Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Oncogene ; 32(4): 479-90, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-22391565

ABSTRACT

Inadvertent mammalian tissue exposures to low doses of ionizing radiation (IR) after radiation accidents, remediation of radioactive-contaminated areas, space travel or a dirty bomb represent an interesting trauma to an organism. Possible low-dose IR-induced bystander effects could impact our evaluation of human health effects, as cells within tissue are not equally damaged after doses of IR ≤10 cGy. To understand tissue responses after low IR doses, we generated a reporter system using the human clusterin promoter fused to firefly luciferase (hCLUp-Luc). Secretory clusterin (sCLU), an extracellular molecular chaperone, induced by low doses of cytotoxic agents, clears cell debris. Low-dose IR (≥2 cGy) exposure induced hCLUp-Luc activity with peak levels at 96 h, consistent with endogenous sCLU levels. As doses increased (≥1 Gy), sCLU induction amplitudes increased and time-to-peak response decreased. sCLU expression was stimulated by insulin-like growth factor-1, but suppressed by p53. Responses in transgenic hCLUp-Luc reporter mice after low IR doses showed that specific tissues (that is, colon, spleen, mammary, thymus and bone marrow) of female mice induced hCLUp-Luc activity more than male mice after whole body (≥10 cGy) irradiation. Tissue-specific, non-linear dose- and time-responses of hCLUp-Luc and endogenous sCLU levels were noted. Colon maintained homeostatic balance after 10 cGy. Bone marrow responded with delayed, but prolonged and elevated expression. Intraperitoneal administration of α-transforming growth factor (TGF)ß1 (1D11), but not control (13C4) antibodies, immediately following IR exposure abrogated CLU induction responses. Induction in vivo also correlated with Smad signaling by activated TGFß1 after IR. Mechanistically, media with elevated sCLU levels suppressed signaling, blocked apoptosis and increased survival of TGFß1-exposed tumor or normal cells. Thus, sCLU is a pro-survival bystander factor that abrogates TGFß1 signaling and most likely promotes wound healing.


Subject(s)
Clusterin/genetics , Gamma Rays , Insulin-Like Growth Factor I/genetics , Transforming Growth Factor beta1/genetics , Tumor Suppressor Protein p53/genetics , Whole-Body Irradiation , Animals , Apoptosis/genetics , Bone Marrow/metabolism , Bone Marrow/radiation effects , Cell Line, Tumor , Clusterin/metabolism , Colon/metabolism , Colon/radiation effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Female , HCT116 Cells , Humans , Insulin-Like Growth Factor I/metabolism , MCF-7 Cells , Male , Mice , Mice, Transgenic , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Promoter Regions, Genetic , Signal Transduction/radiation effects , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Biochem Soc Trans ; 35(Pt 4): 797-801, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17635151

ABSTRACT

We found that Ku70, a known DNA repair factor, has a novel function to bind and inhibit Bax (Bcl-2-associated X protein), a key mediator of apoptosis. Pentapeptides derived from the Bax-binding domain of Ku70 were cell-permeable and protected cells from Bax-mediated apoptosis. These pentapeptides were called BIPs (Bax-inhibiting peptides). BIPs may become a useful therapeutic tool to reduce cellular damage. We also generated BIP mutant pentapeptides that do not inhibit Bax, but retain their cell-penetrating activity. Since both BIPs and BIP mutants are cell-permeable, these peptides were designated CPP5s (cell-penetrating pentapeptides). Among the CPP5s discovered, VPTLK (BIP) and KLPVM (BIP mutant) were confirmed to possess protein transduction activity by examination of the delivery of GFP (green fluorescent protein) into cells by these peptides. The mechanism of cell penetration by CPP5s is not known. CPP5s enter the cell at 0 and 4 degrees C. In preliminary studies, various inhibitors of endocytosis and pinocytosis did not show any significant suppression of CPP5 cell entry. CPP5s have very low toxicity in vitro and in vivo and so may be useful tools in order to develop non-toxic drug-delivery technologies.


Subject(s)
Antigens, Nuclear/physiology , DNA-Binding Proteins/physiology , Oligopeptides/physiology , Protein Sorting Signals/physiology , Protein Transport/physiology , bcl-2-Associated X Protein/antagonists & inhibitors , Animals , Drug Delivery Systems , Humans , Ku Autoantigen
3.
Anticancer Res ; 26(4A): 2707-16, 2006.
Article in English | MEDLINE | ID: mdl-16886681

ABSTRACT

BACKGROUND: The glycoprotein clusterin (CLU) has two known isoforms generated in human cells. A nuclear form of CLU protein (nCLU) is pro-apoptotic, while a secretory form (sCLU) is pro-survival. CLU expression has been associated with tumorigenesis and the progression of various malignancies. MATERIALS AND METHODS: The expression of CLU was studied immunohistochemically in paraffin sections of primary cutaneous malignant melanomas, metastases of malignant melanomas and acquired melanocytic naevi. Using PCR and Western blotting, the expression of CLU was also investigated in various vitamin D-responsive (MeWo, SK-MEL-28) and vitamin D-resistant melanoma cell lines (SK-MEL-5, SK-MEL-25), as well as in normal human melanocytes (NHM), along with 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] treatment. RESULTS: In contrast to acquired melanocytic naevi, CLU immunoreactivity was found in primary cutaneous malignant melanomas and metastases of malignant melanomas in situ. Both CLU protein and RNA were detected in melanoma cell lines and NHM. Treatment with 1,25(OH)2D3 modulated CLU's expression in vitamin D-responsive but not in -resistant melanoma cell lines. CONCLUSION: CLU may be of importance for the progression of malignant melanoma. The growth regulatory effects of 1,25(OH)2D3 in melanoma cell lines may, at least in part, be mediated via modulation of CLU expression.


Subject(s)
Calcitriol/pharmacology , Clusterin/biosynthesis , Melanoma/drug therapy , Melanoma/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Male , Melanoma/pathology , Melanoma/secondary , Middle Aged , Skin Neoplasms/secondary
4.
Cell Death Differ ; 13(1): 12-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16179938

ABSTRACT

Clusterin (CLU) has been implicated in various cell functions involved in carcinogenesis and tumour progression. There are two known CLU protein isoforms generated in human cells. A nuclear form of CLU protein (nCLU) is proapoptotic, and a secretory form (sCLU) is prosurvival. CLU expression has been associated with tumorigenesis of various malignancies, including tumours of prostate, colon, and breast. Furthermore, CLU expression is modulated by many factors that are believed to regulate tumour growth and/or apoptosis, including 1,25-dihydroxyvitamin D3, transforming growth factor beta-1, ultraviolet radiation, and IR. sCLU upregulation appears to be a general molecular stress response. Presently, preliminary results indicate that therapeutic modalities targeting CLU may be effective in cancer treatment. However, such strategies should make sure that nCLU is not eliminated or reduced. This review summarizes our present understanding of the importance of CLU in various physiological functions including tumour growth, and discusses its relevance to future cancer therapy.


Subject(s)
Clusterin/physiology , Neoplasms/etiology , Apoptosis/physiology , Biomarkers, Tumor/genetics , Cell Cycle/physiology , Clusterin/genetics , DNA Repair , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasms/pathology , Neoplasms/physiopathology , Neoplasms/therapy , Prognosis , Protein Isoforms/genetics , Protein Isoforms/physiology
5.
Semin Radiat Oncol ; 11(4): 352-72, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11677660

ABSTRACT

Cellular responses to ionizing radiation (IR) include (a) activation of signal transduction enzymes; (b) stimulation of DNA repair, most notably DNA double strand break (DSB) repair by homologous or nonhomologous recombinatorial pathways; (c) activation of transcription factors and subsequent IR-inducible transcript and protein changes; (d) cell cycle checkpoint delays in G(1), S, and G(2) required for repair or for programmed cell death of severely damaged cells; (e) activation of zymogens needed for programmed cell death (although IR is a poor inducer of such responses in epithelial cells); and (f) stimulation of IR-inducible proteins that may mediate bystander effects influencing signal transduction, DNA repair, angiogenesis, the immune response, late responses to IR, and possibly adaptive survival responses. The overall response to IR depends on the cell's inherent genetic background, as well as its ability to biochemically and genetically respond to IR-induced damage. To improve the anti-tumor efficacy of IR, our knowledge of these pleiotropic responses must improve. The most important process for the survival of a tumor cell following IR is the repair of DNA double strand breaks (DSBs). Using yeast two-hybrid analyses along with other molecular and cellular biology techniques, we cloned transcripts/proteins that are involved in, or presumably affect, nonhomologous DNA double strand break end-joining (NHEJ) repair mediated by the DNA-PK complex. Using Ku70 as bait, we isolated a number of Ku-binding proteins (KUBs). We identified the first X-ray-inducible transcript/protein (xip8, Clusterin (CLU)) that associates with DNA-PK. A nuclear form of CLU (nCLU) prevented DNA-PK-mediated end joining, and stimulated cell death in response to IR or when overexpressed in the absence of IR. Structure-function analyses using molecular and cellular (including green fluorescence-tagged protein trafficking) biology techniques showed that nCLU appears to be an inactive protein residing in the cytoplasm of epithelial cells. Following IR injury, nCLU levels increase and an as yet undefined posttranslational modification appears to alter the protein, exposing nuclear localization sequences (NLSs) and coiled-coil domains. The modified protein translocates to the nucleus and triggers cell death, presumably through its interaction specifically with Ku70. Understanding nCLU responses, as well as the functions of the KUBs, will be important for understanding DSB repair. Knowledge of DSB repair may be used to improve the antitumor efficacy of IR, as well as other chemotherapeutic agents.


Subject(s)
DNA Repair/physiology , DNA, Neoplasm/metabolism , Neoplasms/radiotherapy , Cell Survival , DNA Damage , DNA Repair/genetics , DNA, Neoplasm/genetics , Humans , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Radiation, Ionizing , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 97(11): 5907-12, 2000 May 23.
Article in English | MEDLINE | ID: mdl-10823943

ABSTRACT

Clusterin [CLU, a.k.a. TRPM-2, SGP-2, or ionizing radiation (IR)-induced protein-8 (XIP8)] was implicated in apoptosis, tissue injury, and aging. Its function remains elusive. We reisolated CLU/XIP8 by yeast two-hybrid analyses using as bait the DNA double-strand break repair protein Ku70. We show that a delayed (2-3 days), low-dose (0.02-10 Gy) IR-inducible nuclear CLU/XIP8 protein coimmunoprecipitated and colocalized (by confocal microscopy) in vivo with Ku70/Ku80, a DNA damage sensor and key double-strand break repair protein, in human MCF-7:WS8 breast cancer cells. Overexpression of nuclear CLU/XIP8 or its minimal Ku70 binding domain (120 aa of CLU/XIP8 C terminus) in nonirradiated MCF-7:WS8 cells dramatically reduced cell growth and colony-forming ability concomitant with increased G(1) cell cycle checkpoint arrest and increased cell death. Enhanced expression and accumulation of nuclear CLU/XIP8-Ku70/Ku80 complexes appears to be an important cell death signal after IR exposure.


Subject(s)
Antigens, Nuclear , Cell Death/physiology , DNA Helicases , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Glycoproteins/physiology , Molecular Chaperones , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Adenocarcinoma/pathology , Amino Acid Motifs , Breast Neoplasms/pathology , Cell Nucleus/metabolism , Clusterin , DNA Damage , DNA, Complementary/genetics , DNA, Neoplasm/radiation effects , DNA-Activated Protein Kinase , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/radiation effects , Gene Library , Genes, Reporter , Glycoproteins/chemistry , Glycoproteins/genetics , Green Fluorescent Proteins , Humans , Ku Autoantigen , Luminescent Proteins/genetics , Microscopy, Confocal , Protein Binding , Protein Precursors/metabolism , Protein Serine-Threonine Kinases/physiology , Saccharomyces cerevisiae/genetics , Tumor Cells, Cultured , Two-Hybrid System Techniques
7.
Nucleic Acids Res ; 27(10): 2165-74, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10219089

ABSTRACT

DNA-dependent protein kinase (DNA-PK) plays a critical role in resealing DNA double-stand breaks by non-homologous end joining. Aside from DNA-PK, XRCC4 and DNA ligase IV, other proteins which play a role(s) in this repair pathway remain unknown; DNA-PK contains a catalytic subunit (DNA-PKcs) and a DNA binding subunit (Ku70 and Ku80). We isolated Ku70-binding proteins (KUB1-KUB4) using yeast two-hybrid analyses. Sequence analyses revealed KUB1 to be apolipoprotein J (apoJ), also known as X-ray-inducible transcript 8 (XIP8), testosterone-repressed prostate message-2 (TRPM-2) and clusterin. KUB2 is Ku80. KUB3 and KUB4 are unknown, >10 kb trans-cripts. Interactions of apoJ/XIP8 or KUB3 with Ku70 were confirmed by co-immunoprecipitation analyses in MCF-7:WS8 breast cancer or IMR-90 normal lung fibroblast cells, respectively. The interaction of apoJ/XIP8 with Ku70 was confirmed by far-western analyses. Stable over-expression of full-length apoJ/XIP8 in MCF-7:WS8 caused decreased Ku70/Ku80 DNA end binding that was restored by apoJ/XIP8 monoclonal antibodies. The role of apoJ/XIP8 in ionizing radiation resistance/sensitivity is under investigation.


Subject(s)
Antigens, Nuclear , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , DNA Helicases , DNA-Binding Proteins/metabolism , Molecular Chaperones , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Animals , Base Sequence , Carrier Proteins/genetics , Cell Line , Cloning, Molecular , Clusterin , DNA Probes/genetics , DNA Repair , DNA-Activated Protein Kinase , DNA-Binding Proteins/genetics , Glycoproteins/metabolism , Humans , Ku Autoantigen , Mice , Nuclear Proteins/genetics , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Tumor Cells, Cultured
8.
Virology ; 216(2): 347-56, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8607264

ABSTRACT

Four different transformed cell lines were isolated as a result of independent infection of primary hamster fibroblasts by Rous sarcoma virus (RSV SR-D stocks). These lines differ by the level of their spontaneous metastatic activity: HET-SR-1, HET-SR-8, and HET-SR-10 cell lines induced 70-200 metastatic nodules in the lung and/or lymph nodes of inoculated animals (high metastatic lines, HM). Metastatic activity was not identified after injection of HET-SR cells (low metastatic line, LM). All cell lines contained one copy of integrated and expressed intact RSV provirus. The difference in the amount of v-src protein in cell lines was not correlated with their metastatic potential in vivo. Complete v-srcHM and v-srcLM genes were cloned from corresponding gene libraries and sequenced. In the unique region of both v-src isoforms a GC-rich insert of 60 nucleotides (20 a.a.) was found. The presence of this insert explains the unusual apparent molecular weight of protein encoded by v-srcHM and vsrcLM: 62 kDA. Both genes had 10 identical amino acid changes when compared to the known RSV SR-D v-src sequence. v-srcHM and v-srcLM differ by several amino acid changes. Most of them are localized in the unique domain and the extreme carboxy-terminal region of the of the oncoprotein. Both v-src variants and chimeric v-src with mutually substituted parts were subcloned in a retroviral vector and introduced into avian neuroretina cells. Significant differences in the morphology of transformed neuroretinal cells were associated with the mutations in the carboxy-terminal region of the v-src oncogene. Low metastatic HET-SR cells transfected with v-srcHM and the chimeric gene v-src-LH remarkably increased their metastatic potential. In contrast, this effect was not observed when the same cells were transfected with v-srcLM and the chimeric v-srcHL gene. Specific changes in the distribution of fibronectin matrix typical for high metastatic cells were found in the lines transfected with v-srcHM.


Subject(s)
Avian Sarcoma Viruses/genetics , Cell Transformation, Neoplastic , Cell Transformation, Viral , Genes, src , Mutation , Neoplasm Metastasis/genetics , Neoplasms, Experimental/virology , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Transformed , Chickens , Cloning, Molecular , Cricetinae , DNA, Viral/genetics , Fibronectins/metabolism , Genes, Viral , Mesocricetus , Molecular Sequence Data , Neoplasms, Experimental/genetics , Plasmids , Recombinant Fusion Proteins/genetics , Retina/pathology
9.
Int J Oncol ; 7(3): 453-9, 1995 Sep.
Article in English | MEDLINE | ID: mdl-21552859

ABSTRACT

Previously, it was shown that hamster cells transformed by Rous Sarcoma Virus (RSV) exhibited a decreased expression of the RSV products (including the pp60 src oncogene) when these cells were supertransfected with the N-ras oncogene. To assess the responsibility of the activated N-ras in the modulation of the RSV viral products, a strategy based on two ras antagonists was used; i.e. i) a rap1A/K-rev1 expression vector known for its capacity to revert the K-ras induced transformed phenotype and ii) a plasmid containing antisense N-ras sequence. We present data showing only the plasmid construct containing the N-ras antisense sequense could inhibit expression or N-ras and, at the same time, restore the expression of v-src, up to a level comparable to that of the parental cells. Our results support the idea that some biological switches, triggered and activated through the N-ras oncogene pathway, might modulate the promoter activity of the RSV LTR.

SELECTION OF CITATIONS
SEARCH DETAIL