Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Leukemia ; 29(4): 788-97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25134458

ABSTRACT

Precursor B-lineage acute lymphoblastic leukemia (pre-B ALL) affects hematopoietic development and therefore is associated with immune deficiencies that can be further exacerbated by chemotherapy. It is unclear if and when monoclonal antibodies (mAbs) that stimulate antibody-mediated cellular cytotoxicity (ADCC) can be used for treatment because this depends on the presence of functional effector cells. Here, we used flow cytometry to determine that patient samples at diagnosis, post-induction and relapse contain detectable numbers of CD56+ cells. We were able to selectively expand CD56+ immune effector cells from bone marrow and peripheral blood samples at diagnosis and at various stages of treatment by co-culture with artificial antigen-presenting K562 clone 9.mbIL-21 cells. Amplified CD56+CD3- cells had spontaneous and anti-B cell-activating factor receptor mAb-stimulated ADCC activity against allogeneic ALL cells, which could be further enhanced by IL-15. Importantly, matched CD56+ effector cells also killed autologous ALL cells grown out from leukemia samples of the same patient, through both spontaneous as well as antibody-dependent cellular cytotoxicity. Since autologous cell therapy will not be complicated by graft-versus-host disease, our results show that expanded CD56+ cells could be applied for treatment of pre-B ALL without transplantation, or for purging of bone marrow in the setting of autologous bone marrow transplants.


Subject(s)
Antibodies, Monoclonal/biosynthesis , B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Cytotoxicity, Immunologic , Antineoplastic Agents/therapeutic use , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , CD3 Complex/genetics , CD3 Complex/metabolism , CD56 Antigen/genetics , CD56 Antigen/metabolism , Cell Proliferation/drug effects , Child , Coculture Techniques , Gene Expression , Humans , Interleukin-15/pharmacology , K562 Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Remission Induction
3.
Leukemia ; 26(8): 1786-96, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22373785

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common malignancy affecting children and a major cause of mortality from hematopoietic malignancies in adults. A substantial number of patients become drug resistant during chemotherapy, necessitating the development of alternative modes of treatment. rGel (recombinant Gelonin)/BlyS (B-lymphocyte stimulator) is a toxin-cytokine fusion protein used for selective killing of malignant B-cells expressing receptors for B-cell-activating factor (BAFF/BLyS) by receptor-targeted delivery of the toxin, Gelonin. Here, we demonstrate that rGel/BLyS binds to ALL cells expressing BAFF receptor (BAFF-R) and upon internalization, it induces apoptosis of these cells and causes downregulation of survival genes even in the presence of stromal protection. Using an immunodeficient transplant model for human ALL, we show that rGel/BLyS prolongs survival of both Philadelphia chromosome-positive and negative ALL-bearing mice. Furthermore, we used AMD3100, a CXCR4 antagonist, to mobilize the leukemic cells protected in the bone marrow (BM) microenvironment and the combination with rGel/BLyS resulted in a significant reduction of the tumor load in the BM and complete eradication of ALL cells from the circulation. Thus, a combination treatment with the B-cell-specific fusion toxin rGel/BLyS and the mobilizing agent AMD3100 could be an effective alternative approach to chemotherapy for the treatment of primary and relapsed ALL.


Subject(s)
Antineoplastic Agents/therapeutic use , B-Cell Activating Factor/therapeutic use , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recombinant Fusion Proteins/therapeutic use , Ribosome Inactivating Proteins, Type 1/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Apoptosis/drug effects , B-Cell Activating Factor/administration & dosage , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , Benzylamines , Bone Marrow/pathology , Cell Line, Tumor , Cyclams , Drug Synergism , Gene Expression Regulation, Leukemic/drug effects , Heterocyclic Compounds/pharmacology , Humans , Mice , Mice, Nude , Mice, SCID , NF-kappa B/antagonists & inhibitors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/genetics , Ribosome Inactivating Proteins, Type 1/metabolism , Toxins, Biological/administration & dosage , Toxins, Biological/metabolism , Toxins, Biological/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Leukemia ; 25(8): 1314-23, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21483439

ABSTRACT

The bone marrow (BM) stromal niche can protect acute lymphoblastic leukemia (ALL) cells against the cytotoxicity of chemotherapeutic agents and is a possible source of relapse. The stromal-derived factor-1 (SDF-1)/CXCR4 axis is a major determinant in the crosstalk between leukemic cells and BM stroma. In this study, we investigated the use of AMD11070, an orally available, small-molecule antagonist of CXCR4, as an ALL-sensitizing agent. This compound effectively blocked stromal-induced migration of human ALL cells in culture and disrupted pre-established adhesion to stroma. To examine how to optimally use this compound in vivo, several combinations with cytotoxic drugs were tested in a stromal co-culture system. The best treatment regimen was then tested in vivo. Mice transplanted with murine Bcr/Abl ALL cells survived significantly longer when treated with a combination of nilotinib and AMD11070. Similarly, immunocompromised mice transplanted with human ALL cells and treated with vincristine and AMD11070 had few circulating leukemic cells, normal spleens and reduced human CD19+ cells in the BM at the termination of the experiment. These results show that combined treatment with AMD11070 may be of significant benefit in eradicating residual leukemia cells at locations where they would otherwise be protected by stroma.


Subject(s)
Aminoquinolines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/administration & dosage , Heterocyclic Compounds/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, CXCR4/antagonists & inhibitors , Aminoquinolines/metabolism , Animals , Benzimidazoles/metabolism , Benzylamines , Butylamines , Cell Movement/drug effects , Chemokine CXCL12/antagonists & inhibitors , Cyclams , Heterocyclic Compounds, 1-Ring , Humans , Mice , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction/drug effects , Vincristine/administration & dosage
6.
Leukemia ; 24(4): 813-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20111071

ABSTRACT

Dasatinib is a potent dual Abl/Src inhibitor approved for treatment of Philadelphia chromosome-positive (Ph-positive) leukemias. At a once-daily dose and a relatively short half-life of 3-5 h, tyrosine kinase inhibition is not sustained. However, transient inhibition of K562 leukemia cells with a high-dose pulse of dasatinib or long-term treatment with a lower dose was reported to irreversibly induce apoptosis. Here, the effect of dasatinib on treatment of Bcr/Abl-positive acute lymphoblastic leukemia (ALL) cells was evaluated in the presence of stromal support. Dasatinib eradicated Bcr/Abl ALL cells, caused significant apoptosis and eliminated tyrosine phosphorylation on Bcr/Abl, Src, Crkl and Stat-5. However, treatment of mouse ALL cells with lower doses of dasatinib over an extended period of time allowed the emergence of viable drug-resistant cells. Interestingly, dasatinib treatment increased cell-surface expression of CXCR4, which is important for survival of B-lineage cells, but this did not promote survival. Combined treatment of cells with dasatinib and a CXCR4 inhibitor resulted in enhanced cell death. These results do not support the concept that long-term treatment with low-dose dasatinib monotherapy will be effective in causing irreversible apoptosis in Ph-positive ALL, but suggest that combined treatment with dasatinib and drugs such as AMD3100 may be effective.


Subject(s)
Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/metabolism , Leukemia, Experimental/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Pyrimidines/pharmacology , Thiazoles/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Dasatinib , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, CXCR4/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/radiation effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
7.
Leukemia ; 21(6): 1189-97, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17392819

ABSTRACT

In leukemia patients, resistance to drug treatment develops while the malignant cells can interact with and derive support from their microenvironment, such as bone marrow stroma. To model this process, lymphoblastic leukemia cells from BCR/ABL transgenic mice were treated with the farnesyltransferase inhibitor (FTI) SCH66336 while in coculture with primary mouse embryonic fibroblasts. Coculture with fibroblasts allowed the outgrowth of a subpopulation of drug-resistant lymphoblasts that expressed N-cadherin, a cell-cell adhesion protein that normally is only expressed on specific cell types, including hematopoietic stem cells and fibroblasts. N-cadherin expression promoted increased adhesion of the lymphoblasts to the fibroblasts. Importantly, de novo expression of N-cadherin in parental nonexpressing lymphoblasts using lentiviral transduction increased the ability of the cells to survive FTI treatment. We conclude that FTI drug treatment of Bcr/Abl-positive lymphoblastic leukemia cells that are in contact with a defined microenvironment induces the selective survival of a more primitive subpopulation of leukemia cells that expresses N-cadherin. Experimental drug treatment of cancer cells in model systems that include a microenvironment may reveal novel molecules that contribute to drug resistance and may aid in the design of specific therapies to eradicate more primitive cells.


Subject(s)
Cadherins/physiology , Drug Resistance , Farnesyltranstransferase/antagonists & inhibitors , Fusion Proteins, bcr-abl , Leukemia, Lymphoid/pathology , Animals , Cadherins/genetics , Cell Adhesion , Cell Line, Tumor , Coculture Techniques , Female , Fibroblasts/cytology , Mice , Mice, Transgenic
9.
Leukemia ; 18(1): 23-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14603339

ABSTRACT

Treatment of chronic myelogenous leukemia with a specific inhibitor of the Bcr/Abl tyrosine kinase, imatinib, has shown great promise. However, acute lymphoblastic leukemias that express Bcr/Abl only transiently respond to imatinib. Therefore, alternative treatments for this type of leukemia are urgently needed. Here, we examined the activity of the farnesyltransferase inhibitor SCH66336 as a single chemotherapeutic agent in a nude mouse model representative of very advanced stage Bcr/Abl P190-positive lymphoblastic leukemia/lymphoma. Our results show that oral administration of the inhibitor was able to significantly increase the survival of these mice compared to controls treated with vehicle (P<0.005), and caused marked regression of the tumor burden in the treated mice. Upon prolonged treatment, lymphomas re-emerged and a subset of cells from two of such lymphomas tested was able to survive in the presence of increased concentrations of SCH66336. The same cells, however, remained sensitive towards imatinib. A combination of the two drugs, preceded by a therapy to reduce the initial tumor burden, could be very effective in the treatment of Ph-positive ALL. We conclude that SCH66336, on its own, is remarkably effective in eradicating large numbers of lymphoblastic lymphoma cells and causing visible reduction in tumor size, with minimal toxicity.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Leukemia, Experimental/drug therapy , Lymphoma/drug therapy , Piperidines/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pyridines/therapeutic use , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Benzamides , Disease Models, Animal , Farnesyltranstransferase , Female , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Imatinib Mesylate , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Nude , Mice, Transgenic , Piperazines/therapeutic use , Piperidines/administration & dosage , Piperidines/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/administration & dosage , Pyridines/adverse effects , Pyrimidines/therapeutic use , Survival Rate , Tumor Cells, Cultured/transplantation
10.
Development ; 128(21): 4217-27, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11684658

ABSTRACT

Experiments in cultured cells have implicated the molecular switch Rac in a wide variety of cellular functions. Here we demonstrate that the simultaneous disruption of two negative regulators of Rac, Abr and Bcr, in mice leads to specific abnormalities in postnatal cerebellar development. Mutants exhibit granule cell ectopia concomitant with foliation defects. We provide evidence that this phenotype is causally related to functional and structural abnormalities of glial cells. Bergmann glial processes are abnormal and GFAP-positive astroglia were aberrantly present on the pial surface. Older Abr;Bcr-deficient mice show spontaneous mid-brain glial hypertrophy, which can further be markedly enhanced by kainic acid. Double null mutant astroglia are hyper-responsive to stimulation with epidermal growth factor and lipopolysaccharide and exhibit constitutively increased phosphorylation of p38 mitogen-activated protein kinase, which is regulated by Rac. These combined data demonstrate a prominent role for Abr and Bcr in the regulation of glial cell morphology and reactivity, and consequently in granule cell migration during postnatal cerebellar development in mammals.


Subject(s)
Astrocytes/pathology , Cerebellum/growth & development , Oncogene Proteins/genetics , Protein-Tyrosine Kinases , Proteins/genetics , Proto-Oncogene Proteins , rac GTP-Binding Proteins/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Behavior, Animal , Cerebellum/cytology , Cerebellum/metabolism , GTPase-Activating Proteins , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/metabolism , Gliosis/genetics , Mesencephalon/metabolism , Mesencephalon/pathology , Mesencephalon/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/metabolism , Phosphorylation , Proteins/metabolism , Proto-Oncogene Proteins c-bcr , Purkinje Cells/metabolism , Rhombencephalon/growth & development , Rhombencephalon/metabolism , Rhombencephalon/pathology , p38 Mitogen-Activated Protein Kinases
11.
Int J Mol Med ; 8(2): 127-33, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11445862

ABSTRACT

Racs are involved in the regulation of important cellular processes including mitogenesis. We found that the E3 ubiquitination ligase subunit Cullin-1 interacts with constitutively active Rac3 but not with wild-type Rac3 in yeast. In mammalian cell lysates, Cullin-1 bound to V12Rac3, effector domain mutants V12L37Rac3 and V12H40Rac3, and insert domain deletion mutant V12Rac3DeltaIns(124-135). Cullin-1 also formed a clearly detectable complex with other activated Rac3-related proteins including Rac1, Rac2, Cdc42 and RhoA but not with the distantly related small GTPase Rap1. Since the proteasome is involved in cell cycle control through the programmed degradation of cell cycle proteins, the possible regulation of Rac levels during the cell cycle was examined. However, Rac was expressed at constant levels throughout the cell cycle, and a specific proteasome inhibitor had no effect on Rac protein levels. These combined results indicate that the binding of activated Rac to Cullin-1 does not affect Rac protein levels, nor does it mediate the regulation of mitogenesis by Rac. However, Rac-Cullin-1 interactions may serve to regulate other E3 ligase functions such as subcellular localization. Indeed, activated Rac3 and Cullin-1 co-localized to the perinuclear region of the cell. We also detected complex formation between Rac and the APC component CDC23. These results indicate that Rac may regulate specific proteolytic processes through directed subcellular localization of SCF or APC complexes.


Subject(s)
Cell Cycle Proteins/metabolism , Cullin Proteins , rac GTP-Binding Proteins/metabolism , Animals , Apc8 Subunit, Anaphase-Promoting Complex-Cyclosome , CHO Cells , COS Cells , Cell Cycle Proteins/genetics , Cell Nucleus/metabolism , Chlorocebus aethiops , Cricetinae , Humans , Ubiquitin , rac GTP-Binding Proteins/genetics
12.
Mol Biol Cell ; 12(5): 1457-66, 2001 May.
Article in English | MEDLINE | ID: mdl-11359935

ABSTRACT

Cleft lip and palate syndromes are among the most common congenital malformations in humans. Mammalian palatogenesis is a complex process involving highly regulated interactions between epithelial and mesenchymal cells of the palate to permit correct positioning of the palatal shelves, the remodeling of the extracellular matrix (ECM), and subsequent fusion of the palatal shelves. Here we show that several matrix metalloproteinases (MMPs), including a cell membrane-associated MMP (MT1-MMP) and tissue inhibitor of metalloproteinase-2 (TIMP-2) were highly expressed by the medial edge epithelium (MEE). MMP-13 was expressed both in MEE and in adjacent mesenchyme, whereas gelatinase A (MMP-2) was expressed by mesenchymal cells neighboring the MEE. Transforming growth factor (TGF)-beta3-deficient mice, which suffer from clefting of the secondary palate, showed complete absence of TIMP-2 in the midline and expressed significantly lower levels of MMP-13 and slightly reduced levels of MMP-2. In concordance with these findings, MMP-13 expression was strongly induced by TGF-beta3 in palatal fibroblasts. Finally, palatal shelves from prefusion wild-type mouse embryos cultured in the presence of a synthetic inhibitor of MMPs or excess of TIMP-2 failed to fuse and MEE cells did not transdifferentiate, phenocopying the defect of the TGF-beta3-deficient mice. Our observations indicate for the first time that the proteolytic degradation of the ECM by MMPs is a necessary step for palatal fusion.


Subject(s)
Embryo, Mammalian/metabolism , Matrix Metalloproteinases/metabolism , Palate/embryology , Tissue Inhibitor of Metalloproteinases/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cleft Palate/embryology , Epithelium/metabolism , Extracellular Matrix/metabolism , Female , Fibroblasts/physiology , Humans , Hydroxamic Acids/pharmacology , In Situ Hybridization , Matrix Metalloproteinases/genetics , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Knockout , Organ Culture Techniques , Palate/metabolism , Pregnancy , Protease Inhibitors/pharmacology , Tissue Inhibitor of Metalloproteinases/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinases/genetics , Transforming Growth Factor beta3
13.
Cancer Res ; 61(4): 1398-405, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245441

ABSTRACT

The adapter protein Crkl has been implicated in the abnormal signal transduction pathways activated by the Bcr/Abl oncoprotein, which causes Philadelphia-positive leukemias in humans. To investigate the role of Crkl in tumorigenesis, we have generated transgenic mice that express human Crkl from the CRKL promoter. Western blot analysis showed a 4-6-fold overexpression of transgenic Crkl above endogenous crkl in two lines and increased constitutive complex formation between Crkl and C3G, an exchange factor for the small GTPase Rap1. This was associated with a significant increase in integrin-based motility of transgenic macrophages. Overexpression of Crkl was associated with increased incidence of tumor formation, and Rap1 was activated in a metastatic mammary carcinoma. The coexpression of Crkl and Bcr/Abl in mice transgenic for P190 BCR/ABL and CRKL markedly increased the rapidity of development of leukemia/lymphoma, decreasing the average survival by 3.8 months. These results provide direct evidence that Crkl plays a role in tumor development and is important in the leukemogenesis caused by Bcr/Abl.


Subject(s)
Adaptor Proteins, Signal Transducing , Fusion Proteins, bcr-abl/genetics , Leukemia, Lymphoid/genetics , Nuclear Proteins/physiology , Animals , COS Cells/metabolism , Cell Movement/physiology , Female , Fusion Proteins, bcr-abl/physiology , Lymphoma/genetics , MAP Kinase Signaling System/genetics , Macrophages, Peritoneal/cytology , Male , Mice , Mice, Transgenic , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Phenotype , Promoter Regions, Genetic
14.
Int J Mol Med ; 7(4): 351-7, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254872

ABSTRACT

The Bcr/Abl P190 oncoprotein is responsible for the development of Philadelphia-chromosome positive acute lymphoblastic leukemia (ALL). The Bcr moiety in Bcr/Abl activates the Abl tyrosine kinase, an ingredient essential for the transforming capability of Bcr/Abl. Residues 1-63 of Bcr form an N-terminal oligomerization domain and are key to Abl activation in vitro. Mice transgenic for P190 BCR/ABL reproducibly develop an aggressive B-lineage lymphoblastic leukemia/lymphoma. Here we test the hypothesis that residues 1-63 of Bcr have a major in vivo contribution to the oncogenicity of Bcr/Abl P190 by the generation of mice transgenic for an N-terminal deleted form of P190. We find that although the transgene is expressed in the bone marrow of mice at an early age, the incidence of leukemogenesis is greatly diminished as compared to mice transgenic for non-mutated P190 Bcr/Abl. Sporadic hematological malignancies which did develop showed decreased levels of phosphotyrosine as compared to those of wild-type P190 transgenics, although Ras was activated. These results demonstrate that the Bcr oligomerization domain contributes to the oncogenicity of Bcr/Abl in vivo.


Subject(s)
Fusion Proteins, bcr-abl/physiology , Gene Expression Regulation, Leukemic/genetics , Leukemia, Experimental/genetics , Oncogene Proteins/physiology , Peptide Fragments/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Animals , Fusion Proteins, bcr-abl/genetics , Mice , Mice, Transgenic , Oncogene Proteins/genetics , Peptide Fragments/genetics , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-bcr , Sequence Deletion
15.
Blood ; 97(5): 1399-403, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222386

ABSTRACT

The Philadelphia (Ph) chromosome is found in approximately 3% of pediatric patients with acute lymphoblastic leukemia (ALL) and the percentage markedly increases in adult patients. The prognosis for this class of patients is poor, and no standard chemotherapy combination so far has demonstrated long-term efficacy. The Ph-translocation joins the BCR and ABL genes and leads to expression of a chimeric Bcr/Abl protein with enhanced tyrosine kinase activity. This increase in activity leads to malignant transformation by interference with basic cellular functions such as the control of proliferation, adherence to stroma and extracellular matrix, and apoptosis. One important pathway activated by Bcr/Abl is the Ras pathway. Ras proteins have to undergo a series of posttranslational modifications to become biologically active. The first modification is the farnesylation of the C-terminus catalyzed by farnesyl transferase. We studied the effect of the farnesyl transferase inhibitor SCH66336 in an in vivo murine model of Bcr/Abl-positive acute lymphoblastic leukemia. In the early leukemic phase, mice were randomly assigned to a treatment, a vehicle, and a nontreatment group. The treatment was well tolerated without any detectable side effects. All animals of the control groups died of leukemia/lymphoma within 103 days (range, 18-103 days). In contrast, 80% of the drug-receiving group survived without any signs of leukemia or lymphoma until termination of treatment, after a median treatment period of 200 days (range, 179-232 days). We conclude that farnesyl transferase inhibitor SCH66336 is able to revert early signs of leukemia and significantly prolongs survival in a murine ALL model.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Genes, abl/genetics , Piperidines/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pyridines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Marrow , Farnesyltranstransferase , Genes, abl/drug effects , Mice , Mice, Transgenic , Piperidines/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyridines/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
16.
Free Radic Biol Med ; 29(9): 825-33, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11063908

ABSTRACT

It has been previously reported that the metabolism of reduced glutathione (GSH) by gamma-glutamyltranspeptidase (GGT) in the presence of chelated metals leads to free radical generation and lipid peroxidation (LPO). The present study demonstrates for the first time that an established cell line expressing GGT-rel, a human GGT-related enzyme, metabolizes extracellular GSH to cysteinylglycine (CysGly) in a time-dependent manner when cells were incubated in a medium containing 2.5 mM GSH and 25 mM glycylglycine. Supplementation with 150-165 microM Fe(3+)-EDTA resulted in a reactive oxygen species (ROS) generation process. The resulting data showed a significantly higher level (7.6-fold) of ROS production in the GGT-rel positive cells in comparison with the GGT-rel negative control cells. CysGly and Cys, but not GSH, were responsible for the observed ROS production, as we confirmed by measuring the same process in the presence of Fe(3+)-EDTA and different thiols. A higher iron reduction and an increased LPO level determined by malondialdehyde HPLC measurement were also found in GGT-rel-overexpressing cells compared to GGT-rel negative cells. Our data clearly indicate that in the presence of iron, not only GGT, but also GGT-rel has a pro-oxidant function by generation of a reactive metabolite (CysGly) and must be taken into account as a potential physiopathological oxidation system.


Subject(s)
gamma-Glutamyltransferase/metabolism , 3T3 Cells , Animals , Cysteine/metabolism , Dipeptides/metabolism , Free Radicals/metabolism , Glutathione/metabolism , Glycylglycine/metabolism , Humans , Lipid Peroxidation , Mice , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , gamma-Glutamyltransferase/genetics
17.
Blood ; 96(6): 2226-32, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10979970

ABSTRACT

The deregulated Bcr/Abl tyrosine kinase is responsible for the development of Philadelphia (Ph)-positive leukemia in humans. To investigate the significance of the C-terminal Abl actin-binding domain within Bcr/Abl p190 in the development of leukemia/lymphoma in vivo, mutant p190 DNA constructs were used to generate transgenic mice. Eight founder and progeny mice of 5 different lines were monitored for leukemogenesis. Latency was markedly increased and occurrence decreased in the p190 del C lines as compared with nonmutated p190 BCR/ABL transgenics. Western blot analysis of involved hematologic tissues of the p190 del C transgenics with end-stage disease showed high-level expression of the transgene and tyrosine phosphorylation of Cbl and Hef1/Cas, proteins previously shown to be affected by Bcr/Abl. These results show that the actin-binding domain of Abl enhances leukemia development but does not appear to be an absolute requirement for leukemogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Fusion Proteins, bcr-abl/genetics , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Mutation , Actins/genetics , Actins/metabolism , Animals , Binding Sites , Fusion Proteins, bcr-abl/metabolism , Humans , Leukemia, Experimental/metabolism , Mice , Mice, Transgenic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding
18.
Oncogene ; 19(38): 4362-74, 2000 Sep 07.
Article in English | MEDLINE | ID: mdl-10980612

ABSTRACT

The activated tyrosine kinase, Bcr-abl, is implicated in a number of hematopoietic malignancies. The exact biological mechanism by which the kinases transforms cells is still not well delineated. Previous data has suggested that the inhibition of apoptosis and the deregulation of cell cycle progression as the result of P210Bcr-abl expression might contribute to leukemogenesis. In vitro systems in which Bcr-Abl is over-expressed have concluded that similar growth regulatory pathways are affected as a result of the expression of both P210 and P190Bcr-abl. Here, we utilized an in vitro P190Bcr-abl leukemia mouse model to dissect the early events that contribute to transformation by this isoform of Bcr-Abl. In this mouse model P190Bcr-abl is expressed as a low but physiologically relevant level in that all mice develop pre-B leukemia lymphomas. We show that cell cycle and apoptotic responses to DNA damage are intact in bone marrow and spleen cells of such animals. We also demonstrate a normal induction of p21WAF-1/CIP1 in both hematopoietic and non-hematopoietic tissue as a result of genotoxic stress. We suggest that P190Bcr-abl induced transformation is different than that of P210Bcr-abl.


Subject(s)
Cell Cycle/genetics , Cyclins/metabolism , Fusion Proteins, bcr-abl/genetics , Leukemia/genetics , Animals , Antibodies/pharmacology , Apoptosis/genetics , Apoptosis/radiation effects , Bone Marrow/pathology , Bone Marrow/physiology , Bone Marrow/radiation effects , CD3 Complex/immunology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , Interleukin-2/pharmacology , Lung/pathology , Lung/radiation effects , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Precancerous Conditions , Spleen/pathology , Spleen/physiology , Spleen/radiation effects , Whole-Body Irradiation
19.
Cytogenet Cell Genet ; 89(1-2): 18-23, 2000.
Article in English | MEDLINE | ID: mdl-10894930

ABSTRACT

The closely related small GTP-binding proteins Rac1, Rac2, and Rac3 are part of a larger Rho subfamily of Ras proteins. Because disruption of Ras signaling pathways is relevant to the pathogenesis of a wide variety of cancers, it is important to clearly define the structural and functional characteristics of the participating proteins and their encoding genes. Rho subfamily members are involved in a range of signal transduction pathways relevant to cell growth, differentiation, motility, and stress, and Rac proteins are now recognised as a necessary component of Ras-mediated cellular transformation. We previously mapped RAC3 to chromosome band 17q23--> q25, a region that contains a number of candidate tumour suppressor genes. Because of its oncogenic potential, we have now further refined the location of this gene. Here we confirm that RAC3 maps to chromosome band 17q25.3 and further show that it maps some distance telomeric of a well-characterised minimal breast and ovarian candidate tumour suppressor gene region, BROV. The genomic structure of RAC3, including exon and intron boundaries, is also presented.


Subject(s)
Breast Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Genetic Linkage/genetics , Ovarian Neoplasms/genetics , rac GTP-Binding Proteins/genetics , Amino Acid Sequence , Base Sequence , Chromosome Banding , Chromosomes, Artificial, Yeast/genetics , Cloning, Molecular , Exons/genetics , Female , Genes, Tumor Suppressor/genetics , Humans , In Situ Hybridization, Fluorescence , Introns/genetics , Molecular Sequence Data , Physical Chromosome Mapping , Telomere/genetics , rac GTP-Binding Proteins/chemistry
20.
Cell Tissue Res ; 300(1): 89-95, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10805078

ABSTRACT

After cessation of lactation, the mammary gland undergoes involution, which is characterized by a massive epithelial cell death and proteolytic degradation of the extracellular matrix. Whereas the expression patterns and also the function of TGF-beta isoforms during mammary gland branching morphogenesis and lactation are well understood, their expression during postlactational involution and therefore a possible role in this process is poorly known. In this study we show that TGF-beta3 expression is dramatically induced (>fivefold) during mouse mammary gland involution when compared to that of virgin mouse, reaching a maximal expression level at day 4 after weaning. In contrast, other TGF-beta isoforms do not display significant increase in expression during involution (TGF-beta1, 1.3-fold and TGF-beta2, <1.5-fold) when compared to that of virgin or lactating mice. During mammary gland involution, TGF-beta3 is expressed in the epithelial layer and particularly in myoepithelial cells. A comparison of the kinetics of TGF-beta3 expression to that of programmed cell death and degradation of the basement membrane suggests that TGF-beta3 functions in the remodeling events of the extracellular matrix during the second stage of involution.


Subject(s)
Lactation/physiology , Mammary Glands, Animal/physiology , Transforming Growth Factor beta/genetics , Animals , Apoptosis/physiology , Basement Membrane/cytology , Basement Membrane/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression/physiology , Mice , Mice, Inbred CBA , RNA, Messenger/analysis , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...