Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Lupus ; 25(12): 1288-98, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26964561

ABSTRACT

OBJECTIVE: The objective of this paper is to elucidate the not yet known plasma molecule candidates involved in the induction of tissue factor (TF) expression mediated by ß2GPI-dependent anticardiolipin antibody (aCL/ß2GPI) on monocytes. METHODS: Human serum incubated with FLAG-ß2GPI was applied for affinity chromatography with anti- FLAG antibody. Immunopurified proteins were analyzed by a liquid chromatography coupled with mass spectrometry (LC-MS). TF mRNA induced by the identified molecules on monocytes was also analyzed. RESULTS: Apolipoprotein B100 (APOB) was the only identified serum molecule in the MS search. Oxidized LDL, containing APOB as well as ox-Lig1 (a known ligand of ß2GPI), was revealed as a ß2GPI-binding molecule in the immunoprecipitation assay. TF mRNA was markedly induced by oxidized LDL/ß2GPI complexes with either WBCAL-1 (monoclonal aCL/ß2GPI) or purified IgG from APS patients. The activities of lipoprotein-associated phospholipase A2, one of the component molecules of oxidized LDL, were significantly higher in serum from APS patients than in those from controls. CONCLUSION: APOB (or oxidized LDL) was detected as a major ß2GPI binding serum molecule by LC-MS search. Oxidized LDL/aCL/ß2GPI complexes significantly induced TF expressions on monocytes. These data suggest that complexes of oxidized LDL and aCL/ß2GPI may have a crucial role in the pathophysiology of APS.


Subject(s)
Antibodies, Anticardiolipin/metabolism , Antiphospholipid Syndrome/blood , Apolipoprotein B-100/blood , Lipoproteins, LDL/blood , Thromboplastin/biosynthesis , beta 2-Glycoprotein I/immunology , Animals , HEK293 Cells , Humans , Mice , RAW 264.7 Cells
2.
Br J Cancer ; 111(6): 1054-9, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-24853181

ABSTRACT

In addition to the properties of self-renewal and multipotency, stem cells are characterised by their distinct cell cycle status. Somatic stem cells are maintained in a quiescent state but switch reversibly from quiescence to proliferation as needed. On the other hand, embryonic stem cells and induced pluripotent stem cells proliferate rapidly until the induction of differentiation results in inhibition of cell cycle progression. Uncovering the mechanisms underlying cell cycle control in stem cells should thus provide insight into regulation of the balance between self-renewal and differentiation, a key goal of stem cell biology. Recent research has shown that cancer-initiating cells (CICs), a cell population with stem cell-like properties in cancer, are also quiescent, with this characteristic conferring resistance to anticancer therapies that target dividing cells. Elucidation of the mechanisms of CIC quiescence might therefore be expected to provide a basis for the eradication of cancer. This review summarises our current understanding of the role of F-box and WD40 repeat domain-containing 7 (Fbxw7), a key regulator of the cell cycle, in the maintenance of normal stem cells and CICs, as well as attempts to define future challenges in this field.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle , F-Box Proteins/metabolism , Stem Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Differentiation , F-Box-WD Repeat-Containing Protein 7 , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Stem Cells/cytology
3.
Oncogene ; 32(15): 1921-32, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-22665065

ABSTRACT

The tumor suppressor Fbxw7 (also known as Sel-10, hCdc4, hAgo, or Fbw7) is an F-box protein that functions as the substrate-recognition subunit of an SCF ubiquitin ligase complex and targets a group of oncoproteins for degradation. We now show that Fbxw7 regulates the proliferation and differentiation of keratinocytes by mediating the degradation of c-Myc and Notch proteins. Fbxw7-deficient keratinocytes showed an increased proliferative capacity that was dependent on the accumulation of c-Myc but not on that of Notch. Fbxw7 deficiency also resulted in the premature differentiation of keratinocytes in a manner dependent on both c-Myc and Notch. Although Fbxw7-deficient keratinocytes proliferated excessively in vitro, loss of Fbxw7 did not predispose keratinocytes to the formation of squamous cell carcinoma in vivo induced by the expression of oncogenic Ras, possibly because the stem cell population of keratinocytes becomes exhausted as a result of enhanced Notch activity. Indeed, suppression of Notch signaling by additional ablation of RBP-J in Fbxw7-deficient keratinocytes conferred a more aggressive tumorigenic capacity. Collectively, these results indicate that Fbxw7 controls the proliferation and differentiation of keratinocytes, and that it exerts both inhibitory and stimulatory actions in skin carcinogenesis by counteracting the proliferation-promoting effect of c-Myc and the tumor-suppressive effect of Notch, respectively.


Subject(s)
F-Box Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Keratinocytes/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Notch/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation , Cell Transformation, Neoplastic , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Genes, Tumor Suppressor , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Receptors, Notch/genetics , Skin Neoplasms , Ubiquitin-Protein Ligases/genetics
4.
Oncogene ; 31(15): 1975-87, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-21874044

ABSTRACT

RASSF1A is a key tumor-suppressor gene that is often inactivated in a wide variety of solid tumors. Studies have illustrated that RASSF1A plays vital roles in the regulation of cell-cycle progression and functions as a guardian of mitosis. Nevertheless, the precise mechanism of RASSF1A-dependent regulation of mitosis remains largely unclear. APC/C(Cdc20) is the master switch and regulator of mitosis. The activity of APC/C(Cdc20) is tightly controlled by phosphorylation and specific inhibitors to ensure the sequential ubiquitination of downstream targets. Here, we report on the novel finding of a regulated circuitry that controls the timely expression and hence activity of APC/C(Cdc20) during mitosis. Our study showed that RASSF1A and APC/C(Cdc20) form a molecular relay that regulates the APC/C(Cdc20) activity at early mitosis. We found that RASSF1A inhibits APC/C(Cdc20) function through its D-box motifs. Paradoxically, RASSF1A was also demonstrated to be ubiquitinated by APC/C(Cdc20) in vitro and degraded at prometaphase despite of active spindle checkpoint presence. The first two unique D-boxes at the N-terminal of RASSF1A served as specific degron recognized by APC/C(Cdc20). Importantly, we found that Aurora A and Aurora B directly phosphorylate RASSF1A, a critical step by which RASSF1A switches from being an inhibitor to a substrate of APC/C(Cdc20) during the course of mitotic progression. As a result of RASSF1A degradation, APC/C(Cdc20) can then partially activate the ubiquitination of Cyclin A in the presence of spindle checkpoint. This circuitry is essential for the timely degradation of Cyclin A. To conclude, our results propose a new model for RASSF1A-APC/C(Cdc20) interaction in ensuring the sequential progression of mitosis.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Cycle Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Aurora Kinase B , Aurora Kinases , Cdc20 Proteins , Cyclin A/metabolism , HEK293 Cells , HeLa Cells , Humans , Mitosis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Transfection , Ubiquitination
5.
Cell Death Dis ; 1: e17, 2010.
Article in English | MEDLINE | ID: mdl-21364618

ABSTRACT

As apoptotic pathways are commonly deregulated in breast cancer, exploring how mammary gland cell death is regulated is critical for understanding human disease. We show that primary mammary epithelial cells from protein kinase C delta (PKCδ) -/- mice have a suppressed response to apoptotic agents in vitro. In the mammary gland in vivo, apoptosis is critical for ductal morphogenesis during puberty and involution following lactation. We have explored mammary gland development in the PKCδ -/- mouse during these two critical windows. Branching morphogenesis was altered in 4- to 6-week-old PKCδ -/- mice as indicated by reduced ductal branching; however, apoptosis and proliferation in the terminal end buds was unaltered. Conversely, activation of caspase-3 during involution was delayed in PKCδ -/- mice, but involution proceeded normally. The thymus also undergoes apoptosis in response to physiological signals. A dramatic suppression of caspase-3 activation was observed in the thymus of PKCδ -/- mice treated with irradiation, but not mice treated with dexamethasone, suggesting that there are both target- and tissue-dependent differences in the execution of apoptotic pathways in vivo. These findings highlight a role for PKCδ in both apoptotic and nonapoptotic processes in the mammary gland and underscore the redundancy of apoptotic pathways in vivo.


Subject(s)
Apoptosis , Mammary Glands, Animal/growth & development , Protein Kinase C-delta/physiology , Animals , Caspase 3/metabolism , Cells, Cultured , Dexamethasone/pharmacology , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Thymus Gland/metabolism , Thymus Gland/radiation effects
6.
Oncogene ; 29(12): 1798-809, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20023701

ABSTRACT

The F-box protein Fbxw7 (also known as Fbw7, SEL-10, hCdc4 or hAgo) mediates the ubiquitylation and thereby contributes to the degradation of proteins that positively regulate cell cycle. Conditional ablation of Fbxw7 in mouse embryonic fibroblasts (MEFs) induces cell-cycle arrest accompanied by abnormal accumulation of the intracellular domain of Notch1 (NICD1) and c-Myc. However, the molecular mechanisms by which the accumulation of NICD1 and c-Myc induces cell-cycle arrest have remained unclear. We have now examined the expression of cell-cycle inhibitors in Fbxw7-deficient MEFs and found that the abundance of p27(Kip1) and p57(Kip2) is paradoxically decreased. This phenomenon appears to be attributable to the accumulation of NICD1, given that it was recapitulated by overexpression of NICD1 and blocked by ablation of RBP-J. Conversely, the expression of p16(Ink4a) and p19(ARF) was increased in an NICD1-independent manner in Fbxw7-null MEFs. The increased expression of p19(ARF) was recapitulated by overexpression of c-Myc and abolished by ablation of c-Myc, suggesting that the accumulation of c-Myc is primarily responsible for that of p19(ARF). In contrast, the upregulation of p16(Ink4a) appeared to be independent of c-Myc. These results indicate that cell-cycle inhibitors undergo complex regulation by the Fbxw7-mediated proteolytic system.


Subject(s)
Cell Cycle/physiology , F-Box Proteins/pharmacology , Fibroblasts/cytology , Fibroblasts/physiology , Ubiquitin-Protein Ligases/pharmacology , Animals , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p57/drug effects , Cyclin-Dependent Kinase Inhibitor p57/genetics , DNA Primers , Down-Regulation/drug effects , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Fibroblasts/drug effects , Homeostasis/drug effects , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/pharmacology , RNA Interference , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination
7.
Oncogene ; 27(47): 6164-74, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18641686

ABSTRACT

The F-box protein Fbxw7 mediates the ubiquitylation and consequent degradation of proteins that regulate cell cycle progression, including cyclin E, c-Myc, c-Jun and Notch. Moreover, certain human cancer cell lines harbor loss-of-function mutations in FBXW7 that result in excessive accumulation of Fbxw7 substrates, implicating Fbxw7 in tumor suppression. To elucidate the physiological function of Fbxw7, we conditionally ablated Fbxw7 in mouse embryonic fibroblasts (MEFs). Unexpectedly, loss of Fbxw7 induced cell cycle arrest and apoptosis that were accompanied by abnormal accumulation of the intracellular domain of Notch1 (NICD1). Forced expression of NICD1 in wild-type MEFs recapitulated the phenotype of the Fbxw7-deficient (Fbxw7(Delta/Delta)) MEFs. Conversely, deletion of Rbpj normalized the phenotype of Fbxw7(Delta/Delta) MEFs, indicating that this phenotype is dependent on the Notch1-RBP-J signaling pathway. Deletion of the p53 gene prevented cell cycle arrest but not the induction of apoptosis in Fbxw7(Delta/Delta) cells. These observations suggest that Fbxw7 does not function as an oncosuppressor in MEFs. Instead, it promotes cell cycle progression and cell survival through degradation of Notch1, with loss of Fbxw7 resulting in NICD1 accumulation, cell cycle arrest and apoptosis.


Subject(s)
Apoptosis , Cell Cycle , F-Box Proteins/physiology , Receptor, Notch1/physiology , Ubiquitin-Protein Ligases/physiology , Amyloid Precursor Protein Secretases/physiology , Animals , Cell Proliferation , Cell Survival , F-Box-WD Repeat-Containing Protein 7 , Fibroblasts/cytology , Mice , Proteasome Endopeptidase Complex/physiology , Receptor, Notch1/analysis , Receptor, Notch2/analysis , Receptor, Notch2/physiology , Receptor, Notch3 , Receptors, Notch/analysis , Receptors, Notch/physiology , Signal Transduction , Tumor Suppressor Protein p53/physiology
8.
Oncogene ; 27(22): 3176-85, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18071316

ABSTRACT

The tumor suppressor RASSF1A is inactivated in many human cancers and is implicated in regulation of microtubule stability, cell cycle progression and apoptosis. However, the precise mechanisms of RASSF1A action and their regulation remain unclear. Here we show that Skp2, an oncogenic subunit of the Skp1-Cul1-F-box ubiquitin ligase complex, interacts with, ubiquitinates, and promotes the degradation of RASSF1A at the G1-S transition of the cell cycle. This Skp2-dependent destruction of RASSF1A requires phosphorylation of the latter on serine-203 by cyclin D-cyclin-dependent kinase 4. Interestingly, mutation of RASSF1A-phosphorylation site Ser(203) to alanine results in a delay in cell cycle progression from G1 to S phase. Moreover, enforced expression of Skp2 abolishes the inhibitory effect of RASSF1A on cell proliferation. Finally, the delay in G1-S progression after Skp2 removal is normalized by depletion of RASSF1A. These findings suggest that the Skp2-mediated degradation of RASSF1A plays an important role in cell proliferation and survival.


Subject(s)
Cell Proliferation , G1 Phase , Proteasome Endopeptidase Complex/metabolism , S Phase , S-Phase Kinase-Associated Proteins/physiology , Tumor Suppressor Proteins/metabolism , Ubiquitination , Animals , Cell Survival/genetics , Cells, Cultured , Cyclin D , Cyclin-Dependent Kinase 4/metabolism , Cyclins/metabolism , G1 Phase/genetics , Genes, Tumor Suppressor/physiology , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation , Protein Binding , Protein Processing, Post-Translational/genetics , S Phase/genetics , S-Phase Kinase-Associated Proteins/metabolism , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Ubiquitination/genetics
9.
Oncogene ; 25(56): 7391-400, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-16767160

ABSTRACT

Reversible acetylation on protein lysine residues has been shown to regulate the function of both nuclear proteins such as histones and p53 and cytoplasmic proteins such as alpha-tubulin. To identify novel acetylated proteins, we purified several proteins by the affinity to an anti-acetylated-lysine antibody from cells treated with trichostatin A (TSA). Among the proteins identified, here we report acetylation of the SV40 large T antigen (T-Ag). The acetylation site was determined to be lysine-697, which is located adjacent to the C-terminal Cdc4 phospho-degron (CPD). Overexpression of the CBP acetyltransferase acetylated T-Ag, whereas HDAC1, HDAC3 and SIRT1 bound and deacetylated T-Ag. The acetylation and deacetylation occurred independently of p53, a binding partner of T-Ag, but the acetylation was enhanced in the presence of p53. T-Ag in the cells treated with TSA and NA or the acetylation mimic mutant (K697Q) became unstable in COS-7 cells, suggesting that acetylation regulates stability of T-Ag. Indeed, NIH3T3 cells stably expressing K697Q showed decreased anchorage-independent growth compared with those expressing wild type or the K697R mutant. These results demonstrate that acetylation destabilizes T-Ag and regulates the transforming activity of T-Ag in NIH3T3 cells.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Acetylation , Amino Acid Sequence , Animals , Antigens, Polyomavirus Transforming/chemistry , Base Sequence , COS Cells , Chlorocebus aethiops , Chromatography, High Pressure Liquid , DNA Primers , Histone Deacetylases/metabolism , Mice , Molecular Sequence Data , Sirtuin 1 , Sirtuins/metabolism , Spectrometry, Mass, Electrospray Ionization
10.
Oncogene ; 25(12): 1786-98, 2006 Mar 16.
Article in English | MEDLINE | ID: mdl-16288221

ABSTRACT

The transcription factor E2F mediates cell cycle-dependent expression of genes important for cell proliferation in response to growth stimulation. To further understand the role of E2F, we utilized a sensitive subtraction method to explore new E2F1 targets, which are expressed at low levels and might have been unrecognized in previous studies. We identified 33 new E2F1-inducible genes, including checkpoint genes Claspin and Rad51ap1, and four genes with unknown function required for cell cycle progression. Moreover, we found three groups of E2F1-inducible genes that were not induced by growth stimulation. At least, two groups of genes were directly induced by E2F1, indicating that E2F1 can regulate expression of genes not induced during the cell cycle. One included Neogenin, WASF1 and SGEF genes, which may have a role in differentiation or development. The other was the cyclin-dependent kinase inhibitor p27(Kip1), which was involved in suppression of inappropriate cell cycle progression induced by deregulated E2F. E2F1-responsive regions of these genes were located more upstream than those of typical E2F targets and did not have typical E2F sites. These results indicate that there are groups of E2F1 targets, which are regulated in a distinct manner from that of typical E2F targets.


Subject(s)
Cell Cycle/physiology , E2F1 Transcription Factor/metabolism , Gene Expression Regulation , Oligonucleotide Array Sequence Analysis/methods , Animals , Blotting, Northern , Blotting, Western , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression , Humans
11.
J Biol Chem ; 276(35): 33111-20, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11435423

ABSTRACT

The U box is a domain of approximately 70 amino acids that is present in proteins from yeast to humans. The prototype U box protein, yeast Ufd2, was identified as a ubiquitin chain assembly factor that cooperates with a ubiquitin-activating enzyme (E1), a ubiquitin-conjugating enzyme (E2), and a ubiquitin-protein ligase (E3) to catalyze ubiquitin chain formation on artificial substrates. E3 enzymes are thought to determine the substrate specificity of ubiquitination and have been classified into two families, the HECT and RING finger families. Six mammalian U box proteins have now been shown to mediate polyubiquitination in the presence of E1 and E2 and in the absence of E3. These U box proteins exhibited different specificities for E2 enzymes in this reaction. Deletion of the U box or mutation of conserved amino acids within it abolished ubiquitination activity. Some U box proteins catalyzed polyubiquitination by targeting lysine residues of ubiquitin other than lysine 48, which is utilized by HECT and RING finger E3 enzymes for polyubiquitination that serves as a signal for proteolysis by the 26 S proteasome. These data suggest that U box proteins constitute a third family of E3 enzymes and that E4 activity may reflect a specialized type of E3 activity.


Subject(s)
Fungal Proteins/chemistry , Fungal Proteins/metabolism , Ligases/chemistry , Ligases/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Binding Sites , Brain/metabolism , Cloning, Molecular , Consensus Sequence , Conserved Sequence , DNA, Complementary , Fungal Proteins/genetics , Humans , Ligases/genetics , Lysine , Male , Mice , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Testis/metabolism , Thymus Gland/metabolism , Ubiquitin-Activating Enzymes , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
12.
Biochem Biophys Res Commun ; 282(4): 853-60, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11352628

ABSTRACT

The transition from G1 phase to S phase of the mammalian cell cycle is controlled by many positive and negative regulators, among which cyclin E and p27Kip1, respectively, undergo the most marked changes in concentration at this transition. The abundance of both cyclin E and p27Kip1 is regulated predominantly by posttranslational mechanisms, in particular by proteolysis mediated by the ubiquitin-proteasome pathway. Cyclin E and p27Kip1 each bind to and undergo polyubiquitination by the same ubiquitin ligase, known as SCF(Skp2). The degradation of cyclin E and p27Kip1 is greatly impaired in Skp2-deficient mice, resulting in intracellular accumulation of these proteins. In this article, recent progress in characterization of the molecular mechanisms that control the proteolysis of cyclin E and p27Kip1 is reviewed.


Subject(s)
Cell Cycle Proteins , Cell Cycle , Cyclin E/metabolism , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins , Animals , Cyclin E/physiology , Cyclin-Dependent Kinase Inhibitor p27 , Cysteine Endopeptidases/metabolism , G1 Phase , Ligases/genetics , Ligases/metabolism , Ligases/physiology , Mice , Mice, Knockout , Microtubule-Associated Proteins/physiology , Models, Biological , Multienzyme Complexes/metabolism , Neoplasms/metabolism , Peptide Synthases/metabolism , Polyploidy , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational , S Phase , SKP Cullin F-Box Protein Ligases , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
13.
Biochem Biophys Res Commun ; 284(1): 142-9, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11374883

ABSTRACT

Mice that lack caspase-3, which functions in apoptosis, were generated by gene targeting and shown to undergo hearing loss. The ABR threshold of the caspase-3(-/-) mice was significantly elevated compared to that of caspase-3(+/+) mice at 15 days of age and was progressively elevated further by 30 days. Distortion product otoacoustic emissions were not detectable in caspase-3(-/-) mice at 15 days of age. Caspase-3(-/-) mice exhibited marked degeneration of spiral ganglion neurons and a loss of inner and outer hair cells in the cochlea at 30 days of age, although no such changes were apparent at 15 days. The degenerating neurons manifested features, including cytoplasmic vacuolization, distinct from those characteristic of apoptosis. Spiral ganglion neurons and cochlear hair cells thus appear to require caspase-3 for survival but not for initial development. The mapping of both the human caspase-3 gene and the locus responsible for an autosomal dominant, nonsyndromic form of hearing loss (DFNA24) to chromosome 4q35 suggests that the caspase-3(-/-) mice may represent a model of this human condition.


Subject(s)
Caspases/deficiency , Cochlea/innervation , Deafness/genetics , Neurons/pathology , Aging/pathology , Animals , Auditory Threshold , Caspase 3 , Caspases/biosynthesis , Caspases/genetics , Cell Count , Cell Death/genetics , Cochlea/metabolism , Cochlea/pathology , Deafness/congenital , Deafness/pathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Outer/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Otoacoustic Emissions, Spontaneous/genetics , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Vacuoles/pathology
14.
Int J Mol Med ; 2(4): 403-8, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9857224

ABSTRACT

The mechanism by which radiation induces human peripheral T cell apoptosis is not known. We examined sequential changes in post-irradiated peripheral blood mononuclear cells (PBMC(S)) taken from normal volunteers, by using flow-cytometer and an anti-CD3 monoclonal antibody, annexin V, propidium iodide, anti-Fas antibody, and anti-Fas ligand antibody. After 5 or 10 Gy of irradiation with a 60Co radiation therapy unit, most of the human peripheral T cells showed positivity against annexin V in 15 h, and positivity against propidium iodide in 23 h after irradiation. On a microscopy-video system, approximately 80% of mononuclear cells revealed apoptotic changes in 24 h after irradiation. Because of its proposed role in activation-induced cytotoxicity, we also examined the Fas (CD95/Apo-1) pathway in killing T cells by irradiation. Irradiated PBMC, displayed no increase in surface Fas expression and caspase-3 activity relative to non-irradiated cells. In addition, the anti-Fas ligand failed to eliminate the apoptotic death of PBMC, after irradiation. These results suggest that irradiation induces direct apoptosis of T cells by a Fas-independent mechanism.


Subject(s)
T-Lymphocytes/radiation effects , fas Receptor/biosynthesis , Antibodies, Monoclonal/pharmacology , Apoptosis , Caspase 3/metabolism , Cells, Cultured , Fas Ligand Protein/immunology , Fas Ligand Protein/metabolism , Humans , Microscopy/methods , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Video Recording
SELECTION OF CITATIONS
SEARCH DETAIL