Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Oncol ; 60(3)2022 Mar.
Article in English | MEDLINE | ID: mdl-35103287

ABSTRACT

Oral cancer is a leading cause of cancer­related death worldwide. Current treatment for oral cancer includes surgery, radiotherapy, and chemotherapy; however, their effectiveness is still limited. To identify a new prognostic biomarker and therapeutic target for oral cancer, the Opa interacting protein 5 (OIP5), which plays an essential role in the proper segregation of chromosomes, was examined. Immunohistochemical staining using tissue microarrays indicated that OIP5 was expressed in 120 of 164 (73.2%) oral cancers but was minimally expressed in normal oral tissues. OIP5 expression was significantly associated with poor prognosis in patients with oral cancer. Overexpression of OIP5 enhanced the growth of oral cancer cells, whereas OIP5 knockdown using small interfering RNAs (siRNAs) significantly inhibited cell growth through cell cycle arrest at the G2/M phase. Suppression of OIP5 expression also induced senescence of oral cancer cells. Overall, the findings of the present study suggest that OIP5 may be a candidate prognostic biomarker and therapeutic target in oral cancer.


Subject(s)
Cell Cycle Proteins/analysis , Chromosomal Proteins, Non-Histone/analysis , Mouth Neoplasms/drug therapy , Analysis of Variance , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Cell Cycle Proteins/blood , Cell Cycle Proteins/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/genetics , Chromosomal Proteins, Non-Histone/blood , Chromosomal Proteins, Non-Histone/drug effects , Humans , Mouth Neoplasms/physiopathology
2.
Exp Cell Res ; 405(2): 112658, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34038745

ABSTRACT

Intervertebral disc degeneration (IDD) results from dysregulated metabolism of the extracellular matrix of the nucleus pulposus (NP) and involves the participation of inflammatory factors such as TNF-α. Bromodomain-containing protein 7 (BRD7) shows considerable potential for anti-inflammatory applications. Herein, we investigated the role of BRD7 in IDD. The immunohistochemistry results demonstrated decreased BRD7 expression in severely degenerated human NP tissues compared to those showing mild degeneration. Lentiviruses and adenoviruses were used to knock down or overexpress BRD7 and YAP1, respectively. Our results revealed that BRD7 knockdown promoted matrix degradation and suppressed PI3K and YAP1 expression, while BRD7 overexpression alleviated matrix degradation and promoted YAP1 and PI3K expression. In addition, PI3K inhibition augmented matrix degradation, enhanced apoptosis, and reduced YAP1 expression, whereas YAP1 overexpression promoted matrix synthesis, suppressed apoptosis and promoted PI3K expression. Besides, BRD7 overexpression reversed the reductions in sulfated glycosaminoglycan levels induced by TNF-α, but this effect was blocked by PI3K or YAP1 inhibitors. Moreover, YAP1 and PI3K were shown to interact through coimmunoprecipitation analysis. In summary, our results demonstrate that BRD7 can regulate matrix metabolism and apoptosis in human NP cells through the BRD7-PI3K-YAP1 signaling axis. This study might provide new insights into the prevention and treatment of IDD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/physiology , Extracellular Matrix/metabolism , Intervertebral Disc Degeneration/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis/drug effects , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Humans , Nucleus Pulposus/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism
3.
Aging (Albany NY) ; 12(20): 20835-20861, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33125346

ABSTRACT

OBJECTIVE: Screening for novel prognostic biomarkers and potential therapeutic targets from colorectal cancer microenvironment. RESULTS: 372 genes were overexpressed in colorectal cancer microenvironment, five of which that had the most prognostic powers were enriched in Epithelial-Mesenchymal Transition and cell cycle pathways. For the first time, we showed that SMARCD3 was mainly expressed in CAFs and could be a novel prognostic marker and potential therapeutic target. Function analyses indicated that MSARCD3 might promote CAFs activation and colorectal cancer metastasis through SMARCD3-WNT5A/TGF-ß-MAPK14-SMARCD3 positive feedback loop. Signaling map of SMARCD3 was constructed and several potential drugs that could regulate SMARCD3 were also presented. CONCLUSIONS: SMARCD3 is a novel prognostic biomarker and potential therapeutic target of colorectal cancer, which may promote cancer metastasis through activation of CAFs. METHODS: Colorectal cancer microenvironment related genes were screened based on immune and stromal scores. Function enrichment analyses were performed to show the underlying mechanistic insights of these tumor microenvironment related genes. Kaplan-Meier survival analysis was used for evaluating the prognostic power. Gene-Pathway interaction network analysis and cellular heterogeneity analysis of tumor microenvironment were also performed. Gene set enrichment analysis was performed for signal gene pathway analysis. Protein data from The Cancer Genome Atlas were used for validation.


Subject(s)
Biomarkers, Tumor/genetics , Cancer-Associated Fibroblasts , Chromosomal Proteins, Non-Histone/genetics , Colonic Neoplasms/genetics , Biomarkers, Tumor/biosynthesis , Cancer-Associated Fibroblasts/metabolism , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomal Proteins, Non-Histone/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Survival Rate , Tumor Microenvironment/genetics
4.
Mol Cell ; 67(6): 1001-1012.e6, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28844864

ABSTRACT

BET proteins commonly activate cellular gene expression, yet inhibiting their recruitment paradoxically reactivates latent HIV-1 transcription. Here we identify the short isoform of BET family member BRD4 (BRD4S) as a corepressor of HIV-1 transcription. We found that BRD4S was enriched in chromatin fractions of latently infected T cells, and it was more rapidly displaced from chromatin upon BET inhibition than the long isoform. BET inhibition induced marked nucleosome remodeling at the latent HIV-1 promoter, which was dependent on the activity of BRG1-associated factors (BAF), an SWI/SNF chromatin-remodeling complex with known repressive functions in HIV-1 transcription. BRD4S directly bound BRG1, a catalytic subunit of BAF, via its bromodomain and extraterminal (ET) domain, and this isoform was necessary for BRG1 recruitment to latent HIV-1 chromatin. Using chromatin immunoprecipitation sequencing (ChIP-seq) combined with assay for transposase-accessible chromatin coupled to high-throughput sequencing (ATAC-seq) data, we found that the latent HIV-1 promoter phenotypically resembles endogenous long terminal repeat (LTR) sequences, pointing to a select role of BRD4S-BRG1 complexes in genomic silencing of invasive retroelements.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA, Viral/metabolism , HIV-1/metabolism , Nuclear Proteins/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Virus Latency , Azepines/pharmacology , Cell Cycle Proteins , Chromatin/genetics , Chromatin Assembly and Disassembly/drug effects , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , DNA, Viral/genetics , Dose-Response Relationship, Drug , Down-Regulation , Gene Expression Regulation, Viral , HEK293 Cells , HIV-1/drug effects , HIV-1/genetics , HIV-1/immunology , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Humans , Jurkat Cells , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , RNA Interference , Retroelements , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Time Factors , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transfection , Triazoles/pharmacology , Virus Latency/drug effects
5.
J Biol Chem ; 285(9): 6412-8, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20040597

ABSTRACT

Human solid tumors contain rare cancer side population (SP) cells, which expel the fluorescent dye Hoechst 33342 (H33342) and display cancer stem cell characteristics. Transcriptional profiling of cancer SP cells isolated by H33342 fluorescence analysis is a newly emerging approach to discover cancer stem cell markers and aberrant differentiation pathways. Using Affymetrix expression microarrays and quantitative reverse transcription-PCR, we investigated differential gene expression between SP and non-SP (NSP) cells isolated from human mammary carcinoma cell lines. A total of 136 genes were up-regulated in breast cancer SP relative to NSP cells, one of which was the fetal stem cell factor and Wnt/beta-catenin signaling pathway target SOX17. Strikingly, we discovered that SOX17 was down-regulated by H33342 in a dose-dependent manner. In SP cells, which expel H33342, down-regulation of SOX17 was less pronounced than in NSP cells, which retain H33342. As a result of this, SOX17 displayed a 10-20-fold overexpression in cancer SP relative to NSP cells. Similar results were obtained for further stemness-related genes, namely EPC1 and SPRY1. These findings establish a previously unidentified gene-regulatory impact of H33342 as a novel mechanism responsible for differential gene expression in cancer SP cells. This has significant implications for the future interpretation of cancer SP cells.


Subject(s)
Benzimidazoles/pharmacology , Breast Neoplasms/pathology , Down-Regulation/drug effects , SOXF Transcription Factors/genetics , Biomarkers, Tumor , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/genetics , Female , Fluorescent Dyes , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/drug effects , Membrane Proteins/genetics , Phosphoproteins/drug effects , Phosphoproteins/genetics , Repressor Proteins/drug effects , Repressor Proteins/genetics , SOXF Transcription Factors/drug effects , Stem Cells/chemistry
6.
Clin Cancer Res ; 14(23): 7588-92, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047083

ABSTRACT

Cell cycle checkpoints have long been recognized as important nodes for regulating cell proliferation and maintaining genomic integrity. These checkpoints are often altered in cancer and represent promising points for therapeutic intervention. Until recently, direct targeting of the mitotic checkpoint has been an untapped area for cancer drug discovery. Regulation of the mitotic checkpoint is complex, but many of the critical players have been identified and functionally characterized. A substantial number of these proteins can be localized to the kinetochore, a structure located at the centromeric region of each mitotic chromosome. The kinetochore mediates chromosome attachment to spindle microtubules and subsequent chromosome movement. The mitotic checkpoint monitors microtubule attachment and chromosome position on the mitotic spindle, inhibiting progression into anaphase until proper attachment and metaphase positioning is achieved. Centromere-associated protein E is a kinesin microtubule motor protein that plays an essential role in integrating the mechanics of microtubule-chromosome interactions with mitotic checkpoint signaling, and has emerged as a novel target for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Neoplasms/drug therapy , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Genes, cdc/drug effects , Genes, cdc/physiology , Humans , Mitosis/drug effects , Mitosis/physiology
7.
Zhonghua Bing Li Xue Za Zhi ; 37(2): 124-8, 2008 Feb.
Article in Chinese | MEDLINE | ID: mdl-18681325

ABSTRACT

OBJECTIVE: To study the influence of siRNA inhibition of CENP-A expression on the biological behavior of HepG2 cells. METHODS: Three pairs of 21 bp reverse repeated motifs of CENP-A target sequence with 9 spacer were synthesized and inserted into vector pSilencer 2.1-U6 neo to generate siRNA eukaryotic expression plasmids. After stable transfection into HepG2 cells, cell growth, apoptosis, cell cycles and plate clone forming efficiency were investigated. Expressions of CENP-A mRNA was monitored by the reverse transcriptase polymerase chain reaction (RT-PCR). The protein expression of CENP-A, bcl-2, Bax, p53, p21waf1 and mdm2 were detected by Western-blotting. RESULTS: Two eukaryotic expression plasmids with significant siRNA specific inhibition to the CENP-A gene were created. Compared with control cells, HepG2 cells transfected with the constructs showed G1 phase delay (P < 0.01) and cell number decrease in the S phase (P < 0.001), along with an increased apoptotic rate (P = 0.003), significant increase of Bax expression and decreased bcl-2 expression (P< or =0.001). The protein expressions of p21waf1 was higher and mdm2 was lower than those of the control groups. However, the wild type p53 protein expression was not effected by CENP-A siRNA. CONCLUSIONS: An altered expression of CENP-A may be related to the proliferation of hepatocellular carcinoma through cell cycle regulation involving an altered bcl-2/Bax expression, that may be p53 independent.


Subject(s)
Autoantigens/genetics , Carcinoma, Hepatocellular/pathology , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Neoplastic/drug effects , RNA Interference/drug effects , RNA, Small Interfering/pharmacology , Autoantigens/drug effects , Cell Line, Tumor , Centromere Protein A , Chromosomal Proteins, Non-Histone/drug effects , Hep G2 Cells , Humans , RNA, Small Interfering/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
8.
Exp Mol Pathol ; 78(3): 198-206, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15924871

ABSTRACT

To determine if nuclear factor-kappaB (NF-kB) plays a role in Mallory body (MB) formation, quantitative real-time RT-PCR assay was used to measure liver NF-kappaB1/p105 mRNA levels in 4 different groups of mice. Group 1: mice given IP saline for 15 weeks; group 2: mice fed diethyl 1,4-dihydro-2,4,6,-trimethyl-3,5-pyridinedicarboxylate (DDC) for 10 weeks when MBs were formed; group3: mice fed DDC 10 weeks, then withdrawn 5 weeks when MBs disappeared; group 4: mice fed DDC 10 weeks, withdrawn 4 weeks, then fed DDC+chlormethiazole (CMZ) for 1 week when MBs again formed. The mRNA for p105 NF-kappaB expression was significantly increased in the livers of mice treated with DDC (group 2) and DDC+CMZ (group 4) compared with the control livers (group 1) as well as the drug-withdrawal livers (group 3). Primary cultures of hepatocytes from drug-primed mice (the group 4 mice were withdrawn for another 4 weeks when the MBs had disappeared) were studied. The hepatocytes from drug-primed mice were MB free when isolated and used for primary culture. MBs began to form spontaneously within their cytoplasm after 2-3 days of culture. The NF-kappaB inhibitor (NF-kappaBi), a cell-permeable quinazoline compound that acts as a potent inhibitor of NF-kappaB transcriptional activation, was added to the medium 3 h after planting the cultures of liver cells. No MBs formed in the cells treated with 10 microM, 1 microM, and 0.1 microM NF-kappaBi for 6 days. MBs still formed in the cells treated with 10 nM NF-kappaBi for 6 days. Both DDC-primed and normal control liver cells began to enlarge and elongate after a few hours of culture. In contrast, the cells treated with NF-kappaBi stayed polyhedral in shape just as they appeared prior to culturing. The level of NF-kappaB1/p105 mRNA significantly increased in DDC-primed hepatocytes after 24 h of culture and in normal control hepatocytes after 48 h of culture. In DDC-primed hepatocytes, NF-kappaBi 0.1 muM treatment for 6 days significantly decreased mRNA expression of Src, p105/NF-kappaB1, ERK1, MEKK1, and JNK1/2. In normal control liver cells, NF-kappaBi treatment decreased mRNA expression of Src and JNK1 and stimulated the mRNA expression of p105/NF-kappaB1 and Junk2. NF-kappaBi treatment significantly decreased the total ERK1/2 protein and further decreased the phosphorylated (activated) form of ERK1/2 in the cultured hepatocytes. The results indicate that the p105 NF-kappaB pathway which putatively regulates ERK at both the transcriptional and post-translational levels regulates MB formation by way of changes in gene expression.


Subject(s)
Inclusion Bodies/metabolism , Liver Diseases/pathology , Liver/pathology , Models, Biological , NF-kappa B/metabolism , Protein Precursors/metabolism , Animals , Antigens, Nuclear/drug effects , Antigens, Nuclear/metabolism , Blotting, Western , Cells, Cultured , Chemical and Drug Induced Liver Injury , Chlormethiazole/pharmacology , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Dihydropyridines/toxicity , Enzyme Inhibitors/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Inclusion Bodies/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/metabolism , Liver Diseases/metabolism , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinase 1/metabolism , Male , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/drug effects , NF-kappa B p50 Subunit , Protein Precursors/drug effects , Quinazolines/pharmacology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
9.
J Cell Sci ; 117(Pt 18): 4277-87, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15292402

ABSTRACT

The effect of trichostatin A (TSA)-induced histone acetylation on the interphase chromatin structure was visualized in vivo with a HeLa cell line stably expressing histone H2A, which was fused to enhanced yellow fluorescent protein. The globally increased histone acetylation caused a reversible decondensation of dense chromatin regions and led to a more homogeneous distribution. These structural changes were quantified by image correlation spectroscopy and by spatially resolved scaling analysis. The image analysis revealed that a chromatin reorganization on a length scale from 200 nm to >1 microm was induced consistent with the opening of condensed chromatin domains containing several Mb of DNA. The observed conformation changes could be assigned to the folding of chromatin during G1 phase by characterizing the effect of TSA on cell cycle progression and developing a protocol that allowed the identification of G1 phase cells on microscope coverslips. An analysis by flow cytometry showed that the addition of TSA led to a significant arrest of cells in S phase and induced apoptosis. The concentration dependence of both processes was studied.


Subject(s)
Chromatin/drug effects , Histones/metabolism , Hydroxamic Acids/pharmacology , Protein Synthesis Inhibitors/pharmacology , Acetylation/drug effects , Apoptosis/drug effects , Apoptosis/physiology , Cell Nucleus/drug effects , Cell Nucleus/physiology , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Fractals , G1 Phase/drug effects , G1 Phase/physiology , Genes, cdc/drug effects , Genes, cdc/physiology , HeLa Cells , Histone Deacetylases/metabolism , Histones/genetics , Humans , Image Processing, Computer-Assisted , Interphase/drug effects , Interphase/physiology , Microscopy, Energy-Filtering Transmission Electron , S Phase/drug effects , S Phase/physiology
10.
Neurobiol Dis ; 15(3): 529-33, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15056460

ABSTRACT

In this study, we have shown that a paraneoplastic cerebellar degeneration (PCD)-associated antigen, pcd17, binds to a cell cycle-related protein, MRG15. MRG15 derepresses the E2F-responsive B-myb promoter. The pcd17 antigen inhibits the derepression of the B-myb transcriptional activity by MRG15, and, as a result, pcd17 represses the promoter. Delivery of anti-Purkinje cell antibodies (anti-Yo) into the cells inhibits the repression of B-myb promoter activity by pcd17. Because derepression of the B-myb promoter has been implicated in neuronal death, the results suggest the possible role of the antibodies in the pathogenesis of PCD.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/drug effects , Nerve Degeneration/pathology , Nerve Tissue Proteins/metabolism , Paraneoplastic Cerebellar Degeneration/physiopathology , Purkinje Cells/immunology , Trans-Activators/drug effects , Animals , COS Cells , Chlorocebus aethiops , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , E2F Transcription Factors , Immunoglobulin G/pharmacology , Neoplasm Proteins/immunology , Nerve Degeneration/metabolism , Promoter Regions, Genetic , Purkinje Cells/pathology , Trans-Activators/metabolism , Transcription Factors/metabolism , Two-Hybrid System Techniques
11.
Environ Health Perspect ; 110 Suppl 5: 877-81, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12426151

ABSTRACT

We have studied the effect of gender, genetics, and toxicokinetics on immune parameters in mercury-induced autoimmunity in mice. Data strongly suggest that the mechanism for mercury-induced autoimmunity involves modification of the autoantigen fibrillarin by mercury followed by a T-cell-dependent immune response driven by the modified fibrillarin. Mice with different H-2 haplotypes were treated with (203)HgCl(2) in a dose of 0.5-16 mg Hg/L drinking water for 10 weeks. Whole-body accumulation and renal accumulation of mercury were assessed. Serum antinuclear antibodies were used to evaluate the autoimmune response, and serum immunoglobulin E (IgE) to study effects on T-helper cells of type 2. Strains with a susceptible H-2 haplotype developed autoantibodies to the nucleolar protein fibrillarin (AFA) in a dose-dependent pattern within 2 weeks. The substantially lower whole-body and organ mercury level needed to induce AFA in the susceptible A.SW strain compared with the H-2 congenic B10.S strain demonstrates that genetic factors outside the H-2 region modify the autoimmune response. Mouse strains without the susceptible haplotype did not develop any autoimmune reaction irrespective of dose and organ deposition of mercury. In susceptible mouse strains, males and females had different thresholds for induction of autoimmune reactions. In susceptible strains, serum IgE increased dose dependently and reached a maximum after 1-2.5 weeks. A susceptible H-2 haplotype is therefore a prerequisite for the autoimmune response. Mercury exposure will modulate the response, qualitatively through the existence of dose-related thresholds for autoimmune response and quantitatively as increasing doses cause increasing autoimmune response. Further, gender and non-H-2 genes modulate both the induction and subsequent development of AFA. Induction of IgE seems not to be mechanistically linked to the AFA response.


Subject(s)
Autoimmune Diseases/chemically induced , Autoimmune Diseases/veterinary , Chromosomal Proteins, Non-Histone/biosynthesis , Environmental Pollutants/adverse effects , Environmental Pollutants/immunology , Immunoglobulin E/biosynthesis , Mercury/adverse effects , Mercury/immunology , Animals , Autoimmune Diseases/genetics , Chromosomal Proteins, Non-Histone/drug effects , Disease Models, Animal , Female , Genetic Predisposition to Disease , Haplotypes , Immunoglobulin E/analysis , Kinetics , Male , Mice , Mice, Inbred Strains , Sex Factors
12.
Arch Neurol ; 59(7): 1115-21, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12117359

ABSTRACT

BACKGROUND: Treatment with interferon beta reduces clinical exacerbations in multiple sclerosis (MS) through several immunomodulatory mechanisms that involve the augmentation of programmed cell death (apoptosis) of peripheral T lymphocytes. The expression of survivin, a cell cycle-regulated antiapoptosis protein, is up-regulated in mitogen-stimulated T lymphocytes from patients with MS, and this expression correlates with MS disease activity. OBJECTIVE: To evaluate the effect of interferon beta on the expression of survivin and other apoptosis regulatory molecules in peripheral T lymphocytes from patients with MS. PATIENTS AND METHODS: In a prospective, combined clinical and immunologic study, we evaluated the expression of survivin, Bcl-2 protein, and the death receptor Fas in mitogen-stimulated T lymphocytes from 26 patients with MS, before and serially after treatment with interferon beta-1a. We also investigated the long-term effects of interferon beta-1a on cellular expression of these proteins and T-lymphocyte apoptosis in a cross-sectional study of 19 patients with MS receiving long-term interferon beta-1a therapy. RESULTS: Treatment with interferon beta-1a reduced the expression of survivin in in vitro stimulated T lymphocytes. This reduced expression correlated with augmented T-cell susceptibility to apoptosis and with clinical response to treatment. In contrast, interferon beta-1a therapy did not significantly alter cellular expression of Bcl-2 protein or Fas. This down-regulatory effect of interferon beta-1a on cellular expression of survivin was maintained after long-term therapy. CONCLUSIONS: Our observations suggest that interferon beta exerts a regulatory effect on peripheral T lymphocytes through an antiapoptosis mechanism that involves the down-regulation of cellular survivin expression.


Subject(s)
Adjuvants, Immunologic/pharmacology , Apoptosis/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Interferon-beta/pharmacology , Microtubule-Associated Proteins , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Adjuvants, Immunologic/therapeutic use , Adult , Apoptosis Regulatory Proteins , Chromosomal Proteins, Non-Histone/drug effects , Down-Regulation/drug effects , Female , Humans , Inhibitor of Apoptosis Proteins , Interferon beta-1a , Interferon-beta/therapeutic use , Longitudinal Studies , Male , Neoplasm Proteins , Prospective Studies , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Survivin
13.
Clin Cancer Res ; 8(5): 1234-40, 2002 May.
Article in English | MEDLINE | ID: mdl-12006543

ABSTRACT

PURPOSE: Bexarotene is the first synthetic rexinoid approved for the treatment of all stages of cutaneous T-cell lymphoma (CTCL) however the mechanism of bexarotene action is unknown. We examined the effects of bexarotene on induction of apoptosis and expression of its cognate receptors in well-established CTCL cell lines (MJ, Hut78, and HH). EXPERIMENTAL DESIGN: CTCL cells were treated with 0.1, 1, and 10 microM bexarotene for 24, 48, 72, and 96 h. Apoptosis was determined by flow-cytometry analysis of sub-G(1) hypodiploid nuclei and annexin V binding populations. Apoptosis-associated proteins and retinoid receptors were detected by Western blots. RESULTS: Bexarotene treatment at 1 and 10 microM for 96 h increased the number of cells with sub-G1 populations and annexin V binding in a dose-dependent manner compared with vehicle controls (DMSO) in all three cell lines, respectively. Bexarotene treatment suppressed the expression of retinoid X receptor alpha and retinoic acid receptor alpha proteins in all three lines compared with untreated controls. Bexarotene treatment decreased the protein levels of survivin, activated caspase-3, and cleaved poly(ADP-Ribose) polymerase, but had no obvious effect on expression of Fas/Fas ligand and bcl-2 proteins in all three CTCL lines. CONCLUSIONS: Bexarotene treatment at clinically relevant concentrations causes apoptosis of CTCL cell lines in association with activation of caspase-3 and cleavage of poly(ADP-Ribose) polymerase, as well as down-regulation of retinoid X receptor alpha, retinoic acid receptor alpha, and survivin. These findings support apoptosis as a mechanism for bexarotene therapy in CTCL.


Subject(s)
Apoptosis/drug effects , Lymphoma, T-Cell, Cutaneous/drug therapy , Microtubule-Associated Proteins , Skin Neoplasms/drug therapy , Tetrahydronaphthalenes/pharmacology , Annexin A5/metabolism , Bexarotene , Blotting, Western , Cell Cycle/drug effects , Cell Survival/drug effects , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Inhibitor of Apoptosis Proteins , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , Neoplasm Proteins , Protein Binding/drug effects , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Survivin , Time Factors , Transcription Factors/drug effects , Transcription Factors/metabolism , Tumor Cells, Cultured
14.
Exp Cell Res ; 275(1): 44-53, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11925104

ABSTRACT

Survivin is a member of the inhibitor of apoptosis (IAP) family of negative regulators of programmed cell death that is frequently overexpressed in human tumors. Survivin is not only involved in the regulation of apoptosis, but is also known to play a role in the control of cell cycle progression at the G2/M phase. Survivin is a predominantly cytoplasmic protein expressed in a cell cycle-dependent manner, but the mechanism(s) that determine its nuclear-cytoplasmic localization have not been described. In this study, we report that Survivin is a nuclear shuttling protein that is actively exported from the nucleus via the CRM1-dependent pathway. Nuclear export of Survivin is independent of the export of other shuttling proteins that control the G2/M phase transition, such as cyclin B1 and cdc25. The carboxy-terminal domain of Survivin is both necessary and sufficient for its nuclear export, although this region does not contain a functional leucine-rich nuclear export signal. Differences in the amino acid sequence of this region determine the dramatically different localization of Survivin (in the cytoplasm) and its splicing variant Survivin-DeltaEx3 (in the nucleus). The carboxy-terminal end of Survivin-DeltaEx3 contains a bipartite nuclear localization signal, not present in Survivin, which mediates its strong nuclear accumulation. These data suggest that active transport between the nucleus and cytoplasm may constitute an important regulatory mechanism for Survivin function.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Cytoplasm/metabolism , Karyopherins/metabolism , Microtubule-Associated Proteins , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear , Alternative Splicing , Amino Acid Sequence , Biological Transport, Active , Cell Compartmentation , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/drug effects , Humans , Inhibitor of Apoptosis Proteins , Neoplasm Proteins , Protein Isoforms/chemistry , Protein Isoforms/drug effects , Protein Isoforms/metabolism , Protein Structure, Tertiary , Survivin , Exportin 1 Protein
15.
J Cell Biochem ; 83(2): 342-54, 2001.
Article in English | MEDLINE | ID: mdl-11573250

ABSTRACT

Survivin is a member of the inhibitor of apoptosis (IAP) gene family, containing a single baculovirus IAP repeat (BIR) and no RING finger, that is expressed in many human cancers. Although it has been proposed to be involved in mitotic and cytokinetic processes, its functional subcellular distribution in the cytoplasm and nucleus, and its binding to centrosomes, spindle fibers, and centromeres in relation to these processes, is not fully resolved. We have analyzed the localization of Survivin in normal (Detroit 551, IMR-90) and tumor-derived (HeLa, Saos-2) cell lines, and found that it does colocalize with centrosomes in the cytoplasm during interphase, then moves to centromeres during mitosis, and finally localizes to the midbody spindle fibers during telophase. However, Taxol, a popular microtubule stabilizing agent that is frequently used in the study of these processes, severely disrupted the localization of Survivin. Taxol treatment of cells promoted extensive relocalization of Survivin with alpha-tubulin on microtubules during either interphase or mitosis. Survivin antisense oligonucleotide markedly sensitized HeLa cells to cell death induced by agents acting at the level of cell surface receptor (Fas pathway) or at the level of mitochondria (etoposide). HeLa cell death induced by Survivin antisense oligonucleotide could be partially complemented by Deterin, the Drosophila homolog of Survivin (Jones et al. [2000] J. Biol. Chem. 275:22157-22166). Reciprocally, a chimera of the Deterin BIR domain and Survivin C-terminus could rescue Drosophila Kc cells from death induced by transfection of a human caspase-7-expressing plasmid. These results indicate common components of Survivin and Deterin antiapoptotic action in the vertebrate and invertebrate phyla.


Subject(s)
Apoptosis/physiology , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins , Kinetochores/metabolism , Microtubule-Associated Proteins , Mitosis/physiology , Animals , Apoptosis/drug effects , Cell Division , Cell Line , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/drug effects , Drosophila , HeLa Cells/drug effects , Humans , Inhibitor of Apoptosis Proteins , Insect Proteins/metabolism , Kinetochores/chemistry , Neoplasm Proteins , Oligonucleotides, Antisense/pharmacology , Paclitaxel/pharmacology , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Survivin , Tumor Cells, Cultured
16.
Blood ; 98(7): 2091-100, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567995

ABSTRACT

The inhibitor-of-apoptosis protein survivin is expressed in most cancers and leukemias and during fetal development, but not in most normal adult tissues. Survivin expression was analyzed in umbilical cord blood (UCB) and adult bone marrow CD34(+) cells and in the factor-dependent MO7e cell line; also investigated was whether survivin expression was regulated by hematopoietic growth factors. Survivin messsenger RNA (mRNA) and protein were expressed in fresh UCB and marrow CD34(+) cells. The combination of thrombopoietin, Flt3 ligand, and stem cell factor upregulated survivin expression in CD34(+) cells within 24 hours; survivin expression was cell-cycle related and highest during G2/M, whereas growth-factor withdrawal resulted in decreased survivin expression. Cell-cycle fractionation of UCB CD34(+) with Hoechst-33342/pyronin-Y demonstrated that survivin message was undetectable in freshly isolated G0 cells, but present in G1 cells. After cytokine stimulation, survivin mRNA and protein expression were observed in both G0 and G1 CD34(+) cells as well as in cells that had progressed to S and G2/M phase, indicating that survivin expression is regulated in all phases of the cell cycle. This contrasts with the expression of survivin predominantly during G2/M in cancer cells. In CD34(+) cells and MO7e cells, growth factor-mediated upregulation of survivin was associated with inhibition of apoptosis, and downregulation of survivin was coincident with increased apoptosis. Furthermore, an inverse correlation between survivin and active caspase-3 was observed in CD34(+) cells. These findings demonstrate that survivin is not a cancer-specific antiapoptotic protein and plays a regulatory role in normal adult hematopoiesis.


Subject(s)
Antigens, CD34 , Bone Marrow Cells/metabolism , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/physiology , Cysteine Proteinase Inhibitors/physiology , Fetal Blood/metabolism , Hematopoietic Cell Growth Factors/pharmacology , Microtubule-Associated Proteins , Apoptosis/drug effects , Bone Marrow Cells/cytology , Cell Cycle , Chromosomal Proteins, Non-Histone/metabolism , Cysteine Proteinase Inhibitors/metabolism , Fetal Blood/cytology , Flow Cytometry , Gene Expression Regulation , Hematopoiesis/drug effects , Hematopoietic Cell Growth Factors/physiology , Humans , Inhibitor of Apoptosis Proteins , Neoplasm Proteins , Survivin , T-Lymphocytes/metabolism , Tumor Cells, Cultured
17.
Mol Biol Cell ; 11(8): 2705-17, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10930464

ABSTRACT

This report examines the distribution of an RNA polymerase I transcription factor (upstream binding factor; UBF), pre-rRNA processing factors (nucleolin and fibrillarin), and pre-rRNAs throughout mitosis and postmitotic nucleologenesis in HeLa cells. The results demonstrate that nucleolin, fibrillarin, and pre-rRNAs synthesized at G2/M phase of the previous cell cycle are directly recruited to UBF-associated nucleolar organizer regions (NORs) early in telophase before chromosome decondensation. Unlike the fusion of prenucleolar bodies to the nucleoli, this early recruitment of processing factors and pre-rRNAs is independent of RNA polymerase I transcription. In the absence of polymerase I transcription, the initial localization of nucleolin, fibrillarin, and pre-rRNAs to UBF-associated NORs generates segregated mininucleoli that are similar to the larger ones observed in interphase cells grown under the same conditions. Pre-rRNAs are juxtaposed to UBF-nucleolin-fibrillarin caps that may represent the segregated nucleoli observed by electron microscopy. These findings lead to a revised model of nucleologenesis. We propose that nucleolar formation at the end of mitosis results from direct recruitment of processing factors and pre-rRNAs to UBF-associated NORs before or at the onset of rDNA transcription. This is followed by fusion of prepackaged prenucleolar bodies into the nucleolus. Pre-ribosomal ribonucleoproteins synthesized in the previous cell cycle may contribute to postmitotic nucleologenesis.


Subject(s)
Cell Nucleolus/physiology , Nucleolus Organizer Region/metabolism , Pol1 Transcription Initiation Complex Proteins , RNA Polymerase I/metabolism , Transcription, Genetic/physiology , Cell Cycle/drug effects , Cell Nucleolus/drug effects , Cell Nucleolus/ultrastructure , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Dactinomycin/pharmacology , HeLa Cells , Humans , Models, Biological , Nucleic Acid Synthesis Inhibitors/pharmacology , Nucleolus Organizer Region/drug effects , Phosphoproteins/drug effects , Phosphoproteins/metabolism , RNA Polymerase I/drug effects , RNA Precursors/biosynthesis , RNA Precursors/drug effects , RNA Precursors/metabolism , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , Ribonucleoproteins/drug effects , Ribonucleoproteins/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Nucleolin
18.
Environ Res ; 77(2): 141-8, 1998 May.
Article in English | MEDLINE | ID: mdl-9600807

ABSTRACT

Mercury induces autoantibodies to the nucleolar protein fibrillarin (ANoA) in genetically susceptible (H-2AS) mouse strains. This study examines the importance of mercury toxicokinetics for the induction and strength (titer) of these autoantibodies. Female mice of the inbred strains A.SW and B10.S (H-2AS on the A and C57BL/10 genetic background, respectively) and A.TL and B10.TL (H-2Ak on the A and C57BL/10 background) were treated with 203HgCl2 in a dose of 1, 5, or 16 mg Hg/L drinking water for 56-70 days. Whole-body retention of 203Hg was monitored throughout the experimental period. Mercury accumulation in kidney, liver, heart, spleen, and brain was determined at end of the experiment when blood samples were also obtained for determination of ANoA. The drinking water consumption showed a limited variation between the strains and the dose groups. Therefore, intake of mercury did not vary much between the strains at a given dose level. The whole-body retention of mercury reached steady state after 4-5 weeks. In general, the B10.S and B10.TL strains showed a lower whole-body retention and deposition of mercury in the kidney and the liver, compared with the A.SW and A.TL strains given the same dose of mercury. The B10.S strain showed a threshold for induction of ANoA at 5 mg Hg/L, whereas ANoA were still seen in A.SW mice given 1 mg Hg/L. Taken together, this is compatible with a less efficient elimination of mercury in the A.SW and A.TL strains, which was also supported by the higher ratio between whole-body retention and intake of mercury in these strains. These findings indicate that genes residing outside the H-2 (MHC) complex play an important role for regulating mercury toxicokinetics, the A genes conferring higher accumulation of mercury in the body than the B10 genes. In mice congenic with regard to the susceptible H-2AS haplotype, a highly significant correlation (P<0.01) existed between on the one hand the whole-body retention and organ accumulation of mercury and on the other hand the titer of ANoA. We conclude that mercury toxicokinetics differs significantly among inbred mouse strains. The differences in toxicokinetics are regulated by non-H-2 genes and correlate with the autoimmune response in the genetically susceptible strains: quantitatively as the titer of the ANoA and qualitatively as different threshold doses for induction of ANoA by mercury.


Subject(s)
Autoimmunity/drug effects , Disinfectants/pharmacokinetics , Mercury/pharmacokinetics , Animals , Autoantibodies/drug effects , Autoantibodies/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/immunology , Disease Susceptibility/immunology , Disease Susceptibility/metabolism , Disinfectants/administration & dosage , Disinfectants/toxicity , Dose-Response Relationship, Drug , Drinking , Female , Gene Expression/drug effects , Gene Expression/genetics , Genetic Predisposition to Disease , H-2 Antigens/drug effects , H-2 Antigens/genetics , Mercuric Chloride/administration & dosage , Mercuric Chloride/pharmacokinetics , Mercuric Chloride/toxicity , Mercury/administration & dosage , Mercury/toxicity , Mercury Radioisotopes , Mice , Mice, Inbred A , Mice, Inbred C57BL , Radiation Monitoring , Ribonucleoproteins/drug effects , Ribonucleoproteins/immunology , Solutions/administration & dosage , Solutions/chemistry , Solutions/pharmacokinetics , Species Specificity , Tissue Distribution
19.
Chromosoma ; 105(7-8): 506-14, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9211978

ABSTRACT

Retention of some components within the nucleolus correlates with the presence of rRNA precursors found early in the rRNA processing pathway. Specifically, after most 40S, 38S and 36S pre-rRNAs have been depleted by incubation of Xenopus kidney cells in 0.05 microg/ml actinomycin D for 4 h, only 69% U3 small nucleolar RNA (snoRNA), 68% U14 snoRNA and 72% fibrillarin are retained in the nucleolus as compared with control cells. These nucleolar components are important for processing steps in the pathway that gives rise to 18S rRNA. In contrast, U8 snoRNA, which is used for 5.8S and 28S rRNA production, is fully retained in the nucleolus after actinomycin D treatment. Therefore, U8 snoRNA is in a different category than U3 and U14 snoRNA and fibrillarin. It is proposed that U3 and U14 snoRNA and fibrillarin, but not U8 snoRNA, bind to the external transcribed spacer or internal transcribed spacer 1, and when these binding sites are lost after actinomycin D treatment some of these components cannot be retained in the nucleolus. Other binding sites may also exist, which would explain why only some and not all of these components are lost from the nucleolus.


Subject(s)
Cell Nucleolus/genetics , Dactinomycin/pharmacology , RNA Precursors/drug effects , RNA, Ribosomal/drug effects , RNA, Small Nuclear/metabolism , Animals , Base Sequence , Cell Nucleolus/drug effects , Chromosomal Proteins, Non-Histone/drug effects , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Molecular Sequence Data , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA Precursors/metabolism , RNA, Ribosomal/metabolism , RNA, Small Nuclear/drug effects , Xenopus laevis
20.
J Immunol ; 158(7): 3521-8, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9120314

ABSTRACT

The heavy metal mercury elicits a genetically restricted, anti-nucleolar autoantibody response that targets fibrillarin, a 34-kDa protein component of many small nucleolar ribonucleoprotein particles. The mechanisms by which a toxin such as mercury elicits an autoantibody response that predominantly targets a single intracellular protein autoantigen remain uncertain, but may be prefaced by mercury gaining access to the intracellular environment. Mercury-induced cell death was associated with loss of fibrillarin antigenicity and modification of the molecular properties of fibrillarin as revealed by aberrant migration under nonreducing conditions in SDS-PAGE. Addition of mercury to isolated nuclei also resulted in aberrant migration of fibrillarin, but not other nuclear autoantigens. The sensitivity of the HgCl2-induced modification of fibrillarin to 2-ME, iodoacetamide, and hydrogen peroxide suggested interaction of mercury with the two cysteines in the fibrillarin sequence. This was confirmed by mutation of the cysteines to alanines, which abolished the aberrant migration of fibrillarin in the presence of HgCl2. The modification of the molecular structure of fibrillarin by mercury reduced immunoprecipitation by anti-fibrillarin autoantibodies, pointing to unmodified fibrillarin as the B cell Ag and implicating mercury-modified fibrillarin as the source of T cell antigenicity. These observations demonstrate for the first time that an environmental toxin can alter the physicochemical properties of an autoantigen and may help to explain the antigenic specificity of mercury-induced murine autoimmunity.


Subject(s)
Autoantigens/drug effects , Autoantigens/immunology , Chromosomal Proteins, Non-Histone/immunology , Chromosomal Proteins, Non-Histone/pharmacology , Mercuric Chloride/immunology , Mercuric Chloride/pharmacology , Xenobiotics/immunology , Xenobiotics/pharmacology , Antibodies, Monoclonal/chemistry , Autoantibodies/metabolism , Binding Sites, Antibody , Cell Death/drug effects , Cell Death/immunology , Cell Nucleus/drug effects , Cell Nucleus/immunology , Chromosomal Proteins, Non-Histone/drug effects , Cysteine/physiology , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Epitopes/chemistry , Humans , Subcellular Fractions/drug effects , Subcellular Fractions/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...