Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Endocrinology ; 155(9): 3251-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24971610

ABSTRACT

The SMRT coregulator functions as a dual coactivator and corepressor for estrogen receptor-α (ERα) in a gene-specific manner, and in several studies its elevated expression correlates with poor outcome for breast cancer patients. A specific role of SMRT in breast cancer progression has not been elucidated, but SMRT knock-down limits estradiol-dependent growth of MCF-7 breast cancer cells. In this study, small-interfering RNA (siRNA) and short-hairpin RNA (shRNA) approaches were used to determine the effects of SMRT depletion on growth of ERα-positive MCF-7 and ZR-75-1 breast cancer cells, as well as the ERα-negative MDA-MB-231 breast cancer line. Depletion of SMRT inhibited growth of ERα-positive cells grown in monolayer but had no effect on growth of the ERα-negative cells. Reduced SMRT levels also negatively impacted the anchorage-independent growth of MCF-7 cells as assessed by soft agar colony formation assays. The observed growth inhibitions were due to a loss of estradiol-induced progression through the G1/S transition of the cell cycle and increased apoptosis in SMRT-depleted compared with control cells. Gene expression analyses indicated that SMRT inhibits apoptosis by a coordinated regulation of genes involved in apoptosis. Functioning as a dual coactivator for anti-apoptotic genes and corepressor for pro-apoptotic genes, SMRT can limit apoptosis. Together these data indicate that SMRT promotes breast cancer progression through multiple pathways leading to increased proliferation and decreased apoptosis.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Cell Cycle , Cell Proliferation , Estrogen Receptor alpha/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Estrogen Receptor alpha/genetics , Female , Humans , MCF-7 Cells , Nuclear Receptor Co-Repressor 2/genetics
2.
Mol Cell Biol ; 34(7): 1246-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24449765

ABSTRACT

The silencing mediator of retinoic acid and thyroid hormone receptors (SMRT) is an established histone deacetylase 3 (HDAC3)-dependent transcriptional corepressor. Microarray analyses of MCF-7 cells transfected with control or SMRT small interfering RNA revealed SMRT regulation of genes involved in DNA damage responses, and the levels of the DNA damage marker γH2AX as well as poly(ADP-ribose) polymerase cleavage were elevated in SMRT-depleted cells treated with doxorubicin. A number of these genes are established p53 targets. SMRT knockdown decreased the activity of two p53-dependent reporter genes as well as the expression of p53 target genes, such as CDKN1A (which encodes p21). SMRT bound directly to p53 and was recruited to p53 binding sites within the p21 promoter. Depletion of GPS2 and TBL1, components of the SMRT corepressor complex, but not histone deacetylase 3 (HDAC3) decreased p21-luciferase activity. p53 bound to the SMRT deacetylase activation domain (DAD), which mediates HDAC3 binding and activation, and HDAC3 could attenuate p53 binding to the DAD region of SMRT. Moreover, an HDAC3 binding-deficient SMRT DAD mutant coactivated p53 transcriptional activity. Collectively, these data highlight a biological role for SMRT in mediating DNA damage responses and suggest a model where p53 binding to the DAD limits HDAC3 interaction with this coregulator, thereby facilitating SMRT coactivation of p53-dependent gene expression.


Subject(s)
Histone Deacetylases/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Gene Knockdown Techniques , Humans , MCF-7 Cells , Models, Biological , Mutation , Nuclear Receptor Co-Repressor 2/antagonists & inhibitors , Nuclear Receptor Co-Repressor 2/genetics , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
3.
J Biol Chem ; 288(33): 24020-34, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23814048

ABSTRACT

The role of glucocorticoids in the inhibition of estrogen (17-ß-estradiol (E2))-regulated estrogen receptor (ER)-positive breast cancer cell proliferation is well established. We and others have seen that synthetic glucocorticoid dexamethasone (Dex) antagonizes E2-stimulated endogenous ERα target gene expression. However, how glucocorticoids negatively regulate the ERα signaling pathway is still poorly understood. ChIP studies using ERα- and glucocorticoid receptor (GR)-positive MCF-7 cells revealed that GR occupies several ERα-binding regions (EBRs) in cells treated with E2 and Dex simultaneously. Interestingly, there was little or no GR loading to these regions when cells were treated with E2 or Dex alone. The E2+Dex-dependent GR recruitment is associated with the displacement of ERα and steroid receptor coactivator-3 from the target EBRs leading to the repression of ERα-mediated transcriptional activation. The recruitment of GR to EBRs requires assistance from ERα and FOXA1 and is facilitated by AP1 binding within the EBRs. The GR binding to EBRs is mediated via direct protein-protein interaction between the GR DNA-binding domain and ERα. Limited mutational analyses indicate that arginine 488 located within the C-terminal zinc finger domain of the GR DNA-binding domain plays a critical role in stabilizing this interaction. Together, the results of this study unravel a novel mechanism involved in glucocorticoid inhibition of ERα transcriptional activity and E2-mediated cell proliferation and thus establish a foundation for future exploitation of the GR signaling pathway in the treatment of ER-positive breast cancer.


Subject(s)
Dexamethasone/pharmacology , Estrogen Receptor alpha/metabolism , Receptors, Glucocorticoid/metabolism , Transcription Factor AP-1/metabolism , Amino Acid Sequence , Animals , Arginine/metabolism , Binding Sites/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chromosomes, Human/genetics , Enhancer Elements, Genetic/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Ligands , Models, Biological , Molecular Sequence Data , Nuclear Receptor Coactivator 3/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Stability/drug effects , RNA, Small Interfering/metabolism , Rats , Receptors, Glucocorticoid/chemistry , Transcription, Genetic/drug effects
4.
Biochim Biophys Acta ; 1819(7): 707-15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22406422

ABSTRACT

Forkhead box (FOX) proteins represent a large family of transcriptional regulators unified by their DNA binding domain (DBD) known as a 'forkhead' or 'winged helix' domain. Over 40 FOX genes have been identified in the mammalian genome. FOX proteins share significant sequence similarities in the DBD which allow them to bind to a consensus DNA response element. However, their modes of action are quite diverse as they regulate gene expression by acting as pioneer factors, transcription factors, or both. This review focuses on the mechanisms of chromatin remodeling with an emphasis on three sub-classes-FOXA, FOXO, and FOXP members. FOXA proteins serve as pioneer factors to open up local chromatin structure and thereby increase accessibility of chromatin to factors regulating transcription. FOXP proteins, in contrast, function as classic transcription factors to recruit a variety of chromatin modifying enzymes to regulate gene expression. FOXO proteins represent a hybrid subclass having dual roles as pioneering factors and transcription factors. A subset of FOX proteins interacts with condensed mitotic chromatin and may function as 'bookmarking' agents to maintain transcriptional competence at specific genomic sites. The overall diversity in chromatin remodeling function by FOX proteins is related to unique structural motifs present within the DBD flanking regions that govern selective interactions with core histones and/or chromatin coregulatory proteins. This article is part of a Special Issue entitled: Chromatin in time and space.


Subject(s)
Chromatin Assembly and Disassembly , Forkhead Transcription Factors/physiology , Amino Acid Sequence , Animals , Base Sequence , Conserved Sequence , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Histones/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary
5.
Endocr Relat Cancer ; 18(1): 113-27, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21059860

ABSTRACT

Elevated expression of steroid receptor coactivator-3 (SRC-3), a member of the p160 family of nuclear receptor coactivators, has been implicated in tamoxifen resistance of breast tumors while the involvement of the two other members of this family, SRC-1 and SRC-2, is less well characterized. In this study, using small interfering RNA-based silencing, the role of each SRC coactivator in the growth of the LCC2 estrogen-independent and tamoxifen-resistant breast cancer cell line was evaluated. The loss of SRC-1, SRC-2, or SRC-3 did not significantly alter LCC2 proliferation or cell cycle distribution of 4-hydroxytamoxifen- versus vehicle-treated cells. However, depletion of SRC-2 and SRC-3, but not SRC-1, decreased basal cell proliferation and increased apoptosis. Cell cycle analyses further illustrated the divergent contributions of SRC-2 and SRC-3 with depletion of the former increasing the percentage of cells in the G(0)G(1) and sub-G(0)G(1) phases of cell cycle yet maintaining sensitivity to estradiol and ICI 182 780 antiestrogen, while SRC-3 depletion increased cells in the sub-G(0)G(1) phase and ablated response to estrogen receptor α (ERα) ligands. Surprisingly, the effects of SRC coactivator depletion on ERα transcriptional activity, as measured by luciferase reporter gene, did not correspond to the observed effects on proliferation (e.g. SRC-1 knockdown increases ERα activity). Collectively, these data indicate that SRC control of basal and hormone-regulated proliferations is not solely mediated by ERα, and suggest that targeting growth inhibition by disrupting SRC-2 and SRC-3 function may be an effective approach to inhibit the growth of tamoxifen-resistant breast cancer.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Cell Proliferation , Drug Resistance, Neoplasm/drug effects , Estradiol/pharmacology , Nuclear Receptor Coactivators/physiology , Tamoxifen/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Estradiol/analogs & derivatives , Estrogen Receptor Modulators/pharmacology , Female , Fulvestrant , Humans , Multigene Family/genetics , Multigene Family/physiology , Nuclear Receptor Coactivator 3/genetics , Nuclear Receptor Coactivator 3/metabolism , Nuclear Receptor Coactivator 3/physiology , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism , Tamoxifen/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
6.
Mol Endocrinol ; 24(6): 1187-202, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20392877

ABSTRACT

Although the ability of coactivators to enhance the expression of estrogen receptor-alpha (ERalpha) target genes is well established, the role of corepressors in regulating 17beta-estradiol (E2)-induced gene expression is poorly understood. Previous studies revealed that the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) corepressor is required for full ERalpha transcriptional activity in MCF-7 breast cancer cells, and we report herein the E2-dependent recruitment of SMRT to the regulatory regions of the progesterone receptor (PR) and cyclin D1 genes. Individual depletion of SMRT or steroid receptor coactivator (SRC)-3 modestly decreased E2-induced PR and cyclin D1 expression; however, simultaneous depletion revealed a cooperative effect of this coactivator and corepressor on the expression of these genes. SMRT and SRC-3 bind directly in an ERalpha-independent manner, and this interaction promotes E2-dependent SRC-3 binding to ERalpha measured by co-IP and SRC-3 recruitment to the cyclin D1 gene as measured by chromatin IP assays. Moreover, SMRT stimulates the intrinsic transcriptional activity of all of the SRC family (p160) coactivators. Our data link the SMRT corepressor directly with SRC family coactivators in positive regulation of ERalpha-dependent gene expression and, taken with the positive correlation found for SMRT and SRC-3 in human breast tumors, suggest that SMRT can promote ERalpha- and SRC-3-dependent gene expression in breast cancer.


Subject(s)
Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Coactivator 3/metabolism , Receptors, Progesterone/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cyclin D1/metabolism , Enhancer Elements, Genetic/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Nuclear Receptor Co-Repressor 2/chemistry , Nuclear Receptor Co-Repressor 2/deficiency , Nuclear Receptor Coactivator 2/metabolism , Nuclear Receptor Coactivator 3/genetics , Protein Binding/drug effects , Protein Structure, Tertiary , Receptors, Progesterone/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
7.
Int J Cancer ; 124(8): 1841-51, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19117054

ABSTRACT

B-cell Translocation Gene 2 (BTG2/TIS21/PC3) is an anti-proliferative tumor suppressor gene whose expression is significantly reduced in breast carcinomas, and in MCF-7 and T-47D breast cancer cell lines treated with estradiol (E2). In this study the mechanisms involved in E2 down regulation of BTG2 gene expression were examined. Depletion of ERalpha by siRNA indicated that the receptor is required for E2 down regulation of BTG2 mRNA levels, and cycloheximide experiments indicated that the effect of E2 on BTG2 expression was independent of intermediary protein synthesis. Chromatin immunoprecipitation analyses revealed that ERalpha interacts with the BTG2 promoter in a ligand-independent fashion whereas transfection experiments indicated that ERalpha's DNA and ligand binding domains are required for E2 repression of BTG promoter activity. Surprisingly, histone deacetylase (HDACs) activity is essential for basal expression as evidenced by trichostatin A inhibition of BTG2 mRNA levels. Estradiol treatment did not alter histone H3 acetylation although it did induce displacement of RNA polymerase II from the BTG2 gene. Depletion of the ER specific corepressor REA (Repressor of Estrogen Receptor Activity) significantly abrogated E2-mediated BTG2 repression. Taken together, our results reveal a requirement of HDAC activity for basal BTG2 expression and the ERalpha-REA interaction for estrogen repression of the BTG2 gene. The ability of E2-bound ERalpha and REA to suppress BTG2 expression indicates a positive role for this corepressor in regulation of breast cancer cell proliferation.


Subject(s)
Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/physiology , Base Sequence , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Histone Deacetylases/metabolism , Humans , Ligands , Molecular Sequence Data , Prohibitins , Protein Binding , Protein Structure, Tertiary , Tumor Suppressor Proteins
8.
Endocrinology ; 150(4): 1588-96, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19095746

ABSTRACT

Each of the three members of the p160 steroid receptor coactivator (SRC) family of coactivators (SRC-1, SRC-2 and SRC-3) stimulates estrogen receptor (ER)-alpha function in trans-activation assays. Consequently, we sought to elucidate their contributions to the ER-regulated processes of cell proliferation, apoptosis, and the expression of ERalpha target genes in MCF-7 breast cancer cells. The small interfering RNA depletion of SRC-2 or SRC-3 but not SRC-1 inhibited growth of MCF-7 cells, and this was reflected in decreased cell cycle progression and increased apoptosis in SRC-2- or SRC-3-depleted cells as well as a reduction in ERalpha transcriptional activity measured on a synthetic reporter gene. However, only SRC-3 depletion blocked estradiol stimulated cell proliferation. Depletion of SRC-1 did not affect these events, and together this reveals functional differences between each of the three SRC family coactivators. Regulation of the endogenous ERalpha target gene, c-myc was not affected by depletion of any of the p160 coactivators although depletion of each of them decreased pS2 mRNA expression in estradiol-treated MCF-7 cells. Moreover, progesterone receptor and cyclin D1 gene expression were decreased in SRC-3 small interfering RNA-treated cells. Expression of mRNA and protein levels for the antiapoptotic gene, Bcl-2 was dependent on SRC-3 expression, whereas Bcl-2 protein but not mRNA expression also was sensitive to SRC-1 depletion. Together these data indicate that the closely related p160 coactivators are not functionally redundant in breast cancer cells because they play gene-specific roles in regulating mRNA and protein expression, and they therefore are likely to make unique contributions to breast tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Estrogen Receptor alpha/genetics , Histone Acetyltransferases/physiology , Nuclear Receptor Coactivator 2/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Transcription, Genetic/genetics , Apoptosis/genetics , Blotting, Western , Breast Neoplasms/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cyclin D1/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Histone Acetyltransferases/genetics , Humans , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2/genetics , Nuclear Receptor Coactivator 3 , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Receptors, Progesterone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Transcription Factors/genetics , Transfection
9.
Mol Cell Biol ; 27(17): 5933-48, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17591692

ABSTRACT

Multiple factors influence estrogen receptor alpha (ERalpha) transcriptional activity. Current models suggest that the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) corepressor functions within a histone deactylase-containing protein complex that binds to antiestrogen-bound ERalpha and contributes to negative regulation of gene expression. In this report, we demonstrate that SMRT is required for full agonist-dependent ERalpha activation. Chromatin immunoprecipitation assays demonstrate that SMRT, like ERalpha and the SRC-3 coactivator, is recruited to an estrogen-responsive promoter in estrogen-treated MCF-7 cells. Depletion of SMRT, but not histone deacetylases 1 or 3, negatively impacts estradiol-stimulated ERalpha transcriptional activity, while exogenous expression of SMRT's receptor interaction domains blocks ERalpha activity, indicating a functional interaction between this corepressor and agonist-bound ERalpha. Stimulation of estradiol-induced ERalpha activity by SMRT overexpression occurred in HeLa and MCF-7 cells, but not HepG2 cells, indicating that these positive effects are cell type specific. Similarly, the ability of SMRT depletion to promote the agonist activity of tamoxifen was observed for HeLa but not MCF-7 cells. Furthermore, impairment of agonist-stimulated activity by SMRT depletion is specific to ERalpha and not observed for receptors for vitamin D, androgen, or thyroid hormone. Nuclear receptor corepressor (N-CoR) depletion increased the transcriptional activity of all four tested receptors. SMRT is required for full expression of the ERalpha target genes cyclin D1, BCL-2, and progesterone receptor but not pS2, and its depletion significantly attenuated estrogen-dependent proliferation of MCF-7 cells. Taken together, these data indicate that SMRT, in conjunction with gene-specific and cell-dependent factors, is required for positively regulating agonist-dependent ERalpha transcriptional activity.


Subject(s)
DNA-Binding Proteins/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Repressor Proteins/metabolism , Transcription, Genetic , Animals , Breast Neoplasms , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/genetics , Estrogen Receptor Modulators/metabolism , Estrogen Receptor alpha/genetics , Estrogens/metabolism , Female , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Selective Estrogen Receptor Modulators/metabolism
10.
Mol Endocrinol ; 20(11): 2695-710, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16840538

ABSTRACT

Estrogen receptor-alpha (ERalpha) is a member of the nuclear receptor superfamily of ligand-activated transcription factors. Abundant evidence demonstrates that ERalpha agonists promote, whereas antagonists inhibit, receptor binding to coactivators. In this report we demonstrate that binding of the ICI 182,780 (ICI) pure antiestrogen to ERalpha promotes its interaction with the cAMP response element-binding protein-binding protein (CBP)/p300 but not the p160 family of coactivators, demonstrating the specificity of this interaction. Amino acid mutations within the coactivator binding surface of the ERalpha ligand-binding domain revealed that CBP binds to this region of the ICI-liganded receptor. The carboxy-terminal cysteine-histidine rich domain 3 of CBP, rather than its amino-terminal nuclear interacting domain, shown previously to mediate agonist-dependent interactions of CBP with nuclear receptors, is required for binding to ICI-liganded ERalpha. Chromatin immunoprecipitation assays revealed that ICI but not the partial agonist/antagonist 4-hydroxytamoxifen is able to recruit CBP to the pS2 promoter, and this distinguishes ICI from this class of antiestrogens. Chromatin immunoprecipitation assays for pS2 and cytochrome P450 1B1 promoter regions revealed that ICI-dependent recruitment of CBP, but not receptor, to ERalpha targets is gene specific. ICI treatment did not recruit the steroid receptor coactivator 1 to the pS2 promoter, and it failed to induce the expression of this gene. Taken together, these data indicate that recruitment of the CBP coactivator/cointegrator without steroid receptor coactivator 1 to ERalpha is insufficient to promote transcription of ERalpha target genes.


Subject(s)
Estradiol/analogs & derivatives , Estrogen Receptor alpha/metabolism , p300-CBP Transcription Factors/metabolism , Activating Transcription Factors/metabolism , CREB-Binding Protein/chemistry , CREB-Binding Protein/metabolism , DNA-Binding Proteins , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Fulvestrant , HeLa Cells , Humans , Mutation/physiology , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Receptors, Estrogen/antagonists & inhibitors , Selective Estrogen Receptor Modulators/pharmacology , Tumor Cells, Cultured
11.
Int J Biochem Cell Biol ; 38(8): 1277-89, 2006.
Article in English | MEDLINE | ID: mdl-16507348

ABSTRACT

A neuronal type Ca2+ stimulated nitric oxide synthase was earlier reported by us to be present in the protozoan parasite Leishmania donovani. As part of nitric oxide-cyclic GMP transduction signaling operative in higher eukaryotes and involved in the long-term potentiation, a soluble guanylyl cyclase has also been detected in this lower eukaryote. However, detailed biochemical characterization revealed the enzyme to be Ca2+ modulated and unstimulated by nitric oxide donors as opposed to higher eukaryotes. The possible role of intracellular Ca2+ level in the regulation of guanylyl cyclase activity as well as L. donovani infectivity was explored by measuring the intracellular survival of the parasites in mammalian macrophages after treatments, which decrease or elevate the intracellular Ca2+. Parasites loaded with intracellular Ca2+ chelators displayed significantly decreased infectivity and cyclic GMP level. In contrast, pretreatment with Ca2+ ionophores, which elevated Ca2+ levels in L. donovani, significantly enhanced the cyclic GMP level as well as the infectivity of the parasites. Moreover, treatment with selective inhibitors of soluble guanylyl cyclase also reduced infectivity, even in cases of calcium ionophore-treated parasites. The gene encoding the soluble guanylyl cyclase was cloned, sequenced and over expressed in bacterial system. The recombinant protein showed enzyme characteristics similar to that obtained in L. donovani promastigote cytosol. Together these results suggest a possible link between guanylyl cyclase, intracellular Ca2+ content and parasite infectivity.


Subject(s)
Calcium/metabolism , Guanylate Cyclase/metabolism , Leishmania donovani/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Calcium/pharmacology , Catalysis/drug effects , Cloning, Molecular , Cyclic GMP/metabolism , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Gene Expression Regulation, Enzymologic , Guanosine Triphosphate/pharmacology , Guanylate Cyclase/genetics , Leishmania donovani/genetics , Leishmania donovani/pathogenicity , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Molecular Sequence Data , Nitric Oxide Donors/pharmacology , Protozoan Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
12.
Proc Natl Acad Sci U S A ; 102(5): 1339-44, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15677324

ABSTRACT

Steroid receptor coactivator-3 (SRC-3/AIB1) is a coactivator for nuclear receptors and other transcription factors and an oncogene that contributes to growth regulation and development of mammary and other tumor types. Because of its biological functions, it is important to identify genes regulated by SRC-3. However, because coactivators do not bind DNA directly, extensive work is required to determine whether genes identified by RNA profiling approaches are direct or indirect targets. Here, we report the use of chromatin immunoprecipitation (ChIP)-based assays that involve genomic mapping and computational analyses of immunoprecipitated DNA to identify SRC-3-binding target genes in estradiol (E2)-treated MCF-7 breast cancer cells. We identified 18 SRC-3 genomic binding sites and demonstrated estrogen receptor-alpha (ERalpha) binding to all of them. Both E2-dependent and -independent SRC-3/ERalpha-binding sites were identified. RNA polymerase II ChIP assays were used to determine the correlation between SRC-3 and ERalpha binding and recruitment of the transcriptional machinery. These assays, in conjunction with analyses of RNA obtained from E2-treated cells, lead to the identification of SRC-3/ERalpha-associated genes. The ability of SRC family coactivators to regulate the expression of one of these genes, PARD6B/Par6, was confirmed by using cells individually depleted of SRC-1, SRC-2, or SRC-3 by small interfering RNA. The method described herein can be used to identify genes regulated by non-DNA-binding factors, such as other coactivators or corepressors, as well as DNA-binding transcription factors, and provides information on their binding location that can accelerate further gene characterization.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Acetyltransferases , Cell Line, Tumor , DNA, Neoplasm/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Expressed Sequence Tags , Female , Histone Acetyltransferases , Humans , Neoplasm Proteins/genetics , Nuclear Receptor Coactivator 3 , Oncogene Proteins , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics
13.
J Infect Dis ; 189(6): 1024-34, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-14999606

ABSTRACT

A parasite-specific 51-kDa protein has been isolated from the membrane of macrophages infected with Leishmania donovani, the causative agent of visceral leishmaniasis. Active targeting of doxorubicin to infected macrophages was studied by incorporating it in immunoliposomes prepared by grafting F(ab)'(2) of anti-51-kDa antibody onto the liposomal surface. In a 45-day mouse model of visceral leishmaniasis, complete elimination of spleen parasite burden was achieved by doxorubicin incorporated in immunoliposome (immunodoxosome) at a dose of 250 microg/kg/day that was given for 4 consecutive days. A similar dose of free and liposomal drug (doxosome) had 45% and 84% parasite suppressive effects, respectively. Immunodoxosome and doxosome were generally less toxic than the free drug, as determined by several clinical parameters of cardiotoxicity and liver toxicity. These results not only indicate the potential of doxorubicin as an effective chemotherapeutic agent but also establish the use of immunoliposomes as drug carrier in the therapy of leishmaniasis.


Subject(s)
Antibodies, Protozoan/immunology , Doxorubicin/administration & dosage , Leishmaniasis, Visceral/drug therapy , Animals , Doxorubicin/toxicity , Drug Carriers , Humans , Immunoglobulin Fab Fragments/immunology , Liposomes , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Protozoan Proteins/immunology , Protozoan Proteins/isolation & purification
14.
Eur J Biochem ; 270(18): 3806-13, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12950264

ABSTRACT

Earlier we presented several lines of evidence that a 67-kDa laminin binding protein (LBP) in Leishmania donovani, that is different from the putative mammalian 67-kDa laminin receptor, may play an important role in the onset of leishmaniasis, as these parasites invade macrophages in various organs after migrating through the extracellular matrix. Here we describe the membrane orientation of this Leishmania laminin receptor. Flow cytometric analysis using anti-LBP Ig revealed its surface localization, which was further confirmed by enzymatic radiolabeling of Leishmania surface proteins, autoradiography and Western blotting. Efficient incorporation of LBP into artificial lipid bilayer, as well as its presence in the detergent phase after Triton X-114 membrane extraction, suggests that it may be an integral membrane protein. Limited trypsinization of intact parasite and subsequent immunoblotting of trypsin released material using laminin as primary probe revealed that a major part of this protein harbouring the laminin binding site is oriented extracellularly. Carboxypeptidase Y treatment of the whole cell, as well as the membrane preparation, revealed that a small part of the C-terminal is located in the cytosol. A 34-kDa transmembrane part of LBP could be identified using the photoactive probe, 3-(trifluoromethyl)-3-(m-iodophenyl)diazirine (TID). Partial sequence comparison of the intact protein to that with the trypsin-released fragment indicated that N-terminal may be located extracellularly. Together, these results suggest that LBP may be an integral membrane protein, having significant portion of N-terminal end as well as the laminin binding site oriented extracellularly, a membrane spanning domain and a C-terminal cytosolic end.


Subject(s)
Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Leishmania donovani/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptors, Laminin/chemistry , Receptors, Laminin/metabolism , Animals , Azirines/chemistry , Binding Sites , Carboxypeptidases/metabolism , Flow Cytometry , Humans , Iodine Radioisotopes , Laminin/chemistry , Laminin/metabolism , Leishmania donovani/metabolism , Liposomes/chemistry , Liposomes/metabolism , Peptide Fragments/analysis , Peptide Fragments/chemistry , Photoaffinity Labels/chemistry , Polyethylene Glycols/chemistry , Protein Binding , Protein Structure, Tertiary
15.
Eur J Pharmacol ; 470(1-2): 103-12, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12787838

ABSTRACT

Inflammatory bowel disease is characterized by oxidative and nitrosative stress, leukocyte infiltration and upregulation of proinflammatory cytokines. The aim of the present study was to examine the protective effects of thearubigin, an anti-inflammatory and anti-oxidant beverage derivative, on 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice, a model for inflammatory bowel disease. Intestinal lesions (judged by macroscopic and histological score) were associated with neutrophil infiltration (measured as increase in myeloperoxidase activity in the mucosa), increased serine protease activity (may be involved in the degradation of colonic tissue) and high levels of malondialdehyde (an indicator of lipid peroxidation). Both nitric oxide (NO) and O(2)(-) were increased with concomitant upregulation in the mRNA expression of proinflammatory cytokine response and inducible NO synthase (iNOS). Dose-response studies revealed that pretreatment of mice with thearubigin (40 mg kg(-1) day(-1), i.g. for 10 days) significantly ameliorated the appearance of diarrhoea and the disruption of colonic architecture. Higher dose (100 mg kg(-1)) had comparable effects. This was associated with a significant reduction in the degree of both neutrophil infiltration and lipid peroxidation in the inflamed colon as well as decreased serine protease activity. Thearubigin also reduced the levels of NO and O(2)(-) associated with the favourable expression of T-helper 1 cytokines and iNOS. Consistent with these observations, nuclear factor kappa B (NF-kappa B) activation in colonic mucosa was suppressed in thearubigin-treated mice. The results of this study suggest that thearubigin, the most predominant polyphenol of black tea, exerts beneficial effects in experimental colitis and may, therefore, be useful in the treatment of inflammatory bowel disease.


Subject(s)
Catechin/analogs & derivatives , Catechin/therapeutic use , Colitis/prevention & control , Flavonoids/therapeutic use , Phenols/therapeutic use , Tea , Trinitrobenzenesulfonic Acid/toxicity , Animals , Catechin/pharmacology , Colitis/chemically induced , Colitis/pathology , Dose-Response Relationship, Drug , Female , Flavonoids/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Phenols/pharmacology , Polyphenols
16.
Eur J Biochem ; 269(6): 1622-9, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11895432

ABSTRACT

In the course of trying to understand the pathogenesis of leishmaniasis in relation to extracellular matrix (ECM) elements, laminin, a major ECM protein, has been found to bind saturably and with high affinity to a 67-kDa cell surface protein of Leishmania donovani. This interaction involves a single class of binding sites, which are ionic in nature, conformation-dependent and possibly involves sulfhydryls. Binding activity was significantly enhanced by Zn2+, an effect possibly mediated through Cys-rich zinc finger-like sequences on laminin. Inhibition studies with monoclonals against polypeptide chains and specific peptides with adhesive properties revealed that the binding site was localized in one of the nested zinc finger consensus sequences of B1 chain containing the specific pentapeptide sequence, YIGSR. Furthermore, incubation of L. donovani promastigotes with C(YIGSR)3-NH2 peptide amide or antibody directed against the 67-kDa laminin-binding protein (LBP) induced tyrosine phosphorylation of proteins with a molecular mass ranging from 115 to 130 kDa. These studies suggest a role for LBP in the interaction of parasites with ECM elements, which may mediate one or more downstream signalling events necessary for establishment of infection.


Subject(s)
Laminin/metabolism , Leishmania donovani/metabolism , Membrane Proteins/metabolism , Zinc Fingers , Zinc/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Phosphorylation , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...