Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Leukemia ; 30(2): 390-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26286116

ABSTRACT

We reported that p62 (sequestosome 1) serves as a signaling hub in bone marrow stromal cells (BMSCs) for the formation of signaling complexes, including NFκB, p38MAPK and JNK, that are involved in the increased osteoclastogenesis and multiple myeloma (MM) cell growth induced by BMSCs that are key contributors to multiple myeloma bone disease (MMBD), and demonstrated that the ZZ domain of p62 (p62-ZZ) is required for BMSC enhancement of MMBD. We recently identified a novel p62-ZZ inhibitor, XRK3F2, which inhibits MM cell growth and BMSC growth enhancement of human MM cells. In the current study, we evaluate the relative specificity of XRK3F2 for p62-ZZ, characterize XRK3F2's capacity to inhibit growth of primary MM cells and human MM cell lines, and test the in vivo effects of XRK3F2 in the immunocompetent 5TGM1 MM model. We found that XRK3F2 induces dramatic cortical bone formation that is restricted to MM containing bones and blocked the effects and upregulation of tumor necrosis factor alpha (TNFα), an osteoblast (OB) differentiation inhibitor that is increased in the MM bone marrow microenvironment and utilizes signaling complexes formed on p62-ZZ, in BMSC. Interestingly, XRK3F2 had no effect on non-MM bearing bone. These results demonstrate that targeting p62 in MM models has profound effects on MMBD.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Multiple Myeloma/drug therapy , Osteoclasts/drug effects , Osteogenesis/drug effects , Adaptor Proteins, Signal Transducing/chemistry , Aged , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Multiple Myeloma/pathology , Osteoclasts/physiology , Sequestosome-1 Protein , Tumor Necrosis Factor-alpha/pharmacology
3.
Cell Death Differ ; 19(1): 144-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21660048

ABSTRACT

Autophagy is implicated in regulating cell death in activated T cells, but the underlying mechanism is unclear. Here, we show that inhibition of autophagy via Beclin 1 gene deletion in T cells leads to rampant apoptosis in these cells upon TCR stimulation. Beclin 1-deficient mice fail to mount autoreactive T-cell responses and are resistant to experimental autoimmune encephalomyelitis. Compared with Th17 cells, Th1 cells are much more susceptible to cell death upon Beclin 1 deletion. Cell death proteins are highly increased in Beclin 1-deficient T cells and inhibition of caspases and genetic deletion of Bim reverse apoptosis. In addition, p62/sequestosome 1 binds to caspase-8 but does not control levels of procaspase-8 or other cell death-related proteins. These results establish a direct role of autophagy in inhibiting the programmed cell death through degradation of apoptosis proteins in activated T cells.


Subject(s)
Autophagy , Proteolysis , T-Lymphocytes/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Beclin-1 , Caspase 8/metabolism , Caspases/metabolism , Cell Survival , Heat-Shock Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Sequestosome-1 Protein , Th1 Cells/physiology , Th17 Cells/physiology
4.
Endocrinology ; 142(7): 2898-905, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11416009

ABSTRACT

Pagetic osteoclasts (OCLs) are abnormal in size and contain paramyxoviral-like nuclear inclusions that cross-react with antibodies to measles virus (MV). However, the role that MV infection plays in Paget's disease is unknown, because no animal model of Paget's disease is available. Therefore, we targeted a cellular MV receptor, human CD46 (hCD46), to cells in the OCL lineage in transgenic mice using the mouse tartrate-resistant acid phosphatase (TRAP) gene promoter. In vitro infection of OCL precursors from hCD46 transgenic mice with MV significantly increased OCL formation in bone marrow cultures. The numbers of TRAP-positive mononuclear cells and CFU-GM, the earliest identifiable OCL precursor, were also significantly increased. MV-infected OCLs formed from hCD46 marrow were increased in size, contained markedly increased numbers of nuclei, and had increased bone-resorbing capacity per OCL compared with OCLs formed from marrow of nontransgenic littermates. Furthermore, IL-6 and 24-hydroxylase messenger RNA expression levels were increased in MV-infected hCD46 transgenic mouse bone marrow cultures. Treatment of MV-infected hCD46 marrow cultures with a neutralizing antibody to IL-6 blocked the increased OCL formation seen in these cultures. These data demonstrate that MV infection of OCL precursors results in OCLs that have many features of pagetic OCLs, that the enhanced OCL formation is in part mediated by increased IL-6 expression induced by MV infection, and suggest that the hCD46 transgenic mouse may be a useful model for examining the effects of MV infection on OCL formation in vivo.


Subject(s)
Antigens, CD/metabolism , Measles/pathology , Membrane Glycoproteins/metabolism , Osteitis Deformans/pathology , Osteoclasts/pathology , Stem Cells/pathology , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Animals , Antigens, CD/genetics , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Resorption/physiopathology , Cell Division , Humans , Interleukin-6/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic/genetics , Osteitis Deformans/physiopathology , Osteoclasts/physiology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tartrate-Resistant Acid Phosphatase
5.
J Periodontol ; 71(1): 63-72, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10695940

ABSTRACT

BACKGROUND: Proper formation of cementum, a mineralized tissue lining the tooth root surface, is required for development of a functional periodontal ligament. Further, the presence of healthy cementum is considered to be an important criterion for predictable restoration of periodontal tissues lost as a consequence of disease. Despite the significance of cementum to general oral health, the mechanisms controlling development and regeneration of this tissue are not well understood and research has been hampered by the lack of adequate in vitro experimental models. METHODS: In an effort to establish cementoblast cell populations, without the trappings of a heterogeneous population containing periodontal ligament (PDL) cells, cells were obtained from the root surface of first mandibular molars of OC-TAg transgenic mice. These mice contain the SV40 large T-antigen (TAg) under control of the osteocalcin (OC) promoter. Therefore, only cells that express OC also express TAg and are immortalized in vitro. Based on results of prior in situ studies, OC is expressed by cementoblasts during root development, but not by cells within the PDL. Consequently, when populations are isolated from developing molars using collagenase/trypsin digestion, only cementoblasts, not PDL cells, are immortalized and thus, will survive in culture. RESULTS: The resulting immortalized cementoblast population (OC/CM) expressed bone sialoprotein (BSP), osteopontin (OPN), and OC, markers selective to cells lining the root surface. These cells also expressed type I and XII collagen and type I PTH/PTHrP receptor (PTH1R). In addition to expression of genes associated with cementoblasts, OC/CM cells promoted mineral nodule formation and exhibited a PTHrP mediated cAMP response. CONCLUSIONS: This approach for establishing cementoblasts in vitro provides a model to study cementogenesis as required to enhance our knowledge of the mechanisms controlling development, maintenance, and regeneration of periodontal tissues.


Subject(s)
Cementogenesis , Dental Cementum/cytology , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Adhesion/genetics , Cells, Cultured , Collagen/genetics , Cyclic AMP/metabolism , Dental Cementum/metabolism , Dental Cementum/physiology , Disease Models, Animal , Integrin-Binding Sialoprotein , Mice , Mice, Inbred Strains , Mice, Transgenic , Minerals/metabolism , Odontogenesis/physiology , Osteocalcin/genetics , Osteopontin , Parathyroid Hormone/genetics , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Receptors, Parathyroid Hormone/genetics , Regeneration , Sialoglycoproteins/genetics , Tooth Root/cytology , Tooth Root/physiology
6.
Oncogene ; 19(8): 1114-22, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10713698

ABSTRACT

Several transgenic mouse tumor models were utilized to explore how specific genetic alterations affect the tumor cell response to chemotherapeutic agents in vivo. Specifically, MMTV-ras transgenic mice were interbred to p53 knock-out mice to create a model for assessing the role of p53 in chemotherapeutic responses. In addition, MMTV-ras tumors were compared to MMTV-myc and MMTV-ras/myc tumors. Mice of each genotype reproducibly develop mammary and/or salivary tumors, but tumor growth dynamics vary considerably between genotypes. MMTV-ras/p53-/- tumors exhibit higher S phase fractions than MMTV-ras/p53+/+ tumors, although both tumor types display very low apoptosis levels. In contrast, MMTV-myc tumors exhibit both high S phase fractions and spontaneous apoptosis levels. Tumor-bearing mice of each genotype were treated with either doxorubicin or paclitaxel, and effects on overall tumor growth, cell cycle distribution and apoptosis were evaluated. Surprisingly, neither agent efficiently induced apoptosis in any of the tumor models, including those with wildtype p53. Rather, tumor responses were mediated primarily by changes in cell cycle distribution. However, the spontaneous apoptosis levels did serve as a predictor of tumor growth response, in that only those tumors with high pretreatment apoptosis levels underwent significant regression following treatment with either agent.


Subject(s)
Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Division/drug effects , Cell Division/genetics , Doxorubicin/pharmacology , Female , Genes, ras , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred Strains , Mice, Transgenic , Paclitaxel/pharmacology , Tumor Suppressor Protein p53/genetics
7.
J Clin Oncol ; 17(11): 3631-52, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10550163

ABSTRACT

Ras proteins are guanine nucleotide-binding proteins that play pivotal roles in the control of normal and transformed cell growth and are among the most intensively studied proteins of the past decade. After stimulation by various growth factors and cytokines, Ras activates several downstream effectors, including the Raf-1/mitogen-activated protein kinase pathway and the Rac/Rho pathway. In approximately 30% of human cancers, including a substantial proportion of pancreatic and colon adenocarcinomas, mutated ras genes produce mutated proteins that remain locked in an active state, thereby relaying uncontrolled proliferative signals. Ras undergoes several posttranslational modifications that facilitate its attachment to the inner surface of the plasma membrane. The first-and most critical-modification is the addition of a farnesyl isoprenoid moiety in a reaction catalyzed by the enzyme protein farnesyltransferase (FTase). It follows that inhibiting FTase would prevent Ras from maturing into its biologically active form, and FTase is of considerable interest as a potential therapeutic target. Different classes of FTase inhibitors have been identified that block farnesylation of Ras, reverse Ras-mediated cell transformation in human cell lines, and inhibit the growth of human tumor cells in nude mice. In transgenic mice with established tumors, FTase inhibitors cause regression in some tumors, which appears to be mediated through both apoptosis and cell cycle regulation. FTase inhibitors have been well tolerated in animal studies and do not produce the generalized cytotoxic effects in normal tissues that are a major limitation of most conventional anticancer agents. There are ongoing clinical evaluations of FTase inhibitors to determine the feasibility of administering them on dose schedules like those that portend optimal therapeutic indices in preclinical studies. Because of the unique biologic aspects of FTase, designing disease-directed phase II and III evaluations of their effectiveness presents formidable challenges.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Neoplasms , ras Proteins/physiology , Alkyl and Aryl Transferases/metabolism , Animals , Enzyme Inhibitors/pharmacology , Humans , Mice , Mutation , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/prevention & control , Polyisoprenyl Phosphates/antagonists & inhibitors , Polyisoprenyl Phosphates/metabolism , Protein Prenylation/drug effects , Proto-Oncogene Proteins p21(ras)/physiology , Sesquiterpenes , Signal Transduction/drug effects , ras Proteins/metabolism
8.
Surv Ophthalmol ; 43(6): 508-18, 1999.
Article in English | MEDLINE | ID: mdl-10416793

ABSTRACT

Retinoblastoma, the most common intraocular malignancy ill childhood, has served as a paradigm for the study of genetic mechanisms of oncogenesis. The retinoblastoma susceptibility gene RB1 was the first tumor suppressor gene to be cloned, and genetic and molecular biologic studies of this tumor have greatly expanded the understanding of the mechanics of tumorigenesis. Human retinoblastoma has essentially no naturally occuring animal counterpart. The development of transgenic murine models of retinoblastoma have created an experimental tool for manipulation of a tumor gene system in vivo. These models have also enabled studies of new therapeutic modalities. This review outlines the development of the transgenic murine models of retinoblastoma, together with the genetic mechanisms of retinoblastoma origin. Current therapeutic innovations developed by means of the transgenic models are described.


Subject(s)
Disease Models, Animal , Eye Diseases, Hereditary/pathology , Mice, Transgenic , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Animals , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/therapy , Genes, Retinoblastoma/genetics , Genes, p53/genetics , Humans , Mice , Retinal Neoplasms/genetics , Retinal Neoplasms/therapy , Retinoblastoma/genetics , Retinoblastoma/therapy , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics
9.
Invest Ophthalmol Vis Sci ; 39(13): 2800-5, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9856795

ABSTRACT

PURPOSE: To describe the cell of origin, tumor progression, light and electron microscopic appearance, immunohistochemical properties, and response to frequently used anticancer therapies in two transgenic models of intraocular melanoma. METHODS: Two lines of transgenic mice that develop pigmented intraocular tumors were produced with the SV40 T and t antigens under the control of the mouse tyrosinase gene. Tumors were sequentially studied and characterized by light microscopy, electron microscopy, and immunohistochemistry stains. Tumor response to two cycles of dacarbazine was assessed on the basis of tumor size in one group of animals. Response to external beam irradiation was measured by survival time in other animals. RESULTS: Two lines of transgenic mice developed bilateral intraocular tumors with complete penetrance and without primary cutaneous melanomas. Tumors developed first in the retinal pigment epithelial layer, with subsequent retinal and choroidal invasion, extraocular extension, and metastasis. Tumors stained positive for S-100, HMB-45, and Fas-ligand. Electron microscopy revealed polarization of tumor cells with basement membrane formation, microvilli, immature melanosomes, and abundant endoplasmic reticulum. Dacarbazine significantly reduced tumor size in these mice, and a trend toward dose-dependent decrease in survival was found with external beam irradiation. CONCLUSIONS: Tumors developed from the retinal pigment epithelium. Their histology and growth, however, closely resembled that of human choroidal melanoma. This model may be a useful tool for future studies of endogenous primary pigmented tumors limited to the eye. Response to standard therapies suggests it can serve as a model with which to evaluate therapeutic modalities.


Subject(s)
Melanoma/pathology , Uveal Neoplasms/pathology , Animals , Antigens, Neoplasm , Antigens, Polyomavirus Transforming/metabolism , Dacarbazine/pharmacology , Fas Ligand Protein , Female , Immunohistochemistry , In Situ Hybridization , Male , Melanoma/metabolism , Melanoma/therapy , Melanoma-Specific Antigens , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/metabolism , Pigment Epithelium of Eye/ultrastructure , S100 Proteins/metabolism , Uveal Neoplasms/metabolism , Uveal Neoplasms/therapy
10.
J Biol Chem ; 273(32): 20448-55, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9685399

ABSTRACT

Neu (c-erbB2) is a proto-oncogene product that encodes an epidermal growth factor-like receptor tyrosine kinase. Amplification of wild-type c-Neu and mutational activation of Neu (Neu T) have been implicated in oncogenic transformation of cultured fibroblasts and mammary tumorigenesis in vivo. Here, we examine the relationship between Neu tyrosine kinase activity and caveolin-1 protein expression in vitro and in vivo. Recent studies have suggested that caveolins may function as negative regulators of signal transduction. Our current results show that mutational activation of c-Neu down-regulates caveolin-1 protein expression, but not caveolin-2, in cultured NIH 3T3 and Rat 1 cells. Conversely, recombinant overexpression of caveolin-1 blocks Neu-mediated signal transduction in vivo. These results suggest a reciprocal relationship between c-Neu tyrosine kinase activity and caveolin-1 protein expression. We next analyzed a variety of caveolin-1 deletion mutants to map this caveolin-1-dependent inhibitory activity to a given region of the caveolin-1 molecule. Results from this mutational analysis show that this functional in vivo inhibitory activity is contained within caveolin-1 residues 32-95. In accordance with these in vivo studies, a 20-amino acid peptide derived from this region (the caveolin-1 scaffolding domain) was sufficient to inhibit Neu autophosphorylation in an in vitro kinase assay. To further confirm or refute the relevance of our findings in vivo, we next examined the expression levels of caveolin-1 in mammary tumors derived from c-Neu transgenic mice. Our results indicate that dramatic reduction of caveolin-1 expression occurs in mammary tumors derived from c-Neu-expressing transgenic mice and other transgenic mice expressing downstream effectors of Neu-mediated signal transduction, such as Src and Ras. Taken together, our data suggest that a novel form of reciprocal negative regulation exists between c-Neu and caveolin-1.


Subject(s)
Caveolins , Gene Expression Regulation, Neoplastic/genetics , Membrane Proteins/physiology , Receptor, ErbB-2/physiology , Animals , Caveolin 1/pharmacology , Cell Line , Ceramides/pharmacology , DNA Mutational Analysis , Down-Regulation/physiology , Immunohistochemistry , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Peptide Fragments/pharmacology , Phosphorylation , Proto-Oncogene Mas , Receptor Protein-Tyrosine Kinases/physiology , Sequence Deletion/genetics , Signal Transduction/physiology , Transformation, Genetic/genetics
11.
J Clin Invest ; 102(1): 88-97, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9649561

ABSTRACT

Cellular and molecular characterization of osteoclasts (OCL) has been extremely difficult since OCL are rare cells, and are difficult to isolate in large numbers. We used the tartrate-resistant acid phosphatase promoter to target the bcl-XL and/or Simian Virus 40 large T antigen (Tag) genes to cells in the OCL lineage in transgenic mice as a means of immortalizing OCL precursors. Immunocytochemical studies confirmed that we had targeted Bcl-XL and/or Tag to OCL, and transformed and mitotic OCL were readily apparent in bones from both Tag and bcl-XL/Tag mice. OCL formation in primary bone marrow cultures from bcl-XL, Tag, or bcl-XL/Tag mice was twofold greater compared with that of nontransgenic littermates. Bone marrow cells from bcl-XL/Tag mice, but not from singly transgenic bcl-XL or Tag mice, have survived in continuous culture for more than a year. These cells form high numbers of bone-resorbing OCL when cultured using standard conditions for inducing OCL formation, with approximately 50% of the mononuclear cells incorporated into OCL. The OCL that form express calcitonin receptors and contract in response to calcitonin. Studies examining the proliferative capacity and the resistance of OCL precursors from these transgenic mice to apoptosis demonstrated that the increased numbers of OCL precursors in marrow from bcl-XL/Tag mice was due to their increased survival rather than an increased proliferative capacity compared with Tag, bcl-XL, or normal mice. Histomorphometric studies of bones from bcl-XL/Tag mice also confirmed that there were increased numbers of OCL precursors (TRAP + mononuclear cells) present in vivo. These data demonstrate that by targeting both bcl-XL and Tag to cells in the OCL lineage, we have immortalized OCL precursors that form bone-resorbing OCL with an efficiency that is 300-500 times greater than that of normal marrow.


Subject(s)
Antigens, Polyomavirus Transforming/physiology , Osteoclasts/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Simian virus 40/immunology , Stem Cells/physiology , Acid Phosphatase/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Apoptosis , Calcitonin/pharmacology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Rabbits , Receptors, Calcitonin/physiology , bcl-X Protein
12.
Mol Vis ; 4: 5, 1998 Mar 02.
Article in English | MEDLINE | ID: mdl-9485488

ABSTRACT

PURPOSE: To determine if p53 mediates apoptosis in photoreceptors of retinal degeneration, rd1, mice. METHODS: The rd1/rd1 mice were interbred with p53 null mice to generate p53-/- rd1/rd1 and p53+/+ rd1/rd1 mice. Rates of loss and incidence of apoptosis in rod photoreceptors were analyzed at appropriate ages (postnatal days 12, 14 and 16). RESULTS: The extent and kinetics of photoreceptor cell loss in rd1 mice were nearly indistinguishable in the p53+/+ and p53 null mice. CONCLUSIONS: Photoreceptor cell apoptosis in the rd1 mouse model occurs by a predominantly p53-independent molecular pathway.


Subject(s)
Apoptosis , Genes, p53/physiology , Photoreceptor Cells/pathology , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Aging , Animals , Mice , Mice, Knockout , Mice, Mutant Strains , Photoreceptor Cells/abnormalities , Retina/abnormalities
13.
Mol Cell Biol ; 18(1): 85-92, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418856

ABSTRACT

The farnesyltransferase inhibitor L-744,832 selectively blocks the transformed phenotype of cultured cells expressing a mutated H-ras gene and induces dramatic regression of mammary and salivary carcinomas in mouse mammary tumor virus (MMTV)-v-Ha-ras transgenic mice. To better understand how the farnesyltransferase inhibitors might be used in the treatment of human tumors, we have further explored the mechanisms by which L-744,832 induces tumor regression in a variety of transgenic mouse tumor models. We assessed whether L-744,832 induces apoptosis or alterations in cell cycle distribution and found that the tumor regression in MMTV-v-Ha-ras mice could be attributed entirely to elevation of apoptosis levels. In contrast, treatment with doxorubicin, which induces apoptosis in many tumor types, had a minimal effect on apoptosis in these tumors and resulted in a less dramatic tumor response. To determine whether functional p53 is required for L-744,832-induced apoptosis and the resultant tumor regression, MMTV-v-Ha-ras mice were interbred with p53(-/-) mice. Tumors in ras/p53(-/-) mice treated with L-744,832 regressed as efficiently as MMTV-v-Ha-ras tumors, although this response was found to be mediated by both the induction of apoptosis and an increase in G1 with a corresponding decrease in the S-phase fraction. MMTV-v-Ha-ras mice were also interbred with MMTV-c-myc mice to determine whether ras/myc tumors, which possess high levels of spontaneous apoptosis, have the potential to regress through a further increase in apoptosis levels. The ras/myc tumors were found to respond nearly as efficiently to L-744,832 treatment as the MMTV-v-Ha-ras tumors, although no induction of apoptosis was observed. Rather, the tumor regression in the ras/myc mice was found to be mediated by a large reduction in the S-phase fraction. In contrast, treatment of transgenic mice harboring an activated MMTV-c-neu gene did not result in tumor regression. These results demonstrate that a farnesyltransferase inhibitor can induce regression of v-Ha-ras-bearing tumors by multiple mechanisms, including the activation of a suppressed apoptotic pathway, which is largely p53 independent, or by cell cycle alterations, depending upon the presence of various other oncogenic genetic alterations.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma/genetics , Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Experimental/genetics , Methionine/analogs & derivatives , Salivary Gland Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Carcinoma/pathology , Cell Cycle/drug effects , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase , Female , Genes, ras , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse , Methionine/pharmacology , Methionine/therapeutic use , Mice , Mice, Transgenic , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/pathology
14.
Cancer Res ; 57(4): 600-3, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9044833

ABSTRACT

We have used the MMTV-myc and MMTV-ras transgenic mouse mammary tumor models (T. A. Stewart et al., Cell, 38: 627-637, 1984, and E. Sinn et al., Cell, 49: 465-475, 1987) to evaluate how the c-myc and v-Ha-ras oncogenes influence tumor growth characteristics in vivo. MMTV-myc tumors had much higher levels of spontaneous apoptosis than MMTV-ras tumors, whereas intermediate levels were observed in MMTV-myc/ras tumors. Significant differences in cell cycle characteristics were also observed in tumors from mice of the three genotypes. Tumors from MMTV-myc mice had lower G1 and higher S-phase fractions than MMTV-ras tumors, with intermediate values again observed in the MMTV-myc/ras tumors. Despite these differences, however, tumor growth rates for the different groups were similar. These findings highlight the importance of the balance between cell cycle regulation and cell death in determining the kinetics of tumor growth and indicate that distinct oncogenes can have a profound influence on that balance.


Subject(s)
Apoptosis/genetics , Genes, cdc/physiology , Genes, myc/physiology , Genes, ras/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/genetics , Animals , Cell Cycle/genetics , Cell Division , Genotype , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
15.
Mol Cell Biol ; 17(2): 723-31, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9001226

ABSTRACT

We have used an in vivo tumor model to evaluate the consequences of p53 tumor suppressor protein deficiency in a tissue-specific context. By breeding MMTV-ras transgenic mice, which are highly susceptible to the development of mammary and salivary tumors, with p53(-/-) mice, we generated three classes of animals which contained the MMTV-ras transgene but differed in their p53 functional status (ras/p53(+/+), ras/p53(+/-), or ras/p53(-/-)). ras/p53(-/-) mice developed tumors more rapidly than animals of the other two genotypes; however, the distribution of tumors was unexpectedly altered. Whereas the most frequently observed tumors in ras/p53(+/+) and ras/p53(+/-) mice were of mammary origin, ras/p53(-/-) mice developed primarily salivary tumors. In addition, the mammary and salivary tumors from ras/p53(-/-) mice consistently exhibited a number of unfavorable characteristics, including higher histologic grades, increased growth rates, and extensive genomic instability and heterogeneity, relative to tumors from ras/p53(+/+) mice. Interestingly, the increased growth rates of ras/p53(-/-) tumors appear to be due to impaired cell cycle regulation rather than decreased apoptosis, suggesting that p53-mediated tumor suppression can occur independent of its role in apoptosis.


Subject(s)
Genes, ras/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse , Salivary Gland Neoplasms/genetics , Tumor Suppressor Protein p53/physiology , Tumor Virus Infections/genetics , Aneuploidy , Animals , Apoptosis , Cell Division , Crosses, Genetic , Female , Genetic Heterogeneity , Genotype , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Mice, Transgenic , Salivary Gland Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Virus Infections/pathology
16.
J Bone Miner Res ; 12(12): 2014-23, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9421234

ABSTRACT

Although osteocytes are the most abundant cells in bone, their functional role remains unclear. In part, this is due to lack of availability of osteocyte cell lines which can be studied in vitro. Since others have shown that cell lines can be readily developed from transgenic mice in which the SV40 large T-antigen oncogene is expressed under the control of a promoter which targets the cells of interest, we used this approach to develop an osteocyte cell line. We chose as a promoter osteocalcin, whose expression is essentially limited to bone cells and which is expressed more abundantly in osteocytes than in osteoblasts. From these transgenic mice, we isolated cells from the long bones using sequential collagenase digestion and maintained these cells on collagen-coated surfaces which are optimal for osteocyte maintenance and growth. We describe here the properties of a cell line cloned from these cultures, called MLO-Y4 (for murine long bone osteocyte Y4). The properties of MLO-Y4 cells are very similar to primary osteocytes. Like primary osteocytes and unlike primary osteoblasts, the cell line produces large amounts of osteocalcin but low amounts of alkaline phosphatase. The cells produce extensive, complex dendritic processes and are positive for T-antigen, for osteopontin, for the neural antigen CD44, and for connexin 43, a protein found in gap junctions. This cell line also produces very small amounts of type I collagen mRNA compared with primary osteoblasts. MLO-Y4 cells lack detectable mRNA for osteoblast-specific factor 2, which appears to be a positive marker for osteoblasts but may be a negative marker for osteocytes. This newly established cell line should prove useful for studying the effects of mechanical stress on osteocyte function and for determining the means whereby osteocytes communicate with other bone cells such as osteoblasts and osteoclasts.


Subject(s)
Cell Line , Osteocytes/cytology , Alkaline Phosphatase/metabolism , Animals , Cell Adhesion Molecules/genetics , Collagen/genetics , Connexin 43/genetics , Female , Gene Expression , Hyaluronan Receptors/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteocytes/enzymology , Osteocytes/metabolism , Osteopontin , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sialoglycoproteins/genetics
17.
Arch Ophthalmol ; 114(11): 1376-81, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8906028

ABSTRACT

BACKGROUND: Combined modality therapy for childhood retinoblastoma holds the potential of decreasing treatment-related morbidity while maintaining excellent tumor control rates. OBJECTIVE: To evaluate the efficacy of external beam radiation therapy (EBRT), ferromagnetic hyperthermia (FMH), and the combination of both modalities in the control of ocular tumors in a transgenic murine model of retinoblastoma. METHODS: One hundred sixty-six mouse eyes from 4-week-old animals transgenically positive for simian virus 40 large T antigen were treated with a total dose of 10, 15, 20, 30, 40, 45, or 50 Gy of EBRT in 5-Gy fractions twice daily, with 48 degrees C or 54 degrees C FMH for 20 minutes, or with combined EBRT at 10 or 30 Gy and 48 degrees C or 54 degrees C FMH for 20 minutes. Serial histologic sections, obtained 8 weeks after treatment, were examined for the presence of tumor. RESULTS: The tumor control dose for 50% of eyes (TCD50) treated with EBRT occurred at 27.6 Gy. Ferromagnetic hyperthermia at 48 degrees C cured 30% (6/20) of eyes, while 54 degrees C FMH resulted in a 100% (20/20) cure rate. Combined treatment with 48 degrees C FMH and EBRT exhibited a TCD50 at 3.3 Gy. The thermal enhancement ratio was 8.4. Ferromagnetic hyperthermia at 54 degrees C exhibited tumor cure in all animals, but 25% of eyes were lost owing to secondary treatment complications. CONCLUSIONS: This represents the first documentation of tumor control via EBRT, ocular FMH, and a combination of these treatment modalities in this murine transgenic retinoblastoma model. The extent of treatment synergy in this model suggests that combined treatment application may allow a reduction in total ocular and periocular radiation dose while maintaining excellent local tumor control.


Subject(s)
Eye Neoplasms/therapy , Hyperthermia, Induced , Radiotherapy, High-Energy , Retinoblastoma/therapy , Animals , Antigens, Polyomavirus Transforming/genetics , Combined Modality Therapy , Disease Models, Animal , Dose-Response Relationship, Radiation , Eye Neoplasms/genetics , Eye Neoplasms/pathology , Genes, Retinoblastoma/genetics , Hot Temperature , Iron , Magnetics , Mice , Mice, Transgenic/genetics , Radiotherapy Dosage , Retinoblastoma/genetics , Retinoblastoma/pathology
18.
Arch Ophthalmol ; 114(11): 1385-9, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8906029

ABSTRACT

BACKGROUND: Combined modality therapy in the treatment of retinoblastoma may decrease treatment-related morbidity and second tumor-associated mortality, while maintaining excellent tumor control rates. OBJECTIVE: To evaluate tumor control and potential synergy between intravitreally delivered carboplatin and external beam radiation therapy (EBRT), using a transgenic murine model of spontaneous heritable retinoblastoma. METHODS: Sixty-six mouse eyes from 4-week-old transgenic mice positive for the simian virus 40 large T antigen were evaluated. Thirty-three mice were treated with 5 intravitreal injections of carboplatin (ranging from 0.1-4.0 micrograms) combined with concurrent bilateral EBRT (ranging from 10-30 Gy) delivered in twice daily 5-Gy fractions. All eyes were followed up for treatment complications. Twelve weeks following final treatment, all eyes were enucleated, serial histologic sections obtained, and the eyes examined for the presence of retinoblastoma. RESULTS: No eye treated with 0.1 microgram of carboplatin and EBRT exhibited tumor control. Three (75%) of 4 mice receiving 1.0 microgram of carboplatin combined with 10-Gy EBRT had complete tumor control. Four (100%) of 4 mice receiving 1.0 microgram of carboplatin combined with 30-Gy EBRT had complete tumor control. Nine (100%) of 9 mice receiving 4.0 micrograms of carboplatin in combination with EBRT had complete tumor control. The chemotherapeutic enhancement ratio ranged from 1.07 to 3.24. CONCLUSIONS: Combined administration of intravitreal carboplatin and EBRT enhances local tumor control in murine retinoblastoma. Combining these treatment modalities may allow tumor control in selected patients with retinoblastoma while decreasing treatment-related morbidity and the mutagenic risks associated with radiation and systemic chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Carboplatin/therapeutic use , Eye Neoplasms/therapy , Radiotherapy, High-Energy , Retinoblastoma/diagnostic imaging , Animals , Antigens, Polyomavirus Transforming/genetics , Antineoplastic Agents/administration & dosage , Carboplatin/administration & dosage , Combined Modality Therapy , Disease Models, Animal , Eye Neoplasms/genetics , Eye Neoplasms/pathology , Genes, Retinoblastoma/genetics , Injections , Mice , Mice, Transgenic/genetics , Radiography , Radiotherapy Dosage , Retinoblastoma/genetics , Retinoblastoma/pathology , Vitreous Body
19.
Development ; 122(10): 3319-29, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8898243

ABSTRACT

Targeted expression of oncogenes in transgenic mice can immortalize specific cell types to serve as valuable cultured model systems. Utilizing promoter regions from a set of genes expressed at specific stages of differentiation in a given cell lineage, we demonstrate that targeted oncogenesis can produce cell lines representing sequential stages of development, in essence allowing both spatial and temporal immortalization. Our strategy was based on our production of a committed but immature pituitary gonadotrope cell line by directing expression of the oncogene SV40 T antigen using a gonadotrope-specific region of the human glycoprotein hormone alpha-subunit gene in transgenic mice. These cells synthesize alpha-subunit and gonadotropin-releasing hormone (GnRH) receptor, yet are not fully differentiated in that they do not synthesize the beta-subunits of luteinizing hormone (LH) or follicle-stimulating hormone (FSH). This observation lead to the hypothesis that targeting oncogenesis with promoters that are activated earlier or later in development might immortalize cells that were more primitive or more differentiated, respectively. To test this hypothesis, we used an LHbeta promoter to immortalize a cell that represents a subsequent stage of gonadotrope differentiation (expression of alpha-subunit, GnRH receptor, and LH beta-subunit but not FSH beta-subunit). Conversely, targeting oncogenesis with a longer fragment of the human alpha-subunit gene (which is activated earlier in development) resulted in the immortalization of a progenitor cell that is more primitive, expressing only the alpha-subunit gene. Interestingly, this transgene also immortalized cells of the thyrotrope lineage that express both alpha- and beta-subunits of thyroid-stimulating hormone and the transcription factor GHF-1 (Pit-1). Thus, targeted tumorigenesis immortalizes mammalian cells at specific stages of differentiation and allows the production of a series of cultured cell lines representing sequential stages of differentiation in a given cell lineage.


Subject(s)
Pituitary Gland/cytology , Pituitary Gland/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Carcinogenicity Tests , Cell Differentiation , Female , Gene Expression , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Homeodomain Proteins/metabolism , Humans , LIM-Homeodomain Proteins , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Oncogenes/genetics , Pituitary Hormones, Anterior/genetics , Pituitary Hormones, Anterior/metabolism , Pituitary Neoplasms/pathology , Transcription Factors/metabolism , Tumor Cells, Cultured
20.
J Neurochem ; 66(1): 20-5, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8522954

ABSTRACT

The tyrosine hydroxylase (TH) gene is expressed exclusively in cells and neurons that synthesize and release L-DOPA or catecholamines. To further understand the molecular genetic mechanisms that regulate this cell-type specific expression, a chimeric gene was prepared by linking 3.6 kb of the 5' flanking DNA of the mouse TH gene, including the +1 initiation site for transcription, to an E. coli beta-galactosidase reporter. This fusion gene (TH3.6LAC) was used to prepare transgenic mice, and the tissue distribution of expression of TH3.6LAC was determined by the measurement of beta-galactosidase enzymatic activity and/or by the detection of the transcription product of the chimeric gene by RNase protection assays. In two separate founder lines, TH3.6LAC expression was observed in every region of the brain that was examined, including the olfactory bulb, brainstem, cerebellum, diencephalon, hippocampus, striatum, and cerebral cortex. Expression of TH3.6LAC was observed in the adrenal gland of one founder line but not in the other. TH3.6LAC activation was undetectable in peripheral organs that were examined, including the liver, heart, salivary gland, kidney, lung, and spleen. Although 3.6 kb of the 5' regulatory DNA of the mouse TH gene is sufficient to activate the TH fusion gene in the mouse, it is not enough to restrict its expression to catecholaminergic cells.


Subject(s)
Brain/enzymology , Catecholamines/metabolism , DNA/genetics , Mice/genetics , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Tyrosine 3-Monooxygenase/genetics , Adrenal Glands/enzymology , Animals , Base Sequence , Enzyme Induction , Genes, Reporter , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Organ Specificity , RNA, Messenger/analysis , Recombinant Fusion Proteins/biosynthesis , Transcription, Genetic , Tyrosine 3-Monooxygenase/biosynthesis , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...