Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Oncogene ; 25(38): 5315-25, 2006 Aug 28.
Article in English | MEDLINE | ID: mdl-16936753

ABSTRACT

Despite a number of attempts to improve treatment of ovarian cancer, it remains the most common cause of death from gynecological cancers. Thus, it is very important to identify more effective drugs for treatment and prevention of ovarian cancer. All-trans-retinoic acid (ATRA) has been shown to arrest the growth of ovarian carcinoma cells in G0/G1 and to significantly elevate levels of Rb2/p130 protein, a member of the retinoblastoma family of tumor suppressors. As ATRA treatment leads to a significant increase in the amount of Rb2/p130 protein but not mRNA, the elevated levels of Rb2/p130 protein is likely the result of increased stability. In studies to elucidate the mechanism by which ATRA alters Rb2/p130 stability in ovarian cancer cells, it was determined that PP2A, a serine/threonine phosphatase, binds and dephosphorylates Rb2/p130. Dephosphorylated Rb2/p130 exhibits decreased ubiquitination and thus is not degraded by the proteasome. The sites at which PP2A catalytic subunit (PP2Ac) interacts with Rb2/p130 have been localized to the NLS in the C-terminus of Rb2/p130. These sites are also involved in the interaction of Rb/p130 with importin beta and importin alpha, members of the nuclear transport machinery. It is known that importin alpha recognizes a NLS on a target protein and importin beta binds the nuclear pore complex. Moreover, it has been shown that the binding of importin alpha to NLS significantly decreases with phosphorylation of NLS. In ATRA-treated ovarian carcinoma cells, PP2A binds to Rb2/p130 and dephosphorylates the NLS of Rb2/p130 leading to the interaction of importin alpha with Rb2/p130. Importin beta then binds to the importin alpha-Rb2/p130 complex, leading to the translocation of the Rb2/p130 to the nucleus where it acts to arrest ovarian cancer cells in G1 and suppress proliferation.


Subject(s)
Cell Division/drug effects , Ovarian Neoplasms/pathology , Phosphoprotein Phosphatases/physiology , Retinoblastoma-Like Protein p130/physiology , Tretinoin/therapeutic use , Antineoplastic Agents/pharmacology , Cell Nucleus/enzymology , Female , Humans , Ovarian Neoplasms/drug therapy , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2 , Protein Transport , Retinoids/pharmacology
2.
J Exp Clin Cancer Res ; 23(2): 309-16, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15354417

ABSTRACT

Vitamin A is an essential nutrient important for growth, vision, embryonic development, immune response and reproduction. Various retinoids have been shown to be effective chemotherapeutic and chemopreventive agents for a number of human cancers. Telomeres are nucleoprotein structures found at the end of chromosomes. During cellular division, the telomeres in normal cells shorten progressively and thus, function as a "molecular clock". Telomerase is a ribonucleoprotein complex that extends and maintains telomeres. Activation of telomerase is required for cells to overcome proliferative crisis. Telomerase activation is observed in 90% of human cancers, but not in normal somatic cells. We examined the role of telomerase in mediating the growth suppression of ovarian carcinoma cells by all-trans-retinoic acid (ATRA). Using a number of cell lines with varying levels of growth sensitivity to ATRA, we found that cells that exhibit ATRA-dependant suppression of growth also contained significantly reduced telomerase activity. We also observed a reduction in expression of the telomerase components, hTERT and hTR in ATRA treated ovarian carcinoma cells. Our results suggest that one mechanism by which ATRA acid inhibits cancer cell growth is by suppressing telomerase activity, thereby pushing cells to proliferative crisis.


Subject(s)
Antineoplastic Agents/pharmacology , Down-Regulation/drug effects , Ovarian Neoplasms/enzymology , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Tretinoin/pharmacology , Catalytic Domain , DNA-Binding Proteins , Female , Humans , Ovarian Neoplasms/pathology , RNA , RNA, Long Noncoding , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA, Untranslated/antagonists & inhibitors , RNA, Untranslated/genetics , Telomerase/genetics , Tumor Cells, Cultured
3.
Oncogene ; 20(55): 7935-44, 2001 Nov 29.
Article in English | MEDLINE | ID: mdl-11753676

ABSTRACT

We have examined the effect of all-trans-retinoic acid (RA) on cell cycle gene expression in RA sensitive CA-OV3 and RA resistant SK-OV3 ovarian carcinoma cell lines. Gene expression was analysed by multiprobe RNAse protection, Western blotting and in vitro kinase assays. No differences were observed between RA sensitive and RA resistant ovarian carcinoma cells in the levels of expression of many cell cycle genes including cyclin A, B and E, cdk 2,4 and 6, E2F-1, E2F-2, E2F-3, E2F-4, E2F-5, DP-1 and DP-2. However, RA sensitive CA-OV3 cells expressed higher levels of p53, p27, p21, and p16 compared to RA resistant SK-OV3 cells. In addition, RA treatment of CA-OV3 cells resulted in a significant decrease in hyperphosphorylated RB and RB-2/p130 and corresponding significant increases in the levels of hypophosphorylated and/or partially phosphorylated RB-2/p130 protein and hypophosphorylated RB. Also, RA treatment increased expression of the cdk inhibitor p27 and decreased activity of cdk 2, cdk 4 and cdk 6. Finally, amounts of p27-cyclin E and RB-2/p130-E2F4 complexes were found to increase in CA-OV3 cells growth arrested by RA. These results suggest that the pocket protein pathways are critical targets for retinoid suppression of ovarian carcinoma cell growth.


Subject(s)
Genes, cdc/physiology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Tretinoin/pharmacology , Blotting, Western , Cell Division/drug effects , Cell Division/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Retinoblastoma/genetics , Humans , Macromolecular Substances , Nuclease Protection Assays , Phosphorylation/drug effects , Protein Binding/drug effects , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured
4.
Toxicol Appl Pharmacol ; 174(2): 153-9, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11446831

ABSTRACT

The synthetic retinoid AGN 193109 is a potent pan retinoic acid receptor (RAR) antagonist. Treatment of pregnant mice with a single oral 1 mg/kg dose of this antagonist on day 8 postcoitum results in severe craniofacial (median cleft face or frontonasal deficiency) and eye malformations in virtually all exposed fetuses. Using differential display analysis, we have determined that CYP1A1 mRNA levels are elevated in mouse embryos 6 h following treatment with AGN 193109. Similarly, an elevation in CYP1A1 mRNA levels, protein levels, and aryl hydrocarbon hydoxylase activity occurs in Hepa-1c1c7 cells, with the maximal elevation observed when the cells were treated with 10(-5) M AGN 193109 for 4 to 8 h. Elevation in CYP1A1 mRNA levels in mouse embryos and Hepa-1c1c7 cells does not occur upon treatment with the natural retinoid, all-trans-retinoic acid. Finally, elevation in CYP1A1 mRNA levels was not observed when mutant Hepa-1c1c7 cells, which are defective in either the aryl hydrocarbon receptor (AhR) or aryl hydrocarbon receptor nuclear translocator (ARNT), were treated with AGN 193109. This suggests that the AhR/ARNT pathway and not the RAR/RXR pathway is mediating the elevation of CYP1A1 mRNA levels by AGN 193109, at least in the Hepa-1c1c7 cells. This is the first example of a retinoid that displays the abililty to regulate both the RAR/RXR and AhR/ARNT transcriptional regulatory pathways.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , DNA-Binding Proteins , Embryo, Mammalian/drug effects , Naphthalenes/pharmacology , Receptors, Retinoic Acid/antagonists & inhibitors , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Cells, Cultured , Cytochrome P-450 CYP1A1/drug effects , Cytochrome P-450 CYP1A1/genetics , Embryo, Mammalian/enzymology , Female , Male , Mice , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology
5.
J Cell Biochem ; 81(3): 393-400, 2001.
Article in English | MEDLINE | ID: mdl-11255222

ABSTRACT

Experimental evidence implicates oxidative free radical reactions as central in the processes of neurodegenerative diseases. In particular, cellular interactions with the beta-amyloid protein have been linked to neuron cell death in Alzheimer's disease. Also, uncharacterized dimeric purine moieties have been detected in oxidized DNAs. It has been suggested that inadequate excision-repair of such products plays a functional role in the neurological degeneration observed in familial Alzheimer's disease, Down's syndrome, and xeroderma pigmentosum. Therefore, in order to obtain a reagent to monitor the presence of such products, the purine dimer 8-8-(2'-deoxyguanosyl)-2'-deoxyguanosine-5'-monophosphate was used as a hapten for elicitation of rabbit anti-purine dimer antiserum. This antiserum specifically recognizes various purified 8-8-bideoxyribonucleosides and 8-8-bideoxyribonucleotides. We found that DNA oxidized by the Fenton reaction is specifically recognized by this antiserum. This reagent can therefore be used to demonstrate formation and excision of DNA purine dimers. Moreover, incubation of cultured rat pheochromocytoma PC-12 cells with the beta-amyloid protein resulted in formation of these purine dimers in cellular DNA. These dimers were subsequently removed from cellular DNA. From these results we conclude that the free radicals generated by A beta cause oxidative DNA alterations including purine dimers. Deficient repair of this type of DNA damage might result in neural cell loss via apoptosis. Our findings suggest mechanisms for the roles of beta-amyloid and oxidative free radicals in neurodegenerative diseases and the role of DNA excision-repair in the prevention of lethal neurotoxicity.


Subject(s)
Amyloid beta-Peptides/physiology , DNA/metabolism , Purines/biosynthesis , Animals , DNA/chemistry , DNA Damage , Dimerization , Immune Sera , Molecular Structure , Oxidative Stress , PC12 Cells , Purines/chemistry , Purines/immunology , Rats
6.
J Cell Physiol ; 185(1): 1-20, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10942515

ABSTRACT

Each year, an estimated 26,000 women in the United States are diagnosed with ovarian cancer. During any given year, approximately 14,500 women die from this disease. Ovarian cancer is the seventh most common cancer in women worldwide, after breast, cervix, colon/rectum, stomach, corpus uteri, and lung cancers. In the U.S., ovarian cancer is the second most common gynecologic cancer, and is the fourth leading cause of solid tumor cancer deaths among women. Currently, postoperative chemotherapy of ovarian cancer is still suboptimal. Drug resistance is a common problem resulting in only 20 approximately 30% overall 5-year survival rates. Clearly, continued development of alternative therapeutic strategies is essential for the management of this fatal disease. A number of recent studies have suggested that retinoids may play a potential role as an ovarian cancer chemotherapeutic agent. Retinoids, the natural and synthetic derivatives of vitamin A, have been shown to inhibit the growth of human ovarian cancer cells both in vivo and in culture. This review will initially summarize what is known about the pathological and molecular characteristics of ovarian carcinoma. It will then describe retinoid metabolism and the role of the cellular and nuclear retinoid binding proteins in mediating retinoid action. Following this general review of retinoids and their function, data supporting the role of retinoic acid as a suppresser of ovarian carcinoma cell growth will be presented. Particular attention will be paid to studies suggesting that members of the RB family of proteins and RB2/p130, in particular, are the molecular targets responsible for retinoid mediated inhibition of ovarian carcinoma cell growth. This review will then conclude with a brief discussion of two synthetic retinoids, 4 HPR R(fenretinide) and AHPN/CD437, which have been shown to induce apoptosis in ovarian tumor cells. It will be clear from the studies summarized in this review that retinoids represent a potentially powerful alternative to present chemotherapeutic approaches to the treatment of late stage ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Fenretinide/pharmacology , Fenretinide/therapeutic use , Ovarian Neoplasms/drug therapy , Retinoids/pharmacology , Retinoids/therapeutic use , Cell Differentiation/drug effects , Cell Division/drug effects , Female , Humans , Ovarian Neoplasms/pathology , Ovarian Neoplasms/physiopathology
7.
J Cell Physiol ; 185(1): 61-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10942519

ABSTRACT

Retinoids have great promise in the area of cancer therapy and chemoprevention. These natural and synthetic derivatives of vitamin A have been shown to play an important role in regulating cell differentiation and proliferation. While all-trans-retinoic acid (ATRA) has been demonstrated to inhibit the growth of several ovarian tumor cell lines, other ovarian carcinoma cell lines have been found to be resistant to retinoid dependent growth suppression. Interestingly, a novel synthetic retinoid, CD437 or AHPN, has been demonstrated to inhibit the growth of both ATRA-sensitive (CA-OV3) and ATRA-resistant (SK-OV3) ovarian tumor cell lines as well as to induce apoptosis. The overall goal of this research was to understand the mechanism by which AHPN/CD437 induces apoptosis in ovarian tumor cell lines. Since a number of studies have demonstrated the importance of nuclear receptors (RARs and RXRs) in mediating cellular responses to retinoids, we wished to determine the role of RARs in mediating the AHPN/CD437 response. We modulated RAR level and function by overexpressing either wild type RAR-gamma or a pan dominant negative mutant of all RAR subtypes called RAR-beta (R269Q), or through the use of an RAR-gamma antagonist, MM11253. We found that inhibition of RAR function reduced but did not eliminate induction of apoptosis in both CA-OV3 and SK-OV3 cells by AHPN/CD437. Likewise, overexpression of wild type RAR-gamma was found to increase apoptosis after treatment with AHPN/CD437. Our results suggest that in ovarian carcinomas, AHPN/CD437 induced apoptosis is mediated at least in part via an RAR pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Receptors, Retinoic Acid/metabolism , Retinoids/pharmacology , Antineoplastic Agents/therapeutic use , Female , Humans , Ovarian Neoplasms/metabolism , Retinoids/therapeutic use , Signal Transduction/drug effects , Tumor Cells, Cultured
8.
Arch Biochem Biophys ; 380(2): 339-46, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10933889

ABSTRACT

The biological actions of retinoic acid (RA) are mediated by retinoic acid receptors (RARalpha, -beta, and -gamma) and retinoid X receptors (RXRalpha, -beta, and -gamma). Although the ligand-binding domains of RARs and RXRs have been suggested to share the same novel folding pattern, the ligand-binding pockets of each of the retinoid receptors must have unique structural features since it has been possible to develop RAR subtype-selective and RXR-selective retinoids. We have previously demonstrated the importance for RA binding and RA-dependent transactivation of Arg(276) in RARalpha and Arg(278) in RARgamma; however, in RARbeta Arg(269) functions in conjunction with Lys(220). Here we have examined the role of the hydroxyl group of RARgamma Ser(289) and its homologous amino acid residues in RARalpha (Ser(287)) and RARbeta (Ser(280)) alone and in conjunction with their respective RARgamma Arg(278) homologs for RA binding and RA-dependent transactivation activity. The hydroxyl group of this Ser in all three RARs was found by itself not to be important for RA binding and RA-dependent transactivation activity. However, in RARalpha and RARgamma this Ser appears to play a small role in conjunction with Arg(276) and Arg(278), respectively, for these activities. Alternatively, strong synergism was observed in RARbeta between Ser(280) and Arg(269) for RA-binding and RA-dependent transactivation activity. This provides further evidence that the mechanism of interaction between the carboxylate group of retinoids and the amino acid residues in the ligand binding pocket of RARbeta is different from that of RARalpha and RARgamma.


Subject(s)
Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Animals , Base Sequence , Binding Sites/genetics , DNA Primers/genetics , In Vitro Techniques , Kinetics , Mice , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Receptors, Retinoic Acid/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoic Acid Receptor alpha , Serine/chemistry , Transcriptional Activation , Retinoic Acid Receptor gamma
9.
J Cell Biochem ; 77(4): 604-14, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10771516

ABSTRACT

The biological actions of retinoic acid (RA) are mediated by retinoic acid receptors (RARalpha, beta, and gamma) and retinoid X receptors (RXR alpha, beta, and gamma). Each of the RARs is expressed as four to seven different isoforms. Four isoforms of RAR beta (beta1, beta2, beta3, and beta4), which differ only in their N-terminal sequence (A domain) have been described. These RARbeta isoforms display a specific pattern of expression in developing and adult animals and are highly evolutionarily conserved suggesting that they mediate distinct cellular effects of vitamin A. Experiments were performed to examine directly the RA-binding activity, transactivation activity, and anti-AP1 activity of each of these four RARbeta isoforms. The results demonstrate that RARbeta1, beta2, and beta3 bind RA with a similar K(d) value, have a similar EC(50) value in RA-dependent transactivation assays and inhibit AP1 activity to a similar level. By contrast, RARbeta4 has an elevated K(d) for RA, an increased EC(50) value in RA-dependent transactivation assays and does not display the ability to inhibit AP1 activity. This provides additional evidence that at least one RAR isoform, RARbeta4, may mediate distinct activities within a cell. Furthermore, these data suggest that the presence of an A domain in RARbeta is important for modulating these activities of RARs.


Subject(s)
Receptors, Retinoic Acid/metabolism , Transcription Factor AP-1/metabolism , Transcriptional Activation , Tretinoin/metabolism , Alitretinoin , Animals , Blotting, Western , Dose-Response Relationship, Drug , Kinetics , Mice , Protein Binding , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Structure, Tertiary , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Transfection
10.
Oncogene ; 19(11): 1457-65, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10723137

ABSTRACT

Retinoids have been shown to inhibit the growth of many human tumor cells including breast, ovarian and squamous cell carcinoma (SCC). While the exact mechanism of retinoid mediated growth suppression is not known, a role for the retinoic acid receptors (RARs) and retinoid X receptors (RXRs) has been established in both the breast and ovarian tumor cell models. We set out to determine if modulation of RAR/RXR function would alter the retinoid sensitivity of oral SCC cells. We found that the growth of SCC cells was significantly inhibited by treatment with either all-trans-retinoic acid (trans-RA) or the synthetic, conformationally restricted RARgamma selective retinoids MM11254 and MM11389. In order to demonstrate a role for RAR/RXR function in this process, stable oral SCC cell clones constitutively overexpressing the dominant negative mutant RARbeta2 (R269Q) were prepared and shown to exhibit reduced RAR/RXR transcriptional transactivation activity. We found that oral SCC cells exhibiting reduced RAR/RXR function became resistant to growth inhibition by all-trans-RA, MM11254 and MM11389. Likewise, treatment of oral SCC cells with the RARgamma antagonist MM11253 was found to block the ability of MM11254 and MM11389 to inhibit SCC cell growth. Thus, modulation of RAR function through the use of RAR-gamma selective agonists, an RAR-gamma selective antagonist or a pan-RAR dominant negative mutant significantly alters the growth inhibitory response of oral SCC cells to retinoids.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Growth Inhibitors/pharmacology , Mouth Neoplasms/metabolism , Receptors, Retinoic Acid/physiology , Retinoids/pharmacology , Arginine/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , Cell Division/genetics , Gene Transfer Techniques , Glutamine/genetics , Growth Inhibitors/genetics , Growth Inhibitors/metabolism , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mutagenesis, Site-Directed , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoids/chemical synthesis , Tretinoin/pharmacology , Tumor Cells, Cultured , Retinoic Acid Receptor gamma
11.
J Biol Chem ; 273(51): 34016-21, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9852056

ABSTRACT

The diverse biological actions of retinoic acid (RA) are mediated by retinoic acid receptors (RARalpha, beta and gamma) and retinoid X receptors (RXR alpha, beta, and gamma). Although the ligand-binding domains of RARs share the same novel folding pattern, many RAR subtype-specific retinoids have been synthesized indicating that the ligand-binding pocket of each RAR subtype has unique features. Previously we have demonstrated the importance for RA binding and RA-dependent transactivation of Arg276 of RARalpha alone and in RARbeta Arg269 in conjunction with Lys220. In this study, we have examined the role of the homologous amino acid residues (Lys229 and Arg278) in RARgamma for these activities. Like RARalpha but dissimilar to RARbeta, Arg278 in RARgamma alone was found to play an important role in RA binding and RA-dependent transactivation. Since Lys236 in RARgamma was suggested from the crystal structure of holo-RARgamma to interact with RA, we also examined its role and that of its homologs in RARalpha and RARbeta. Despite the suggestion from the crystal structure, neither Lys236 nor its homologs in RARalpha and RARbeta play a role in the binding of RA or RA-dependent transactivation. It is likely that Lys236 in RARgamma and its homologs in RARalpha and RARbeta are solvent exposed rather than pointing into the RA-binding pocket.


Subject(s)
Arginine , Lysine , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Amino Acid Substitution , Animals , Binding Sites , DNA Primers , Kinetics , Mice , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Retinoic Acid Receptor gamma
12.
Am J Pathol ; 153(2): 469-80, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9708807

ABSTRACT

Retinoids are effective growth modulators of human ovarian carcinoma cell lines. Their effects are mediated by nuclear retinoic acid receptors (RARs) and retinoid X receptors (RXRs), which are transcriptional factors and members of the steroid/thyroid receptor superfamily. To our knowledge, until now, the cellular distribution of RAR proteins in human ovarian tumor specimens is unknown. This study provides new data on the differential cellular localization of RAR alpha protein in 16 serous adenocarcinomas originating from the ovaries, fallopian tubes, and the peritoneum. Using an affinity-purified antiserum specific for RAR alpha and a monoclonal antibody recognizing the full-length estrogen receptor molecule (clone 6F11), we performed immunohistochemistry on frozen tissue sections and examined the relationship between RAR alpha and estrogen receptor protein expression by comparing the percentage of immunostained tumor cells for either receptor. Our findings indicate a strong linear relationship between the percentages of RAR alpha- and estrogen receptor-labeled tumor cells as determined by linear regression analysis (P < 0.005, r = 0.825). A modest inverse relationship was found between the percentage of RAR alpha-positive tumor cells and histological grade, attesting to a differentiation-dependent trend (P < 0.04). No significant relationship was found between RAR alpha-labeled cells and clinical stage (P = 0.139), site of tumor origin (ovaries versus fallopian tubes versus peritoneum) (P = 0.170), and primary versus metastatic lesion (P = 0.561). Thus, serous adenocarcinomas are capable of expressing RAR alpha and estrogen receptor despite high histological grade and advanced stage of neoplastic disease. Compared with the heterogeneous localization of RAR alpha in cancer cells, there was widespread RAR alpha immunoreactivity in tumor-infiltrating lymphocytes, vascular endothelial cells, and stromal fibroblasts, underscoring the value of immunohistochemistry in the accurate determination of RAR/(RXR) content in tumor specimens.


Subject(s)
Adenocarcinoma/metabolism , Fallopian Tube Neoplasms/metabolism , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/metabolism , Receptors, Estrogen/metabolism , Receptors, Retinoic Acid/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Blotting, Western , Fallopian Tube Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Retinoic Acid Receptor alpha
13.
Mol Biol Rep ; 25(1): 1-12, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9540062

ABSTRACT

Maximal transcription of a prototypical cell cycle controlled histone H4 gene requires a proliferation-specific in vivo genomic protein/DNA interaction element, Site II. Three sequence-specific transcription factors interact with overlapping recognition motifs within Site II: interferon regulatory factor IRF-2 (HiNF-M), the putative H4 subtype-specific protein H4TF-2 (HiNF-P), and HiNF-D which represents a complex of the homeodomain protein CDP/cut, CDC2, cyclin A and pRB. However, natural sequence variation in the Site II sequences of different human H4 genes abolishes binding of specific trans-acting factors; the functional consequences of these variations have not been investigated. To address the precise contribution of H4 promoter factors to the level of H4 gene transcription, we performed a systematic mutational analysis of Site II transcriptional motifs. These mutants were tested for ability to bind each of the Site II cognate proteins, and subsequently evaluated for ability to confer H4 transcriptional activity using chimeric H4 promoter/CAT fusion constructs in different cell types. We also analyzed the effect of over-expressing IRF-2 on CAT reporter gene expression driven by mutant H4 promoters and assessed H4 transcriptional control in cells nullizygous for IRF-1 and IRF-2. Our results show that the recognition sequence for IRF-2 (HiNF-M) is the dominant component of Site II and modulates H4 gene transcription levels by 3 fold. However, the overlapping recognition sequences for IRF-2 (HiNF-M), H4TF-2 (HiNF-P) and CDP/cut (HiNF-D) together modulate H4 gene transcription levels by at least an order of magnitude. Thus, maximal activation of H4 gene transcription during the cell cycle in vivo requires the integrated activities of multiple transcription factors at Site II. We postulate that the composite organization of Site II supports responsiveness to multiple signalling pathways modulating the activities of H4 gene transcription factors during the cell cycle. Variations in Site II sequences among different H4 genes may accommodate differential regulation of H4 gene expression in cells and tissues with unique phenotypic properties.


Subject(s)
Cell Cycle/genetics , DNA-Binding Proteins/metabolism , Histones/genetics , Repressor Proteins , Transcription Factors/metabolism , Transcriptional Activation/genetics , Animals , Base Sequence , Cell Line , Cricetinae , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , HeLa Cells , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Molecular Sequence Data , Mutation , Phosphoproteins/genetics , Phosphoproteins/physiology , Promoter Regions, Genetic/genetics , Rats , Recombinant Fusion Proteins
14.
J Cell Biochem ; 68(3): 378-88, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9518263

ABSTRACT

We have used conformationally restricted retinoids to investigate the role of individual RAR subtypes and RXR in mediating the growth response of ovarian tumor cells to retinoids. Our results show that treatment of all-trans-RA-sensitive CAOV-3 cells with retinoids that bind and activate a single RAR or RXR led to a partial inhibition of growth. Treatment of all-trans-RA- resistant SKOV-3 cells did not alter growth. Maximum inhibition of growth, comparable to that observed following treatment with natural retinoids such as all-trans-RA and 9-cis-RA, was obtained only following treatment with a combination of an RAR-selective compound and an RXR-selective one. These results suggest that activation of both RAR and RXR classes is required in order to obtain maximum inhibition of ovarian tumor cell growth by retinoids. In addition, one compound, AHPN, was found to inhibit both RA-sensitive CAOV-3 and RA-resistant SKOV-3 cells. Further study of the effects of this retinoid showed that AHPN acts through an apoptotic pathway. Taken together, our results suggest that retinoids may serve as effective anti-proliferative agents in the treatment of ovarian cancer.


Subject(s)
Ovarian Neoplasms/pathology , Retinoids/chemistry , Retinoids/pharmacology , Apoptosis/drug effects , Cell Transformation, Neoplastic/drug effects , Dose-Response Relationship, Drug , Female , Humans , Ligands , Molecular Conformation , Receptors, Retinoic Acid , Retinoid X Receptors , Retinoids/administration & dosage , Structure-Activity Relationship , Time Factors , Transcription Factors , Tumor Cells, Cultured
15.
Oncogene ; 17(22): 2839-49, 1998 Dec 03.
Article in English | MEDLINE | ID: mdl-9879990

ABSTRACT

Retinoids have been shown to inhibit the growth of a number of human tumor cells, including several ovarian adenocarcinoma cell lines. All-trans retinoic acid (RA) is an effective growth suppressor of CA-OV3 cells but not SK-OV3 cells. Since the effects of RA are known to be mediated via the nuclear receptors (RARs and RXRs), we initially compared levels of the various RARs and RXRs in the CA-OV3 and SK-OV3 cell lines. The RA resistant SK-OV3 cells expressed reduced levels of RAR-alpha and RXR-alpha. Furthermore, induction of RAR-alpha by RA was impaired in the RA resistant SK-OV3 cells as was RARE binding and RARE-dependent transcriptional activity. These results suggested that changes in the amounts and/or activity of RARs and/or RXR-alpha could determine the growth response of ovarian tumor cells to RA. This was confirmed by modulating the levels of RARs and RXR-alpha in the SK-OV3 cells using the LacSwitch inducible expression system. Stably transfected clones of RA resistant SK-OV3 cells exhibited a significant inhibition of growth by RA treatment when RAR-alpha was induced. Overexpression of both RAR-alpha and RXR-alpha resulted in a level of growth inhibition nearly equal to that exhibited by the RA sensitive CA-OV3 cell line. Similar results were obtained when a combination of RXR-alpha and either RAR-beta or RAR-gamma was overexpressed in SK-OV3 cells. Our results show that the nuclear receptors and RXR-alpha play a critical role in mediating growth suppression by RA in ovarian cancer cells.


Subject(s)
Ovarian Neoplasms/pathology , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , Blotting, Western , Cell Division/drug effects , DNA, Neoplasm/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression/drug effects , Humans , Receptors, Retinoic Acid/genetics , Response Elements/genetics , Retinoic Acid Receptor alpha , Retinoid X Receptors , Transcription Factors/genetics , Transcriptional Activation/drug effects , Transfection , Tumor Cells, Cultured , Retinoic Acid Receptor gamma
16.
Exp Cell Res ; 232(2): 277-86, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9168803

ABSTRACT

We prepared single cell clones from two ovarian carcinoma cell lines, CA-OV3 and SK-OV3, and analyzed the effect of all-trans-RA treatment on cell division, DNA synthesis, and cell cycle stage distribution of these single cell clones. Our results show that despite the well-known heterogeneous nature of these cell lines, all single cell clones of SK-OV3 cells are resistant to the growth inhibitory effects of all-trans-RA. In contrast, all single cell clones of CA-OV3 cells were growth inhibited by all-trans-RA. However, the extent of growth inhibition did vary somewhat from clone to clone. Additional studies employing flow cytometry showed that all-trans-RA blocked CA-OV3 cell cycle progression in the G1 stage. Finally, all-trans-RA was able to inhibit G1 progression in growth-arrested CA-OV3 cells following stimulation with fetal bovine serum, insulin, IGF-1, or estrogen. Since each of these growth factors is known to act via distinct signal transduction pathways, our results suggest that all-trans-RA blocks G1 progression by targeting a downstream process or event which occurs at a point after the insulin/IGF-1, estrogen, and serum signal transduction pathways converge.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , G1 Phase/drug effects , Growth Inhibitors/pharmacology , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Tretinoin/pharmacology , Animals , Cattle , Cell Division/drug effects , DNA Replication/drug effects , DNA, Neoplasm/biosynthesis , Estradiol/pharmacology , Female , Fetal Blood/physiology , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured/drug effects
17.
J Cell Biochem ; 65(2): 245-53, 1997 May.
Article in English | MEDLINE | ID: mdl-9136081

ABSTRACT

The proto-oncogenes c-jun and junD are closely related transcriptional factors with opposing actions on cell growth and division. Expression of c-jun rapidly increases as cells enter the cell cycle. Levels of c-jun are also increased in the early stages of experimental cardiac hypertrophy and failure but expression decreases with time. In contrast, junD accumulates in quiescent cells. Expression in end-stage cardiomyopathy has not been studied. Steady-state levels of c-jun and junD mRNA were determined in failing human myocardium (obtained at the time of cardiac transplantation) and in control myocardium from patients who died of noncardiac causes. Relative expression was normalized for glyceraldehyde-3-phosphate dehydrogenase expression. Levels of junD were almost four-fold depressed in myocardium from myopathic hearts (2.1 +/- 0.27, x +/- SE; n = 20) vs. the controls (7.7 +/- 1.1; n = 3). Levels of c-jun were similar in both myopathic and control hearts. Relative expression of beta-myosin heavy chain was the same in both myopathic and control hearts. Levels of junD were still found to be depressed in the myopathic hearts after normalization for myosin heavy chain gene expression. We conclude that c-jun and junD are differentially regulated in end-stage human cardiomyopathy with expression of junD being decreased while relative levels of c-jun mRNA remain unchanged. Further studies are needed to determine the role of junD down-regulation in the development and/or maintenance of the abnormalities present in end-stage heart disease.


Subject(s)
Cardiomyopathies/metabolism , Gene Expression , Genes, jun , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Male , Middle Aged , Myocardial Ischemia/metabolism , RNA, Messenger/metabolism
18.
J Biol Chem ; 272(17): 11244-9, 1997 Apr 25.
Article in English | MEDLINE | ID: mdl-9111026

ABSTRACT

The diverse biological actions of retinoic acid (RA) are mediated by retinoic acid receptors (RARs) and retinoid X receptors. Although it has been suggested that the ligand binding domains (LBDs) of RARs share the same novel folding pattern, many RAR subtype-specific agonists and antagonists have been synthesized demonstrating that the LBD of each RAR subtype has unique features. We have examined the role of several positively charged amino acid residues located in the LBD of RARalpha in RA binding. These results are compared with previously published data for the homologous mutations in RARbeta. Lys227 of RARalpha does not appear to be important for RA binding or RA-dependent transactivation, whereas the homologous residue in RARbeta, Lys220, plays an important synergistic role with Arg269 in these two activities. In addition, Arg276 of RARalpha, like its homologous residue Arg269 of RARbeta, was found to play an important role in the binding of RA most likely by interacting with the carboxylate group of RA. However, the orientation of and electronic environment associated with Arg276 in RARalpha appears to be different from that of Arg269 in RARbeta, thus contributing to the uniqueness of the ligand binding pocket of each receptor.


Subject(s)
Amino Acids, Diamino/metabolism , Receptors, Retinoic Acid/metabolism , Retinoids/metabolism , Transcriptional Activation , Amino Acids, Diamino/genetics , Animals , Arginine/genetics , Arginine/metabolism , Binding Sites/genetics , Lysine/genetics , Lysine/metabolism , Mice , Mutation , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Stereoisomerism , Tretinoin/metabolism , Vitamin A/metabolism
19.
J Biol Chem ; 272(2): 746-53, 1997 Jan 10.
Article in English | MEDLINE | ID: mdl-8995359

ABSTRACT

The diverse biological functions of retinoic acid (RA) are mediated through retinoic acid receptors (RARs) and retinoid X receptors. RARs contain a high affinity binding site for RA which is sensitive to treatment with sulfhydryl modification reagents. In an attempt to identify which Cys residues are important for this loss of binding, we created three site-specific RARbeta mutants: C228A, C258A, and C267A. The affinity for RA of all three mutant receptors was in the range of that of the wild type protein, suggesting that none of these Cys residues are critical for RA binding. Rather, these modified Cys residue(s) function to sterically hinder RA binding; however, the modified Cys residues critical for the inhibition of binding differ depending on the reagent employed. Only modification of Cys228 is necessary to inhibit RA binding when RARbeta is modified by reagents which transfer large bulky groups while both Cys228 and Cys267 must be modified when a small functional group is transferred. These data suggest that both Cys228 and Cys267 but not Cys258 lie in the ligand binding pocket of RARbeta. However, Cys228 lies closer to the opening of the RARbeta ligand binding pocket whereas Cys267 lies more deeply buried.


Subject(s)
Cysteine/chemistry , Receptors, Retinoic Acid/metabolism , Sulfhydryl Reagents/metabolism , Binding Sites , Blotting, Western , Dithionitrobenzoic Acid/pharmacology , Ethylmaleimide/pharmacology , Hydroxymercuribenzoates/pharmacology , Mutagenesis, Site-Directed , Plasmids , Polymerase Chain Reaction , Protease Inhibitors/pharmacology , Retinoic Acid Receptor alpha , Structure-Activity Relationship , Tretinoin/metabolism , Retinoic Acid Receptor gamma
20.
Exp Cell Res ; 237(1): 118-26, 1997 Nov 25.
Article in English | MEDLINE | ID: mdl-9417874

ABSTRACT

We wished to determine the effect of altering the levels or functional activity of retinoid receptors, in particular retinoic acid receptor-alpha (RAR-alpha) and retinoid X receptor-alpha (RXR-alpha) on the growth sensitivity of ovarian tumor cells to all-trans-retinoic acid (all-trans-RA). We found that CA-OV3 cells could be made resistant to all-trans-RA growth inhibition by overexpressing RAR-beta(R269Q), an efficient dominant negative mutant which inhibits the function of all RAR subtypes. Antisense technology was then used to prepare stable transfectants of the retinoid-sensitive ovarian carcinoma cell line CA-OV3 in which expression of RAR-alpha, RXR-alpha, or both RAR-alpha and RXR-alpha was reduced. The effect of all-trans-RA on ovarian tumor cell growth was determined by MTT assay, autoradiographic analysis of DNA synthesis, and anchorage-independent colony formation in soft agar. Our results show that cell lines expressing reduced levels of either RAR-alpha alone or RXR-alpha alone exhibited a small decrease in sensitivity to growth inhibition by all-trans-RA. However, maximum RA resistance was obtained in cell lines in which the levels of both RAR-alpha and RXR-alpha were reduced. These results demonstrate the importance of both retinoid nuclear receptors and retinoid-X receptors in general, and RAR-alpha and RXR-alpha in particular, as mediators of ovarian carcinoma cell growth inhibition by retinoids.


Subject(s)
Antineoplastic Agents/toxicity , Receptors, Retinoic Acid/physiology , Transcription Factors/physiology , Tretinoin/toxicity , Animals , Cell Division/drug effects , Cell Nucleus/metabolism , Female , Humans , Isopropyl Thiogalactoside/pharmacology , Mice , Ovarian Neoplasms , RNA, Antisense/metabolism , RNA, Messenger/metabolism , Receptors, Retinoic Acid/biosynthesis , Recombinant Proteins/biosynthesis , Retinoic Acid Receptor alpha , Retinoid X Receptors , Transcription Factors/biosynthesis , Transcription, Genetic , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...