Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 281
Filter
Add more filters










Publication year range
1.
J Intern Med ; 283(1): 16-44, 2018 01.
Article in English | MEDLINE | ID: mdl-28940884

ABSTRACT

Platelet-derived growth factor (PDGF) isoforms and their receptors have important roles during embryogenesis, particularly in the development of various mesenchymal cell types in different organs. In the adult, PDGF stimulates wound healing and regulates tissue homeostasis. However, overactivity of PDGF signalling is associated with malignancies and other diseases characterized by excessive cell proliferation, such as fibrotic conditions and atherosclerosis. In certain tumours, genetic or epigenetic alterations of the genes for PDGF ligands and receptors drive tumour cell proliferation and survival. Examples include the rare skin tumour dermatofibrosarcoma protuberance, which is driven by autocrine PDGF stimulation due to translocation of a PDGF gene, and certain gastrointestinal stromal tumours and leukaemias, which are driven by constitute activation of PDGF receptors due to point mutations and formation of fusion proteins of the receptors, respectively. Moreover, PDGF stimulates cells in tumour stroma and promotes angiogenesis as well as the development of cancer-associated fibroblasts, both of which promote tumour progression. Inhibitors of PDGF signalling may thus be of clinical usefulness in the treatment of certain tumours.


Subject(s)
Carcinogenesis , Neoplasms , Neovascularization, Pathologic/metabolism , Platelet-Derived Growth Factor , Receptors, Platelet-Derived Growth Factor , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction
3.
Oncogene ; 32(47): 5409-20, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-23524585

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive brain malignancy characterized by high heterogeneity and invasiveness. It is increasingly accepted that the refractory feature of GBM to current therapies stems from the existence of few tumorigenic cells that sustain tumor growth and spreading, the so-called glioma-initiating cells (GICs). Previous studies showed that cytokines of the bone morphogenetic protein (BMP) family induce differentiation of the GICs, and thus act as tumor suppressors. Molecular pathways that explain this behavior of BMP cytokines remain largely elusive. Here, we show that BMP signaling induces Smad-dependent expression of the transcriptional regulator Snail in a rapid and sustained manner. Consistent with its already established promigratory function in other cell types, we report that Snail silencing decreases GBM cell migration. Consequently, overexpression of Snail increases GBM invasiveness in a mouse xenograft model. Surprisingly, we found that Snail depletes the GBM capacity to form gliomaspheres in vitro and to grow tumors in vivo, both of which are important features shared by GICs. Thus Snail, acting downstream of BMP signaling, dissociates the invasive capacity of GBM cells from their tumorigenic potential.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Neoplastic Stem Cells/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics , Smad1 Protein/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Smad4 Protein/genetics , Smad5 Protein/genetics , Snail Family Transcription Factors , Transcription Factors/genetics
4.
Oncogene ; 28(35): 3121-31, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19543319

ABSTRACT

Earlier we used a glioma model to identify loci in the mouse genome, which were repeatedly targeted by platelet-derived growth factor (PDGF)-containing Moloney murine leukemia viruses. The gene Prkg2, encoding cyclic guanosine monophosphate (cGMP)-dependent protein kinase II, cGKII, was tagged by retroviral insertions in two brain tumors. The insertions were both situated upstream of the kinase domain and suggested creating a truncated form of the cGKII protein. We transfected different human glioma cell lines with Prkg2 and found an overall reduction in colony formation and cell proliferation compared with controls transfected with truncated Prkg2 (lacking the kinase domain) or empty vector. All glioma cells transfected with the cGKII phosphorylate vasodilator-stimulated phosphoprotein, VASP, after cGMP analog treatment. Glioma cell lines positive for the Sox9 transcription factor showed reduced Sox9 expression when Prkg2 was stably transfected. When cGKII was activated by cGMP analog treatment, Sox9 was phosphorylated, Sox9 protein expression was suppressed and the glioma cell lines displayed loss of cell adhesion, inhibition of Akt phosphorylation and G1 arrest. Sox9 repression by siRNA was similarly shown to reduce glioma cell proliferation. Expression analysis of stem and glial lineage cell markers also suggests that cGKII induces differentiation of glioma cell lines. These findings describe an anti-proliferative role of cGKII in human glioma biology and would further explain the retroviral tagging of the cGKII gene during brain tumor formation in PDGF-induced tumors.


Subject(s)
Cell Proliferation , Cyclic GMP-Dependent Protein Kinases/physiology , Glioma/pathology , Proto-Oncogene Proteins c-akt/metabolism , SOX9 Transcription Factor/metabolism , Cell Adhesion/physiology , Cell Culture Techniques , Cell Line, Tumor , Culture Media, Serum-Free , Cyclic GMP-Dependent Protein Kinase Type II , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Genetic Vectors , Humans , Phosphorylation , Retroviridae/genetics , SOX9 Transcription Factor/genetics , Time Factors , Transfection
5.
Oncogene ; 28(12): 1537-48, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19219070

ABSTRACT

SOX5 is a member of the high-mobility group superfamily of architectural non-histone proteins involved in gene regulation and maintenance of chromatin structure in a wide variety of developmental processes. Sox5 was identified as a brain tumor locus in a retroviral insertional mutagenesis screen of platelet-derived growth factor B (PDGFB)-induced mouse gliomas. Here we have investigated the role of Sox5 in PDGFB-induced gliomagenesis in mice. We show that Sox5 can suppress PDGFB-induced glioma development predominantly upon Ink4a-loss. In human glioma cell lines and tissues, we found very low levels of SOX5 compared with normal brain. Overexpression of Sox5 in human glioma cells led to a reduction in clone formation and inhibition of proliferation. Combined expression of Sox5 and PDGFB in primary brain cell cultures caused decreased proliferation and an increased number of senescent cells in the Ink4a-/- cells only. Protein analyses showed a reduction in the amount and activation of Akt and increased levels of p27(Kip1) upon Sox5 expression that was dominant to PDGFB signaling and specific to Ink4a-/- cells. Upon inhibition of p27(Kip1), the effects of Sox5 on proliferation and senescence could be reversed. Our data suggest a novel pathway, where Sox5 may suppress the oncogenic effects of PDGFB signaling during glioma development by regulating p27(Kip1) in a p19(Arf)-dependent manner, leading to acute cellular senescence.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/physiology , Glioma/prevention & control , Proto-Oncogene Proteins c-sis/physiology , SOXD Transcription Factors/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p27/physiology , Glioma/pathology , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mice , Proto-Oncogene Proteins c-sis/antagonists & inhibitors , Tumor Cells, Cultured
6.
Oncogene ; 26(43): 6289-96, 2007 Sep 20.
Article in English | MEDLINE | ID: mdl-17438529

ABSTRACT

In a subset of gliomas, the platelet-derived growth factor (PDGF) signaling pathway is perturbed. This is usually an early event occurring in low-grade tumors. In high-grade gliomas, the subsequent loss of the INK4a-ARF locus is one of the most common mutations. Here, we dissected the separate roles of Ink4a and Arf in PDGFB-induced oligodendroglioma development in mice. We found that there were differential functions of the two tumor suppressor genes. In tumors induced from astrocytes, both Ink4a-loss and Arf-loss caused a significantly increased incidence compared to wild-type mice. In tumors induced from glial progenitor cells there was a slight increase in tumor incidence in Ink4a-/- mice and Ink4a-Arf-/- mice compared to wild-type mice. In both progenitor cells and astrocytes, Arf-loss caused a pronounced increase in tumor malignancy compared to Ink4a-loss. Hence, Ink4a-loss contributed to tumor initiation from astrocytes and Arf-loss caused tumor progression from both glial progenitor cells and astrocytes. Results from in vitro studies on primary brain cell cultures suggested that the PDGFB-induced activation of the mitogen-activated protein kinase pathway via extracellular signal-regulated kinase was involved in the initiation of low-grade oligodendrogliomas and that the additional loss of Arf may contribute to tumor progression through increased levels of cyclin D1 and a phosphoinositide 3-kinase-dependent activation of p70 ribosomal S6 kinase causing a strong proliferative response of tumor cells.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/deficiency , Oligodendroglioma/metabolism , Oligodendroglioma/pathology , Proto-Oncogene Proteins c-sis/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Progression , Mice , Mice, Knockout , Oligodendroglioma/genetics , Proto-Oncogene Proteins c-sis/genetics , Signal Transduction , Survival Rate , Tissue Culture Techniques
7.
Biochem Biophys Res Commun ; 290(1): 349-58, 2002 Jan 11.
Article in English | MEDLINE | ID: mdl-11779176

ABSTRACT

The simultaneous presence of the EGFR and its ligand TGF-alpha in human tumor tissues suggests that autocrine TGF-alpha stimulation drives tumor growth. Here we show that autocrine TGF-alpha stimulation does cause increased tumor growth in vivo, an effect that was proven to be mediated via EGFR activation, and that this TGF-alpha/EGFR autocrine loop was accessible to an EGFR specific tyrosine kinase inhibitor. Clones of the EGFR expressing glioma cell line U-1242 MG were transfected with TGF-alpha cDNA using a tetracycline-inhibitory system for gene expression. TGF-alpha expression was inhibited by the presence of tetracycline, and subcutaneous tumors forming from cell lines injected into nude mice could be inhibited by feeding mice tetracycline. We confirmed that TGF-alpha mRNA and protein were present in these tumors and that, subsequently, the endogenous EGFR was activated. Tumor growth could be inhibited by an EGFR specific tyrosine kinase inhibitor of the type 4-(3-chloroanilino)-6,7-dimethoxy-quinazoline, administered daily by intraperitoneal injection, thereby interrupting the autocrine loop.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Transforming Growth Factor alpha/metabolism , Tyrphostins/pharmacology , Animals , Blotting, Northern , Blotting, Western , Cell Division/drug effects , DNA, Complementary/metabolism , Enzyme Inhibitors/pharmacology , Glioma/drug therapy , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Quinazolines , RNA/metabolism , RNA, Messenger/metabolism , Tetracycline/pharmacology , Time Factors , Transfection , Tumor Cells, Cultured , Tyrosine/metabolism
8.
Mech Dev ; 107(1-2): 181-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11520676

ABSTRACT

The intermediate filament glial fibrillary acidic protein (GFAP) constitutes the major cytoskeletal protein in astrocytes (J. Neuroimmunol. 8 (1985) 203) and is traditionally referred to as a specific marker for astrocytes. To identify early glial precursors, we created GFAPpromoter-lacZ transgenic mice, using a 1.8kb 5' fragment of human GFAP. The expression of the transgene was first detected in the neuroepithelium at embryonic day 9.5. It was further found in the ventricular zone of the developing telencephalon, in the cerebellar primordium, trigeminal ganglia, and radial glia. Later, scattered beta-gal+ cells were seen in pons, brain stem and glia limitans. The results indicate that GFAP activity is regulated in a region-specific manner during central nervous system (CNS) development and that the gene is turned on in different cell types independently.


Subject(s)
Central Nervous System/embryology , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/genetics , Nerve Tissue Proteins , Promoter Regions, Genetic , Animals , Brain/embryology , Brain/metabolism , Central Nervous System/metabolism , Cloning, Molecular , Epithelial Cells/metabolism , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Intermediate Filament Proteins/analysis , Mice , Mice, Transgenic , Nestin , Neuroglia/metabolism , Transgenes , Vimentin/analysis
9.
Breast Cancer Res Treat ; 61(3): 197-210, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10965996

ABSTRACT

Mammary spindle-cell tumours and sarcomas seem to be restricted to dogs and humans. Two cell lines from spontaneous primary canine mammary spindle-cell tumours (CMT-U304 and CMT-U309) and two cell lines from spontaneous primary canine mammary osteosarcomas (CMT-U334 and CMT-U335) were established to study the mesenchymal phenotypes of mammary tumours in the female dog. The cells from the spindle-cell tumours expressed cytokeratin, vimentin and smooth muscle actin filaments. When these cells were inoculated subcutaneously into female and male nude mice they formed different types of mesenchymal tumours such as spindle-cell tumours, fibroma and rhabdomyoid tumours (n = 6/8). The cells from the osteosarcomas expressed vimentin filaments and also formed different types of mesenchymal tumours such as chondroid, rhabdomyoid, smooth muscle-like and spindle-cell tumours (n = 6/10). The cell lines CMT-U304, CMT-U309 and CMT-U335 had receptors for progesterone but none of the four cell lines had receptors for estrogen. All four cell lines and their corresponding primary tumours showed identical allelic patterns in microsatellite analysis. By in situ hybridization with genomic DNA we could verify that all formed tumours but one were of canine origin. Our results support the hypothesis that canine mammary tumours are derived from pluripotent stem cells.


Subject(s)
Bone Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Mammary Neoplasms, Animal/pathology , Osteosarcoma/pathology , Stem Cells/pathology , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/ultrastructure , Carcinogenicity Tests , Cell Transformation, Neoplastic/metabolism , Cytoskeletal Proteins/metabolism , DNA, Neoplasm/analysis , Dogs , Female , Immunohistochemistry , In Situ Hybridization , Male , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Nude , Microsatellite Repeats , Neoplasm Transplantation , Osteosarcoma/metabolism , Osteosarcoma/ultrastructure , Phenotype , Receptors, Progesterone/metabolism , Stem Cells/metabolism , Tumor Cells, Cultured
10.
Exp Cell Res ; 259(1): 293-9, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10942601

ABSTRACT

Overexpression of Met is a common finding in thyroid carcinomas. Recently, we reported on overexpression and ligand-independent constitutive activation of Met in anaplastic thyroid carcinoma cells. In the present study we have investigated a putative mechanism for this phenomenon. Cell lines with constitutively activated Met expressed both TGF-alpha mRNA and protein. Western blot analysis revealed expression of receptors for epidermal growth factor (EGFR) in all carcinoma cell lines; in tumor cells with elevated levels of TGF-alpha mRNA there was a constitutive tyrosine phosphorylation of the EGFRs. Preincubation of carcinoma cells with suramin decreased EGFR activation and downregulated Met expression as well as the ligand-independent phosphorylation of Met. Similar results were obtained with a EGFR tyrosine kinase inhibitor, AG 1478. The MEK inhibitor U0126 had an even more pronounced effect compared to AG 1478, indicating a Ras/MAPK-mediated signal in the regulation of Met expression and activation. Inhibition of EGFR signaling also decreased proliferation of the anaplastic thyroid carcinoma cells. Thus, aberrant activation of EGFRs may lead to an overexpression and activation of Met, which may be of importance for the malignant phenotype of anaplastic thyroid carcinomas.


Subject(s)
Carcinoma , ErbB Receptors/physiology , MAP Kinase Kinase Kinase 1 , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-met/genetics , Thyroid Neoplasms , Antineoplastic Agents/pharmacology , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-met/metabolism , Quinazolines , RNA, Messenger/analysis , Suramin/pharmacology , Transforming Growth Factor alpha/genetics , Tumor Cells, Cultured , Tyrphostins/pharmacology
11.
J Neurosci Res ; 60(2): 245-56, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10740230

ABSTRACT

We have studied the effect of induced glial fibrillary acidic protein (GFAP) on motility, cell morphology, and proliferation of two originally GFAP-negative human glioma cell lines. Glioma cell lines U-1242 MG and U-251 MG sp subclone 3A were transfected with a vector system that allows for an inducible GFAP expression. This experimental system creates an "on/off" situation in which GFAP expression is suppressed by tetracycline. Inducible expression of GFAP in the absence of tetracycline was confirmed by immunofluorescence staining and Northern and Western blotting. The study showed that forced GFAP expression resulted in an inhibition of cell motility measured as the phagokinetic track area of individual cells seeded sparsely on a surface covered with gold particles. It also resulted in a change in cell morphology, with extended cell processes, and it was associated with a low fraction of cells in S-phase. We conclude that the down-regulation of GFAP expression that is often seen in gliomas in vivo may be an important parameter of tumor progression related mainly to the motile and thereby invasive properties of malignant glioma cells.


Subject(s)
Brain Neoplasms/metabolism , Cell Division/physiology , Cell Movement/physiology , Glial Fibrillary Acidic Protein/metabolism , Glioma/metabolism , Brain Neoplasms/physiopathology , Cell Size , Cloning, Molecular , Glioma/physiopathology , Humans , Tumor Cells, Cultured
12.
Int J Cancer ; 85(6): 819-28, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10709103

ABSTRACT

Three phenotypically different clonal human glioma cell lines were injected stereotactically into nude-rat brains, to determine their individual growth potential and to establish an in vivo system in which different therapeutic modalities could be tested. As assessed by serial sectioning, microscopic evaluation, and computer analysis, the mean approximate tumour volume after 3-7 weeks in vivo was 0.42 mm(3) for U-343 MG, 2.6 mm(3) for U-343 MGa Cl2:6, and 50.3 mm(3) for U-343 MGa 31L. When compared with the initial injected cell volume, only U-343 MGa 31L had increased in size, U-343 MGa Cl2:6 remained approximately the same but showed a certain proliferative potential, and U-343 MG regressed. Thus, only U-343 MGa 31L cells had high tumorigenic potential, invaded and replaced brain tissue in every direction, while U-343 MGa Cl2:6 cells grew in sheet-like tumour extensions along white-matter nerve-fibre tracts, in this respect mimicking foetal astrocytes. The tumorigenic potential of the U-343 MGa 31L cell clone was associated with a variable phenotype, as observed when the in vivo and in vitro characteristics were compared. The in vivo phenotype was characterized by the loss of GFAP immunoreactivity, the gain of heterogeneously distributed cellular tenascin, fibronectin, and laminin, but absence of extracellularly deposited material, and by the formation of irregular vessels. It appears that the intrinsic capacity of glioma cells to adapt to in vivo conditions is decisive for their tumorigenicity in the brain, rather than any single phenotypic property in itself. Moreover, the 2 glioma cell clones best suited for in vitro growth were no longer tumorigenic.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Neoplasm Transplantation , Animals , Brain , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Clone Cells , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Glioma/blood supply , Glioma/metabolism , Humans , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Male , Phenotype , Rats , Rats, Nude , Tumor Cells, Cultured
13.
Int J Cancer ; 85(3): 398-406, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10652433

ABSTRACT

In human gliomas, platelet-derived growth factor (PDGF) ligand and receptor mRNA are often co-expressed, which suggests the presence of an autocrine loop. To further investigate the significance of PDGF stimulation in brain tumors, we used a previously developed mouse tumor model, in which malignant brain tumors of neuroepithelial origin were induced by injecting a murine retrovirus containing the human PDGF B-chain gene into the brains of neonatal mice. In the present investigation, we have characterized a cell line established from such an experimentally induced tumor in an INK4a-/- mouse. Cultured tumor cells expressed nestin and NG2 chondroitin sulfate proteoglycan and are thus most likely derived from an oligodendrocyte precursor cell. Tumor cells produced PDGF-B protein and displayed constitutively activated PDGF alpha receptors. Autocrine receptor activation could be blocked with the specific PDGF receptor tyrosine kinase inhibitor CGP 57148B, which led to almost complete inhibition of cell proliferation, which was much less affected by a PDGF B-chain aptamer that inhibits binding of PDGF-B to PDGF receptors and is unlikely to be able to pass through the plasma membrane. Our results imply an important role for PDGF autocrine stimulation in both initiation and progression of a subtype of gliomas.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Nerve Tissue Proteins , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Autocrine Communication , Benzamides , Blotting, Northern , Blotting, Western , Brain Neoplasms/chemically induced , Carrier Proteins/genetics , Cell Division , Cyclin-Dependent Kinase Inhibitor p16 , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Glioma/chemically induced , Humans , Imatinib Mesylate , Intermediate Filament Proteins/biosynthesis , Mice , Mice, Inbred Strains , Mice, Nude , Nestin , Piperazines/pharmacology , Precipitin Tests , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-sis/administration & dosage , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Tumor Cells, Cultured
14.
Int J Cancer ; 85(2): 211-22, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10629080

ABSTRACT

While platelet-derived growth factor, PDGF, is not regularly expressed in mesenchymal tissues, PDGF B mRNA is often found in tumors derived from these tissues. PDGF B protein is also present, but the protein levels in individual tumors do not appear to correlate well with those of the mRNA. PDGF B is homologous to the v-sis oncogene of simian sarcoma virus (SSV), and certain deletions confined to 3; and 5; non-coding sequences of PDGF B mRNA are consistently found in tumors induced by SSV and by a PDGF B retrovirus. Part of exon 1, including a non-coding GC-rich regulatory domain and the signal sequence as well as a 149 nucleotide long AU-rich stretch in the 3; non-coding region, are often lost. We hoped that this information could provide a clue to defective regulatory mechanisms present in human tumors and have searched for such mRNA variants in human sarcoma cell lines and soft tissue tumors. We identified a splice variant of PDGF B mRNA that, similar to v-sis, lacks the 149 nucleotide stretch and introduces an anomalous splice point between exons 6 and 7. This weakly abundant mRNA variant was co-expressed with the regular PDGF B mRNA, but its presence failed to show any association with increased levels of immunohistochemically detectable PDGF B protein. Instead, the highest levels of cellular PDGF B protein were found in samples with mRNAs showing exon 1 divergence. The changes in the 5; end of the mRNA were not accompanied by any genomic aberrations.


Subject(s)
Alternative Splicing , Exons , Neoplasms/genetics , Proto-Oncogene Proteins c-sis/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , 3' Untranslated Regions , 3T3 Cells , Animals , Antibodies, Monoclonal/immunology , Base Sequence , Blotting, Northern , CHO Cells , Cricetinae , DNA, Neoplasm , Exons/genetics , Genes, sis , Humans , Immunologic Techniques , Mice , Molecular Sequence Data , Neoplasms/pathology , Proto-Oncogene Proteins c-sis/analysis , Proto-Oncogene Proteins c-sis/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
15.
Cell Biol Int ; 23(5): 351-8, 1999.
Article in English | MEDLINE | ID: mdl-10579901

ABSTRACT

The gas3/PMP22 gene product is a dual function protein, involved in both peripheral nerve myelination and cell proliferation. gas 3/PMP22 is highly expressed in myelinating Schwann cells and is required for normal PNS development. In addition, a more general function for gas3 is suggested by its expression in non-neural tissues and upregulation by growth arrest in cultured rodent fibroblasts. In the present work, the expression of the gas3 gene has been studied in human fibroblasts. We have confirmed that gas3 mRNA is upregulated when cells are serum starved or grown to high cell density (G0 arrest). When quiescent cells were stimulated by serum or platelet-derived growth factor-BB (PDGF-BB), gas3 mRNA was down regulated. In contrast, we found that the expression of gas3 mRNA was neither upregulated in senescent cells nor in cells arrested in G1 using Lovastatin. Thus, high expression of gas3 is not related to growth inhibition in general, but more probably to the G0 growth arrest state. Furthermore, we found that in two malignant fibrous histiocytoma cell lines, gas3 expression was lower than in normal fibroblasts, suggesting an altered regulation of the gas3 gene in transformed cells.


Subject(s)
Fibrosarcoma , Myelin Proteins/genetics , Resting Phase, Cell Cycle/physiology , Skin/cytology , Aged , Antineoplastic Agents/pharmacology , Blotting, Northern , Cellular Senescence/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Infant , Kinetics , Lovastatin/pharmacology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology
16.
Physiol Rev ; 79(4): 1283-316, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10508235

ABSTRACT

Platelet-derived growth factor (PDGF) is a major mitogen for connective tissue cells and certain other cell types. It is a dimeric molecule consisting of disulfide-bonded, structurally similar A- and B-polypeptide chains, which combine to homo- and heterodimers. The PDGF isoforms exert their cellular effects by binding to and activating two structurally related protein tyrosine kinase receptors, denoted the alpha-receptor and the beta-receptor. Activation of PDGF receptors leads to stimulation of cell growth, but also to changes in cell shape and motility; PDGF induces reorganization of the actin filament system and stimulates chemotaxis, i.e., a directed cell movement toward a gradient of PDGF. In vivo, PDGF has important roles during the embryonic development as well as during wound healing. Moreover, overactivity of PDGF has been implicated in several pathological conditions. The sis oncogene of simian sarcoma virus (SSV) is related to the B-chain of PDGF, and SSV transformation involves autocrine stimulation by a PDGF-like molecule. Similarly, overproduction of PDGF may be involved in autocrine and paracrine growth stimulation of human tumors. Overactivity of PDGF has, in addition, been implicated in nonmalignant conditions characterized by an increased cell proliferation, such as atherosclerosis and fibrotic conditions. This review discusses structural and functional properties of PDGF and PDGF receptors, the mechanism whereby PDGF exerts its cellular effects, and the role of PDGF in normal and diseased tissues.


Subject(s)
Platelet-Derived Growth Factor/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Animals , Disease , Embryonic and Fetal Development , Humans , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/pharmacology , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/genetics , Signal Transduction
17.
Endocrinology ; 140(9): 4300-10, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10465304

ABSTRACT

Thyroid growth and function are intricately regulated by both positive and negative factors. In the present study, we have investigated the expression of transforming growth factor-beta (TGF-beta) super-family members and their receptors in normal porcine thyroid follicle cells. In tissue sections of porcine thyroids, we observed an expression of TGF-beta1, activin A, and bone morphogenetic protein (BMP)-7 proteins. The staining was localized to the follicular epithelium. In affinity cross-linking experiments, TGF-beta1 was found to bind to heteromeric complexes of TGF-beta type I and type II receptors, and activin A bound most efficiently to heteromeric complexes of activin type IB and type II receptors. We were unable to detect any BMP receptors (BMPRs) in attempts to perform affinity cross-linking with BMP-7. However, expression of BMPR-IA and BMPR-II messenger RNA (mRNA) was detected by Northern blot analysis. Both TGF-beta1 and activin A, but not BMP-7, increased the phosphorylation of Smad2, induced nuclear translocation of Smad2, Smad3, and Smad4, and inhibited thyrocyte cell growth as well as TSH-stimulated cAMP response. TGF-beta1 was more potent, compared with activin A, to induce these cellular responses. Taken together, our findings indicate a role for several members of the TGF-beta family in regulation of thyroid growth and function.


Subject(s)
Inhibins/metabolism , Receptors, Growth Factor/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Thyroid Gland/metabolism , Transforming Growth Factor beta/metabolism , Activin Receptors , Activins , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Division/physiology , Cells, Cultured , DNA-Binding Proteins/physiology , Ligands , Reference Values , Signal Transduction/physiology , Smad2 Protein , Smad3 Protein , Swine , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/physiology , Trans-Activators/physiology
18.
Mol Cell Endocrinol ; 153(1-2): 79-90, 1999 Jul 20.
Article in English | MEDLINE | ID: mdl-10459856

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine. In the present study we have investigated the expression of TGF-beta receptors (TbetaR's) and SMAD proteins in non-neoplastic and neoplastic thyroid follicle cells. We found expression of all TbetaR's (type I, II and III) and SMAD proteins analysed (Smad2, Smad3, Smad4, Smad6 and Smad7). Five out of six human anaplastic thyroid carcinoma cell lines were growth inhibited by addition of TGF-beta1, and therefore considered to be TGF-responsive. One cell line however, HTh 7, did not respond to TGF-beta1 with growth inhibition, induction of the extracellular matrix protein fibronectin or immediate early genes junB, Smad6 and Smad7 mRNA. Analysis of the TGF-beta intracellular signalling pathway in HTh 7 cells showed that receptors were capable of signalling, e.g. Smad2 phosphorylation and SMAD nuclear translocation. In summary, our data shows abundant expression of TGF-beta signalling components in thyroid follicle cells, and the escape from TGF-beta sensitivity in one anaplastic thyroid carcinoma despite an apparently functional TGF-beta/SMAD-signalling pathway, indicating a novel mechanism for TGF-beta insensitivity.


Subject(s)
Activin Receptors, Type I , Carcinoma/pathology , Protein Serine-Threonine Kinases/physiology , Receptors, Transforming Growth Factor beta/physiology , Thyroid Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Carcinoma/metabolism , Cell Division , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression , Genes, myc , Humans , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/analysis , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad2 Protein , Smad3 Protein , Smad4 Protein , Smad6 Protein , Smad7 Protein , Thyroid Neoplasms/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection , Tumor Cells, Cultured
19.
Thyroid ; 9(1): 7-11, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10037069

ABSTRACT

The aim of the present study was to investigate the N-terminal part (the translated part of exon 1) of the human thyrotropin receptor (TSHR) for the presence of mutations. Patients with Graves' disease (n = 160) and healthy controls (blood donors; n = 140) were screened using single-stranded conformational polymorphism (SSCP) in combination with restriction enzyme digestion for the two previously known mutations in this part of the receptor, viz. D36H and P52T TSHR-variants. We did not find any novel mutation in this region. However, D36H and P52T variants were found both in the TSHR of Graves' patients and in the healthy controls. The overall frequency of the D36H-receptor variant was 5.0% (15/300) and of the P52T-receptor, 7.3% (22/300). There was no major difference in the frequency for either of the TSHR alleles between the 2 groups. Thus, these 2 polymorphic variants of the TSHR seem to occur in a relatively high frequency in the population.


Subject(s)
Receptors, Thyrotropin/genetics , Alleles , Amino Acid Substitution , Aspartic Acid/genetics , Exons , Female , Gene Frequency , Genetic Testing , Graves Disease/genetics , Heterozygote , Histidine/genetics , Humans , Male , Mutation , Polymorphism, Single-Stranded Conformational , Proline/genetics , Restriction Mapping , Threonine/genetics
20.
Cancer Res ; 58(23): 5275-9, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9850047

ABSTRACT

In existing mouse models for malignant brain tumors, genes with no proven pathogenical relevance for humans have been used. Coexpression of platelet-derived growth factor (PDGF) and PDGF receptors suggests an autocrine mechanism of growth factor stimulation in the development of brain tumors in man. A murine retrovirus coding for the PDGF B-chain was, therefore, used to induce brain tumors in mice. Of 35 mice who received injections, 15 developed brain tumors of oligo- or monoclonal origin. They coexpressed PDGF B-chain and alpha-receptor mRNA, as expected, from an autocrine mechanism of transformation. Most tumors displayed characteristics of glioblastoma multiforme or of a primitive neuroectodermal tumor, and the consistent expression of nestin suggested that they were all derived from an immature neuroglial progenitor. The results show that an autocrine mechanism of transformation may be an initial or early event in neuro-oncogenesis. The present model provides an ideal system for studies of genetic mechanisms involved in the development of brain tumors.


Subject(s)
Brain Neoplasms/etiology , Moloney murine leukemia virus/genetics , Platelet-Derived Growth Factor/physiology , Proto-Oncogene Proteins/physiology , 3T3 Cells/metabolism , Animals , Brain Neoplasms/pathology , Brain Neoplasms/virology , Clone Cells , Disease Models, Animal , Female , Male , Mice , Moloney murine leukemia virus/metabolism , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-sis , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha , Receptors, Platelet-Derived Growth Factor/biosynthesis , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/physiology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...