Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 425
Filter
1.
Cell Mol Immunol ; 18(3): 711-722, 2021 03.
Article in English | MEDLINE | ID: mdl-32728200

ABSTRACT

In cancer, myeloid cells have tumor-supporting roles. We reported that the protein GPNMB (glycoprotein nonmetastatic B) was profoundly upregulated in macrophages interacting with tumor cells. Here, using mouse tumor models, we show that macrophage-derived soluble GPNMB increases tumor growth and metastasis in Gpnmb-mutant mice (DBA/2J). GPNMB triggers in the cancer cells the formation of self-renewing spheroids, which are characterized by the expression of cancer stem cell markers, prolonged cell survival and increased tumor-forming ability. Through the CD44 receptor, GPNMB mechanistically activates tumor cells to express the cytokine IL-33 and its receptor IL-1R1L. We also determined that recombinant IL-33 binding to IL-1R1L is sufficient to induce tumor spheroid formation with features of cancer stem cells. Overall, our results reveal a new paracrine axis, GPNMB and IL-33, which is activated during the cross talk of macrophages with tumor cells and eventually promotes cancer cell survival, the expansion of cancer stem cells and the acquisition of a metastatic phenotype.


Subject(s)
Fibrosarcoma/pathology , Hyaluronan Receptors/metabolism , Interleukin-33/metabolism , Lung Neoplasms/pathology , Macrophages/immunology , Membrane Glycoproteins/metabolism , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Cell Proliferation , Fibrosarcoma/etiology , Fibrosarcoma/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Interleukin-33/genetics , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred DBA , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Sarcoma, Experimental/etiology , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
PLoS Genet ; 15(4): e1008039, 2019 04.
Article in English | MEDLINE | ID: mdl-30970016

ABSTRACT

The SWI/SNF-family chromatin remodeling protein ATRX is a tumor suppressor in sarcomas, gliomas and other malignancies. Its loss of function facilitates the alternative lengthening of telomeres (ALT) pathway in tumor cells, while it also affects Polycomb repressive complex 2 (PRC2) silencing of its target genes. To further define the role of inactivating ATRX mutations in carcinogenesis, we knocked out atrx in our previously reported p53/nf1-deficient zebrafish line that develops malignant peripheral nerve sheath tumors and gliomas. Complete inactivation of atrx using CRISPR/Cas9 was lethal in developing fish and resulted in an alpha-thalassemia-like phenotype including reduced alpha-globin expression. In p53/nf1-deficient zebrafish neither peripheral nerve sheath tumors nor gliomas showed accelerated onset in atrx+/- fish, but these fish developed various tumors that were not observed in their atrx+/+ siblings, including epithelioid sarcoma, angiosarcoma, undifferentiated pleomorphic sarcoma and rare types of carcinoma. These cancer types are included in the AACR Genie database of human tumors associated with mutant ATRX, indicating that our zebrafish model reliably mimics a role for ATRX-loss in the early pathogenesis of these human cancer types. RNA-seq of p53/nf1- and p53/nf1/atrx-deficient tumors revealed that down-regulation of telomerase accompanied ALT-mediated lengthening of the telomeres in atrx-mutant samples. Moreover, inactivating mutations in atrx disturbed PRC2-target gene silencing, indicating a connection between ATRX loss and PRC2 dysfunction in cancer development.


Subject(s)
Sarcoma, Experimental/etiology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , X-linked Nuclear Protein/deficiency , X-linked Nuclear Protein/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Carcinogenesis/genetics , Carcinogenesis/metabolism , Disease Models, Animal , Erythropoiesis , Female , Gene Knockout Techniques , Globins/genetics , Humans , Loss of Function Mutation , Male , Neurofibromin 1/deficiency , Neurofibromin 1/genetics , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Telomere Homeostasis/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
3.
Photochem Photobiol Sci ; 18(1): 129-139, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30357250

ABSTRACT

Although infrared radiation (IR) represents more than 50% of the solar radiation reaching the Earth's surface, this waveband has been hardly investigated in terms of tumourigenesis. The objective of the present study was to investigate the influence of IR on ultraviolet B (UVB)-induced carcinogenesis in male and female wild type mice. For this purpose, male and female C57BL/6N mice were subjected to a long-term irradiation protocol. Mice were irradiated once neonatally and from the age of eight weeks for 36 weeks with a cumulative dose of 576 kJ m-2 UVB and/or 78 895 kJ m-2 IR. In order to resemble natural sun irradiation, exposure to physiological doses of UVB and IR was performed simultaneously. Mice were screened for arising lesions twice a week. Lesions were excised and histologically diagnosed. Kaplan-Meier analyses were carried out and lesion counts and cumulated hazard rates for the development of lesions in the UVB and IR + UVB-exposed groups in male and female mice were compared. We found that IR-exposure did not change the number of epithelial malignant tumours in UVB-exposed wild type mice. In combination with IR there was a tendency of more tumours with increased malignancy: 23 vs. seven spindle cell shaped sarcomas and seven vs. two MelanA+/S100+ tumours in groups of 35 C57BL/6 mice. IR did not influence UVB-induced carcinogenesis differently in male and female mice. However, comparing UVB and sham irradiated animals irrespective of IR exposure, UVB-induced non-epithelial tumours arose significantly earlier in male mice than in female mice.


Subject(s)
Carcinogenesis/radiation effects , Infrared Rays/adverse effects , Neoplasms, Radiation-Induced/etiology , Sarcoma, Experimental/etiology , Skin Neoplasms/etiology , Skin/radiation effects , Ultraviolet Rays/adverse effects , Animals , Female , Humans , Kaplan-Meier Estimate , MART-1 Antigen/analysis , Male , Mice , Mice, Inbred C57BL , S100 Proteins/analysis , Sarcoma, Experimental/pathology , Sex Factors , Skin/pathology , Skin Neoplasms/pathology
4.
JCI Insight ; 3(20)2018 10 18.
Article in English | MEDLINE | ID: mdl-30333301

ABSTRACT

Cancer results from the accumulation of genetic mutations in a susceptible cell of origin. We and others have also shown that injury promotes sarcoma development, but how injury cooperates with genetic mutations at the earliest stages of tumor formation is not known. Here, we utilized dual recombinase technology to dissect the complex interplay of the timing of KrasG12D activation, p53 deletion, and muscle injury in sarcomagenesis using a primary mouse model of soft tissue sarcoma. When mutations in oncogenic Kras and p53 are separated by 3 weeks, few sarcomas develop without injury. However, the transformation potential of these tumor-initiating cells can be unmasked by muscle injury. In the absence of Kras mutations, injury of the muscle with global deletion of p53 results in sarcomas with amplification of chromosomal regions encompassing the Met or Yap1 gene. These findings demonstrate a complex interplay between the timing of genetic mutations and perturbations in the tumor microenvironment, which provides insight into the earliest stages of sarcoma development.


Subject(s)
Cell Transformation, Neoplastic/genetics , Muscle Neoplasms/etiology , Muscle, Skeletal/injuries , Sarcoma, Experimental/etiology , Wounds and Injuries/complications , Animals , Cell Line, Tumor , DNA Nucleotidyltransferases/genetics , Disease Models, Animal , Integrases/genetics , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Time Factors , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics
5.
Toxicol Pathol ; 45(6): 774-785, 2017 08.
Article in English | MEDLINE | ID: mdl-29046139

ABSTRACT

The use of immunohistochemical (IHC) staining in determining and/or confirming the cellular origin of poorly differentiated sarcomas was evaluated in this study. Sarcomatous neoplasms were evaluated in a research study conducted in 2 strains of p53+/- haploinsufficient mice. The most common neoplasms were undifferentiated sarcomas, followed by osteosarcomas and rhabdomyosarcomas (RMSs). The RMSs were poorly differentiated and appeared similar to the pleomorphic, or adult type, RMS of humans. All sarcomas stained positive by IHC for the mesenchymal cell intermediate filament vimentin. The RMSs were identified by positive IHC staining for myogenin, a transcription factor specific to skeletal muscle. Osteosarcomas were easily identifiable on hematoxylin and eosin-stained slides; no generally accepted IHC stain specific for bone is presently available. Some of the undifferentiated sarcomas contained numerous macrophages that stained positive for F4/80, a macrophage marker; the positive-staining cells were considered to be infiltrating macrophages. One-third of the neoplasms observed in this study were associated with subcutaneous implanted electronic microchips used for animal identification. Based upon histopathologic evaluation and IHC staining, it was not possible to distinguish neoplasms associated with subcutaneous microchips from neoplasms not associated with microchips.


Subject(s)
Haploinsufficiency/genetics , Rhabdomyosarcoma/pathology , Sarcoma, Experimental/pathology , Tumor Suppressor Protein p53/genetics , Animals , Immunohistochemistry , Male , Mice, Knockout , Rhabdomyosarcoma/etiology , Rhabdomyosarcoma/genetics , Sarcoma, Experimental/etiology , Sarcoma, Experimental/genetics
6.
Int J Radiat Biol ; 88(6): 457-65, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22502876

ABSTRACT

BACKGROUND: Resistance to radiation therapy remains a serious impediment to cancer therapy. We previously reported heterogeneity for clonogenic survival when testing in vitro radiation resistance among single-cell derived clones from a human mesenchymal cancer stem cell model (hMSC). Here we aimed to determine whether this heterogeneity persisted in tumours established from these clones, and whether the response to radiation treatment was principally governed by cell-intrinsic qualities or by factors pertaining to the tumour microenvironment, such as the degree of hypoxia and vascularization. METHODS: Immune-deficient female mice were implanted on the backs with cells from one of the clones. The subsequent tumours were subjected to either radiation treatment or had the tumour microenvironment assayed, when they reached 400 mm³. Radiation was given as a single fraction of 0-15 Gy and the degree of tumour control and time to three times the treatment volume were noted. Tumours used for the microenvironmental assay had intratumoral hypoxia measured by the Eppendorf oxygen electrode and pimonidazole staining, and the extent of vascularization determined by a microvasculature density assay using endothelial-specific staining. RESULTS: All microenvironmental assays indicated a similar degree of hypoxia and vascularization for the selected clones. Nonetheless, the tumours responded differently to radiation treatment since the BB3 clone displayed tumour control at 5, 10 and 15 Gy, whereas tumour control was not seen below 15 Gy with the CE8 clone. CONCLUSION: For tumours that displayed similar degrees of oxygenation and vascularization, the clone-specific in vitro radiation resistance could predict the in vivo response to radiation treatment. These results favor the hypothesis that intrinsic genetic factors can govern radiation resistance in this cancer stem cell model.


Subject(s)
Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/pathology , Radiation Tolerance , Sarcoma, Experimental/etiology , Sarcoma, Experimental/radiotherapy , Tumor Microenvironment/radiation effects , Animals , Cell Line, Tumor , Cell Survival/radiation effects , Female , Humans , Hypoxia/metabolism , Hypoxia/radiotherapy , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Nude , Neoplastic Stem Cells/radiation effects , Oxygen/metabolism , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/pathology , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
7.
PLoS One ; 7(3): e33711, 2012.
Article in English | MEDLINE | ID: mdl-22438986

ABSTRACT

Expression of Piwi proteins is confined to early development and stem cells during which they suppress transposon migration via DNA methylation to ensure genomic stability. Piwi's genomic protective function conflicts with reports that its human ortholog, Hiwi, is expressed in numerous cancers and prognosticates shorter survival. However, the role of Hiwi in tumorigenesis has not been examined. Here we demonstrate that (1) over-expressing Hiwi in sarcoma precursors inhibits their differentiation in vitro and generates sarcomas in vivo; (2) transgenic mice expressing Hiwi (mesodermally restricted) develop sarcomas; and (3) inducible down-regulation of Hiwi in human sarcomas inhibits growth and re-establishes differentiation. Our data indicates that Hiwi is directly tumorigenic and Hiwi-expressing cancers may be addicted to Hiwi expression. We further show that Hiwi associated DNA methylation and cyclin-dependent kinase inhibitor (CDKI) silencing is reversible along with Hiwi-induced tumorigenesis, via DNA-methyltransferase inhibitors. Our studies reveal for the first time not only a novel oncogenic role for Hiwi as a driver of tumorigenesis, but also suggest that the use of epigenetic agents may be clinically beneficial for treatment of tumors that express Hiwi. Additionally, our data showing that Hiwi-associated DNA hyper-methylation with subsequent genetic and epigenetic changes favoring a tumorigenic state reconciles the conundrum of how Hiwi may act appropriately to promote genomic integrity during early development (via transposon silencing) and inappropriately in adult tissues with subsequent tumorigenesis.


Subject(s)
Argonaute Proteins/genetics , Argonaute Proteins/physiology , DNA Methylation/genetics , Sarcoma/etiology , Animals , Base Sequence , Cell Differentiation , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor Proteins/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , DNA Methylation/physiology , Down-Regulation , Gene Expression Profiling , Gene Silencing , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Transgenic , Protein Array Analysis , Sarcoma/genetics , Sarcoma/physiopathology , Sarcoma/therapy , Sarcoma, Experimental/etiology , Sarcoma, Experimental/genetics , Sarcoma, Experimental/physiopathology , Tumor Stem Cell Assay
8.
J Radiat Res ; 53(1): 101-9, 2012.
Article in English | MEDLINE | ID: mdl-22302050

ABSTRACT

Radiotherapy is an integral part of overall cancer therapy. One of the most serious adverse effects of irradiation concern, for long-term survivors, the development of post-radiation sarcoma (PRS) in healthy tissues located within the irradiated area. PRS have bad prognosis and are often detected at a late stage. Therefore, it is obvious that the early detection PRS is a key-point and the development of preclinical models is worthy to evaluate innovative diagnostic and therapeutic procedures. The aim of this study was to develop a spontaneous rodent model of PRS and to evaluate the potency of Positron Emission Tomography (PET) for early detection. Fifteen Wistars rats were irradiated unilateraly on the hindlimb with a single dose of 30 Gy. Sequential analysis was based on observational staging recordings, Computerized Tomography (CT) scanning and PET. Tumors were removed and, histopathological and immunochemistry analyses were performed. Among the irradiated rats, 12 sarcomas (80%) were detected. All tumors occurred naturallty within the irradiated hindlimb and were highly aggressive since most tumors (75%) were successfully transplanted and maintained by serial transplantation into nude mice. Upon serial staging recordings, using PET, was found to enable the detection of PRS earlier after irradiation than with the other methods (i.e. 11.9 ± 1.8 vs 12.9 ± 2.6 months). These results confirmed the interest of experimental models of PRS for the preclinical evaluation of innovative diagnostic strategies and confirmed the potency of PET for early detection of PRS. This preclinical model of PRS can also be proposed for the evaluation of therapeutic strategies.


Subject(s)
Models, Animal , Neoplasms, Radiation-Induced/diagnostic imaging , Positron-Emission Tomography , Sarcoma, Experimental/diagnostic imaging , Soft Tissue Neoplasms/diagnostic imaging , Animals , Biomarkers, Tumor/analysis , Cobalt Radioisotopes , Early Diagnosis , Fluorine Radioisotopes , Fluorodeoxyglucose F18 , Hindlimb , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplasms, Radiation-Induced/pathology , Radiopharmaceuticals , Rats , Rats, Wistar , Sarcoma, Experimental/etiology , Sarcoma, Experimental/pathology , Soft Tissue Neoplasms/etiology , Soft Tissue Neoplasms/pathology , Tomography, X-Ray Computed
9.
Virchows Arch ; 454(2): 195-201, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19096872

ABSTRACT

Sarcomagenesis, in contrast to carcinogenesis, is poorly understood. Microsatellite instability has been implicated in the development of many cancers, in particular those associated with chronic inflammatory conditions. In an experimental animal model, rats developed not only a peri-implantational chronic inflammatory reaction, but also malignant mesenchymal tumors in response to different biomaterials. Therefore, it was the aim of our study to test if the development of biomaterial-induced sarcomas is characterized by a mutator phenotype. A multiplex-PCR approach was designed to screen biomaterial-induced sarcomas for the presence of microsatellite instability. Seven different microsatellite loci were tested in ten tumors for microsatellite instability using a fluorochrome-labelled multiplex-PCR and subsequent fragment analysis. All tumors provided a microsatellite-stable phenotype at all loci tested. Our data suggest that microsatellite instability is rarely or not at all a feature of malignant transformation of biomaterial-induced soft tissue tumors. Thus, there is no evidence that a mutator phenotype is a hallmark of biomaterial-induced sarcomagenesis.


Subject(s)
Biocompatible Materials/adverse effects , Microsatellite Instability , Sarcoma, Experimental/etiology , Animals , Phenotype , Polymerase Chain Reaction , Rats , Rats, Inbred F344
10.
Biochemistry (Mosc) ; 73(7): 763-75, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18707584

ABSTRACT

This review deals with the contemporary investigations of carcinogenesis induced by foreign bodies. The main attention is given to the interactions of macrophages with an implanted foreign body and their possible role in tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , Foreign Bodies/complications , Animals , Macrophages/immunology , Macrophages/metabolism , Mice , Rats , Sarcoma, Experimental/etiology , Sarcoma, Experimental/pathology
11.
Eur J Med Chem ; 43(4): 741-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17658668

ABSTRACT

The synthesis of a ligand containing a nitrobenzyl group as bioreductive pharmacophore and the preparation of the corresponding technetium and rhenium complexes are presented. (99m)Tc labelling was performed in high yield (>90%) by ligand substitution using fac-[(99m)Tc(CO)(3)(H(2)O)(3)](+) as precursor. The structure of the technetium complex was established by chromatographic comparison with the analogous rhenium compound which was fully characterized by elemental analysis, spectroscopic methods and X-ray crystallography. Reduction potential of the rhenium complex was in the characteristic range for bioreductive compounds. Biodistribution in normal mice was characterized by fast blood and soft tissue depuration and combined excretion via the hepatobiliary and urinary systems. Tumour uptake was low, probably due to low lipophilicity but tumour/muscle ratios were favourable as a consequence of high excretion.


Subject(s)
Organotechnetium Compounds/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Rhenium/chemistry , Animals , Cell Hypoxia , Crystallography, X-Ray , Diagnostic Imaging , In Vitro Techniques , Liver/metabolism , Lung/metabolism , Mice , Models, Molecular , Molecular Structure , Organotechnetium Compounds/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/etiology , Sarcoma, Experimental/metabolism , Spectrophotometry, Infrared , Structure-Activity Relationship , Tissue Distribution , Xenograft Model Antitumor Assays
12.
Neoplasma ; 54(2): 173-9, 2007.
Article in English | MEDLINE | ID: mdl-17319793

ABSTRACT

Excessive bone formation occurring in such conditions as paravertebral ligamentous ossification, hallux osteophytes or some neoplastic tumors, presents a significant problem, both epidemiological and clinical. Since pathogenesis of this disorder is still unclear, we studied its mechanism in experimental model utilizing inducible orthotopic osteogenesis. Periosteal bone apposition stimulated by Moloney sarcoma is characterized by unusually high volume of new bone tissue appearing subperiosteally in the bone adjacent to the tumor. Genes engaged in this growth have not been characterized so far. Here we show the results of mRNA Representation Difference Analysis in Moloney sarcoma, which reveal high expression of four genes coding extracellular matrix proteins: osteopontin, fibronectin, stromelysin-1 and tenascin. These findings suggest that the uncommon dynamics of the Moloney sarcoma-induced osteogenesis depends on high expression of these extracellular matrix proteins.


Subject(s)
Fibronectins/metabolism , Matrix Metalloproteinase 3/metabolism , Moloney murine sarcoma virus/physiology , Osteogenesis/physiology , Osteopontin/metabolism , Periosteum/virology , Sarcoma, Experimental/etiology , Tenascin/metabolism , Animals , Blotting, Northern , Mice , Mice, Inbred BALB C
13.
Carcinogenesis ; 28(1): 191-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16857722

ABSTRACT

Chronic inflammation is a recognized risk factor for human cancer at various sites because of persistent oxidative and nitrative tissue damage. Trp53+/- mice show the predisposition to tumor development, such as sarcomas and lymphomas, compared with Trp53+/+ mice. We investigated the effects of chronic inflammation, especially oxidative and nitrative stress, induced by subcutaneous implantation of a plastic plate (10 x 5 x 1 mm) as a foreign body on tumorigenesis in Trp53+/- and Trp53+/+ mice. The plastic plates were implanted at the age of about 11 weeks. Thirty out of 38 Trp53+/- mice (79%) developed sarcomas around the implant (mean time of tumor appearance was 45.8 +/- 12.0 weeks of age), whereas only one of 10 Trp53+/+ mice with an implant (10%) developed a tumor, at 56 weeks. No sarcomas developed at a sham-operation site. Two of 10 Trp53+/- mice with no implant (20%) also developed three sarcomas spontaneously at 77, 81 and 84 weeks. Increased immunostaining for markers of oxidative and nitrative stress (8-oxo-7,8-dihydro-2'-deoxyguanosine, 8-nitroguanine and 3-nitrotyrosine) and expression of inducible nitric oxide synthase in tumor cells and inflammatory cells were detected in implant-induced sarcomas compared with spontaneous sarcomas in Trp53+/- mice. Furthermore, p53 loss of heterozygosity was observed in 26 out of 29 implant-induced sarcomas (90%). These results indicate that implanted foreign bodies significantly enhanced sarcoma development in Trp53+/- mice, and this may be associated with increased oxidaive and nitrative stress. Loss of the remaining wild-type p53 allele and loss of p53 function appears to be, at least in part, underlying molecular mechanisms during the development of sarcomas at the implantation site in Trp53+/- mice. Such implant-induced sarcoma development in Trp53+/- mice could be useful for studying molecular mechanisms and developing new strategies for chemoprevention in human carcinogenesis induced by chronic inflammation and/or foreign bodies.


Subject(s)
Foreign Bodies/metabolism , Guanine/analogs & derivatives , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress , Sarcoma, Experimental/etiology , Tumor Suppressor Protein p53/physiology , Animals , Guanine/metabolism , Humans , Inflammation/complications , Inflammation/metabolism , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Tumor Suppressor Protein p53/genetics
14.
J Biol Chem ; 279(6): 4877-86, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14612455

ABSTRACT

The Mdm2 gene is amplified in approximately one-third of human sarcomas and overexpressed in a variety of other human cancers. Mdm2 functions as an oncoprotein, in part, by acting as a negative regulator of the p53 tumor suppressor protein. Multiple spliced forms of Mdm2 transcripts have been observed in human tumors; however, the contribution of these variant transcripts to tumorigenesis is unknown. In this report, we isolate alternative splice forms of Mdm2 transcripts from sarcomas that spontaneously arise in Mdm2-overexpressing mice, including Mdm2-b, the splice form most commonly observed in human cancers. Transduction of Mdm2-b into a variety of cell types reveals that Mdm2-b promotes p53-independent cell growth, inhibits apoptosis, and up-regulates the RelA subunit of NFkappaB. Furthermore, expression of Mdm2-b induces tumor formation in transgenic mice. These results identify a p53-independent role for Mdm2 and determine that an alternate spliced form of Mdm2 can contribute to formation of cancer via a p53-independent mechanism. These findings also provide a rationale for the poorer prognosis of those patients presenting with tumors harboring multiple Mdm2 transcripts.


Subject(s)
Alternative Splicing , Nuclear Proteins , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Sarcoma, Experimental/etiology , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Apoptosis , Base Sequence , Cell Division , DNA, Complementary/genetics , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , NIH 3T3 Cells , Proto-Oncogene Proteins c-mdm2 , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Sequence Homology, Amino Acid , Transfection
15.
Inhal Toxicol ; 14(7): 685-703, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12122570

ABSTRACT

Cellulose fibers, along with many other organic fibers, are durable. Therefore, if inhaled, they have the potential to persist within the lung, and may then cause disease. Here we report the effects of injecting high-purity cellulose fibers into the abdominal cavity of rats. A respirable fraction of cellulose fiber was collected from an aerosol of a thermo-mechanically-processed wood pulp. A sample of respirable crocidolite asbestos, known to produce mesotheliomas in rats, was used as a positive control. Total doses of 10(6), 10(7), 10(8), or 10(9) WHO fibers were injected intraperitoneally as 3 weekly aliquots. A negative control was provided by phosphate-buffered saline used to suspend the fibers for injection. There were 50 rats per treatment group except for the 10(8) and 10(9) fibers crocidolite groups which were reduced to 26 rats because of the expectation of high tumor incidence in these groups. The two higher doses of crocidolite asbestos caused greatly reduced survival compared to the saline controls. With cellulose there was a much less marked effect on survival. In the highest dose cellulose group, multiple large nodules (granulomas) and widespread adhesions (bands of new tissue connecting organs to each other and to the abdominal wall) were present in all animals. Granulomas were not observed in the 10(9) fibers crocidolite group. More than 80% of animals in the 10(8) and 10(9) crocidolite asbestos groups had mesotheliomas, a type of tumor sometimes observed in people exposed to asbestos. In contrast, there were only 2 animals in the cellulose groups with mesothelioma tumors, 1 in the 10(7) and 1 in the 10(8) groups. However, 9 (18%) of the 10(9) cellulose group had malignant tumors that, in contrast to the usual pattern of mesothelioma development following treatment with mineral fibers in rats, showed no obvious involvement of mesothelial tissues, were not associated with blood-stained ascites fluid, and were thus classified as sarcomas. This study has demonstrated that a high dose of cellulose fibers is capable of producing tumors when injected into the abdominal cavity of rats.


Subject(s)
Carcinogens/toxicity , Cellulose/toxicity , Peritoneal Neoplasms/etiology , Sarcoma, Experimental/etiology , Air Pollutants, Occupational/toxicity , Animals , Asbestos, Crocidolite/toxicity , Carcinogenicity Tests , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Maximum Tolerated Dose , Mesothelioma/etiology , Mineral Fibers/toxicity , Particle Size , Pleural Neoplasms/etiology , Rats , Rats, Wistar
16.
Cancer Res ; 62(5): 1489-95, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11888925

ABSTRACT

AUF1/heterogeneous nuclear ribonucleoprotein D (hnRNPD) binds to adenylate uridylate-rich elements contained in the 3' untranslated region of many short-lived mRNAs. This binding has been shown in vitro to control the stability of adenylate uridylate-rich element-containing mRNAs, including mRNAs encoding proto-oncogenes, cytokines, or other signaling molecules. However, no studies have yet been undertaken to identify the mRNAs subject to AUF1-mediated regulation in vivo. The purpose of our study was to investigate the biological functions of AUF1. Thus, we derived transgenic (Tg) mice, which overexpress one isoform of AUF1, the p37(AUF1). Mice of the three Tg lines analyzed exhibit altered levels of expression of several target mRNAs, such as c-myc, c-jun, c-fos, granulocyte macrophage colony-stimulating factor, and tumor necrosis factor alpha. The Tg line with the highest amount of Tg p37(AUF1) protein developed sarcomas. The tumors strongly expressed AUF1 Tg protein and Cyclin D1. Taken together, our data show that: (a) AUF1 is a key regulatory factor of gene expression in vivo; and (b) the deregulation of this heterogeneous nuclear ribonucleoprotein leads to tumorigenesis.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D , RNA-Binding Proteins/genetics , Sarcoma, Experimental/etiology , Actins/genetics , Animals , Cyclin D1/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Heterogeneous Nuclear Ribonucleoprotein D0 , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , RNA, Messenger/analysis
17.
Int J Radiat Biol ; 75(10): 1251-6, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10549601

ABSTRACT

PURPOSE: In view of current interest in the biological effects of amplitude-modulated microwaves arising from the rapid development of mobile communications, the effects of low-level microwaves on cancer development were investigated using a rat sarcoma model. MATERIALS AND METHODS: Two-month-old female Sprague-Dawley rats were treated by injection of benzo(a)pyrene and irradiated with GSM (Global System for Mobile)-modulated 900-MHz microwaves in an anechoic chamber at 55 or 200 microW cm(-2) (75 and 270 mW kg(-1) average whole-body SAR, 2h daily for 2 weeks). Rats were exposed from day 20, 40 or 75 after carcinogen injection. Additional groups of rats were sham-exposed in a second anechoic chamber. Anti-phosphatidylinositol autoantibody levels were evaluated in sera to monitor malignant transformation. RESULTS: Microwave exposure had no effect on the development of tumours. No acceleration or delays in tumour onset were observed. Animal survival was not modified and serum autoantibody levels were similar in exposed and sham-exposed groups. CONCLUSION: Low-level GSM microwave exposure of rat bearing benzo(a)pyrene-induced tumours had no effect on auto-antibody levels, tumour appearance and survival. The low exposure levels used here correspond to exposure limits for whole-body exposure of humans.


Subject(s)
Microwaves/adverse effects , Sarcoma, Experimental/etiology , Animals , Autoantibodies/blood , Benzo(a)pyrene/toxicity , Carcinogens/toxicity , Female , Rats , Rats, Sprague-Dawley , Sarcoma, Experimental/chemically induced , Sarcoma, Experimental/immunology , Survival Rate , Telecommunications
18.
Microvasc Res ; 58(3): 312-28, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10527772

ABSTRACT

We studied microvessel organization in five brain tumor models (ENU, MSV, RG-2, S635cl15, and D-54MG) and normal brain, including microvessel diameter (LMVD), intermicrovessel distance (IMVD), microvessel density (MVD), surface area (S(v)), and orientation. LMVD and IMVD were larger and MVD was lower in tumors than normal brain. S(v) in tumors overlapped normal brain values and orientation was random in both tumors and brain. ENU and RG-2 tumors and brain were studied by electron microscopy. Tumor microvessel wall was thicker than that of brain. ENU and normal brain microvessels were continuous and nonfenestrated. RG-2 microvessels contained fenestrations and endothelial gaps; the latter had a maximum major axis of 3.0 microm. Based on anatomic measurements, the pore area of RG-2 tumors was estimated at 7.4 x 10(-6) cm(2) g(-1) from fenestrations and 3.5 x 10(-5) cm(2) g(-1) from endothelial gaps. Increased permeability of RG-2 microvessels to macromolecules is most likely attributable to endothelial gaps. Three microvessel populations may occur in brain tumors: (1) continuous nonfenestrated, (2) continuous fenestrated, and (3) discontinuous (with or without fenestrations). The first group may be unique to brain tumors; the latter two are similar to microvessels found in systemic tumors. Since structure-function properties of brain tumor microvessels will affect drug delivery, studies of microvessel function should be incorporated into clinical trials of brain tumor therapy, especially those using macromolecules.


Subject(s)
Brain Neoplasms/blood supply , Animals , Brain/blood supply , Brain Neoplasms/etiology , Carcinogens/toxicity , Endothelium, Vascular/pathology , Ethylnitrosourea/toxicity , Female , Intercellular Junctions/pathology , Microcirculation/pathology , Microcirculation/physiopathology , Neovascularization, Pathologic , Pregnancy , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Retroviridae Infections/etiology , Retroviridae Infections/pathology , Sarcoma Viruses, Murine , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/etiology , Tumor Cells, Cultured , Tumor Virus Infections/etiology , Tumor Virus Infections/pathology
19.
Toxicol Pathol ; 27(5): 519-27, 1999.
Article in English | MEDLINE | ID: mdl-10528631

ABSTRACT

Heterozygous p53+/- transgenic mice are being studied for utility as a short-term alternative model to the 2-yr rodent carcinogenicity bioassay. During a 26-wk study to assess the potential carcinogenicity of oxymetholone using p-cresidine as a positive control, glass/polypropylene microchips (radio transponder identification devices) were subcutaneously implanted into male and female p53+/- mice. During week 15, the first palpable mass was clinically observed at an implant site. This rapidly growing mass virtually quadrupled in size by week 25. Microscopic examination of all implant sites revealed that 18 of 177 animals had a subcutaneous histologically malignant sarcoma. The neoplasms were characterized as undifferentiated sarcomas unrelated to drug treatment, as indicated by the relatively even distribution among dose groups, including controls. An unusual preneoplastic mesenchymal change characterized by the term "mesenchymal dysplasia" was present in most groups and was considered to be a prodromal change to sarcoma development. The tumors were observed to arise from dysplastic mesenchymal tissue that developed within the tissue capsule surrounding the transponder. The preneoplastic changes, including mesenchymal dysplasia, appeared to arise at the transponder's plastic anchoring barb and then progressed as a neoplasm to eventually surround the entire microchip. Capsule membrane endothelialization, inflammation, mesenchymal basophilia and dysplasia, and sarcoma were considered unequivocal preneoplastic/neoplastic responses to the transponder and were not related to treatment with either oxymetholone or p-cresidine.


Subject(s)
Genes, p53/genetics , Polypropylenes/adverse effects , Sarcoma, Experimental/pathology , Transducers/adverse effects , Anabolic Agents/toxicity , Animals , Carcinogens/toxicity , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Female , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxymetholone/toxicity , Sarcoma, Experimental/etiology , Sarcoma, Experimental/genetics , Skin/drug effects , Skin/pathology , Skin/ultrastructure , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Survival Analysis
20.
J Immunol ; 163(8): 4224-31, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10510359

ABSTRACT

Proteins involved in class I MHC (MHC-I) Ag processing, such as the TAP, are deficient in some human tumor cells. This suggests that antitumor responses by CD8 T cells provide selection pressure to favor outgrowth of cells with defective processing of tumor Ags. Nonetheless, this evidence is only correlative, and controlled in vivo experiments have been lacking to demonstrate that TAP deficiency promotes survival of tumor cells. To explore the role of Ag processing defects in tumor progression, matched panels of TAP1-positive and TAP1-negative tumor cell lines were generated from a parental transformed murine fibroblast line. Inoculation of C57BL/6 mice with TAP1-negative cells produced large and persistent tumors. In contrast, TAP1-positive cells did not generate lasting tumors, although small tumors were detected transiently and regressed spontaneously. Both TAP1-positive and TAP1-negative cells produced tumors in athymic mice, confirming that TAP-dependent differences in tumorigenicity were due to T cell-dependent immune responses. Inoculation of C57BL/6 mice with mixtures of TAP1-positive and TAP1-negative cells produced tumors composed exclusively of TAP1-negative cells, indicating in vivo selection for cells with TAP deficiency. Thus, loss of TAP function allows some tumor cells to avoid T cell-dependent elimination, resulting in selection for tumor cells with deficient Ag processing.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antigen Presentation/immunology , Cell Transformation, Neoplastic/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Antigen Presentation/genetics , Cell Division/immunology , Cell Line, Transformed , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Histocompatibility Antigens Class I/biosynthesis , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , RNA, Messenger/biosynthesis , Sarcoma, Experimental/etiology , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology , T-Lymphocytes/immunology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...