Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
Oncogene ; 35(37): 4836-45, 2016 09 15.
Article in English | MEDLINE | ID: mdl-26876210

ABSTRACT

Rad54 and Mus81 mammalian proteins physically interact and are important for the homologous recombination DNA repair pathway; however, their functional interactions in vivo are poorly defined. Here, we show that combinatorial loss of Rad54 and Mus81 results in hypersensitivity to DNA-damaging agents, defects on both the homologous recombination and non-homologous DNA end joining repair pathways and reduced fertility. We also observed that while Mus81 deficiency diminished the cleavage of common fragile sites, very strikingly, Rad54 loss impaired this cleavage to even a greater extent. The inefficient repair of DNA double-strand breaks (DSBs) in Rad54(-/-)Mus81(-/-) cells was accompanied by elevated levels of chromosome missegregation and cell death. Perhaps as a consequence, tumor incidence in Rad54(-/-)Mus81(-/-) mice remained comparable to that in Mus81(-/-) mice. Our study highlights the importance of the cooperation between Rad54 and Mus81 for mediating DNA DSB repair and restraining chromosome missegregation.


Subject(s)
DNA Helicases/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Neoplasms/genetics , Nuclear Proteins/genetics , Animals , Chromosomes/genetics , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair/genetics , Homologous Recombination/genetics , Humans , Mice , Mice, Knockout , Neoplasms/pathology
2.
Oncogene ; 34(14): 1780-9, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-24858046

ABSTRACT

BLM is a DNA helicase important for the restart of stalled replication forks and for homologous recombination (HR) repair. Mutations of BLM lead to Bloom Syndrome, a rare autosomal recessive disorder characterized by elevated levels of sister chromatid exchanges (SCEs), dwarfism, immunodeficiency, infertility and increased cancer predisposition. BLM physically interacts with MUS81, an endonuclease involved in the restart of stalled replication forks and HR repair. Herein we report that loss of Mus81 in Blm hypomorph mutant mice leads to infertility, and growth and developmental defects that are not observed in single mutants. Double mutant cells and mice were hypersensitive to Mitomycin C and γ-irradiation (IR) compared with controls and their repair of DNA double-strand breaks (DSBs) mediated by HR pathway was significantly defective, whereas their non-homologous-end-joining repair was elevated compared with controls. We also demonstrate the importance of the loss of the nuclease activity of Mus81 in the defects observed in Mus81(-/-) and double mutant cells. Exacerbated IR-induced chromosomal aberration was observed in double mutant mice and despite their reduced SCE levels, these mutants showed increased tumorigenesis risks. Our data highlight the importance of Mus81 and Blm in DNA DSB repair pathways, fertility, development and cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA End-Joining Repair/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Lymphoma/genetics , RecQ Helicases/genetics , Animals , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/radiation effects , Chromosome Aberrations/chemically induced , Chromosome Aberrations/radiation effects , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , Gamma Rays/adverse effects , Lymphoma/chemically induced , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitomycin/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology
3.
Cell Death Differ ; 20(6): 812-22, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23449389

ABSTRACT

The serine threonine kinase checkpoint kinase 2 (CHK2) is a DNA damage checkpoint protein important for the ATM-p53 signaling pathway. In addition to its phosphorylation, CHK2 is also ubiquitylated, and both post-translational modifications are important for its function. However, although the mechanisms that regulate CHK2 phosphorylation are well established, those that control its ubiquitylation are not fully understood. In this study, we demonstrate that the ubiquitin E3 ligase PIRH2 (p53-induced protein with a RING (Really Interesting New Gene)-H2 domain) interacts with CHK2 and mediates its polyubiquitylation and proteasomal degradation. We show that the deubiquitylating enzyme USP28 forms a complex with PIRH2 and CHK2 and antagonizes PIRH2-mediated polyubiquitylation and proteasomal degradation of CHK2. We also provide evidence that CHK2 ubiquitylation by PIRH2 is dependent on its phosphorylation status. Cells deficient in Pirh2 displayed accumulation of Chk2 and enhanced hyperactivation of G1/S and G2/M cell-cycle checkpoints. This hyperactivation was, however, no longer observed in Pirh2-/-Chk2-/- cells, providing evidence for the importance of Chk2 regulation by Pirh2. These findings indicate that PIRH2 has central roles in the ubiquitylation of Chk2 and its turnover and in the regulation of its function.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Checkpoint Kinase 2 , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination
4.
Cell Death Differ ; 16(2): 331-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19008920

ABSTRACT

Bcl-2 associated factor 1 (Bclaf1) is a nuclear protein that was originally identified in a screen of proteins that interact with the adenoviral bcl-2 homolog E1B19K. Overexpression of Bclaf1 was shown to result in apoptosis and transcriptional repression that was reversible in the presence of Bcl-2 or Bcl-x(L). Furthermore, antiapoptotic members, but not proapoptotic members of the Bcl-2 protein family, were shown to interact with Bclaf1 and prevent its localization to the nucleus. Bclaf1 has also recently been identified as a binding partner for Emerin, a nuclear membrane protein that is mutated in X-linked recessive Emery-Dreifuss muscular dystrophy. To ascertain the in vivo function of Bclaf1, we have generated mice that carry a targeted mutation of the bclaf1 locus. In this study, we show that Bclaf1 is required for proper spatial and temporal organization of smooth muscle lineage during the saccular stage of lung development. We also show that Bclaf1 is dispensable for thymocyte development but is essential for peripheral T-cell homeostasis. Despite its postulated role as a proapoptotic protein, Bclaf1-deficient cells did not show any defect in cell death linked to development or after exposure to various apoptotic stimuli. Our findings show a critical role for Bclaf1 in developmental processes independent of apoptosis.


Subject(s)
DNA-Binding Proteins/physiology , Lung/growth & development , Lymphocytes/immunology , Repressor Proteins/physiology , Animals , Apoptosis , DNA-Binding Proteins/genetics , Homozygote , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Repressor Proteins/genetics , bcl-2-Associated X Protein/metabolism
5.
Apoptosis ; 12(4): 791-800, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17245644

ABSTRACT

Previously, we analyzed mice lacking either caspase-2 or caspase-3 and documented a role for caspase-2 in developmental and chemotherapy-induced apoptosis of oocytes. Those data also revealed dispensability of caspase-3, although we found this caspase critical for ovarian granulosa cell death. Because of the mutual interdependence of germ cells and granulosa cells, herein we generated caspase-2 and -3 double-mutant (DKO) mice to evaluate how these two caspases functionally relate to each other in orchestrating oocyte apoptosis. No difference was observed in the rate of spontaneous oocyte apoptosis between DKO and wildtype (WT) females. In contrast, the oocytes from DKO females were more susceptible to apoptosis induced by DNA damaging agents, compared with oocytes from WT females. This increased sensitivity to death of DKO oocytes appears to be a specific response to DNA damage, and it was associated with a compensatory upregulation of caspase-12. Interestingly, DKO oocytes were more resistant to apoptosis induced by methotrexate (MTX) than WT oocytes. These results revealed that in female germ cells, insults that directly interfere with their metabolic status (e.g. MTX) require caspase-2 and caspase-3 as obligatory executioners of the ensuing cell death cascade. However, when DNA damage is involved, and in the absence of caspase-2 and -3, caspase-12 becomes upregulated and mediates apoptosis in oocytes.


Subject(s)
Apoptosis/physiology , Caspase 12/metabolism , Caspase 3/metabolism , Cysteine Endopeptidases/metabolism , Oocytes/enzymology , Animals , Antibiotics, Antineoplastic/metabolism , Caspase 12/genetics , Caspase 2 , Caspase 3/genetics , Cell Shape , Cells, Cultured , Cysteine Endopeptidases/genetics , Doxorubicin/metabolism , Female , Lymphocyte Activation , Lymphocytes/cytology , Mice , Mice, Knockout , Oocytes/cytology , Oocytes/physiology , Phenotype , Protease Inhibitors/metabolism , Signal Transduction/physiology , Spleen/cytology , Thymus Gland/cytology
6.
Cell Death Differ ; 12(1): 25-37, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15499374

ABSTRACT

CD95 apoptosis resistance of tumor cells is often acquired through mutations in the death domain (DD) of one of the CD95 alleles. Furthermore, Type I cancer cells are resistant to induction of apoptosis by soluble CD95 ligand (CD95L), which does not induce efficient formation of the death-inducing signaling complex (DISC). Here, we report that tumor cells expressing a CD95 allele that lacks a functional DD, splenocytes from heterozygous lpr(cg) mice, which express one mutated CD95 allele, and Type I tumor cells stimulated with soluble CD95L can all die through CD95 when protein synthesis or nuclear factor kappa B is inhibited. This noncanonical form of CD95-mediated apoptosis is dependent on the enzymatic activity of procaspase-8 but does not involve fully processed active caspase-8 subunits. Our data suggest that it is possible to overcome the CD95 apoptosis resistance of many tumor cells that do not efficiently form a DISC through noncanonical activation of the caspase-8 proenzyme.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , fas Receptor/physiology , Alleles , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspase 8 , Caspase Inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Dactinomycin/pharmacology , Death Domain Receptor Signaling Adaptor Proteins , Drug Resistance, Neoplasm , Enzyme Activation , Fas Ligand Protein , Humans , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred C3H , Mitochondria/drug effects , Mitochondria/metabolism , Mutation , NF-kappa B/antagonists & inhibitors , Oligopeptides/pharmacology , Receptors, Tumor Necrosis Factor/metabolism , fas Receptor/genetics
7.
Annu Rev Genet ; 35: 209-41, 2001.
Article in English | MEDLINE | ID: mdl-11700283

ABSTRACT

The development of cancer requires multiple genetic alterations perturbing distinct cellular pathways. In human cancers, these alterations often arise owing to mutations in tumor-suppressor genes whose normal function is to either inhibit the proliferation, apoptosis, or differentiation of cells, or maintain their genomic integrity. Mouse models for tumor suppressors frequently provide definitive evidence for the antitumorigenic functions of these genes. In addition, animal models permit the identification of previously unsuspected roles of these genes in development and differentiation. The availability of null and tissue-specific mouse mutants for tumor-suppressor genes has greatly facilitated our understanding of the mechanisms leading to cancer. In this review, we describe mouse models for tumor-suppressor genes.


Subject(s)
Genes, Tumor Suppressor , Models, Animal , Animals , Apoptosis , Cell Cycle , DNA Damage , DNA Repair , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Neoplasms, Experimental/genetics
8.
Nature ; 410(6828): 549-54, 2001 Mar 29.
Article in English | MEDLINE | ID: mdl-11279485

ABSTRACT

Programmed cell death is a fundamental requirement for embryogenesis, organ metamorphosis and tissue homeostasis. In mammals, release of mitochondrial cytochrome c leads to the cytosolic assembly of the apoptosome-a caspase activation complex involving Apaf1 and caspase-9 that induces hallmarks of apoptosis. There are, however, mitochondrially regulated cell death pathways that are independent of Apaf1/caspase-9. We have previously cloned a molecule associated with programmed cell death called apoptosis-inducing factor (AIF). Like cytochrome c, AIF is localized to mitochondria and released in response to death stimuli. Here we show that genetic inactivation of AIF renders embryonic stem cells resistant to cell death after serum deprivation. Moreover, AIF is essential for programmed cell death during cavitation of embryoid bodies-the very first wave of cell death indispensable for mouse morphogenesis. AIF-dependent cell death displays structural features of apoptosis, and can be genetically uncoupled from Apaf1 and caspase-9 expression. Our data provide genetic evidence for a caspase-independent pathway of programmed cell death that controls early morphogenesis.


Subject(s)
Apoptosis/physiology , Flavoproteins/physiology , Membrane Proteins/physiology , Mitochondria/physiology , Animals , Apoptosis Inducing Factor , Apoptotic Protease-Activating Factor 1 , Caspase 9 , Caspases/metabolism , Cell Differentiation , Chimera , Embryo, Mammalian/cytology , Embryonic and Fetal Development/physiology , Female , Flavoproteins/genetics , Gene Targeting , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Morphogenesis , Proteins/physiology , Recombination, Genetic , Stem Cells
9.
Mol Cell Biol ; 20(15): 5454-68, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10891486

ABSTRACT

Many apoptotic signaling pathways are directed to mitochondria, where they initiate the release of apoptogenic proteins and open the proposed mitochondrial permeability transition (PT) pore that ultimately results in the activation of the caspase proteases responsible for cell disassembly. BNIP3 (formerly NIP3) is a member of the Bcl-2 family that is expressed in mitochondria and induces apoptosis without a functional BH3 domain. We report that endogenous BNIP3 is loosely associated with mitochondrial membrane in normal tissue but fully integrates into the mitochondrial outer membrane with the N terminus in the cytoplasm and the C terminus in the membrane during induction of cell death. Surprisingly, BNIP3-mediated cell death is independent of Apaf-1, caspase activation, cytochrome c release, and nuclear translocation of apoptosis-inducing factor. However, cells transfected with BNIP3 exhibit early plasma membrane permeability, mitochondrial damage, extensive cytoplasmic vacuolation, and mitochondrial autophagy, yielding a morphotype that is typical of necrosis. These changes were accompanied by rapid and profound mitochondrial dysfunction characterized by opening of the mitochondrial PT pore, proton electrochemical gradient (Deltapsim) suppression, and increased reactive oxygen species production. The PT pore inhibitors cyclosporin A and bongkrekic acid blocked mitochondrial dysregulation and cell death. We propose that BNIP3 is a gene that mediates a necrosis-like cell death through PT pore opening and mitochondrial dysfunction.


Subject(s)
Cell Death/genetics , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Apoptosis Inducing Factor , Apoptotic Protease-Activating Factor 1 , Bongkrekic Acid/pharmacology , Caspase 3 , Caspase 9 , Caspases/genetics , Caspases/metabolism , Cell Death/drug effects , Cell Line , Cyclosporine/pharmacology , Cytochrome c Group/metabolism , DNA Fragmentation , Fibroblasts/pathology , Fibroblasts/ultrastructure , Flavoproteins/metabolism , HeLa Cells , Humans , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mitochondria/drug effects , Necrosis , Permeability , Proteins/genetics , Proteins/metabolism , Reactive Oxygen Species/metabolism
10.
Cell Res ; 10(4): 267-78, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11191349

ABSTRACT

Apoptosis or programmed cell death (PCD) is an evolutionarily conserved cellular process that is essential for normal development and homeostasis of multicellular organisms. Defects in the apoptosis signaling result in many diseases including autoimmune diseases and cancer. The apoptosis signaling pathway was first described genetically in the nematode Caenorhabditis elegans which serves as a framework for the more complex apoptotic pathways that exist in mammals. In this review, we will discuss the apoptotic pathways that are emerging in mammals as elucidated by studies of gene-targeted mutant mice.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Mutation , Signal Transduction , Animals , Apoptosis/genetics , Caspases/genetics , Mice , Mice, Knockout/genetics
11.
Nat Immunol ; 1(1): 77-82, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10881179

ABSTRACT

Brca1 (breast cancerl, early onset) deficiency results in early embryonic lethality. As Brca1 is highly expressed in the T cell lineage, a T cell-specific disruption of Brca1 was generated to assess the role of Brca1 in relation to T lymphocyte development. We found that thymocyte development in Brca1-/- mice was impaired not as a result of V(D)J T cell receptor (TCR) recombination but because thymocytes had increased expression of tumor protein p53. Chromosomal damage accumulation and abnormal cell death were observed in mutant cells. We found that cell death inhibitor Bcl-2 overexpression, or p53-/- backgrounds, completely restored survival and development of Brca1-/- thymocytes; peripheral T cell numbers were not totally restored in Brcal-/- p53-/- mice; and that a mutant background for p21 (cyclin-dependent kinase inhibitor 1A) did not restore Brca1-/- thymocyte development, but partially restored peripheral T cell development. Thus, the outcome of Brca1 deficiency was dependent on cellular context, with the major defects being increased apoptosis in thymocytes, and defective proliferation in peripheral T cells.


Subject(s)
BRCA1 Protein/genetics , BRCA1 Protein/immunology , Gene Rearrangement, T-Lymphocyte/immunology , T-Lymphocytes/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation/immunology , Mice , Mice, Knockout , T-Lymphocytes/cytology
12.
J Immunol ; 163(9): 4909-16, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10528193

ABSTRACT

Caspase-3 is essential for Fas-mediated apoptosis in vitro. We investigated the role of caspase-3 in Fas-mediated cell death in vivo by injecting caspase-3-deficient mice with agonistic anti-Fas Ab. Wild-type controls died rapidly of fulminant hepatitis, whereas the survival of caspase-3-/- mice was increased due to a delay in hepatocyte cell death. Bcl-2 expression in the liver was dramatically decreased in wild-type mice following anti-Fas injection, but was unchanged in caspase-3-/- mice. Hepatocytes from anti-Fas-injected wild-type, but not caspase-3-/-, mice released cytochrome c into the cytoplasm. Western blotting confirmed the lack of caspase-3-mediated cleavage of Bcl-2. Presumably the presence of intact Bcl-2 in caspase-3-/- hepatocytes prevents the release of cytochrome c from the mitochondria, a required step for the mitochondrial death pathway. We also show by Western blot that Bcl-xL, caspase-9, caspase-8, and Bid are processed by caspase-3 in injected wild-type mice but that this processing does not occur in caspase-3-/- mice. This study thus provides novel in vivo evidence that caspase-3, conventionally known for its downstream effector function in apoptosis, also modifies Bcl-2 and other upstream proteins involved in the regulation of Fas-mediated apoptosis.


Subject(s)
Apoptosis/immunology , Caspases/physiology , Liver/enzymology , Liver/immunology , fas Receptor/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Caspase 3 , Caspases/genetics , Cytochrome c Group/metabolism , In Situ Nick-End Labeling , Injections, Intraperitoneal , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational/immunology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Staining and Labeling , Survival Analysis , fas Receptor/immunology
13.
Immunol Rev ; 169: 283-302, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10450525

ABSTRACT

Apoptosis, or programmed cell death (PCD), is the subject of much current investigative interest. Developing embryos and many adult organ systems require the tight coupling of cellular proliferation and PCD to ensure proper organogenesis and optimal tissue function. Over the past decade, our knowledge of the genetic basis underlying the execution of apoptosis in mammals has progressed enormously, thanks largely to groundbreaking studies performed in the nematode Caenorhabditis elegans. In contrast, the components of the signaling apparatus that links the various death stimuli and the receptors they stimulate to the execution mechanism remain relatively unknown. It is only in the past 4 years that studies of signal transduction via members of the tumor necrosis factor (TNF) receptor superfamily have identified a plethora of novel signaling proteins, including molecules that are directly involved in apoptosis signaling, and others that regulate the induction of cell death. This two-part review focuses on the biology of apoptosis and signaling through members of the TNF receptor superfamily as revealed by the study of gene-targeted "knockout" mice. These genetic mutant animals are invaluable tools not only for confirming or refuting a proposed function of a particular gene in an in vivo setting, but also for uncovering novel functions for a gene that were not anticipated from conventional in vitro experiments. In the field of apoptosis, as for many other areas of biomedical research, knockout mice and cell lines can be used as models for studying human disease, with the ultimate goal of developing therapeutic strategies.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/genetics , Apoptosis/immunology , Receptors, Tumor Necrosis Factor/genetics , Animals , Apoptotic Protease-Activating Factor 1 , Carrier Proteins/genetics , Carrier Proteins/immunology , Caspases/genetics , Caspases/immunology , Fas-Associated Death Domain Protein , Gene Targeting , Genes, bcl-2 , Humans , Mice , Mice, Knockout , Models, Biological , Proteins/genetics , Proteins/immunology , Signal Transduction , TNF Receptor-Associated Factor 6
14.
Science ; 284(5411): 156-9, 1999 Apr 02.
Article in English | MEDLINE | ID: mdl-10102818

ABSTRACT

The ability of p53 to promote apoptosis in response to mitogenic oncogenes appears to be critical for its tumor suppressor function. Caspase-9 and its cofactor Apaf-1 were found to be essential downstream components of p53 in Myc-induced apoptosis. Like p53 null cells, mouse embryo fibroblast cells deficient in Apaf-1 and caspase-9, and expressing c-Myc, were resistant to apoptotic stimuli that mimic conditions in developing tumors. Inactivation of Apaf-1 or caspase-9 substituted for p53 loss in promoting the oncogenic transformation of Myc-expressing cells. These results imply a role for Apaf-1 and caspase-9 in controlling tumor development.


Subject(s)
Apoptosis , Caspases/physiology , Genes, p53 , Neoplasms, Experimental/pathology , Proteins/physiology , Animals , Apoptotic Protease-Activating Factor 1 , Caspase 9 , Caspases/genetics , Cell Division , Cell Transformation, Neoplastic , Cells, Cultured , Cytochrome c Group/metabolism , Genes, myc , Genes, ras , Mice , Mice, Nude , Mitochondria/metabolism , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Proteins/genetics , Tumor Suppressor Protein p53/metabolism
15.
Cell ; 94(6): 739-50, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9753321

ABSTRACT

Apoptosis is essential for the precise regulation of cellular homeostasis and development. The role in vivo of Apaf1, a mammalian homolog of C. elegans CED-4, was investigated in gene-targeted Apaf1-/- mice. Apaf1-deficient mice exhibited reduced apoptosis in the brain and striking craniofacial abnormalities with hyperproliferation of neuronal cells. Apaf1-deficient cells were resistant to a variety of apoptotic stimuli, and the processing of Caspases 2, 3, and 8 was impaired. However, both Apaf1-/- thymocytes and activated T lymphocytes were sensitive to Fas-induced killing, showing that Fas-mediated apoptosis in these cells is independent of Apaf1. These data indicate that Apaf1 plays a central role in the common events of mitochondria-dependent apoptosis in most death pathways and that this role is critical for normal development.


Subject(s)
Apoptosis/physiology , Brain/cytology , Brain/embryology , Caspases , Mitochondria/enzymology , Proteins/genetics , Animals , Apoptotic Protease-Activating Factor 1 , Brain Chemistry/physiology , Caspase 2 , Caspase 3 , Caspase 8 , Caspase 9 , Cells, Cultured , Cysteine Endopeptidases/metabolism , Cytochrome c Group/metabolism , Embryo, Mammalian/abnormalities , Enzyme Precursors/metabolism , Fibroblasts/cytology , Fibroblasts/enzymology , Gene Expression , Head/abnormalities , Membrane Potentials/physiology , Mice , Mice, Knockout , Phenotype , Proteins/immunology , Proteins/metabolism , Skin Abnormalities , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/radiation effects , Thymus Gland/chemistry , Thymus Gland/cytology , Thymus Gland/radiation effects , Ultraviolet Rays , fas Receptor/physiology
16.
Cell ; 94(3): 339-52, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9708736

ABSTRACT

Mutation of Caspase 9 (Casp9) results in embryonic lethality and defective brain development associated with decreased apoptosis. Casp9-/- embryonic stem cells and embryonic fibroblasts are resistant to several apoptotic stimuli, including UV and gamma irradiation. Casp9-/- thymocytes are also resistant to dexamethasone- and gamma irradiation-induced apoptosis, but are surprisingly sensitive to apoptosis induced by UV irradiation or anti-CD95. Resistance to apoptosis is accompanied by retention of the mitochondrial membrane potential in mutant cells. In addition, cytochrome c is translocated to the cytosol of Casp9-/- ES cells upon UV stimulation, suggesting that Casp9 acts downstream of cytochrome c. Caspase processing is inhibited in Casp9-/- ES cells but not in thymocytes or splenocytes. Comparison of the requirement for Casp9 and Casp3 in different apoptotic settings indicates the existence of at least four different apoptotic pathways in mammalian cells.


Subject(s)
Apoptosis/genetics , Caspases , Cysteine Endopeptidases/physiology , Signal Transduction/genetics , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase 9 , Cell Line , Cerebral Cortex/abnormalities , Cysteine Endopeptidases/genetics , Cytochrome c Group/metabolism , Dexamethasone/pharmacology , Embryo, Mammalian , Enzyme Activation/genetics , Fibroblasts/cytology , Fibroblasts/enzymology , Gamma Rays , Gene Expression Regulation, Developmental , Lymphocyte Activation , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mitochondria/enzymology , Organ Specificity/genetics , Prosencephalon/abnormalities , Spleen/cytology , Spleen/immunology , Spleen/radiation effects , Stem Cells , Thymus Gland/cytology , Thymus Gland/enzymology
17.
Genes Dev ; 12(6): 806-19, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9512515

ABSTRACT

Caspases are fundamental components of the mammalian apoptotic machinery, but the precise contribution of individual caspases is controversial. CPP32 (caspase 3) is a prototypical caspase that becomes activated during apoptosis. In this study, we took a comprehensive approach to examining the role of CPP32 in apoptosis using mice, embryonic stem (ES) cells, and mouse embryonic fibroblasts (MEFs) deficient for CPP32. CPP32(ex3-/-) mice have reduced viability and, consistent with an earlier report, display defective neuronal apoptosis and neurological defects. Inactivation of CPP32 dramatically reduces apoptosis in diverse settings, including activation-induced cell death (AICD) of peripheral T cells, as well as chemotherapy-induced apoptosis of oncogenically transformed CPP32(-/-) MEFs. As well, the requirement for CPP32 can be remarkably stimulus-dependent: In ES cells, CPP32 is necessary for efficient apoptosis following UV- but not gamma-irradiation. Conversely, the same stimulus can show a tissue-specific dependence on CPP32: Hence, TNFalpha treatment induces normal levels of apoptosis in CPP32 deficient thymocytes, but defective apoptosis in oncogenically transformed MEFs. Finally, in some settings, CPP32 is required for certain apoptotic events but not others: Select CPP32(ex3-/-) cell types undergoing cell death are incapable of chromatin condensation and DNA degradation, but display other hallmarks of apoptosis. Together, these results indicate that CPP32 is an essential component in apoptotic events that is remarkably system- and stimulus-dependent. Consequently, drugs that inhibit CPP32 may preferentially disrupt specific forms of cell death.


Subject(s)
Apoptosis/physiology , Caspases , Cell Nucleus/metabolism , Cysteine Endopeptidases/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , CD3 Complex/pharmacology , Caspase 3 , Cell Death/physiology , Cell Division/physiology , Cysteine Endopeptidases/deficiency , Cysteine Endopeptidases/genetics , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/physiology , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Embryonic and Fetal Development , Female , Gene Expression/genetics , Gene Expression/physiology , Longevity/genetics , Longevity/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Mutation/genetics , Mutation/physiology , Neutrophils/physiology , Osmotic Pressure , Stem Cells/radiation effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology , Tumor Cells, Cultured/radiation effects , Ultraviolet Rays , fas Receptor/pharmacology
18.
J Mammary Gland Biol Neoplasia ; 3(4): 431-45, 1998 Oct.
Article in English | MEDLINE | ID: mdl-10819537

ABSTRACT

In humans, the inheritance of mutations in the breast cancer susceptibility genes BRCA1 and BRCA2 increases the risk of developing breast and ovarian cancer. To study their biological function and to create animal models for these cancer susceptibility genes, several strains of mice mutated in the homologous genes Brca1 and Brca2 have been generated by gene targeting. Analyses of these "knock-out" mouse mutants have provided invaluable knowledge about the function of these genes. Brca1 and Brca2 null mutants are similar in phenotype: mutations in both genes result in embryonic lethality and the developing embryos show signs of a cellular proliferation defect associated with activation of the p53 pathway. The significance of this activation, as well as the role of these cancer susceptibility genes in DNA damage repair, is discussed.


Subject(s)
BRCA1 Protein/physiology , Embryonic and Fetal Development , Genes, BRCA1 , Neoplasm Proteins/physiology , Transcription Factors/physiology , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , BRCA2 Protein , Breast Neoplasms/genetics , Female , Fetal Death , Gene Expression Regulation, Developmental , Genes, p53 , Humans , Mice , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
19.
Nat Genet ; 16(3): 298-302, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9207798

ABSTRACT

Mutations in the mouse Brca1 gene cause lethality at different embryonic stages. We have shown that Brca1 mutant embryos, in which the fifth and sixth exons of Brca1 are deleted die before E7.5 and show decreased cellular proliferation. Brca1 mutants also show decreased expression of mdm2, a gene encoding an inhibitor of p53 activity. Thus, we have proposed that the reduction in mdm2 expression in Brca1 (5-6) mutants might lead to increased p53 activity. Consistent with this finding, the expression of p21, which encodes a G1 cell cycle inhibitor and is a target for p53 transcriptional activation was dramatically increased in the Brca1 (5-6) mutants, suggesting that impaired cellular proliferation could be due to a G1 cell-cycle arrest, caused by increased p21 levels. To test this hypothesis, we generated mice double mutant for Brca1 (5-6) and p53, or Brca1 (5-6) and p21. Mutation in either p53 or p21 prolonged the survival of Brca1 (5-6) mutant embryos from E7.5 to E9.5. The development of most Brca1 (5-6): p21 double-mutant embryos was comparable to that of their wild-type littermates, although no mutant survived past E10.5. The fact that mutation of neither p53 nor p21 completely rescued Brca1 (5-6) embryos suggests that their lethality is likely due to a multi-factorial process.


Subject(s)
Cyclins/genetics , Embryonic and Fetal Development , Genes, BRCA1/genetics , Genes, p53/genetics , Mutation , Animals , BRCA1 Protein/metabolism , Blotting, Southern , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA Probes , Embryo, Mammalian/metabolism , Female , Genotype , Male , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Phenotype , Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism
20.
Genes Dev ; 11(10): 1242-52, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9171369

ABSTRACT

Mutations of the tumor suppressor gene BRCA2 are associated with predisposition to breast and other cancers. Homozygous mutant mice in which exons 10 and 11 of the Brca2 gene were deleted by gene targeting (Brca2(10-11)) die before day 9.5 of embryogenesis. Mutant phenotypes range from severely developmentally retarded embryos that do not gastrulate to embryos with reduced size that make mesoderm and survive until 8.5 days of development. Although apoptosis is normal, cellular proliferation is impaired in Brca2(10-11) mutants, both in vivo and in vitro. In addition, the expression of the cyclin-dependent kinase inhibitor p21 is increased. Thus, Brca2(10-11) mutants are similar in phenotype to Brca1(5-6) mutants but less severely affected. Expression of either of these two genes was unaffected in mutant embryos of the other. This study shows that Brca2, like Brca1, is required for cellular proliferation during embryogenesis. The similarity in phenotype between Brca1 and Brca2 mutants suggests that these genes may have cooperative roles or convergent functions during embryogenesis.


Subject(s)
Cell Division/genetics , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Neoplasm Proteins/genetics , Transcription Factors/genetics , Animals , BRCA2 Protein , DNA-Binding Proteins/genetics , Embryonic and Fetal Development/genetics , Female , Heterozygote , Mesoderm , Mice , Mice, Mutant Strains , Proto-Oncogene Proteins p21(ras)/genetics , Rad51 Recombinase , Trophoblasts
SELECTION OF CITATIONS
SEARCH DETAIL