Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Ann Oncol ; 29(2): 405-417, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29092024

ABSTRACT

Background: Male breast cancer (BC) is rare, managed by extrapolation from female BC. The International Male BC Program aims to better characterize and manage this disease. We report the results of part I, a retrospective joint analysis of cases diagnosed during a 20-year period. Methods: Patients with follow-up and tumor samples, treated between 1990 and 2010, in 93 centers/9 countries. Samples were centrally analyzed in three laboratories (the United Kingdom, the Netherlands and the United States). Results: Of 1822 patients enrolled, 1483 were analyzed; 63.5% were diagnosed between 2001 and 2010, 57 (5.1%) had metastatic disease (M1). Median age at diagnosis: 68.4 years. Of 1054 M0 cases, 56.2% were node-negative (N0) and 48.5% had T1 tumors; 4% had breast conserving surgery (BCS), 18% sentinel lymph-node biopsy; half received adjuvant radiotherapy; 29.8% (neo)adjuvant chemotherapy and 76.8% adjuvant endocrine therapy (ET), mostly tamoxifen (88.4%). Per central pathology, for M0 tumors: 84.8% ductal invasive carcinomas, 51.5% grade 2; 99.3% estrogen receptor (ER)-positive; 81.9% progesterone receptor (PR)-positive; 96.9% androgen receptor (AR)-positive [ER, PR or AR Allred score ≥3]; 61.1% Ki67 expression low (<14% positive cells); using immunohistochemistry (IHC) surrogates, 41.9% were Luminal-A-like, 48.6% Luminal-B-like/HER-2-negative, 8.7% HER-2-positive, 0.3% triple negative. Median follow-up: 8.2 years (0.0-23.8) for all, 7.2 years (0.0-23.2), for M0, 2.6 years (0.0-12.7) for M1 patients. A significant improvement over time was observed in age-corrected BC mortality. BC-specific-mortality was higher for men younger than 50 years. Better overall (OS) and recurrence-free survival (RFS) were observed for highly ER+ (P = 0.001), highly PR+ (P = 0.002), highly AR+ disease (P = 0.019). There was no association between OS/RFS and HER-2 status, Ki67, IHC subtypes nor grade. Conclusions: Male BC is usually ER, PR and AR-positive, Luminal B-like/HER2-negative. Of note, 56% patients had T1 tumors but only 4% had BCS. ER was highly positive in >90% of cases but only 77% received adjuvant ET. ER, PR and AR were associated with OS and RFS, whereas grade, Ki67 and IHC surrogates were not. Significant improvement in survival over time was observed.


Subject(s)
Breast Neoplasms, Male , Adult , Aged , Biomarkers, Tumor/analysis , Breast Neoplasms, Male/mortality , Breast Neoplasms, Male/pathology , Breast Neoplasms, Male/surgery , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Proportional Hazards Models , Retrospective Studies
2.
Curr Opin Cell Biol ; 3(2): 224-9, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1715728

ABSTRACT

This review describes three biological processes in which there is evidence for single cells being able to measure elapsed time. We describe the work that has led to this view, and review more recent work that has provided new insights into possible mechanisms for the measurement of time.


Subject(s)
Biological Clocks , Cell Differentiation , Cell Survival/physiology , Animals , Fetal Hemoglobin/genetics , Fetal Hemoglobin/physiology , Fibroblasts/cytology , Humans , Oligodendroglia/cytology
3.
Oncogene ; 26(38): 5577-86, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17369861

ABSTRACT

Neuropilin-1 (NRP1) functions as a coreceptor through interaction with plexin A1 or vascular endothelial growth factor (VEGF) receptor during neuronal development and angiogenesis. NRP1 potentiates the signaling pathways stimulated by semaphorin 3A and VEGF-A in neuronal and endothelial cells, respectively. In this study, we investigate the role of tumor cell-expressed NRP1 in glioma progression. Analyses of human glioma specimens (WHO grade I-IV tumors) revealed a significant correlation of NRP1 expression with glioma progression. In tumor xenografts, overexpression of NRP1 by U87MG gliomas strongly promoted tumor growth and angiogenesis. Overexpression of NRP1 by U87MG cells stimulated cell survival through the enhancement of autocrine hepatocyte growth factor/scatter factor (HGF/SF)/c-Met signaling. NRP1 not only potentiated the activity of endogenous HGF/SF on glioma cell survival but also enhanced HGF/SF-promoted cell proliferation. Inhibition of HGF/SF, c-Met and NRP1 abrogated NRP1-potentiated autocrine HGF/SF stimulation. Furthermore, increased phosphorylation of c-Met correlated with glioma progression in human glioma biopsies in which NRP1 is upregulated and in U87MG NRP1-overexpressing tumors. Together, these data suggest that tumor cell-expressed NRP1 promotes glioma progression through potentiating the activity of the HGF/SF autocrine c-Met signaling pathway, in addition to enhancing angiogenesis, suggesting a novel mechanism of NRP1 in promoting human glioma progression.


Subject(s)
Glioma/pathology , Hepatocyte Growth Factor/physiology , Neuropilin-1/physiology , Signal Transduction/physiology , Animals , Butadienes/pharmacology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Hepatocyte Growth Factor/pharmacology , Humans , Immunoblotting , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neuropilin-1/genetics , Neuropilin-1/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/physiology , RNA, Small Interfering/genetics , Transfection , Transplantation, Heterologous , Tumor Burden
4.
Cancer Res ; 55(13): 2746-51, 1995 Jul 01.
Article in English | MEDLINE | ID: mdl-7796398

ABSTRACT

Primary cortical astrocytes were isolated from normal (+/+), heterozygous (+/-), or homozygous (-/-) p53-knockout mice. The normal astrocytes grew slowly and underwent crisis after limited division, while the homozygously defective cells grew rapidly and without contact inhibition. These -/- cells could not initially form colonies in soft agarose but acquired this capability after 10 passages in FCS or basic fibroblast growth factor but not epidermal growth factor. Almost all -/- astrocytes weakly expressed glial fibrillary acidic protein at passage 10 and were also A2B5+ when cultured in basic fibroblast growth factor. Most heterozygous cells resembled normal ones; however, some survived crisis, grew rapidly, and formed colonies. Outgrowing cells had all lost the wild-type p53 allele. These molecular and cellular events mimic the early stages of human brain tumors, suggest a role for p53 in the earliest stages of disease progression, and provide an experimental system to analyze the effects of other tumor-specific mutations in the disease process.


Subject(s)
Astrocytes/cytology , Cerebral Cortex/cytology , Genes, p53 , Animals , Animals, Newborn , Cell Transformation, Neoplastic , Female , Heterozygote , Male , Mice , Mice, Knockout
5.
Cancer Res ; 59(1): 35-43, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9892180

ABSTRACT

A novel putative tumor suppressor gene and member of the NF2/ERM/ 4.1 superfamily was isolated using Differential Display PCR (DDPCR) on primary lung tumors. When reintroduced into nonexpressing non-small cell lung carcinoma cell lines, this gene, named DAL-1 (for Differentially expressed in Adenocarcinoma of the Lung), was shown to suppress growth. In addition, significantly reduced expression (>50%) of DAL-1 was measured in 39 primary non-small cell lung carcinoma tumors as compared with patient-matched normal lung tissue. Immunocytochemical staining with a polyclonal anti-DAL-1 antibody localized the protein to the plasma membrane, particularly at cell-cell contact points, a pattern reminiscent of other members of the protein 4.1 superfamily including ezrin and NF2. The data suggest DAL-1 is a novel membrane-associated protein with potential to play an important role in the origin and progression of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cytoskeletal Proteins , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Lung Neoplasms/genetics , Membrane Proteins/genetics , Neuropeptides , Tumor Suppressor Proteins , Amino Acid Sequence , Genes, Neurofibromatosis 2 , Humans , Microfilament Proteins , Molecular Sequence Data , Phosphoproteins/genetics , Sequence Analysis , Sequence Homology, Amino Acid
6.
Cancer Res ; 56(21): 5079-86, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8895767

ABSTRACT

Alterations of the EGFR gene occur frequently in human gliomas where the most common is an in-frame deletion of exons 2-7 from the extracellular domain, resulting in a truncated mutant receptor (deltaEGFR or de 2-7 EGFR). We previously demonstrated that introduction of deltaEGFR into human U87MG glioblastoma cells (U87MG.deltaEGFR) conferred remarkably enhanced tumorigenicity in vivo. Here, we show by cell-mixing experiments that the enhanced tumorigenicity conferred by deltaEGFR is attributable to a growth advantage intrinsic to cells expressing the mutant receptor. We analyzed the labeling index of the proliferation markers Ki-67 and bromodeoxyuridine and found that tumors derived from U87MG.deltaEGFR cells had significantly higher labeling indexes than those of tumors derived from U87MG cells that were either naive, expressed kinase-deficient mutants of deltaEGFR, or overexpressed exogenous wild-type EGFR. We also utilized terminal deoxynucleotidyl transferase-mediated nick end-labeling assays and showed that the apoptotic index of U87MG.deltaEGFR tumors was more than 4-fold lower than that of parental U87MG tumors. This decrease in cell death was inversely correlated with the expression level of Bcl-X(L), a negative regulator of apoptosis, which was more than 3-fold higher in U87MG.deltaEGFR-derived tumors than in those derived from parental cells. Similar observations were obtained in vitro in serum-free conditions. These results suggest that deltaEGFR exerts its pronounced enhancement of glioblastoma tumorigenicity by stimulating proliferation and inhibiting apoptosis and that the effects are directly attributable to its constitutively active signal.


Subject(s)
Apoptosis , ErbB Receptors/physiology , Glioblastoma/pathology , Animals , Cell Division , ErbB Receptors/genetics , Glioblastoma/genetics , Humans , Mice , Mice, Inbred BALB C , Mutation , Proto-Oncogene Proteins c-bcl-2/analysis , Tumor Cells, Cultured
7.
Cell Signal ; 12(11-12): 769-79, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11152963

ABSTRACT

Expression of the src homology 3 (SH3)-encoding, expressed in tumorigenic astrocytes (SETA) gene is associated with astrocyte transformation in culture and tumors in the adult brain. SETA binds to the apoptosis regulator apoptosis-linked gene 2 (ALG-2) interacting protein 1 (AIP1), and modulates apoptosis in astrocytes. The predicted protein structure of SETA revealed two SH3 domains, while related proteins were reported to have three. Here we report the identification of an additional SH3 domain N-terminal to the previously identified SETA sequence. Yeast two-hybrid screening of a p53(-/-) astrocyte cDNA library with this SH3 domain identified a novel gene, SETA binding protein 1 (SB1), with 55% amino acid identity to the renal tumor antigen, NY-REN-45. In vitro confrontation and co-immunoprecipitation experiments confirmed the binding of SB1 to SETA. Evidence that SETA binds to the CD2 protein, the proto-oncogene c-Cbl, and the signal transduction molecule Grb2, and can dimerize via its C-terminal coiled coil (CC) domain is also presented.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases , src Homology Domains , Amino Acid Sequence , Animals , Antigens, Neoplasm/chemistry , Astrocytes/metabolism , CD2 Antigens/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cells, Cultured , Dimerization , Exons/genetics , GRB2 Adaptor Protein , Gene Deletion , Gene Library , Mice , Molecular Sequence Data , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Precipitin Tests , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-cbl , Rats , Sequence Alignment , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , Two-Hybrid System Techniques
8.
Neuro Oncol ; 2(1): 1-5, 2000 01.
Article in English | MEDLINE | ID: mdl-11302248

ABSTRACT

The prognosis for most patients with astrocytic glioma is poor, and postoperative life expectancy has not significantly improved in the last decade despite advances in diagnosis, surgery, and adjuvant therapy. Progress has been made, however, in cataloging the genetic alterations that occur in these tumors. Studying the allelic changes using loss of heterozygosity analysis has proven to be a reliable and rapid way of identifying genetic alterations fundamental to the pathology of this disease. In this study, we used a series of fluorescent-labeled markers and a new horizontal ultrathin gel electrophoresis technology (HUGE; GeneSys Technologies, Inc.) to analyze loss of heterozygosity on 11p15 in a series of 24 matched normal/tumor glioma pairs that included both anaplastic astrocytomas and glioblastomas. These studies significantly narrowed the region harboring a putative 11p15.5 glioma-associated gene and further suggest that a second gene involved in the pathogenesis of brain tumors may exist, centromeric, in bands 11p15.5-p15.4.


Subject(s)
Alleles , Astrocytoma/genetics , Brain Neoplasms/genetics , Chromosomes, Human, Pair 11/genetics , Electrophoresis, Polyacrylamide Gel/methods , Gene Deletion , Glioblastoma/genetics , DNA Fragmentation , Humans , Loss of Heterozygosity
9.
Neuro Oncol ; 2(1): 6-15, 2000 01.
Article in English | MEDLINE | ID: mdl-11302255

ABSTRACT

Differential display polymerase chain reaction analysis was used to compare five differentiation states of the O-2A progenitor-like cell line CG4: progenitor cells and cells at 12 h or 4 days after the induction of differentiation into oligodendrocytes or astrocytes. This led to the identification of 52 sequence tags that were expressed differentially with cellular phenotype. One sequence was upregulated during differentiation of CG4 cells and represented a novel gene that we named SETA (SH3 domain-containing gene expressed in tumorigenic astrocytes). This gene encodes an SH3 domain-containing adapter protein with sequence similarity to the CD2AP (CD2 adapter protein) and CMS (Cas ligand with multiple Src homology) genes. SETA mRNA was expressed at high levels in the developing rat brain but was barely detectable in the normal adult rat or human brain. However, SETA mRNA was found in approximately one half of the human gliomas tested, including astrocytomas grades II, III, and IV, as well as oligodendrogliomas, mixed oligoastrocytomas, and human glioma-derived cell lines. A rat glioma generated by treatment with the alkylating carcinogen ethylnitrosourea on postnatal day 1 and a derived cell line also expressed SETA mRNA. Furthermore, in an in vitro model of astrocytoma progression based on p53-/- astrocytes, expression of SETA was restricted to cells that are tumorigenic.


Subject(s)
Astrocytes/physiology , Central Nervous System Neoplasms/etiology , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Brain/embryology , Brain/growth & development , Brain/metabolism , Brain Neoplasms/metabolism , Cell Line , DNA, Complementary/genetics , Data Display , Embryonic and Fetal Development , Glioma/metabolism , Humans , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Neuroglia/cytology , Neuroglia/metabolism , Oligodendroglia/cytology , Polymerase Chain Reaction , Rats , Stem Cells/cytology
10.
Brain Res ; 861(2): 354-62, 2000 Apr 10.
Article in English | MEDLINE | ID: mdl-10760497

ABSTRACT

Dorsal root ganglion (DRG) neurons show a transient peak expression of acetylcholinesterase (AChE) during periods of axonal outgrowth prior to synaptogenesis, suggesting that AChE has a non-enzymatic role during development. We have previously shown that perturbation of cell surface AChE in cultured embryonic rat DRG neurons results in decreased neurite outgrowth and neurite detachment. In this report, we demonstrate a direct correlation between endogenous AChE content and neurite outgrowth in primary DRG neurons. Adenoviral vectors were constructed using full-length rat AChE(T) cDNA in either the sense or antisense orientations to overexpress or knock down AChE expression, respectively. Treatment with the sense-expressing vector produced a 2.5-fold increase in AChE expression and a 2-fold increase in neurite length compared with either untreated or null virus-treated control cells. Conversely, treatment with the antisense-expressing vector reduced AChE expression by 40% and resulted in a reduction in neurite length of similar magnitude. We also observed that overexpression of AChE resulted in greater branching at the distal tips of each primary neurite as well as an increase in cell body size. These findings further indicate that AChE expressed on the axonal surface of developing DRG neurons may modulate their adhesive properties and thereby support axonal development.


Subject(s)
Acetylcholinesterase/metabolism , Ganglia, Spinal/metabolism , Neurites/metabolism , Acetylcholinesterase/genetics , Adenoviridae/genetics , Animals , Genetic Vectors/genetics , Neurons/metabolism , Rats , Tumor Cells, Cultured
16.
Curr Protoc Neurosci ; Chapter 3: Unit 3.4, 2001 May.
Article in English | MEDLINE | ID: mdl-18428468

ABSTRACT

Oligodendrocytes are the myelinating cells of the central nervous system, and are avidly studied by investigators interested in neural cell lineages, myelination, multiple sclerosis, and oligodendroglioma. This unit presents a strategy for manipulating the division and differentiation of oligodendrocyte progenitors by isolating O-2A progenitors from optic nerve and then purifying and expanding them by culture in a defined medium supplemented with platelet-derived growth factor (PDGF) plus basic fibroblast growth factor (bFGF). Cells are then allowed to differentiate through removal of the growth factors. A protocol is also provided for monitoring the cell type composition of the cultures by immunohistochemistry. In addition to making it possible to generate large numbers of cells, this approach allows access to intermediate differentiation stages. Cells can thus be studied as they differentiate into oligodendrocytes, making the actual process of differentiation open to examination.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Oligodendroglia/cytology , Stem Cells/cytology , Animals , Animals, Newborn , Cell Differentiation/physiology , Cells, Cultured , Humans , Oligodendroglia/physiology , Optic Nerve/cytology , Optic Nerve/growth & development , Rats , Stem Cells/physiology
17.
Dev Biol ; 162(2): 525-38, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8150211

ABSTRACT

When stimulated by platelet-derived growth factor (PDGF), oligodendrocyte-type-2 astrocyte (O-2A) progenitors derived from perinatal rat optic nerves undergo a limited number of cell divisions before clonally related cells synchronously and symmetrically differentiate into nondividing oligodendrocytes. The duration of this mitotic period is thought to be controlled by a cell-intrinsic biological clock. Thus, in the presence of PDGF, the measurement of time by the biological clock is intimately coupled to the control of division and differentiation. In contrast, O-2A progenitors grown in the presence of PDGF plus basic fibroblast growth factor (bFGF) divide indefinitely in the absence of differentiation and so do not exhibit a limited period of division. We have tested whether growth in PDGF plus bFGF alters the duration of the limited period of division O-2A progenitors exhibit in response to PDGF alone. Accordingly, O-2A progenitors were grown in the presence of PDGF plus bFGF for varying lengths of time, before being switched to conditions that promote timed differentiation (PDGF but not bFGF). Increasing duration of culture in PDGF plus bFGF led to a gradual shortening of the period for which O-2A progenitors were subsequently responsive to PDGF alone, until eventually all cells differentiated without dividing after switching. In contrast, a short exposure to bFGF was not sufficient to cause a similar alteration in the pattern of differentiation. These results indicate that O-2A progenitors prevented from undergoing timed differentiation nevertheless retain the ability to measure elapsed time, implying that the biological clock in this cell type can be uncoupled from differentiation. Furthermore, they demonstrate that the biological clock does not impose an absolute limit on the number of divisions that an O-2A progenitor can undergo. In contrast with existing hypotheses, our observations suggest that the molecular mechanism that controls timed differentiation must consist of at least two components, with the clock itself being in some manner distinct from mechanisms that limit cell division and/or directly regulate differentiation.


Subject(s)
Astrocytes/cytology , Oligodendroglia/cytology , Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Optic Nerve/embryology , Platelet-Derived Growth Factor/pharmacology , Rats , Receptors, Platelet-Derived Growth Factor/analysis , Time Factors
18.
Glia ; 8(1): 12-9, 1993 May.
Article in English | MEDLINE | ID: mdl-8509161

ABSTRACT

The inhibition of differentiation of oligodendrocyte-type-2 astrocyte (O-2A) progenitors into oligodendrocytes caused by basic fibroblast growth factor (bFGF) can be overcome by non-O-2A lineage cells present in the optic nerve and by astrocytes purified from cerebral cortices. Although purified O-2A progenitors grown in the presence of bFGF for up to 6 days were inhibited from differentiating into oligodendrocytes, O-2A progenitors growing in heterogeneous optic nerve cultures did not show a similar inhibition of differentiation. The factor(s) responsible for overriding the inhibitory effects of bFGF appeared to be secreted by astrocytes, as extensive generation of oligodendrocytes was seen in cultures of purified O-2A progenitors exposed to bFGF+ medium conditioned by purified astrocytes (ACM). In addition, purified O-2A progenitors displayed a remarkable sensitivity to bFGF, which extended at least down to concentrations of 0.03 ng/ml, a concentration of < 2 x 10(-12) M. At a bFGF concentration of just 0.1 ng/ml, this mitogen still promoted DNA synthesis in as many O-2A progenitors as in cultures exposed to 1-30 ng/ml of this growth factor, but exhibited a reduced ability to promote DNA synthesis in oligodendrocytes. In addition, although concentrations of bFGF as low as 0.03 ng/ml were a potent stimulator of DNA synthesis in O-2A progenitors, application of this amount of bFGF no longer inhibited the differentiation of progenitors into oligodendrocytes as effectively as application of higher bFGF concentrations. Thus, the induction of DNA synthesis by bFGF can be uncoupled from the inhibition of differentiation.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Astrocytes/physiology , Cell Differentiation/physiology , Cerebral Cortex/physiology , Fibroblast Growth Factor 2/pharmacology , Oligodendroglia/cytology , Optic Nerve/cytology , Stem Cells/cytology , Animals , Cell Communication , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Separation , Cells, Cultured , DNA Replication/drug effects , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/analysis , Kinetics , Oligodendroglia/drug effects , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Time Factors
19.
J Biol Chem ; 275(25): 19275-81, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10858458

ABSTRACT

Expression of the src homology 3 (SH3) domain-containing expressed in tumorigenic astrocytes (SETA) gene is associated with the tumorigenic state in astrocytes. SETA encodes a variety of adapter proteins containing either one or two SH3 domains, as suggested by the sequence heterogeneity of isolated cDNAs. Using both SH3 domains in a yeast two-hybrid screen of a glial progenitor cell cDNA library, we isolated the rat homolog of the ALG-2-interacting protein 1 or ALG-2-interacting protein X (AIP1/Alix). In vitro confrontation experiments showed that the SH3-N domain of SETA interacted with the proline-rich C terminus of AIP1. In co-immunoprecipitation experiments, SETA and AIP1 interacted and could form a complex with apoptosis-linked gene 2 protein. Endogenous SETA and AIP1 proteins showed similar patterns of staining in primary rat astrocytes. Misexpression of a variety of SETA protein isoforms in these astrocytes revealed that they localized to the actin cytoskeleton. Furthermore, SETA proteins containing the SH3-N domain were able to sensitize astrocytes to apoptosis induced by UV irradiation. Expression of the isolated SH3-N domain had the greatest effect in these experiments, indicating that interference in the interaction between endogenous SETA and AIP1 sensitizes astrocytes to apoptosis in response to DNA damage.


Subject(s)
Apoptosis/physiology , Astrocytes/cytology , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Neoplasm Proteins/physiology , Nerve Tissue Proteins/physiology , Actins/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Astrocytes/radiation effects , DNA, Complementary , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Binding , Rats , Sequence Homology, Amino Acid , Two-Hybrid System Techniques , Ultraviolet Rays , Xenopus , src Homology Domains
20.
Glia ; 15(3): 308-27, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8586466

ABSTRACT

Mutation of the p53 gene is among the most common lesions in a variety of human tumors, including those of the central nervous system. In most instances, mutation of one p53 allele is followed by loss of the remaining wild-type allele, resulting in cells with a complete absence of functional wild-type p53 protein. However, in some situations, such as at initiation of spontaneously arising gliomas or as the germline configuration of patients with the Li-Fraumeni syndrome, cells clearly carry both wild-type and mutant p53 alleles. These observations lead to the hypothesis that p53 mutations can give rise to loss of tumor suppressor functions as well as to gain of oncogenic transformation capabilities. In this review, we define the types of mutations that occur in the p53 gene in various glial tumors, contrast that with the spectra described in other human tumor types, and discuss the biochemistry and physiology of the p53 protein and its ability to regulate and be regulated by other gene products. We use this information to propose roles for p53 in the initiation and progression of human gliomas.


Subject(s)
Brain Neoplasms/genetics , Genes, p53 , Glioma/genetics , Amino Acid Sequence , Brain Neoplasms/pathology , Cell Death , Cell Division , DNA Damage , Germ-Line Mutation , Glioma/pathology , Humans , Molecular Sequence Data , Mutation , Transcription, Genetic , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL