibitor, reduces the expression of p-Smad2/3 and LC3-II and induces apoptosis in bort-resistant CAFs. A combination of bort and LY2109761 synergistically induces apoptosis of RPMI8226 co-cultured with bort-resistant CAFs. These data define a key role for CAFs in bort resistance of plasma cells and provide the basis for a novel targeted therapeutic approach.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Multiple Myeloma/drug therapy , Pyrazoles/pharmacology , Pyrroles/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Aged , Aged, 80 and over , Animals , Autophagy/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Drug Combinations , Drug Resistance, Neoplasm/genetics , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Plasma Cells/drug effects , Plasma Cells/metabolism , Plasma Cells/pathology , Primary Cell Culture , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Survival Analysis , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
5.
Leukemia ; 28(4): 904-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23995611

ABSTRACT

The role of cancer-associated fibroblasts (CAFs) has not been previously studied in multiple myeloma (MM). Here, cytofluorimetric analysis revealed higher proportions of bone marrow (BM) CAFs in patients with active MM (both at diagnosis and relapse) compared with patients in remission or those with monoclonal gammopathy of undetermined significance or deficiency anemia (controls). CAFs from MM patients produced increased levels of transforming growth factor-ß, interleukin-6, stromal cell-derived factor-1α, insulin-like growth factor-1, vascular endothelial growth factor and fibroblast growth factor-2 and displayed an activated and heterogeneous phenotype, which supported their origin from resident fibroblasts, endothelial cells and hematopoietic stem and progenitor cells via the endothelial-mesenchymal transition as well as mesenchymal stem cells via the mesenchymal transition, as both of these processes are induced by MM cells and CAFs. Active MM CAFs fostered chemotaxis, adhesion, proliferation and apoptosis resistance in MM cells through cytokine signals and cell-to-cell contact, which were inhibited by blocking CXCR4, several integrins and fibronectin. MM cells also induced the CAFs proliferation. In syngeneic 5T33MM and xenograft mouse models, MM cells induced the expansion of CAFs, which, in turn, promoted MM initiation and progression as well as angiogenesis. In BM biopsies from patients and mice, nests of CAFs were found in close contact with MM cells, suggesting a supportive niche. Therefore, the targeting of CAFs in MM patients may be envisaged as a novel therapeutic strategy.


Subject(s)
Bone Marrow Cells/physiology , Fibroblasts/physiology , Multiple Myeloma/pathology , Aged , Aged, 80 and over , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Chemokine CXCL12/physiology , Disease Progression , Epithelial-Mesenchymal Transition , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenotype
10.
Leukemia ; 23(10): 1894-903, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19494837

ABSTRACT

Multiple myeloma (MM) is a B-cell malignancy, which often remains incurable because of the development of drug resistance governed by the bone marrow (BM) microenvironment. Novel treatment strategies are therefore urgently needed. In this study, we evaluated the anti-MM activity of JNJ-26481585, a novel 'second-generation' pyrimidyl-hydroxamic acid-based histone deacetylase inhibitor, using the syngeneic murine 5TMM model of MM. In vitro, JNJ-26481585 induced caspase cascade activation and upregulation of p21, resulting in apoptosis and cell cycle arrest in the myeloma cells at low nanomolar concentrations. Similar results could be observed in BM endothelial cells using higher concentrations, indicating the selectivity of JNJ-26481585 toward cancer cells. In a prophylactic and therapeutic setting, treatment with JNJ-26481585 resulted in an almost complete reduction of the tumor load and a significant decrease in angiogenesis. 5T2MM-bearing mice also developed a MM-related bone disease, characterized by increased osteoclast number, development of osteolytic lesions and a reduction in cancellous bone. Treatment of these mice with JNJ-264815 significantly reduced the development of bone disease. These data suggest that JNJ-26481585 has a potent anti-MM activity that can overcome the stimulatory effect of the BM microenvironment in vivo making this drug a promising new anti-MM agent.


Subject(s)
Disease Models, Animal , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Animals , Apoptosis/drug effects , Blotting, Western , Bone Diseases/drug therapy , Bone Diseases/etiology , Bone Marrow/drug effects , Bone Marrow/pathology , Cell Cycle/drug effects , Mice , Mice, Inbred C57BL , Multiple Myeloma/blood supply , Neovascularization, Pathologic/prevention & control , Osteolysis/drug therapy , Osteolysis/etiology
11.
Br J Cancer ; 98(12): 1966-74, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18521088

ABSTRACT

Aplidin is an antitumour drug, currently undergoing phase II evaluation in different haematological and solid tumours. In this study, we analysed the antimyeloma effects of Aplidin in the syngeneic 5T33MM model, which is representable for the human disease. In vitro, Aplidin inhibited 5T33MMvv DNA synthesis with an IC(50) of 3.87 nM. On cell-cycle progression, the drug induced an arrest in transition from G0/G1 to S phase, while Western blot showed a decreased cyclin D1 and CDK4 expression. Furthermore, Aplidin induced apoptosis by lowering the mitochondrial membrane potential, by inducing cytochrome c release and by activating caspase-9 and caspase-3. For the in vivo experiment, 5T33MM-injected C57Bl/KaLwRij mice were intraperitoneally treated with vehicle or Aplidin (90 microg kg(-1) daily). Chronic treatment with Aplidin was well tolerated and reduced serum paraprotein concentration by 42% (P<0.001), while BM invasion with myeloma cells was decreased by 35% (P<0.001). Aplidin also reduced the myeloma-associated angiogenesis to basal values. This antiangiogenic effect was confirmed in vitro and explained by inhibition of endothelial cell proliferation and vessel formation. These data indicate that Aplidin is well tolerated in vivo and its antitumour and antiangiogenic effects support the use of the drug in multiple myeloma.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Depsipeptides/therapeutic use , Disease Models, Animal , Multiple Myeloma/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Cycle , DNA Replication/drug effects , Depsipeptides/pharmacology , Mice , Mice, Inbred C57BL , Multiple Myeloma/pathology , Peptides, Cyclic , Rats
13.
Curr Pharm Biotechnol ; 7(6): 415-21, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17168657

ABSTRACT

Thalidomide and the analogues lenalidomide (CC-5013, Revlimid) and CC-4047 (Actimid) belong to the family of immunomodulatory drugs (IMiDs). These agents have anti-angiogenic properties, modulate TNFalpha and IL-12 secretion, co-stimulate T cells, increase NK cell toxicity and have direct anti-tumour effects. These characteristics have made of them promising drugs for treatment of relapsed, refractory and newly diagnosed MM. It seems that lenalidomide and CC-4047 are more powerful in inhibiting TNFalpha and have, except for myelosuppression, less side effects than Thal. Combination of IMiDs with dexamethasone, bortezomib and with chemotherapeutic agents are tested in numerous trials, which will help in determining the optimal combination and administration schedule of different molecules to take advantage of non-overlapping toxicities and synergisms between those agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Cytokines/immunology , Immunologic Factors/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Thalidomide/analogs & derivatives , Thalidomide/administration & dosage , Animals , Apoptosis/drug effects , Humans , Treatment Outcome
14.
Leukemia ; 20(10): 1870-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16900214

ABSTRACT

The cell surface expression of CD9, a glycoprotein of the tetraspanin family influencing several processes including cell motility and metastasis, inversely correlates with progression in several solid tumors. In the present work, we studied the expression and role of CD9 in multiple myeloma (MM) biology using the 5T33MM mouse model. The 5T33MMvitro cells were found to be CD9 negative. Injection of these cells in mice caused upregulation of CD9 expression, while reculturing them resulted in downregulation of CD9. Coculturing of CD9-negative 5T33MMvitro cells with BM endothelial cells (BMECs) resulted in a partial retrieval of CD9. Laser microdissection followed by real-time polymerase chain reaction and immunohistochemistry performed on bone sections of 5T33MMvivo diseased mice demonstrated strong local expression of CD9 on MM cells in contact with BMEC compared to MM cells further away. These findings were also confirmed by immunohistochemistry in MM patients. Neutralizing anti-CD9 antibodies inhibited transendothelial invasion of CD9-expressing human MM5.1 and murine 5T33MMvivo cells. In conclusion, we provide evidence that CD9 expression by the MM cells is upregulated in vivo by close interaction of the cells with BMEC and that CD9 is involved in transendothelial invasion, thus possibly mediating homing and/or spreading of the MM cells.


Subject(s)
Antigens, CD/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Membrane Glycoproteins/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Animals , Antigens, CD/genetics , Biopsy , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Communication , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Multiple Myeloma/physiopathology , Neoplasm Invasiveness , Tetraspanin 29 , Up-Regulation
15.
Histol Histopathol ; 20(4): 1227-50, 2005 10.
Article in English | MEDLINE | ID: mdl-16136504

ABSTRACT

Multiple myeloma (MM) is a malignant disease that results from an excess of monotypic plasma cells in the bone marrow (BM). This malignancy is characterised by complex karyotypic aberrancies. In 60% of all MM there are recurrent primary translocations involving the heavy chain gene locus. The MM cells strongly interact with the BM microenvironment, which is composed of endothelial cells, stromal cells, osteoclasts, osteoblasts, immune cells, fat cells and the extracellular matrix. This interaction is responsible for the specific homing in the BM, the proliferation and survival of the MM cells, the resistance of MM cells to chemotherapy, the development of osteolysis, immunodeficiency and anaemia. New therapeutic agents target both the MM, as well as the interaction MM cell - BM microenviroment.


Subject(s)
Bone Marrow Cells/physiology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Animals , Humans , Multiple Myeloma/epidemiology , Multiple Myeloma/physiopathology
16.
Leukemia ; 19(9): 1628-33, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16015389

ABSTRACT

Multiple myeloma (MM) is an incurable B-cell cancer characterised by the monoclonal proliferation of tumour cells in the bone marrow (BM). It has been described that matrix metalloproteinases (MMPs) and especially MMP-9 is secreted by MM cells. In this study, we investigated the possibility to exploit MMP-9 activity to activate prodrugs and to target MM cells as a new tumour-specific therapy. Cleavage of the prodrug EV1-FITC by MMP-9 resulted in release of fluorescence which can be used as a measure of prodrug activation. The 5T33MM mouse model was used in this proof-of-principle study. The prodrug was activated in a higher amount by addition to MMP-9-producing 5T33MMvv cells, homogenates from tumour-bearing organs (BM, spleen) and isolated 5T33MM-diseased BM and spleen cells compared to non-MMP-9-producing 5T33MMvt cells and homogenates/cells from non-tumour-bearing organs/mice, as measured by fluorescence release. This fluorescence release could be inhibited by the MMP-2/MMP-9-specific inhibitor, CTT. Activation of the prodrug in the 5T33MM spleen and BM homogenates was confirmed by chromatography. EV1-fluorescein isothiocyanate injection into 5T33MM-diseased animals resulted in a higher fluorescence release by the isolated BM and spleen cells compared to injection into healthy animals. In conclusion, MMP-9 activity can be used to activate prodrugs that target MM.


Subject(s)
Bone Marrow Cells/drug effects , Fluoresceins/therapeutic use , Matrix Metalloproteinase 9/metabolism , Multiple Myeloma/drug therapy , Oligopeptides/therapeutic use , Prodrugs/therapeutic use , Animals , Biotransformation , Bone Marrow Cells/metabolism , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Fluoresceins/chemical synthesis , Fluoresceins/metabolism , Fluorescence , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Inbred C57BL , Molecular Conformation , Multiple Myeloma/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Organ Specificity/drug effects , Prodrugs/chemical synthesis , Prodrugs/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/metabolism
17.
Virchows Arch ; 445(1): 27-35, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15168117

ABSTRACT

The aim of the study was to compare the angiogenic status, potential qualitative differences in microvessels and carbonic anhydrase IX expression in bone-marrow (BM) metastases and different haematological tumours at time of diagnosis. The microvessel density (MVD), endothelial-cell proliferation (ECP) and carbonic anhydrase IX (CA IX) immunoreactivity were determined on 210 trephine biopsies from 57 patients with multiple myeloma (MM), 13 with acute myeloid leukaemia (AML), 48 with chronic myeloproliferative syndrome (CMPS), 26 with chronic lymphocytic leukaemia (CLL), 25 with epithelial BM metastases, 18 with monoclonal gammopathy of undetermined significance (MGUS) and from a control group composed of 23 patients without haematological neoplasm. There was an increased MVD and ECP in epithelial BM metastases, MM, AML, CMPS and in a part of CLL. While an ECP greater than 0 was detected in 72% of MM, 75% of CMPS and 92% of AML, it was invariably observed (100%) in the BM metastases. The absence of ECP together with a MVD comparable with the control group in our MGUS cases supports the view that MGUS is a pre-angiogenic condition. Qualitative differences in microvessels were associated with growth patterns in MM and CLL and were observed between the different entities of CMPS. In one-third of the epithelial BM metastases, there was a focal CA IX immunoreactivity, which was never observed in the haematological diseases.


Subject(s)
Antigens, Neoplasm/metabolism , Bone Marrow Neoplasms/secondary , Carbonic Anhydrases/metabolism , Endothelium, Vascular/pathology , Hematologic Neoplasms/pathology , Neovascularization, Pathologic/pathology , Paraproteinemias/pathology , Biomarkers, Tumor/metabolism , Biopsy , Bone Marrow Neoplasms/enzymology , Capillaries/pathology , Capillaries/physiopathology , Carbonic Anhydrase IX , Cell Division , Hematologic Neoplasms/enzymology , Humans , Immunohistochemistry , Microcirculation , Paraproteinemias/enzymology
18.
Histol Histopathol ; 19(3): 935-50, 2004 07.
Article in English | MEDLINE | ID: mdl-15168356

ABSTRACT

Increased microvessel density (MVD) has been observed in the bone marrow (BM) of patients with multiple myeloma (MM), acute lymphoblastic leukaemia, acute myeloid leukaemia, and myelodysplastic and myeloproliferative syndrome. The MVD is the net result of cumulative phases of angiogenesis and angio-regression and is as such not an indicator of the ongoing angiogenesis at the time of biopsy. There is, therefore, a need for additional methods that allow the estimation of ongoing angiogenesis. Double immunostainings for CD34 and Ki-67 can be used on paraffin-embedded tissue to determine the endothelial proliferation fraction. The BM endothelial cells, as a component of the BM stroma, have a close interaction with the malignant cells. In MM, for example, they are involved in the specific homing and are a source of paracrine growth factors. Targeting the BM microvessels will not only influence the nutrient and oxygen supply, but will in addition reduce the growth stimuli provided by the EC.


Subject(s)
Bone Marrow/physiopathology , Endothelium, Vascular/pathology , Hematologic Neoplasms/pathology , Hematologic Neoplasms/physiopathology , Myeloproliferative Disorders/pathology , Neovascularization, Pathologic , Angiogenesis Inhibitors/therapeutic use , Animals , Antigens, CD34/analysis , Bone Marrow/blood supply , Bone Marrow/immunology , Bone Marrow/pathology , Cell Division , Cell Hypoxia , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Microcirculation , Multiple Myeloma/blood supply , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Multiple Myeloma/physiopathology
19.
Leukemia ; 18(5): 976-82, 2004 May.
Article in English | MEDLINE | ID: mdl-14999296

ABSTRACT

The migration of multiple myeloma (MM) cells from the circulation into the bone marrow (BM) implicates that they must have the capacity to cross the BM endothelium including the subendothelial basement membrane. In this study, human CD138+ MM cells were immunomagnetically isolated from BM samples of MM patients and their invasion through Matrigel, that is, a reconstituted basement membrane, was determined. We demonstrated that primary MM cells have the capacity to transmigrate through basement membrane and that this invasiveness was considerably increased when assessed on Matrigel filters coated with BM endothelial cells (EC) (4LHBMEC line) (transendothelial invasion). The isolated MM cells were shown by zymography to secrete matrix metalloproteinase (MMP)-9 and anti-MMP-9 antibodies inhibited transendothelial invasion, indicating that MMP-9 is involved in this process. BM EC were found to increase the MMP-9 secretion in MM cells, indicating that EC enhance MM cell invasion through stimulation of MMP-9 secretion. BM EC were found to produce hepatocyte growth factor (HGF), and this cytokine also stimulated MMP-9 secretion in MM cells, while anti-HGF antibodies significantly inhibited EC-stimulated MM cell invasion. In summary, our findings provide evidence that MM cell-BM EC interactions enhance the invasion of human MM cells through stimulation of MMP-9 secretion.


Subject(s)
Bone Marrow Cells/physiology , Endothelial Cells/physiology , Hepatocyte Growth Factor/physiology , Matrix Metalloproteinase 9/physiology , Multiple Myeloma/pathology , Cells, Cultured , Humans , Multiple Myeloma/enzymology , Neoplasm Invasiveness , Up-Regulation
20.
Br J Cancer ; 90(5): 1076-83, 2004 Mar 08.
Article in English | MEDLINE | ID: mdl-14997210

ABSTRACT

Insulin-like growth factor-1 (IGF-1) has been described as an important factor in proliferation, cell survival and migration of multiple myeloma (MM) cells. Angiogenesis correlates with development and prognosis of the MM disease. Vascular endothelial growth factor (VEGF) is one of the prominent factors involved in this process. The different functions of IGF-1 were investigated in the 5TMM mouse model with emphasis on proliferation, migration and VEGF secretion, and the signalling pathways involved. Western Blot analysis revealed that ERK1/2 and Akt (PKB) were activated after IGF-1 stimulation. The activation of ERK1/2 was reduced by the PI3K inhibitor Wortmannin, implying that the PI3K pathway is involved in its activation. Insulin-like growth factor-1 induced an increase in DNA synthesis in MM cells, which was mediated by a PI3K/Akt-MEK/ERK pathway. Insulin-like growth factor-1 enhanced F-actin assembly and this process was only PI3K mediated. Stimulation by IGF-1 of VEGF production was reduced by PD98059, indicating that only the MEK-ERK pathway is involved in IGF-1-stimulated VEGF production. In conclusion, IGF-1 mediates its multiple effects on MM cells through different signal transduction pathways. In the future, we can study the potential in vivo effects of IGF-1 inhibition on tumour growth and angiogenesis in MM.


Subject(s)
Chemotaxis/drug effects , Insulin-Like Growth Factor I/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Division/drug effects , Cell Movement/drug effects , Disease Models, Animal , Down-Regulation , Flavonoids/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Multiple Myeloma/metabolism , Neovascularization, Pathologic/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...