Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1208279, 2023.
Article in English | MEDLINE | ID: mdl-37397253

ABSTRACT

Fetal Alcohol Spectrum Disorder (FASD) arises from maternal consumption of alcohol during pregnancy affecting 2%-5% of the Western population. In Xenopus laevis studies, we showed that alcohol exposure during early gastrulation reduces retinoic acid (RA) levels at this critical embryonic stage inducing craniofacial malformations associated with Fetal Alcohol Syndrome. A genetic mouse model that induces a transient RA deficiency in the node during gastrulation is described. These mice recapitulate the phenotypes characteristic of prenatal alcohol exposure (PAE) suggesting a molecular etiology for the craniofacial malformations seen in children with FASD. Gsc +/Cyp26A1 mouse embryos have a reduced RA domain and expression in the developing frontonasal prominence region and delayed HoxA1 and HoxB1 expression at E8.5. These embryos also show aberrant neurofilament expression during cranial nerve formation at E10.5 and have significant FASD sentinel-like craniofacial phenotypes at E18.5. Gsc +/Cyp26A1 mice develop severe maxillary malocclusions in adulthood. Phenocopying the PAE-induced developmental malformations with a genetic model inducing RA deficiency during early gastrulation strongly supports the alcohol/vitamin A competition model as a major molecular etiology for the neurodevelopmental defects and craniofacial malformations seen in children with FASD.

2.
Can J Physiol Pharmacol ; 87(10): 859-72, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20052012

ABSTRACT

The human prolactin-inducible protein/gross cystic disease fluid protein-15 (hPIP/GCDFP-15) is a secretory glycoprotein found primarily in apocrine tissues including the breast and salivary glands. With largely unknown functions, PIP has been implicated in breast cancer and metastasis, host defense processes and T lymphocyte apoptosis. To begin to address PIP function in vivo, we generated the PIP null mouse (Pip-/-). Additionally, to determine the effect of the loss of PIP on gene expression and to gain insight into some of the molecular mechanisms underlying PIP function, microarray analysis of the submandibular gland was also undertaken. Pip-/- mice developed normally with no overt differences in behaviour or gross morphology and were fertile. However, histological examination of 3-month-old Pip-/- mice sometimes showed enlarged submandibular lymph nodes, lymphocytic aggregations within the prostate lobes, and enlarged medulla in the thymus. Functional analysis of gene expression revealed sets of multiple differentially expressed genes associated with cell death and survival, lipid metabolism, inflammation, immune disease, and cancer, as a consequence of mPIP abrogation. Taken together, these studies lend support to an immunomodulatory role for PIP in vivo and provide further insights into potentially novel signaling pathways and regulatory networks for PIP.


Subject(s)
Proteins/genetics , Submandibular Gland/metabolism , Animals , Blotting, Western , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Immunohistochemistry , Lacrimal Apparatus/metabolism , Male , Mice , Mice, Knockout , Mutation , Oligonucleotide Array Sequence Analysis , Proteins/physiology , RNA/biosynthesis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Saliva/metabolism
3.
Blood ; 96(9): 3249-55, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11050010

ABSTRACT

Deficiency of folate or vitamin B(12) (cobalamin) causes megaloblastic anemia, a disease characterized by pancytopenia due to the excessive apoptosis of hematopoietic progenitor cells. Clinical and experimental studies of megaloblastic anemia have demonstrated an impairment of DNA synthesis and repair in hematopoietic cells that is manifested by an increased percentage of cells in the DNA synthesis phase (S phase) of the cell cycle, compared with normal hematopoietic cells. Both folate and cobalamin are required for normal de novo synthesis of thymidylate and purines. However, previous studies of impaired DNA synthesis and repair in megaloblastic anemia have concerned mainly the decreased intracellular levels of thymidylate and its effects on nucleotide pools and misincorporation of uracil into DNA. An in vitro model of folate-deficient erythropoiesis was used to study the relationship between the S-phase accumulation and apoptosis in megaloblastic anemia. The results indicate that folate-deficient erythroblasts accumulate in and undergo apoptosis in the S phase when compared with control erythroblasts. Both the S-phase accumulation and the apoptosis were induced by folate deficiency in erythroblasts from p53 null mice. The complete reversal of the S-phase accumulation and apoptosis in folate-deficient erythroblasts required the exogenous provision of specific purines or purine nucleosides as well as thymidine. These results indicate that decreased de novo synthesis of purines plays as important a role as decreased de novo synthesis of thymidylate in the pathogenesis of megaloblastic anemia.


Subject(s)
Anemia, Megaloblastic/genetics , Apoptosis , Cell Cycle/physiology , DNA Replication , Erythroblasts/physiology , Genes, p53 , Anemia, Megaloblastic/pathology , Anemia, Megaloblastic/physiopathology , Animals , Cells, Cultured , DNA Repair , Erythroblasts/pathology , Folic Acid/metabolism , Kinetics , Mice , Mice, Inbred Strains , Mice, Knockout , S Phase , Vitamin B 12/metabolism
4.
Nat Genet ; 24(2): 175-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10655065

ABSTRACT

The gene FUS (also known as TLS (for translocated in liposarcoma) and hnRNP P2) is translocated with the gene encoding the transcription factor ERG-1 in human myeloid leukaemias. Although the functions of wild-type FUS are unknown, the protein contains an RNA-recognition motif and is a component of nuclear riboprotein complexes. FUS resembles a transcription factor in that it binds DNA, contributes a transcriptional activation domain to the FUS-ERG oncoprotein and interacts with several transcription factors in vitro. To better understand FUS function in vivo, we examined the consequences of disrupting Fus in mice. Our results indicate that Fus is essential for viability of neonatal animals, influences lymphocyte development in a non-cell-intrinsic manner, has an intrinsic role in the proliferative responses of B cells to specific mitogenic stimuli and is required for the maintenance of genomic stability. The involvement of a nuclear riboprotein in these processes in vivo indicates that Fus is important in genome maintenance.


Subject(s)
B-Lymphocytes/immunology , Ribonucleoproteins/metabolism , Animals , Animals, Newborn , Bone Marrow Cells/immunology , Chimera , Crosses, Genetic , Female , Genotype , Heterogeneous-Nuclear Ribonucleoproteins , Humans , Liver/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA-Binding Protein FUS , RNA-Binding Proteins/metabolism , Restriction Mapping , Ribonucleoproteins/deficiency , Ribonucleoproteins/genetics , Spleen/immunology
5.
Oncogene ; 17(9): 1119-30, 1998 Sep 03.
Article in English | MEDLINE | ID: mdl-9764822

ABSTRACT

Erythroleukemia induced by the anemia strain of Friend virus occurs in two stages. The first stage results in rapid expansion of pre-leukemic proerythroblasts (FVA cells) dependent on erythropoietin (Epo) for differentiation and survival in vitro. The second stage is characterized by emergence of erythroleukemic clones (MEL cells) which typically bear activation of the ets-oncogene, PU.1/spi.1, and loss of functional p53. We developed a Friend virus-sensitive, p53-deficient mouse model to investigate the biological advantage conferred by p53-loss during tumor progression. Here we report p53 was not required for cell survival or growth arrest during differentiation of FVA cells, nor was p53 required for induction of apoptosis upon Epo withdrawal. However, we detected induction of the p21Cip1 cyclin-dependent kinase inhibitor gene during differentiation, which was markedly enhanced in the presence of p53. p53-dependent expression of p21Cip1 occurred in the absence of an increase in p53 mRNA and protein levels and was specific for p21Cip1, since expression of gadd45, mdm-2, cyclin G and bax were unaffected by p53. In contrast, treatment of FVA cells with DNA damaging agents led to rapid accumulation of p53 protein resulting in transcription of multiple p53-regulated genes, leading to either apoptosis or growth arrest, depending on the agent used. These data demonstrate that p53-dependent activities during differentiation of preleukemic erythroblasts are distinct from those observed in response to genotoxic agents. We propose that enhancement of p53-dependent gene expression during differentiation may represent a tumor suppressor function which is necessary to monitor differentiation of preleukemic cells and which is selected against during tumor progression.


Subject(s)
DNA Damage/physiology , Friend murine leukemia virus , Leukemia, Erythroblastic, Acute/physiopathology , Tumor Suppressor Protein p53/physiology , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cell Division/drug effects , Cell Division/genetics , Cell Division/radiation effects , Cell Survival/drug effects , Cell Survival/physiology , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , DNA Damage/drug effects , DNA Damage/radiation effects , Dactinomycin/pharmacology , Disease Progression , Erythroblasts/cytology , Erythroblasts/drug effects , Erythroblasts/radiation effects , Erythropoietin/pharmacology , Female , G1 Phase/drug effects , G1 Phase/genetics , G1 Phase/radiation effects , Gene Expression/genetics , Genes, p53/drug effects , Genes, p53/genetics , Genes, p53/radiation effects , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Erythroblastic, Acute/virology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mutation/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Transcriptional Activation/radiation effects , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Nat Genet ; 16(4): 338-44, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9241269

ABSTRACT

Most mammalian genes will soon be characterized as cDNA sequences with little information about their function. To utilize this sequence information for large-scale functional studies, a gene trap retrovirus shuttle vector has been developed to disrupt genes expressed in murine embryonic stem (ES) cells. A library of mutant clones was isolated, and regions of genomic DNA adjacent to 400 independent provirus inserts were cloned and sequenced. The flanking sequences, designated 'promoter-proximal sequence tags', or PSTs, identified 63 specific genes and anonymous cDNAs disrupted as a result of virus integration. The efficiency of tagged sequence mutagenesis suggests that many of the 10,000-20,000 genes expressed in ES cells can be targeted, providing defined mutations for the analysis of gene functions in vivo. In addition, PSTs provide the first expressed sequence tags derived from genomic DNA, and define gene features such as exon boundaries and promoters that are missing from cDNA sequences.


Subject(s)
Genetic Techniques , Genetic Vectors , Mutagenesis , Animals , Base Sequence , DNA, Complementary , Databases, Factual , Forecasting , Gene Expression , Gene Targeting , Humans , Mice , Molecular Sequence Data , Stem Cells
7.
Cell ; 84(4): 543-50, 1996 Feb 23.
Article in English | MEDLINE | ID: mdl-8598041

ABSTRACT

H2-M is a nonconventional major histocompatibility complex (MHC) class II molecule that has been implicated in the loading of peptides onto conventional class II molecules. We generated mice with a targeted mutation in the H2-Ma gene, which encodes a subunit for H2-M. Although the mutant mice express normal class II cell surface levels, these are structurally distinct from the compact SDS-resistant complexes expressed by wild-type cells and are predominantly bound by class II-associated invariant chain peptides (CLIPs). Cells from these animals are unable to present intact protein antigens to class II-restricted T cells and show reduced capacity to present exogenous peptides. Numbers of mature CD4+ T lymphocytes in mutant mice are reduced 3- to 4-fold and exhibit altered reactivities. Overall, this phenotype establishes an important role for H2-M in regulating MHC class II function in vivo and supports the notion that self-peptides contribute to the specificity of T cell positive selection.


Subject(s)
Antigen Presentation/immunology , Histocompatibility Antigens Class II/genetics , T-Lymphocytes/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Biological Transport/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , Gene Expression/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Lymphocyte Count , Membrane Proteins/immunology , Mice , Mice, Mutant Strains , Peptides/immunology , Peptides/metabolism , Phenotype , Protein Binding/immunology , Spleen/cytology , T-Lymphocytes/metabolism , T-Lymphocytes/ultrastructure
9.
Oncogene ; 10(1): 109-15, 1995 Jan 05.
Article in English | MEDLINE | ID: mdl-7529916

ABSTRACT

Wild type p53 can induce cell cycle arrest at specific points in the cell cycle, in particular G1/S, an ability lost by most p53 mutants. We have previously reported that p53 mutant genes can rescue REF52 cells from ras-induced growth arrest and that over expression of wild type p53 inhibits cell growth in these cells. In this paper we examined whether p53 can also induce cell cycle arrest at the G2/M boundary of the cell cycle. To accomplish this we used the REF52 cell line and the temperature sensitive p53val135 mutant allele. Cells were enriched in the late G1 and early S phases before the temperature shift. REF52 cells expressing mutant-p53val135 alone with an activated H-ras gene arrest primarily at the G1/S and G2/M parts of the cell cycle at the restrictive temperature, as determined by flow cytometry analysis. These results suggest that the anti-proliferative activity of p53 may be involved in regulation of the cell cycle at the G2/M restriction point as well as transit through G1/S and initiation of DNA synthesis.


Subject(s)
G2 Phase/genetics , Genes, p53 , Mitosis/genetics , Animals , Cell Division/genetics , Cell Line , DNA , Epitopes/genetics , Genes, p53/immunology , Hot Temperature , Rats
10.
Mol Cell Biol ; 14(6): 4183-92, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8196656

ABSTRACT

Erythropoietin (Epo) inhibits apoptosis in murine proerythroblasts infected with the anemia-inducing strain of Friend virus (FVA cells). We have shown that the apoptotic process in FVA cell populations deprived of Epo is asynchronous as a result of a heterogeneity in Epo dependence among individual cells. Here we investigated whether apoptosis in FVA cells correlated with cell cycle phase or stabilization of p53 tumor suppressor protein. DNA analysis in nonapoptotic FVA cell subpopulations cultured without Epo demonstrated little change in the percentages of cells in G1,S, and G2/M phases over time. Analysis of the apoptotic subpopulation revealed high percentages of cells in G1 and S, with few cells in G2/M at any time. When cells were sorted from G1 and S phases prior to culture without Epo, apoptotic cells appeared at the same rate in both populations, indicating that no prior commitment step had occurred in either G1 or S phase. Steady-state wild-type p53 protein levels were very low in FVA cells compared with control cell lines and did not accumulate in Epo-deprived cultures; however, p53 protein did accumulate when FVA cells were treated with the DNA-damaging agent actinomycin D. These data indicate that erythroblast apoptosis caused by Epo deprivation (i) occurs throughout G1 and S phases and does not require cell cycle arrest, (ii) does not have a commitment event related to cell cycle phase, and (iii) is not associated with conformational changes or stabilization of wild-type p53 protein.


Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , DNA/biosynthesis , Erythropoietin/pharmacology , Genes, p53 , Hematopoietic Stem Cells/cytology , Tumor Suppressor Protein p53/biosynthesis , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/physiology , Cell Line , Cells, Cultured , DNA Damage , Flow Cytometry , Friend murine leukemia virus/genetics , G1 Phase/drug effects , G1 Phase/physiology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Kinetics , Mice , Mice, Inbred Strains , Protein Conformation , S Phase/drug effects , S Phase/physiology , Thymidine/metabolism , Time Factors , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/chemistry
11.
Mol Cell Biol ; 11(3): 1344-52, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1996096

ABSTRACT

Overexpression of an activated ras gene in the rat embryo fibroblast line REF52 results in growth arrest at either the G1/S or G2/M boundary of the cell cycle. Both the DNA tumor virus proteins simian virus 40 large T antigen and adenovirus 5 E1a are able to rescue ras induced lethality and cooperate with ras to fully transform REF52 cells. In this report, we present evidence that the wild-type activity of the tumor suppressor gene p53 is involved in the negative growth regulation of this model system. p53 genes encoding either a p53Val-135 or p53Pro-193 mutation express a highly stable p53 protein with a conformation-dependent loss of wild-type activity and the ability to eliminate any endogenous wild-type p53 activity in a dominant negative manner. In cotransfection assays, these mutant p53 genes are able to rescue REF52 cells from ras-induced growth arrest, resulting in established cell lines which express elevated levels of the ras oncoprotein and show morphological transformation. Full transformation, as assayed by tumor formation in nude mice, is found only in the p53Pro-193-plus-ras transfectants. These cells express higher levels of the ras protein than do the p53Val-135-plus-ras-transfected cells. Transfection of REF52 cells with ras alone or a full-length genomic wild-type p53 plus ras results in growth arrest and lethality. Therefore, the selective event for p53 inactivation or loss during tumor progression may be to overcome a cell cycle restriction induced by oncogene overexpression (ras). These results suggest that a normal function of p53 may be to mediate negative growth regulation in response to ras or other proliferative inducing signals.


Subject(s)
Cell Cycle , Genes, p53 , Genes, ras , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cell Line , Cell Transformation, Neoplastic , Cloning, Molecular , Gene Expression , Mutation , Rats , Transfection
12.
J Virol ; 62(12): 4752-5, 1988 Dec.
Article in English | MEDLINE | ID: mdl-2846884

ABSTRACT

The Friend virus-transformed erythroleukemic cell line DP16-9B4 has undergone a complex rearrangement of the p53 oncogene and lacks any detectable expression of the p53 protein. We report here characterization of both p53 alleles in this cell line and identify independent integrations of Friend murine leukemia virus sequences into the coding region of both alleles.


Subject(s)
Alleles , Friend murine leukemia virus/genetics , Leukemia, Erythroblastic, Acute/microbiology , Neoplasm Proteins/genetics , Oncogenes , Phosphoproteins/genetics , Animals , Blotting, Southern , Cloning, Molecular , DNA Restriction Enzymes , DNA, Viral/analysis , Gene Expression Regulation , Nucleic Acid Hybridization , Tumor Cells, Cultured , Tumor Suppressor Protein p53
SELECTION OF CITATIONS
SEARCH DETAIL
...