Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Mol Neurobiol ; 56(6): 4566-4581, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30353492

ABSTRACT

The role of astrocytes is becoming increasingly important to understanding how glioblastoma (GBM) tumor cells diffusely invade the brain. Yet, little is known of the contribution of extracellular vesicle (EV) signaling in GBM/astrocyte interactions. We modeled GBM-EV signaling to normal astrocytes in vitro to assess whether this mode of intercellular communication could support GBM progression. EVs were isolated and characterized from three patient-derived GBM stem cells (NES+/CD133+) and their differentiated (diff) progeny cells (NES-/CD133-). Uptake of GBM-EVs by normal primary astrocytes was confirmed by fluorescence microscopy, and changes in astrocyte podosome formation and gelatin degradation were measured. Quantitative mass spectrometry-based proteomics was performed on GBM-EV stimulated astrocytes. Interaction networks were generated from common, differentially abundant proteins using Ingenuity® (Qiagen Bioinformatics) and predicted upstream regulators were tested by qPCR assays. Podosome formation and Cy3-gelatin degradation were induced in astrocytes following 24-h exposure to GBM-stem and -diff EVs, with EVs released by GBM-stem cells eliciting a greater effect. More than 1700 proteins were quantified, and bioinformatics predicted activations of MYC, NFE2L2, FN1, and TGFß1 and inhibition of TP53 in GBM-EV stimulated astrocytes that were then confirmed by qPCR. Further qPCR studies identified significantly decreased Δ133p53 and increased p53ß in astrocytes exposed to GBM-EVs that might indicate the acquisition of a pro-inflammatory, tumor-promoting senescence-associated secretory phenotype (SASP). Inhibition of TP53 and activation of MYC signaling pathways in normal astrocytes exposed to GBM-EVs may be a mechanism by which GBM manipulates astrocytes to acquire a phenotype that promotes tumor progression.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/metabolism , Extracellular Vesicles/metabolism , Glioblastoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Aged , Cell Differentiation , Cell Line, Tumor , Cellular Senescence , Extracellular Vesicles/ultrastructure , Gelatin/metabolism , Humans , Male , Middle Aged , Nanoparticles/ultrastructure , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Particle Size , Phenotype , Podosomes/metabolism , Protein Isoforms/metabolism , Proteolysis , Proteome/metabolism
2.
Sci Rep ; 8(1): 11393, 2018 07 30.
Article in English | MEDLINE | ID: mdl-30061574

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an untreatable, progressive, neurodegenerative disease specifically affecting motor neurons. Recently, the tyrosine kinase receptor EphA4 was directly implicated in ALS disease progression. We report that a long-lived mutated form of the EphA4 antagonist EphA4-Fc (mutEphA4-Fc), which blocks EphA4 binding to its ligands and inhibits its function, significantly improved functional performance in SOD1G93A ALS model mice, as assessed by rotarod and hind-limb grip strength tests. Further, heterozygous motor neuron-specific EphA4 gene deletion in SOD1G93A mice promoted significant improvement in functional performance during the disease course and a delay in disease onset relative to control mice. Importantly, mice in the heterozygous deletion group showed significantly improved survival of motor neurons and architecture of endplates of neuromuscular junctions compared with control and homozygous EphA4-deletion groups. Our novel results show that EphA4 signalling directly regulates motor neuron survival and that mutEphA4-Fc is a promising therapeutic candidate to slow disease progression in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Motor Neurons/metabolism , Motor Neurons/pathology , Receptor, EphA4/metabolism , Signal Transduction , Superoxide Dismutase-1/metabolism , Animals , Cell Survival , Disease Models, Animal , Heterozygote , Mice, Knockout , Mutation/genetics
3.
Leukemia ; 31(8): 1779-1787, 2017 08.
Article in English | MEDLINE | ID: mdl-27922598

ABSTRACT

The human EphA3 gene was discovered in a pre-B acute lymphoblastic leukemia (pre-B-ALL) using the EphA3-specific monoclonal antibody (mAb), IIIA4, which binds and activates both human and mouse EphA3. We use two models of human pre-B-ALL to examine EphA3 function, demonstrating effects on pre-B-cell receptor signaling. In therapeutic targeting studies, we demonstrated antitumor effects of the IIIA4 mAb in EphA3-expressing leukemic xenografts and no antitumor effect in the xenografts with no EphA3 expression providing evidence that EphA3 is a functional therapeutic target in pre-B-ALL. Here we show that the therapeutic effect of the anti-EphA3 antibody was greatly enhanced by adding an α-particle-emitting 213Bismuth payload.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptor, EphA3/immunology , Animals , Bismuth , Cell Line, Tumor , Humans , Immunotherapy , Mice , Receptor, EphA3/metabolism , Xenograft Model Antitumor Assays
4.
Br J Cancer ; 114(2): 188-98, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26657652

ABSTRACT

BACKGROUND: Resistance to temozolomide (TMZ) greatly limits chemotherapeutic effectiveness in glioblastoma (GBM). Here we analysed the ability of the Inhibitor-of-apoptosis-protein (IAP) antagonist birinapant to enhance treatment responses to TMZ in both commercially available and patient-derived GBM cells. METHODS: Responses to TMZ and birinapant were analysed in a panel of commercial and patient-derived GBM cell lines using colorimetric viability assays, flow cytometry, morphological analysis and protein expression profiling of pro- and antiapoptotic proteins. Responses in vivo were analysed in an orthotopic xenograft GBM model. RESULTS: Single-agent treatment experiments categorised GBM cells into TMZ-sensitive cells, birinapant-sensitive cells, and cells that were insensitive to either treatment. Combination treatment allowed sensitisation to therapy in only a subset of resistant GBM cells. Cell death analysis identified three principal response patterns: Type A cells that readily activated caspase-8 and cell death in response to TMZ while addition of birinapant further sensitised the cells to TMZ-induced cell death; Type B cells that readily activated caspase-8 and cell death in response to birinapant but did not show further sensitisation with TMZ; and Type C cells that showed no significant cell death or moderately enhanced cell death in the combined treatment paradigm. Furthermore, in vivo, a Type C patient-derived cell line that was TMZ-insensitive in vitro and showed a strong sensitivity to TMZ and TMZ plus birinapant treatments. CONCLUSIONS: Our results demonstrate remarkable differences in responses of patient-derived GBM cells to birinapant single and combination treatments, and suggest that therapeutic responses in vivo may be greatly affected by the tumour microenvironment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Dipeptides/pharmacology , Glioblastoma/pathology , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Animals , Blotting, Western , Caspase 8/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/drug effects , Flow Cytometry , Humans , In Vitro Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Phase-Contrast , Neoplasm Transplantation , Temozolomide , Xenograft Model Antitumor Assays
6.
Br J Cancer ; 111(7): 1255-61, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25144626

ABSTRACT

The dismal outlook for patients with the most aggressive and common form of adult brain cancer, glioblastoma (GBM), motivates a search for new therapeutic strategies and targets for this aggressive disease. Here we review the findings to date on the role of Eph family receptor tyrosine kinases and their ephrin ligands in brain cancer. Expression of the Eph family of cell surface proteins is generally downregulated to very low levels in normal adult tissues making them particularly attractive for directed therapeutic targeting. Recent Eph targeting studies in pre-clinical models of GBM have been very encouraging and may provide an avenue to treat these highly refractory aggressive tumours.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Receptors, Eph Family/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Ephrins/physiology , Glioblastoma/drug therapy , Humans , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Signal Transduction
7.
Leukemia ; 26(5): 918-26, 2012 May.
Article in English | MEDLINE | ID: mdl-22116550

ABSTRACT

Improved survival of patients with acute lymphoblastic leukemia (ALL) has emerged from identifying new prognostic markers; however, 20% of children still suffer recurrence. Previously, the altered expression of Fat1 cadherin has been implicated in a number of solid tumors. In this report, in vitro analysis shows that Fat1 protein is expressed by a range of leukemia cell lines, but not by normal peripheral blood (PB) and bone marrow (BM) cells from healthy donors. In silico analysis of expression of array data from clinical leukemias found significant levels of Fat1 transcript in 11% of acute myeloid leukemia, 29% and 63% of ALL of B and T lineages, respectively, and little or no transcript present in normal PB or BM. Furthermore, in two independent studies of matched diagnosis-relapse of precursor B-cell (preB) ALL pediatric samples (n=32 and n=27), the level of Fat1 mRNA expression was prognostic at the time of diagnosis. High Fat1 mRNA expression was predictive of shorter relapse-free and overall survival, independent of other traditional prognostic markers, including white blood cell count, sex and age. The data presented demonstrate that Fat1 expression in preB-ALL has a role in the emergence of relapse and could provide a suitable therapeutic target in high-risk preB-ALL.


Subject(s)
Cadherins/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Cadherins/genetics , Child , Genes, Tumor Suppressor , Humans , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Recurrence , Survival Analysis
8.
Br J Cancer ; 100(7): 1095-102, 2009 Apr 07.
Article in English | MEDLINE | ID: mdl-19277044

ABSTRACT

Aberrant expression of Eph and ephrin proteins has well-established functions in oncogenesis and tumour progression. We describe EphA1 expression in 6 colorectal cancer (CRC) cell lines, 18 controls and 125 CRC specimens. In addition, a well-characterised cohort of 53 paired normal colon and CRCs was also assessed. Expression of EphA1 mRNA was assessed by quantitative real-time PCR and correlated with protein expression by flow cytometry, immunoprecipitation, western blotting and immunohistochemistry. Significant upregulation (2- to 10-fold) of EphA1 was seen in over 50% of cases (P=0.005) whereas many of the remainder showed downregulation of EphA1. Intriguingly, EphA1 over-expression was more prevalent in stage II compared to stage III CRCs (P=0.02). Low EphA1 expression significantly correlated with poor survival (P=0.02). Epigenetic silencing appeared to explain the loss of EphA1 expression as methylation of the EphA1 CpG island strongly correlated with low EphA1 expression (P<0.01). Furthermore, EphA1 re-expression could be induced by treatment with demethylating agents. Our findings identify EphA1 as a potential prognostic marker in CRC. Although therapies targeting high EphA1 expression seem plausible in CRC, the loss of expression in advanced disease suggests a potential risk that targeted therapy, by selecting for loss of expression, might contribute to disease progression.


Subject(s)
Colorectal Neoplasms/genetics , Epigenesis, Genetic , Gene Silencing , Receptor, EphA1/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Colon/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/mortality , CpG Islands , DNA Methylation , Decitabine , Humans , Immunohistochemistry , Polymerase Chain Reaction , Promoter Regions, Genetic , Receptor, EphA1/analysis
9.
Br J Cancer ; 92(4): 760-9, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15685234

ABSTRACT

The Testisin gene (PRSS21) encodes a glycosylphosphatidylinositol (GPI)-linked serine protease that exhibits testis tissue-specific expression. Loss of Testisin has been implicated in testicular tumorigenesis, but its role in testis biology and tumorigenesis is not known. Here we have investigated the role of CpG methylation in Testisin gene inactivation and tested the hypothesis that Testisin may act as a tumour suppressor for testicular tumorigenesis. Using sequence analysis of bisulphite-treated genomic DNA, we find a strong relationship between hypermethylation of a 385 bp 5' CpG rich island of the Testisin gene, and silencing of the Testisin gene in a range of human tumour cell lines and in 100% (eight/eight) of testicular germ cell tumours. We show that treatment of Testisin-negative cell lines with demethylating agents and/or a histone deacetylase inhibitor results in reactivation of Testisin gene expression, implicating hypermethylation in Testisin gene silencing. Stable expression of Testisin in the Testisin-negative Tera-2 testicular cancer line suppressed tumorigenicity as revealed by inhibition of both anchorage-dependent cell growth and tumour formation in an SCID mouse model of testicular tumorigenesis. Together, these data show that loss of Testisin is caused, at least in part, by DNA hypermethylation and histone deacetylation, and suggest a tumour suppressor role for Testisin in testicular tumorigenesis.


Subject(s)
CpG Islands , DNA Methylation , DNA, Neoplasm/metabolism , Gene Silencing , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Testicular Neoplasms/metabolism , Adult , Animals , Cell Line, Tumor , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors , Humans , Immunohistochemistry , Male , Membrane Proteins , Mice , Mice, SCID , Orchiectomy , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Testicular Neoplasms/genetics , Testicular Neoplasms/surgery , Transplantation, Heterologous
10.
Sci STKE ; 2001(112): re20, 2001 Dec 11.
Article in English | MEDLINE | ID: mdl-11741094

ABSTRACT

Interactions between Eph receptors and their ligands the ephrin proteins are critically important in many key developmental processes. Emerging evidence also supports a role for these molecules in postembryonic tissues, particularly in pathological processes, including tissue injury and tumor metastasis. We review the signaling mechanisms that allow the 14 Eph and nine ephrin proteins to deliver intracellular signals that regulate cell shape and movement. What emerges is that the initiation of these signals is critically dependent on which Eph and ephrin proteins are expressed, the level of their expression, and, in some cases, which splice variants are expressed. Diversity at the level of initial interaction and in the downstream signaling processes regulated by Eph-ephrin signaling provides a subtle, versatile system of regulation of intercellular adhesion, cell shape, and cell motility.


Subject(s)
Gene Expression Regulation, Developmental , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Animals , Gene Expression Regulation, Developmental/physiology , Humans , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics
11.
Immunol Cell Biol ; 79(5): 419-28, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564149

ABSTRACT

The contribution of synovial cells to the pathogenesis of rheumatoid arthritis (RA) is only partly understood. Monoclonal antibody (mAb) 1D5 is one of very few mAb ever raised against RA synovial cells in order to study the biology of these cells. Studies on the expression pattern and structural features of the 1D5 Ag suggest that 1D5 recognizes human vascular cell adhesion molecule-1 (VCAM-1), which is an intercellular adhesion molecule. Vascular cell adhesion molecule-1 may be involved in a number of crucial intercellular interactions in RA.


Subject(s)
Antibodies, Monoclonal/immunology , Arthritis, Rheumatoid/immunology , Synovial Membrane/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Antibodies, Monoclonal/isolation & purification , Antigens, Surface/chemistry , Antigens, Surface/immunology , Antigens, Surface/metabolism , Arthritis, Rheumatoid/physiopathology , Cell Line , Flow Cytometry , Humans , Intercellular Adhesion Molecule-1/immunology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Synovial Membrane/cytology , Synovial Membrane/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Tissue Extracts , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/chemistry , Vascular Cell Adhesion Molecule-1/immunology
12.
Exp Hematol ; 29(9): 1109-16, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11532352

ABSTRACT

OBJECTIVE: The aim of this study was to determine the function of primitive hematopoietic stem cells (PHSC) at phases G(0) and G(1) of the cell cycle. MATERIALS AND METHODS: A combination of supravital dyes rhodamine123 (Rh), Hoechst33342 (Ho), and pyronin (PY) was used to isolate the G(0) and G(1) subsets of PHSC. A competitive repopulation assay was used to evaluate their in vivo function. RESULTS: We confirmed that the Rh(lo)Lin(-)Kit(+)Sca-1(+) PHSC were relatively quiescent when compared with the more mature Rh(hi)Lin(-)Kit(+)Sca-1(+) HSC and Rh(hi)Lin(-)Kit(+)Sca-1(-) progenitors. In addition, cells with Rh(lo)Lin(-)Kit(+)Sca-1(+), Rh(lo)Ho(lo)Lin(-)Sca-1(+), or Rh(lo)Ho(sp)Lin(-)Sca-1(+) phenotypes identified the same cell population. We further subfractionated the Rh(lo)Ho(lo/sp)Lin(-)Sca-1(+) PHSC using PY into PY(lo) and PY(hi) subsets. Limiting dilution analysis revealed that the frequency of long-term in vivo competitive repopulating units (CRU) of the PY(lo)Rh(lo)Ho(lo/sp) PHSC was 1 in 10 cells, whereas there was at least a three-fold lower frequency in those isolated at the G(1) phase (PY(hi)). We found a dose-dependent PY-mediated cytotoxicity that at moderate concentration affected most of the murine hematopoietic compartment but spared the early HSC compartment. CONCLUSION: Our data confirm that the HSC compartment is hierarchically ordered on the basis of quiescence and further extend this concept to PY-mediated cytotoxicity. PY supravital dye can be used to reveal functional heterogeneity within the Rh(lo)Ho(lo/sp) PHSC population but is of limited use in dissecting the relatively more mature hematopoietic stem/progenitor cell population.


Subject(s)
Benzimidazoles , Fluorescent Dyes , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Pyronine , Rhodamine 123 , Animals , Bone Marrow Cells/cytology , Cell Death/drug effects , Cell Death/radiation effects , Flow Cytometry , G1 Phase/physiology , Hematopoiesis , Immunophenotyping , Mice , Mice, Inbred C57BL , Photosensitizing Agents/pharmacology , Pyronine/pharmacology , Resting Phase, Cell Cycle/physiology
13.
Growth Factors ; 18(4): 303-17, 2001.
Article in English | MEDLINE | ID: mdl-11519828

ABSTRACT

The Eph family of receptor tyrosine kinases plays a crucial role during development and is implicated in oncogenesis. Using a partial cDNA clone of an Eph-related kinase (Esk) we isolated the complete coding region of a gene which we show to be murine EphA1 by both structural and functional criteria. The chromosomal localization is shown to be syntenic to hEphA1 and the genomic organization also shows distinct features found in the hEphA1 gene. Functionally, in keeping with findings for the human homologue, both soluble recombinant and "native" mEphA1 show preferential binding to ephrin A1. However, we also observed significant binding to other A-type ligands as has been observed for other Eph receptors. We analysed the expression of mEphA1 mRNA by in situ hybridization on tissue sections. mEphA1 was expressed in epithelial elements of skin, adult thymus, kidney and adrenal cortex. Taken together with previous Northern blotting data these results suggest that mEphA1 is expressed widely in differentiated epithelial cells.


Subject(s)
Receptor Protein-Tyrosine Kinases/genetics , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cloning, Molecular , Cricetinae , DNA, Complementary/genetics , Ephrin-A1 , Epithelium/enzymology , Gene Expression , Humans , In Situ Hybridization , Mice , Molecular Sequence Data , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA1 , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility , Species Specificity
14.
J Comp Neurol ; 436(2): 248-62, 2001 Jul 23.
Article in English | MEDLINE | ID: mdl-11438928

ABSTRACT

The Eph family of receptor tyrosine kinases and their ligands, the ephrins, are important regulators of axon guidance and cell migration in the developing nervous system. Inactivation of the EphA4 gene results in axon guidance defects of the corticospinal tract, a major descending motor pathway that originates in the cortex and terminates at all levels of the spinal cord. In this investigation, we report that although the initial development of the corticospinal projection is normal through the cortex, internal capsule, cerebral peduncle, and medulla in the brain of EphA4 deficient animals, corticospinal axons exhibit gross abnormalities when they enter the gray matter of the spinal cord. Notably, many corticospinal axons fail to remain confined to one side of the spinal cord during development and instead, aberrantly project across the midline, terminating ipsilateral to their cells of origin. Given the possible repulsive interactions between EphA4 and one of its ligands, ephrinB3, this defect could be consistent with a loss of responsiveness by corticospinal axons to ephrinB3 that is expressed at the spinal cord midline. Furthermore, we show that EphA4 deficient animals exhibit ventral displacement of the mature corticospinal termination pattern, suggesting that developing corticospinal axons, which may also express ephrinB3, fail to be repelled from areas of high EphA4 expression in the intermediate zone of the normal spinal cord. Taken together, these results suggest that the dual expression of EphA4 on corticospinal axons and also within the surrounding gray matter is very important for the correct development and termination of the corticospinal projection within the spinal cord.


Subject(s)
Biotin/analogs & derivatives , Body Patterning/genetics , Cell Differentiation/genetics , Fetal Proteins/deficiency , Growth Cones/metabolism , Neuronal Plasticity/genetics , Pyramidal Tracts/abnormalities , Receptor Protein-Tyrosine Kinases/deficiency , Age Factors , Animals , Biotin/pharmacokinetics , Carbocyanines/pharmacokinetics , Dextrans/pharmacokinetics , Ephrin-B3 , Fetal Proteins/genetics , Fetal Proteins/metabolism , Fetus , Fluorescent Dyes/pharmacokinetics , Functional Laterality/genetics , Growth Cones/ultrastructure , Immunohistochemistry , Medulla Oblongata/abnormalities , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Pyramidal Tracts/cytology , Pyramidal Tracts/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA4
15.
Neuron ; 29(1): 73-84, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11182082

ABSTRACT

The EphA4 receptor tyrosine kinase regulates the formation of the corticospinal tract (CST), a pathway controlling voluntary movements, and of the anterior commissure (AC), connecting the neocortical temporal lobes. To study EphA4 kinase signaling in these processes, we generated mice expressing mutant EphA4 receptors either lacking kinase activity or with severely downregulated kinase activity. We demonstrate that EphA4 is required for CST formation as a receptor for which it requires an active kinase domain. In contrast, the formation of the AC is rescued by kinase-dead EphA4, suggesting that in this structure EphA4 acts as a ligand for which its kinase activity is not required. Unexpectedly, the cytoplasmic sterile-alpha motif (SAM) domain is not required for EphA4 functions. Our findings establish both kinase-dependent and kinase-independent functions of EphA4 in the formation of major axon tracts.


Subject(s)
Axons/enzymology , Fetal Proteins/metabolism , Pyramidal Tracts/embryology , Pyramidal Tracts/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Brain Stem/cytology , Brain Stem/embryology , Brain Stem/enzymology , Ephrin-A4 , Ephrin-B2 , Fetal Proteins/deficiency , Fetal Proteins/genetics , In Situ Hybridization , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , Molecular Sequence Data , Motor Cortex/cytology , Motor Cortex/embryology , Motor Cortex/enzymology , Organ Specificity , Prosencephalon/cytology , Prosencephalon/embryology , Prosencephalon/enzymology , Protein Structure, Tertiary/genetics , Pyramidal Tracts/cytology , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphA4 , Signal Transduction/genetics , Temporal Lobe/cytology , Temporal Lobe/embryology
16.
Growth Factors ; 17(4): 287-300, 2000.
Article in English | MEDLINE | ID: mdl-10801077

ABSTRACT

PURPOSE: To investigate the pharmacokinetics and tolerability of recombinant human interleukin-4 (rhuIL-4), administered by daily subcutaneous injection, in patients with advanced cancer. PATIENTS AND METHODS: Fourteen patients with advanced cancer treated with rhuIL-4 at escalating dose levels of 0.25, 1.0 and 5.0 microg/kg/day, on days 1, 8-17, and 28-57. The primary endpoints of the study were toxicity of rhuIL-4 and the determination of the pharmacokinetics of rhuIL-4 when given by subcutaneous injection. Secondary endpoints included effects on blood counts, hematopoietic cell precursors, and various immunologic parameters. RESULTS: rhuIL-4 was well tolerated at all three dose levels. Detectable serum levels of IL-4 were found in patients at the 1.0 and 5.0 microg/kg/day dose levels. Peak serum IL-4 levels were achieved about 2 h after injection and IL-4 was still detectable 8 h after injection. No grade 4 toxicities were observed and grade 3 toxicities were confined to fever, headache and raised hepatic alkaline phosphatase. No consistent hematological or immunologic effects were observed. Although therapeutic efficacy was not an endpoint, one complete response (Hodgkin's disease) was observed. One patient with chronic lymphocytic leukemia progressed on therapy. CONCLUSION: rhuIL-4 up to 5.0 microg/kg/day is well tolerated when given by subcutaneous injection. Biologically relevant serum IL-4 levels can be achieved and sustained for at least 8 h after a single injection.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Interleukin-4/pharmacokinetics , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cytotoxicity, Immunologic , Escherichia coli/genetics , Female , Humans , Injections, Subcutaneous , Interleukin-4/adverse effects , Interleukin-4/chemistry , Interleukin-4/pharmacology , Male , Middle Aged , Neoplasms/immunology , Recombinant Proteins/adverse effects , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
17.
Cancer Res ; 60(4): 808-14, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706085

ABSTRACT

The Wilms' tumor suppressor gene, WT1, encodes a transcription of the Cys2-His2 zinc finger type. Loss of WT1 gene function has been implicated in the development of malignancies including Wilms' tumor and acute leukemias. We have shown previously that ectopic expression of WT1 +KTS isoforms in murine M1 leukemic cells spontaneously induces monocytic differentiation without the requirement for external differentiation-inducing stimuli. To determine whether these observed effects in vitro corresponded to a reduction in tumorigenicity in vivo, parental M1, control M1.Neo, and M1.WT1 +KTS cells were transplanted into C.B-17 scid/scid mice, and the growth and metastatic behavior of the cell lines were monitored for a period of 20 weeks. Mice inoculated either s.c. on the flank or directly into the peritoneal cavity, with M1 cells stably expressing WT1 +KTS isoforms exhibited a marked decrease in tumor formation compared with control groups. Moreover, tumors arising in mice after the injection of M1.WT1 +KTS cells exhibited a loss in ectopic WT1 protein expression. Confirmation that the tumors arose from M1.WT1 +KTS cells was achieved by the amplification of the introduced transgene from tumor samples and indicates that the tumorigenicity of leukemic M1 cells in these animals correlates with a loss in WT1 expression. This investigation is the first to demonstrate the tumor-suppressive effects of WT1 expression in a leukemic cell line, further advancing the notion that WT1 acts as a differentiation-promoting gene during hematopoiesis and that loss of functional WT1 expression may contribute to leukemogenesis in vivo.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Tumor Suppressor/physiology , Leukemia, Experimental/prevention & control , Transcription Factors/genetics , Animals , Female , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Tumor Cells, Cultured , WT1 Proteins
18.
Blood ; 94(7): 2477-86, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10498621

ABSTRACT

The Eph family of receptor tyrosine kinases (RTK) has restricted temporal and spatial expression patterns during development, and several members are also found to be upregulated in tumors. Very little is known of the promoter elements or regulatory factors required for expression of Eph RTK genes. In this report we describe the identification and characterization of the EphA3 gene promoter region. A region of 86 bp located at -348 bp to -262 bp upstream from the transcription start site was identified as the basal promoter. This region was shown to be active in both EphA3-expressing and -nonexpressing cell lines, contrasting with the widely different levels of EphA3 expression. We noted a region rich in CpG dinucleotides downstream of the basal promoter. Using Southern blot analyses with methylation-sensitive restriction enzymes and bisulfite sequencing of genomic DNA, sites of DNA methylation were identified in hematopoietic cell lines which correlated with their levels of EphA3 gene expression. We showed that EphA3 was not methylated in normal tissues but that a subset of clinical samples from leukemia patients showed extensive methylation, similar to that observed in cell lines. These results suggest that DNA methylation may be an important mechanism regulating EphA3 transcription in hematopoietic tumors.


Subject(s)
Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/metabolism , Leukemia/genetics , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/genetics , 5' Untranslated Regions/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA Methylation , Dinucleoside Phosphates/metabolism , Exons , Female , Fetal Blood/metabolism , Humans , Molecular Sequence Data , Pancreas/metabolism , Placenta/metabolism , Polymerase Chain Reaction , Pregnancy , Receptor, EphA3 , Tumor Cells, Cultured
19.
Mech Dev ; 83(1-2): 77-94, 1999 May.
Article in English | MEDLINE | ID: mdl-10381569

ABSTRACT

Eph receptor tyrosine kinases (RTK) and their ephrin ligands are involved in the transmission of signals which regulate cytoskeletal organisation and cell migration, and are expressed in spatially restricted patterns at discrete phases during embryogenesis. Loss of function mutants of Eph RTK or ephrin genes result in defects in neuronal pathfinding or cell migration. In this report we show that soluble forms of human EphA3 and ephrin-A5, acting as dominant negative inhibitors, interfere with early events in zebrafish embryogenesis. Exogenous expression of both proteins results in dose-dependent defects in somite development and organisation of the midbrain-hindbrain boundary and hindbrain. The nature of the defects as well as the distribution and timing of expression of endogenous ligands/receptors for both proteins suggest that Eph-ephrin interaction is required for the organisation of embryonic structures by coordinating the cellular movements of convergence during gastrulation.


Subject(s)
Gastrula/metabolism , Membrane Proteins/metabolism , Multigene Family/physiology , Proteins/metabolism , Animals , Cell Movement , Dose-Response Relationship, Drug , Embryo, Nonmammalian/anatomy & histology , Ephrin-A1 , Ephrin-A3 , Ephrin-A5 , Ephrin-B1 , Gene Expression Regulation, Developmental , Genes, Dominant , Humans , Kinetics , Membrane Proteins/analysis , RNA, Messenger/pharmacology , Time Factors , Zebrafish/embryology
20.
Proc Natl Acad Sci U S A ; 95(22): 13248-53, 1998 Oct 27.
Article in English | MEDLINE | ID: mdl-9789074

ABSTRACT

Members of the Eph family of tyrosine kinase receptors have been implicated in the regulation of developmental processes and, in particular, axon guidance in the developing nervous system. The function of the EphA4 (Sek1) receptor was explored through creation of a null mutant mouse. Mice with a null mutation in the EphA4 gene are viable and fertile but have a gross motor dysfunction, which is evidenced by a loss of coordination of limb movement and a resultant hopping, kangaroo-like gait. Consistent with the observed phenotype, anatomical studies and anterograde tracing experiments reveal major disruptions of the corticospinal tract within the medulla and spinal cord in the null mutant animals. These results demonstrate a critical role for EphA4 in establishing the corticospinal projection.


Subject(s)
Fetal Proteins/genetics , Fetal Proteins/physiology , Gene Expression Regulation, Developmental , Medulla Oblongata/embryology , Movement Disorders/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/physiology , Spinal Cord/embryology , Animals , Base Sequence , Gait , Genotype , Homozygote , Medulla Oblongata/physiopathology , Mice , Mice, Knockout , Molecular Sequence Data , Movement Disorders/enzymology , Nerve Fibers/pathology , Nerve Fibers/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Pathways/embryology , Neural Pathways/physiopathology , Polymerase Chain Reaction , Receptor, EphA4 , Recombination, Genetic , Restriction Mapping , Spinal Cord/physiopathology , Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL