Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
BMC Cancer ; 18(1): 970, 2018 Oct 11.
Article in English | MEDLINE | ID: mdl-30309325

ABSTRACT

BACKGROUND: Prior small studies have shown increased expression of sperm protein 17 (Sp17) in epithelial ovarian cancer (EOC) tissue and suggest Sp17 as a potential biomarker for EOC. However, how Sp17 expression varies with histology, grade, and stage of EOC and its expression in other ovarian neoplasms has not been defined. It is unknown whether patients with EOC have elevated serum Sp17 levels or if Sp17 expression is associated with survival outcomes. METHODS: The study included 982 patients with benign, borderline, and malignant ovarian neoplasms and normal ovary. There were 878 patients with tissue only, 39 with serum only, and 65 with matching serum and tissue. Immunohistochemical (IHC) staining with anti-Sp17 antibody was performed on tissue specimens and the intensity scored as weak, moderate, or strong. A sandwich enzyme-linked immunosorbent assay (ELISA) was performed to measure Sp17 sera concentrations. RESULTS: Sp17 expression was most commonly seen in serous cystadenomas (83%) and serous borderline tumors (100%). Of the 773 EOC specimens, 223 (30%) expressed Sp17. Grade and histology were significantly associated with Sp17 expression among EOC specimens (p < 0.001) on both univariate and multivariable analysis, with grade 1 serous adenocarcinomas showing the highest expression (51%). Sp17 expression was limited in other benign and non-epithelial malignant neoplasms. Neither Sp17 tissue expression nor serum concentration correlated with survival outcomes. Serum concentrations were higher in patients with Sp17 tissue expression, and the highest concentrations were noted among patients with serous and clear cell adenocarcinomas. CONCLUSIONS: Sp17 is highly expressed in benign, borderline, and low grade malignant serous ovarian neoplasms and can be quantified in serum. Sp17 expression may have diagnostic significance in this subset of patients.


Subject(s)
Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial/metabolism , Carrier Proteins/metabolism , Cystadenoma, Serous/metabolism , Ovarian Neoplasms/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, Surface/blood , Biomarkers, Tumor/blood , Calmodulin-Binding Proteins , Carcinoma, Ovarian Epithelial/pathology , Carrier Proteins/blood , Cell Line, Tumor , Child , Cystadenoma, Serous/pathology , Female , Humans , Membrane Proteins , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/pathology , Prognosis , Retrospective Studies , Survival Analysis , Up-Regulation , Young Adult
2.
Papillomavirus Res ; 2: 21-30, 2016 12.
Article in English | MEDLINE | ID: mdl-27182559

ABSTRACT

Human papillomavirus type 16 (HPV16) infections are intra-epithelial, and thus, HPV16 is known to interact with Langerhans cells (LCs), the resident epithelial antigen-presenting cells (APCs). The current paradigm for APC-mediated induction of T cell anergy is through delivery of T cell receptor signals via peptides on MHC molecules (signal 1), but without costimulation (signal 2). We previously demonstrated that LCs exposed to HPV16 in vitro present HPV antigens to T cells without costimulation, but it remained uncertain if such T cells would remain ignorant, become anergic, or in the case of CD4+ T cells, differentiate into Tregs. Here we demonstrate that Tregs were not induced by LCs presenting only signal 1, and through a series of in vitro immunizations show that CD8+ T cells receiving signal 1 + 2 from LCs weeks after consistently receiving signal 1 are capable of robust effector functions. Importantly, this indicates that T cells are not tolerized but instead remain ignorant to HPV, and are activated given the proper signals.


Subject(s)
Antigen Presentation , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Human papillomavirus 16/immunology , Immune Tolerance , Langerhans Cells/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , CD8-Positive T-Lymphocytes/immunology , Cellular Senescence/immunology , Chemokines/immunology , Costimulatory and Inhibitory T-Cell Receptors/immunology , Cytokines/immunology , Humans , Interleukin-2/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology
3.
J Gen Virol ; 97(2): 422-434, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26555393

ABSTRACT

Herpes simplex virus (HSV) was originally implicated in the aetiology of cervical cancer, and although high-risk human papillomavirus (HPV) is now the accepted causative agent, the epidemiological link between HSV and HPV-associated cancers persists. The annexin A2 heterotetramer (A2t) has been shown to mediate infectious HPV type 16 (HPV16) uptake by human keratinocytes, and secretory leukocyte protease inhibitor (SLPI), an endogenous A2t ligand, inhibits HPV16 uptake and infection. Interestingly, HSV infection induces a sustained downregulation of SLPI in epithelial cells, which we hypothesized promotes HPV16 infection through A2t. Here, we show that in vitro infection of human keratinocytes with HSV-1 or HSV-2, but not with an HSV-1 ICP4 deletion mutant that does not downregulate SLPI, leads to a >70% reduction of SLPI mRNA and a >60% decrease in secreted SLPI protein. Consequently, we observed a significant increase in the uptake of HPV16 virus-like particles and gene transduction by HPV16 pseudovirions (two- and 2.5-fold, respectively) in HSV-1- and HSV-2-infected human keratinocyte cell cultures compared with uninfected cells, whereas exogenously added SLPI reversed this effect. Using a SiMPull (single-molecule pulldown) assay, we demonstrated that endogenously secreted SLPI interacts with A2t on epithelial cells in an autocrine/paracrine manner. These results suggested that ongoing HSV infection and resultant downregulation of local levels of SLPI may impart a greater susceptibility for keratinocytes to HPV16 infection through the host cell receptor A2t, providing a mechanism that may, in part, provide an explanation for the aetiological link between HSV and HPV-associated cancers.


Subject(s)
Host-Pathogen Interactions , Human papillomavirus 16/physiology , Keratinocytes/virology , Secretory Leukocyte Peptidase Inhibitor/metabolism , Simplexvirus/physiology , Virus Internalization , Cell Line , Down-Regulation , Humans
4.
Papillomavirus Res ; 1: 12-21, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26665182

ABSTRACT

Human papillomaviruses (HPV) establish persistent infections because of evolved immune evasion mechanisms, particularly HPV-mediated suppression of the immune functions of Langerhans cells (LC), the antigen presenting cells of the epithelium. Polyinosinic-polycytidilic acid (Poly-I:C) is broadly immunostimulatory with the ability to enhance APC expression of costimulatory molecules and inflammatory cytokines resulting in T cell activation. Here we investigated the activation of primary human LC derived from peripheral blood monocytes after exposure to HPV16 virus like particles followed by treatment with stabilized Poly-I:C compounds (s-Poly-I:C), and their subsequent induction of HPV16-specific T cells. Our results indicate that HPV16 particles alone were incapable of inducing LC activation as demonstrated by the lack of costimulatory molecules, inflammatory cytokines, chemokine-directed migration, and HPV16-specific CD8+ T cells in vitro. Conversely, s-Poly-I:C caused significant upregulation of costimulatory molecules and induction of chemokine-directed migration of LC that were pre-exposed to HPV16. In HLA-A*0201-positive donors, s-Poly-I:C treatment was able to induce CD8+ T cell immune responses against HPV16-derived peptides. Thus, s-Poly-I:C compounds are attractive for translation into therapeutics in which they could potentially mediate clearance of persistent HPV infection.

5.
Clin Immunol ; 161(2): 197-208, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26360252

ABSTRACT

Human papillomavirus (HPV)-mediated suppression of Langerhans cell (LC) function can lead to persistent infection and development of cervical intraepithelial neoplasia (CIN). Women with HPV-induced high-grade CIN2/3 have not mounted an effective immune response against HPV, yet it is unknown if LC-mediated T cell activation from such women is functionally impaired against HPV. We investigated the functional activation of in vitro generated LC and their ability to induce HPV16-specific T cells from CIN2/3 patients after exposure to HPV16 followed by treatment with stabilized Poly-I:C (s-Poly-I:C). LC from patients exposed to HPV16 demonstrated a lack of costimulatory molecule expression, inflammatory cytokine secretion, and chemokine-directed migration. Conversely, s-Poly-I:C caused significant phenotypic and functional activation of HPV16-exposed LC, which resulted in de novo generation of HPV16-specific CD8(+) T cells. Our results highlight that LC of women with a history of persistent HPV infection can present HPV antigens and are capable of inducing an adaptive T cell immune response when given the proper stimulus, suggesting that s-Poly-I:C compounds may be attractive immunomodulators for LC-mediated clearance of persistent HPV infection.


Subject(s)
Human papillomavirus 16/immunology , Langerhans Cells/immunology , Lymphocyte Activation/immunology , Papillomavirus Infections/immunology , Poly I-C/immunology , Uterine Cervical Dysplasia/immunology , Uterine Cervical Dysplasia/virology , Adult , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , DNA, Viral/immunology , Female , Humans , Langerhans Cells/virology , Middle Aged , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology
6.
J Antimicrob Chemother ; 70(6): 1686-90, 2015.
Article in English | MEDLINE | ID: mdl-25712315

ABSTRACT

OBJECTIVES: High-risk human papillomavirus (HPV) infection leads to the development of several human cancers that cause significant morbidity and mortality worldwide. HPV type 16 (HPV16) is the most common of the cancer-causing genotypes and gains entry to the basal cells of the epithelium through a non-canonical endocytic pathway that involves the annexin A2/S100A10 heterotetramer (A2t). A2t is composed of two annexin A2 monomers bound to an S100A10 dimer and this interaction is a potential target to block HPV16 infection. Here, recently identified small molecule inhibitors of A2t (A2ti) were investigated for their ability to prevent HPV16 infection in vitro. METHODS: A2ti were added to HeLa cells in increasing concentrations prior to the addition of HPV16. Cytotoxicity was evaluated via trypan blue exclusion. HPV16 pseudovirion infection and fluorescently labelled HPV16 capsid internalization was measured with flow cytometry. RESULTS: A2ti blocked HPV16 infection by 100% without substantial cellular toxicity or reduction in cell growth. Furthermore, A2ti blocked HPV16 entry into epithelial cells by 65%, indicating that the observed inhibition of HPV16 infection is in part due to a block in entry and that non-infectious entry may occur in the absence of A2t binding. CONCLUSIONS: These results demonstrate that targeting A2t may be an effective strategy to prevent HPV16 infection.


Subject(s)
Annexin A2/antagonists & inhibitors , Antiviral Agents/pharmacology , Endocytosis/drug effects , Human papillomavirus 16/physiology , Virus Internalization/drug effects , Cell Survival/drug effects , HeLa Cells , Humans
7.
Prostate ; 75(3): 280-91, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25399517

ABSTRACT

BACKGROUND: LIGHT, a ligand for lymphotoxin-ß receptor (LTßR) and herpes virus entry mediator, is predominantly expressed on activated immune cells and LTßR signaling leads to the recruitment of lymphocytes. The interaction between LIGHT and LTßR has been previously shown to activate immune cells and result in tumor regression in a virally-induced tumor model, but the role of LIGHT in tumor immunosuppression or in a prostate cancer setting, where self antigens exist, has not been explored. We hypothesized that forced expression of LIGHT in prostate tumors would shift the pattern of immune cell infiltration toward an anti-tumoral milieu, would inhibit T regulatory cells (Tregs) and would induce prostate cancer tumor associated antigen (TAA) specific T cells that would eradicate tumors. METHODS: Real Time PCR was used to evaluate expression of forced LIGHT and other immunoregulatory genes in prostate tumors samples. For in vivo studies, adenovirus encoding murine LIGHT was injected intratumorally into TRAMP-C2 prostate cancer cell tumor bearing mice. Chemokine and cytokine concentrations were determined by multiplex ELISA. Flow cytometry was used to phenotype tumor infiltrating lymphocytes and expression of LIGHT on the tumor cell surface. Tumor-specific lymphocytes were quantified via ELISpot assay. Treg induction and Treg suppression assays determined Treg functionality after LIGHT treatment. RESULTS: LIGHT in combination with a therapeutic vaccine, PSCA TriVax, reduced tumor burden. LIGHT expression peaked within 48 hr of infection, recruited effector T cells that recognized mouse prostate stem cell antigen (PSCA) into the tumor microenvironment, and inhibited infiltration of Tregs. Tregs isolated from tumor draining lymph nodes had impaired suppressive capability after LIGHT treatment. CONCLUSION: Forced LIGHT treatment combined with PSCA TriVax therapeutic vaccination delays prostate cancer progression in mice by recruiting effector T lymphocytes to the tumor and inhibiting Treg mediated immunosuppression. Prostate 75:280-291, 2015. © 2014 Wiley Periodicals, Inc.


Subject(s)
Cancer Vaccines/immunology , Prostate/metabolism , Prostatic Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Immune Tolerance , Immunosuppression Therapy , Male , Mice , Prostate/immunology , Prostate/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
8.
Virology ; 452-453: 279-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24606705

ABSTRACT

Human papillomavirus (HPV) has evolved mechanisms that allow it to evade the human immune system. Studies have shown HPV-mediated suppression of activation of Langerhans cells (LC) is a key mechanism through which HPV16 evades initial immune surveillance. However, it has not been established whether high- and low-risk mucosal and cutaneous HPV genotypes share a common mechanism of immune suppression. Here, we demonstrate that LC exposed to capsids of HPV types 18, 31, 45, 11, (alpha-papillomaviruses) and HPV5 (beta-papillomavirus) similarly suppress LC activation, including lack of costimulatory molecule expression, lack of cytokine and chemokine secretion, lack of migration, and deregulated cellular signaling. In contrast, HPV1 (mu-papillomavirus) induced costimulatory molecule and cytokine upregulation, but LC migration and cellular signaling was suppressed. These results suggest that alpha and beta HPV genotypes, and partially a mu genotype, share a conserved mechanism of immune escape that enables these viruses to remain undetected in the absence of other inflammatory events.


Subject(s)
Down-Regulation , Langerhans Cells/immunology , Papillomaviridae/genetics , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Biological Evolution , Cytokines/immunology , Genotype , Humans , Langerhans Cells/virology , Papillomaviridae/classification , Papillomavirus Infections/virology
9.
Recife; s.n; 2014. 183 p. ilus, tab.
Thesis in Portuguese | LILACS | ID: lil-719864

ABSTRACT

A leishmaniose visceral canina (LVC) e uma doença parasitária causada por protozoários do gênero Leishmania, principalmente por Leishmania infantum. A epidemiologia da doença varia de região para região e o entendimento dos fatores associados à infecção em cães pode ajudar na elaboração de medidas de controle mais específicas. O diagnóstico sorológico da infecção sofreu mudanças importantes nos últimos anos com a introdução do TR-DPP® e do estabelecimento de novos critérios de diagnóstico (TR-DPP® + EIE-LVC) pelo Ministério da Saúde. Dentro desse contexto, no presente estudo objetivou-se estudar a epidemiologia da LVC no município de Goiana, estado de Pernambuco, nordeste do Brasil. Para tal, realizaram-se testes sorológicos (TR-DPP® e EIE-LVC) e análise clínico-epidemiológica em 360 cães semi e domiciliados, de ambos os sexos, raças e idades variadas, nos distritos de Atapuz, Tejucupapo e Pontas de Pedra no referido município. No TR-DPP®47 (13,1 por cento) animais foram reagentes, onde se observou associação significativa dos resultados com os seguintes sinais clínicos: alopecia, lesões na pele paresia e linfonodomegalia. Já no EIE-LVC 21 (5,8 por cento) animais foram reagentes, havendo associação significativa entre a classificação clínica dos animais, condição corporal, alopecia, lesões na pele, secreção ocular, paresia e linfonodomegalia. Já de acordo com o critério do Ministério da Saúde do Brasil, apenas 15 (4,2 por cento) animais foram classificados como positivos. De fato, verificou-se uma fraca concordância (Kappa = 0,39) entre os dois testes sorológicos. Conclui-se que a LVC encontra-se estabelecida em Goiana e que o uso do TR-DPP® como teste de triagem e do EIE-LVC como teste confirmatório pode levar a perda de cães infectados, uma vez que cães positivos do TR-DPP® são negativos no EIE-LVC e vice-versa.


Subject(s)
Antigens, Viral/isolation & purification , Antigens, Viral , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Hepacivirus , HIV , Human T-lymphotropic virus 1 , Microspheres , Vaccines, Synthetic , Donor Selection , Sensitivity and Specificity , Serologic Tests
10.
PLoS One ; 7(8): e43519, 2012.
Article in English | MEDLINE | ID: mdl-22927980

ABSTRACT

Mucosotropic, high-risk human papillomaviruses (HPV) are sexually transmitted viruses that are causally associated with the development of cervical cancer. The most common high-risk genotype, HPV16, is an obligatory intracellular virus that must gain entry into host epithelial cells and deliver its double stranded DNA to the nucleus. HPV capsid proteins play a vital role in these steps. Despite the critical nature of these capsid protein-host cell interactions, the precise cellular components necessary for HPV16 infection of epithelial cells remains unknown. Several neutralizing epitopes have been identified for the HPV16 L2 minor capsid protein that can inhibit infection after initial attachment of the virus to the cell surface, which suggests an L2-specific secondary receptor or cofactor is required for infection, but so far no specific L2-receptor has been identified. Here, we demonstrate that the annexin A2 heterotetramer (A2t) contributes to HPV16 infection and co-immunoprecipitates with HPV16 particles on the surface of epithelial cells in an L2-dependent manner. Inhibiting A2t with an endogenous annexin A2 ligand, secretory leukocyte protease inhibitor (SLPI), or with an annexin A2 antibody significantly reduces HPV16 infection. With electron paramagnetic resonance, we demonstrate that a previously identified neutralizing epitope of L2 (aa 108-120) specifically interacts with the S100A10 subunit of A2t. Additionally, mutation of this L2 region significantly reduces binding to A2t and HPV16 pseudovirus infection. Furthermore, downregulation of A2t with shRNA significantly decreases capsid internalization and infection by HPV16. Taken together, these findings indicate that A2t contributes to HPV16 internalization and infection of epithelial cells and this interaction is dependent on the presence of the L2 minor capsid protein.


Subject(s)
Annexin A2/chemistry , Annexin A2/metabolism , Capsid Proteins/metabolism , Human papillomavirus 16/physiology , Oncogene Proteins, Viral/metabolism , Protein Multimerization , S100 Proteins/chemistry , S100 Proteins/metabolism , Amino Acid Sequence , Annexin A2/genetics , Annexin A2/immunology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Epithelial Cells/virology , Epitopes/chemistry , Epitopes/immunology , Gene Knockdown Techniques , HeLa Cells , Human papillomavirus 16/metabolism , Humans , Molecular Sequence Data , Mutation , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Protein Structure, Quaternary , RNA, Small Interfering/genetics , Receptors, Cell Surface , S100 Proteins/genetics , S100 Proteins/immunology , Substrate Specificity
11.
Rev. bras. hematol. hemoter ; 31(5): 375-383, 2009. ilus, tab
Article in Portuguese | LILACS | ID: lil-533583

ABSTRACT

A leucemia de células T no adulto (ATL) é causada pelo vírus linfotrópico de células T (HTLV-1). Contudo, apenas 2 por cento-5 por cento dos indivíduos infectados desenvolvem a ATL e somente 40-60 anos após a infecção. Um fator de risco para adquirir a doença é a via da transmissão do vírus pela amamentação e durante o parto, sugerindo que a criança já é portadora do vírus. Desde a descoberta do vírus, em 1980, vários artigos científicos foram publicados descrevendo as manifestações clínicas, biologia do vírus e alterações intracelulares induzidas pelo vírus. Esta revisão visa explorar alguns aspectos da relação entre HTLV-1 e a leucemia de células T do Adulto.


The human T-lymphotropic virus (HTLV-1) is known to be the etiologic agent of adult T-cell leukemia (ATL). Only 2-5 percent of infected individuals develop ATL and even then only 40-60 years after infection. One risk factor to develop ATL is the transmission of the virus by breastfeeding and during delivery, suggesting that infants of infected mothers are already carriers of the virus. Since the discovery of the virus in 1980 many scientific papers have been published describing the clinical manifestations, biology of the virus and the intracellular alterations induced by the virus. This review aims to explore some aspects of the relationship between HTLV-1 and ATL.


Subject(s)
Humans , Leukemia, T-Cell , Human T-lymphotropic virus 1
12.
Cancer Res ; 68(1): 106-14, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18172302

ABSTRACT

The focus of this study is on the expression and regulation of the estrogen-regulated breast cancer and salivary gland expression (BASE) gene that may function as a breast cancer marker. In MCF7 cells, BASE is repressed by estrogen in an estrogen receptor alpha (ER alpha)-dependent manner. Promoter analysis of the BASE gene led to the identification of a 2-kb upstream enhancer that harbors binding sites for ER alpha and FoxA1. The recruitment of both ER alpha and FoxA1 to this region was shown by chromatin immunoprecipitation analysis. Furthermore, mutation studies and knockdown experiments show a clear separation between gene expression mediated by FoxA1 and ER alpha-dependent gene regulation. Additionally, we provide information on BASE expression in human breast tumor samples.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Binding Sites , Cell Line, Tumor , Chromatin Immunoprecipitation , Down-Regulation , Enhancer Elements, Genetic , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Mutation , Promoter Regions, Genetic
13.
Mol Endocrinol ; 22(2): 361-79, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17962383

ABSTRACT

Estradiol (E2) is believed to modulate physiological functions relevant to osteoblast biology through the actions of estrogen receptors (ERs) that in turn regulate the expression of target genes. The molecular effects of estrogen action in bone remain to be fully elucidated. This study reports a genome-wide molecular and computational analysis of the interaction between ER and regulatory elements on the DNA of target genes in human primary osteoblasts. Of approximately 54,000 gene probes surveyed in this study, a total of 375 genes were up-regulated and 418 genes were down-regulated on exposure to E2, with only 46 of these being direct target genes after 24 h, as determined by concomitant cycloheximide treatment. Computational analysis discovered several pathways where E2 co-regulates multiple functionally linked components. Examination of the genomic sequence of IGF binding protein 4 located ER response elements within the first intron. Using by chromatin immunoprecipitation, we show a site- and cell-specific recruitment of transcription factors to this newly identified regulatory region. Transient transfection studies revealed that this intronic region acts as a functional promoter in human osteoblasts. Taken together, this analysis provides a comprehensive gene transcription profile and identifies several genes of potential physiological importance in controlling estrogen-mediated signaling in primary osteoblasts.


Subject(s)
Estrogens/pharmacology , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Insulin-Like Growth Factor Binding Protein 4/genetics , Osteoblasts/drug effects , Binding Sites , Cells, Cultured , Chromatin Immunoprecipitation , Cycloheximide/pharmacology , Estradiol/pharmacology , Humans , Insulin-Like Growth Factor Binding Protein 4/metabolism , Introns/genetics , Models, Biological , Oligonucleotide Array Sequence Analysis , Osteoblasts/cytology , Osteoblasts/metabolism , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects , Transfection
14.
Oncogene ; 24(31): 4894-907, 2005 Jul 21.
Article in English | MEDLINE | ID: mdl-15870696

ABSTRACT

Valproate (VPA) and trichostatin A (TSA), inhibitors of zinc-dependent deacetylase activity, induce reduction in the levels of mRNA encoding oestrogen receptor-alpha (ERalpha), resulting in subsequent clearance of ERalpha protein from breast and ovarian cell lines. Inhibition of oestrogen signalling may account for the endocrine disorders, menstrual abnormalities, osteoporosis and weight gain that occur in a proportion of women treated with VPA for epilepsy or for bipolar mood disorder. Transcriptome profiling revealed that VPA and TSA also modulate the expression of, among others, key regulatory components of the cell cycle. Meta-analysis of genes directly responsive to oestrogen indicates that VPA and TSA have a generally antioestrogenic profile in ERalpha positive cells. Concomitant treatment with cycloheximide prevented most of these changes in gene expression, including downregulation of ERalpha mRNA, indicating that a limited number of genes signal a hyperacetylated state within cells. Three members of the NAD-dependent deacetylases, the sirtuins, are upregulated by VPA and by TSA and sirtuin activity contributes to loss of ERalpha expression. However, prolonged inhibition of the sirtuins by sirtinol also induces loss of ERalpha from cells. Mechanistically, we show that VPA invokes reversible promoter shutoff of the ERalpha, pS2 and cyclin D1 promoters, by inducing recruitment of methyl cytosine binding protein 2 (MeCP2) with concomitant exclusion of the maintenance methylase DNMT1. Furthermore, we demonstrate that, in the presence of VPA, local DNA methylation, deacetylation and demethylation of activated histones and recruitment of inhibitory complexes occurs on the pS2 promoter.


Subject(s)
Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Transcription, Genetic/drug effects , Valproic Acid/pharmacology , Base Sequence , Breast Neoplasms , Cell Line, Tumor , DNA Primers , Enzyme Inhibitors/pharmacology , Female , Gene Silencing , Genetic Markers , Humans , Kinetics , Polymerase Chain Reaction , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
15.
EMBO J ; 23(18): 3653-66, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15343269

ABSTRACT

Unliganded (apo-) estrogen receptor alpha (ERalpha, NR3A1) is classically considered as transcriptionally unproductive. Reassessing this paradigm demonstrated that apo-human ERalpha (ERalpha66) and its N-terminally truncated isoform (ERalpha46) are both predominantly nuclear transcription factors that cycle on the endogenous estrogen-responsive pS2 gene promoter in vivo. Importantly, isoform-specific consequences occur in terms of poising the promoter for transcription, as evaluated by determining (i) the engagement of several cofactors and the resulting nucleosomal organization; and (ii) the CpG methylation state of the pS2 promoter. Although transcriptionally unproductive, cycling of apo-ERalpha66 prepares the promoter to respond to ligand, through sequentially targeting chromatin remodeling complexes and general transcription factors. Additionally, apo-ERalpha46 recruits corepressors, following engagement of cofactors identical to those recruited by apo-ERalpha66. Together, these data describe differential activities of ERalpha isoforms. Furthermore, they depict the maintenance of a promoter in a repressed state as a cyclical process that is intrinsically dependent on initial poising of the promoter.


Subject(s)
Apoproteins/genetics , Estrogen Receptor alpha/genetics , Membrane Proteins/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Apoproteins/metabolism , Biological Clocks , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chromatin/metabolism , Chromatin Immunoprecipitation , CpG Islands , DNA Methylation , Epigenesis, Genetic/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Growth Substances/metabolism , Humans , Ligands , Membrane Proteins/metabolism , Nucleosomes/metabolism , Presenilin-2 , Protein Isoforms , Transcription Factors/metabolism , Transcriptional Activation
16.
Cell ; 115(6): 751-63, 2003 Dec 12.
Article in English | MEDLINE | ID: mdl-14675539

ABSTRACT

Transcriptional activation of a gene involves an orchestrated recruitment of components of the basal transcription machinery and intermediate factors, concomitant with an alteration in local chromatin structure generated by posttranslational modifications of histone tails and nucleosome remodeling. We provide here a comprehensive picture of events resulting in transcriptional activation of a gene, through evaluating the estrogen receptor-alpha (NR3A1) target pS2 gene promoter in MCF-7 cells. This description integrates chromatin remodeling with a kinetic evaluation of cyclical networks of association of 46 transcription factors with the promoter, as determined by chromatin immunoprecipitation assays. We define the concept of a "transcriptional clock" that directs and achieves the sequential and combinatorial assembly of a transcriptionally productive complex on a promoter. Furthermore, the unanticipated findings of key roles for histone deacetylases and nucleosome-remodeling complexes in limiting transcription implies that transcriptional activation is a cyclical process that requires both activating and repressive epigenetic processes.


Subject(s)
Promoter Regions, Genetic/genetics , Proteins/metabolism , Receptors, Estrogen/metabolism , Transcriptional Activation/genetics , Biological Clocks/genetics , Cell Line, Tumor , Epigenesis, Genetic/genetics , Estrogen Receptor alpha , Estrogens/metabolism , Histone Deacetylases/genetics , Humans , Nucleosomes/genetics , Proteins/genetics , Receptors, Estrogen/genetics , Trefoil Factor-1 , Tumor Suppressor Proteins
17.
Mol Cell ; 11(3): 695-707, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12667452

ABSTRACT

We present an integrated model of hERalpha-mediated transcription where both unliganded and liganded receptors cycle on estrogen-responsive promoters. Using ChIP, FRAP, and biochemical analysis we evaluate hERalpha at several points in these cycles, establishing the ubiquitination status and subnuclear distribution of hERalpha, its mobility, the kinetics of transcriptional activation, and the cyclic recruitment of E3 ligases and the 19S regulatory component of the proteasome. These experiments, together with an evaluation of the inhibition of transcription and proteasome action, demonstrate that proteasome-mediated degradation and hERalpha-mediated transactivation are inherently linked and act to continuously turn over hERalpha on responsive promoters. Cyclic turnover of hERalpha permits continuous responses to changes in the concentration of estradiol.


Subject(s)
Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Signal Transduction , Blotting, Western , Cell Nucleus/metabolism , Chromatin/metabolism , Cysteine Endopeptidases/metabolism , Dose-Response Relationship, Drug , Estradiol/metabolism , Estrogen Receptor alpha , Estrogens/metabolism , Humans , Kinetics , Ligands , Models, Biological , Multienzyme Complexes/metabolism , Oligonucleotides/pharmacology , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Proteasome Endopeptidase Complex , Protein Binding , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
18.
Mol Cell ; 10(5): 1019-32, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12453411

ABSTRACT

The functional interplay between different domains of estrogen receptor-alpha (ERalpha, NR3A1) is responsible for the overall properties of the full-length protein. We previously identified an interaction between the N-terminal A and C-terminal domains, which we demonstrate here to repress ligand-independent transactivation and transrepression abilities of ERalpha. Using targeted mutations based on ERalpha structural models, we determine the basis for this interaction that defines a regulatory interplay between ERalpha A domain, corepressors, and ERalpha Helix 12 for binding to the same C-terminal surface. We propose a dynamic model where binding of different ligands influences the A/D-F domain interaction and results in specific functional outcomes. This model gives insights into the dynamic properties of full-length ERalpha and into the structure of unliganded ERalpha.


Subject(s)
Receptors, Estrogen/metabolism , Amino Acid Sequence , Estrogen Receptor alpha , Gene Silencing , Genes, Reporter , Glutathione Transferase/metabolism , HeLa Cells , Humans , Ligands , Models, Genetic , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Biosynthesis , Protein Conformation , Protein Isoforms , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Estrogen/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Software , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured , beta-Galactosidase/metabolism
19.
Endocrinology ; 143(9): 3397-404, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12193552

ABSTRACT

The role of estrogens in the development and physiology of the male reproductive tract remains provocative, with a growing body of evidence suggesting that estrogens are able to influence normal testis development and physiology, through their classical receptors, estrogen receptor (ER)-alpha and ER-beta. We describe the identification and characterization of a new promoter that is involved in the expression of ER-alpha in the epididymis and in testis. This promoter lies on chromosome 6q25.1, approximately 16 kb upstream of the first coding exon of ER-alpha. Sequence analysis indicates that this promoter has a conventional TATA box and GC box but no upstream CAAT sequence. Alternative splicing results in at least two species of mRNA encoding ER-alpha being synthesized from this promoter. Transcription profiling of human tissues shows that, among those tested, this promoter is predominantly active only in testis and epididymal tissues. Transient transfection assays using this new promoter in a number of cell lines indicate that the region we have identified functions as a promoter and that tissue-specific regulation is likely to be dependent on inhibitory sequences greater than 1 kb upstream of the transcription start site.


Subject(s)
Epididymis/metabolism , Gene Expression , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Testis/metabolism , Aged , Alternative Splicing , Base Sequence , Cells, Cultured , Chromosome Mapping , Chromosomes, Human, Pair 6 , Estrogen Receptor alpha , Humans , Male , Molecular Sequence Data , RNA , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology , TATA Box , Transfection
20.
J Biol Chem ; 277(29): 26244-51, 2002 Jul 19.
Article in English | MEDLINE | ID: mdl-12011094

ABSTRACT

The human estrogen receptor-alpha (hER alpha) gene is a complex genomic unit exhibiting alternative splicing and promoter usage in a tissue-specific manner. During the investigation of new hER alpha mRNA variants by rapid amplification of 5' cDNA ends, we identified a cDNA in which the acceptor site of exon 1A, into which the different leader exons are normally alternatively spliced, was spliced accurately the 3' extremity of exon 1A (scrambled 1A-->1A hER alpha cDNA). Reverse transcription-PCR and S1 nuclease mapping analysis revealed that 1A-->1A hER alpha transcripts were not circular RNAs constituted by exon 1A only but corresponded to linear polyadenylated hER alpha RNAs composed of the eight coding exons of the hER alpha gene and characterized by a duplication of exon 1A. Genomic Southern blot experiments excluded the hypothesis of duplication of hER alpha exon 1A in the human genome. Therefore, these data suggested that 1A-->1A hER alpha transcripts were likely generated by trans-splicing. The production of such transcripts by trans-splicing of pre-mRNAs generated from a chimeric gene formed by a single hER alpha exon 1A, exon 2, and their flanking intronic regions was demonstrated in transient transfection experiments. Therefore, in addition to the alternative cis-splicing, the hER alpha gene is also subject to natural trans-splicing.


Subject(s)
RNA, Messenger/genetics , Receptors, Estrogen/genetics , Trans-Splicing , Alternative Splicing , Blotting, Southern , Cell Line , Estrogen Receptor alpha , Female , Humans , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...