Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Toxicol Appl Pharmacol ; 255(1): 65-75, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21683088

RESUMO

Exposure during early development to chemicals with hormonal action may be associated with weight gain during adulthood because of altered body homeostasis. It is known that organotins affect adipose mass when exposure occurs during fetal development, although no knowledge of effects are available for exposures after birth. Here we show that the environmental organotin tributyltin chloride (TBT) exerts adipogenic action when peripubertal and sexually mature mice are exposed to the chemical. The duration and extent of these effects depend on the sex and on the dose of the compound, and the effects are relevant at doses close to the estimated human intake (0.5µg/kg). At higher doses (50-500µg/kg), TBT also activated estrogen receptors (ERs) in adipose cells in vitro and in vivo, based on results from acute and longitudinal studies in ERE/luciferase reporter mice. In 3T3-L1 cells (which have no ERs), transiently transfected with the ERE-dependent reporter plus or minus ERα or ERß, TBT (in a dose range of 1-100nM) directly targets each ER subtype in a receptor-specific manner through a direct mechanism mediated by ERα in undifferentiated preadipocytic cells and by ERß in differentiating adipocytes. The ER antagonist ICI-182,780 inhibits this effect. In summary, the results of this work suggest that TBT is adipogenic at all ages and in both sexes and that it might be an ER activator in fat cells. These findings might help to resolve the apparent paradox of an adipogenic chemical being also an estrogen receptor activator by showing that the two apparently opposite actions are separated by the different doses to which the organism is exposed.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Receptores de Estrogênio/efeitos dos fármacos , Compostos de Trialquitina/toxicidade , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Animais , Dietilestilbestrol/farmacologia , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/fisiologia
2.
J Control Release ; 294: 165-175, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30557650

RESUMO

Extracellular vesicles (EVs), are naturally occurring cargo delivery tools with the potential to be used as drug vehicles of single agents or combination therapies. We previously demonstrated that human lung cancer cell-derived EVs could be used for the systemic delivery of oncolytic virus (OVs) and chemotherapy drugs such as paclitaxel (PTX), leading to enhanced anti-tumor effects in nude mice. In the current work, we evaluated the biodistribution of EVs by using bioluminescence and fluorescence imaging technologies, thus proving the ability of these EVs-formulations to specifically target the neoplasia, while leaving other body tissues unaffected. Moreover, in vivo imaging of NFκB activation in an immunocompetent reporter mouse model allowed to demonstrate the selective ability of EVs to induce tumor-associated inflammatory reactions, which are characterized by immunogenic cell death and CD3+/CD4+/CD8+ T-cell infiltration. While EVs have the potential to induce a systemic immune reaction by pro-inflammatory cytokines, our study provides compelling evidences of a localized inflammatory effect in the peritumoral area. Collectively, our findings strongly support the systemic administration of EVs formulations with OVs alone or in combination with chemotherapy agents as a novel strategy aimed at treating primary and metastatic cancers.


Assuntos
Adenoviridae , Antineoplásicos/administração & dosagem , Vesículas Extracelulares , Vírus Oncolíticos , Paclitaxel/administração & dosagem , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Terapia Combinada , Linfócitos do Interstício Tumoral/imunologia , Masculino , Camundongos Transgênicos , Neoplasias/imunologia , Neoplasias/terapia , Distribuição Tecidual
3.
J Neuroendocrinol ; 30(2)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28650102

RESUMO

Most of the ageing-associated pathologies are coupled with a strong inflammatory component that accelerates the progress of the physiopathological functional decline related to ageing. The currently available pharmacological tools for the control of neuroinflammation present several side effects that restrict their application, particularly in chronic disorders. The discovery of the potential anti-inflammatory action exerted by endogenous oestrogens, as well as the finding that activation of oestrogen receptor α results in a significant decrease of inflammation at the cellular level and in models of inflammatory diseases, prompted us to embark in a series of studies aimed at the generation of reporter systems, allowing us to (i) understand the anti-inflammatory action of oestrogens at molecular level; (ii) evaluate the extent to which the action of this steroid hormone was relevant in models of pathologies characterised by a strong inflammatory component; and (iii) investigate the efficacy of novel, synthetic oestrogens endowed with anti-inflammatory activity. Accordingly, we conceived the NFκB-luc2 reporter mouse, a model characterised by dual reporter genes for fluorescence and bioluminescence imaging under the control of a synthetic DNA able to bind the transcription factor nuclear factor kappa B, the master regulator of the expression of most of the cytokines responsible for the initial phase of acute inflammation. Here, we summarise the philosophy that has driven our research in the past years, as well as some of the results obtained so far.


Assuntos
Encéfalo/metabolismo , Encefalite/metabolismo , Animais , Citocinas/metabolismo , Luciferases/metabolismo , Medições Luminescentes , Camundongos , NF-kappa B/metabolismo
4.
J Control Release ; 283: 223-234, 2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-29864473

RESUMO

Standard of care for cancer is commonly a combination of surgery with radiotherapy or chemoradiotherapy. However, in some advanced cancer patients this approach might still remaininefficient and may cause many side effects, including severe complications and even death. Oncolytic viruses exhibit different anti-cancer mechanisms compared with conventional therapies, allowing the possibility for improved effect in cancer therapy. Chemotherapeutics combined with oncolytic viruses exhibit stronger cytotoxic responses and oncolysis. Here, we have investigated the systemic delivery of the oncolytic adenovirus and paclitaxel encapsulated in extracellular vesicles (EV) formulation that, in vitro, significantly increased the transduction ratio and the infectious titer when compared with the virus and paclitaxel alone. We demonstrated that the obtained EV formulation reduced the in vivo tumor growth in animal xenograft model of human lung cancer. Indeed, we found that combined treatment of oncolytic adenovirus and paclitaxel encapsulated in EV has enhanced anticancer effects both in vitro and in vivo in lung cancer models. Transcriptomic comparison carried out on the explanted xenografts from the different treatment groups revealed that only 5.3% of the differentially expressed genes were overlapping indicating that a de novo genetic program is triggered by the presence of the encapsulated paclitaxel: this novel genetic program might be responsible of the observed enhanced antitumor effect. Our work provides a promising approach combining anticancer drugs and viral therapies by intravenous EV delivery as a strategy for the lung cancer treatment.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Vesículas Extracelulares , Neoplasias Pulmonares/terapia , Vírus Oncolíticos , Paclitaxel/administração & dosagem , Animais , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Baço/efeitos dos fármacos , Baço/patologia
5.
Mol Cell Endocrinol ; 246(1-2): 69-75, 2006 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-16388894

RESUMO

Non-invasive imaging of reporter gene expression using different imaging modalities is increasing its role for the in vivo assessment of molecular processes. Multimodality imaging protocols overcome limitations to a single imaging modality and provide a thorough view of specific processes, often allowing a quantitative measurement and direct visualization of the process in a specific target organ or tissue. The use of the right reporter gene for the development of animal models and the characterization of its expression in different conditions and tissues is fundamental for basic, translational and future pharmacological applications of a given model. This paper summarizes the major steps in the development and evaluation of a specific animal model for in vivo molecular imaging studies and describes the first example of an animal model designed for the in vivo assessment of a specific receptor activity and its possible evolution towards multimodality imaging analysis.


Assuntos
Diagnóstico por Imagem/métodos , Diagnóstico por Imagem/tendências , Modelos Animais , Biologia Molecular/métodos , Biologia Molecular/tendências , Animais , Genes Reporter/genética , Vetores Genéticos , Camundongos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo
6.
Maturitas ; 54(4): 315-20, 2006 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-16753274

RESUMO

In the latest few years, the merging of imaging and animal engineering technologies has led to the generation of innovative tools that provide the opportunity to look into the dynamics of specific molecular events in living animals during their entire life under a completely renewed perspective. These tools will have a profound impact not only on basic research, but also on drug discovery and development allowing to depict the activity of any therapeutic agents in all their designed targets as well as in the organs where they may cause undesired effects. Along this research line, our laboratory has recently described the first animal model reporting the state of activity of estrogen receptors (ERs) in real time: the ERE-luc reporter mouse. The application of optical imaging to the ERE-luc has allowed an unprecedented in depth view of estrogen signaling in all of its target tissues. For example, the analysis of the state of activity of ERs in the physiological setting of the estrous cycle has provided compelling evidence that hormone-independent mechanisms are responsible for activating ERs in non-reproductive organs. This discovery may pave the way to a rational basis for the development of novel, more selective and effective treatments for menopause.


Assuntos
Receptores de Estrogênio/metabolismo , Animais , Feminino , Genes Reporter , Luciferases/metabolismo , Camundongos , Modelos Animais
7.
Oncogene ; 20(7): 775-87, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11314012

RESUMO

v-ErbA is a mutated variant of thyroid hormone receptor (TRalpha/NR1A1) borne by the Avian Erythroblastosis virus causing erythroleukemia. TRalpha is known to activate transcription of specific genes in the presence of its cognate ligand, T3 hormone, while in its absence it represses it. v-ErbA is unable to bind ligand, and hence is thought to contribute to leukemogenesis by actively repressing erythroid-specific genes such as the carbonic anhydrase II gene (CA II). In the prevailing model, v-ErbA occludes liganded TR from binding to its cognate elements and constitutively interacts with the corepressors NCoR/SMRT. We previously identified a v-ErbA responsive element (VRE) within a DNase I hypersensitive region (HS2) located in the second intron of the CA II gene. We now show that HS2 fulfils all the requirements for a genuine enhancer that functions independent of its orientation and position with a profound erythroid-specific activity in normal erythroid progenitors (T2ECs) and in leukemic erythroid cell lines. We find that the HS2 enhancer activity is governed by two adjacent GATA-factor binding sites. v-ErbA as well as unliganded TR prevent HS2 activity by nullifying the positive function of factors bound to GATA-sites. However, v-ErbA, in contrast to TR, does not convey active repression to silence the transcriptional activity intrinsic to a heterologous tk promoter. We propose that depending on the sequence and context of the binding site, v-ErbA contributes to leukemogenesis by occluding liganded TR as well as unliganded TR thereby preventing activation or repression, respectively.


Assuntos
Anidrases Carbônicas/genética , Elementos Facilitadores Genéticos , Leucemia Eritroblástica Aguda/genética , Proteínas Oncogênicas v-erbA/genética , Receptores dos Hormônios Tireóideos/genética , Animais , Sequência de Bases , Sítios de Ligação , Galinhas , Células Precursoras Eritroides , Regulação Leucêmica da Expressão Gênica , Humanos , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Ligação Proteica , Células Tumorais Cultivadas
8.
Oncogene ; 12(2): 457-62, 1996 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-8570225

RESUMO

N-myc expression is negatively regulated by retinoic acid (RA) which induces the growth arrest and differentiation of neuroblastoma (NB) cells. However, it has not been completely defined whether N-Myc promotes growth and/or antagonises neuronal differentiation of NB cells or whether the down regulation of N-myc occurs as a consequence of the onset of differentiation. By transfecting an N-myc gene construct into these cells, we found that the constitutive overexpression of N-myc stimulated proliferation in RA containing medium and, although these cells were still responsive to RA, they were no longer able to differentiate. Since N-Myc functions appear to be mediated by heterodimerization with Max, the ectopic overexpression of max in NB cells was also investigated. In contrast to N-Myc, Max strongly induced the differentiation by enhancing the effects of RA. Max-transfected cells rapidly arrested growth and differentiated fully within a few days of RA treatment. These findings suggest that the relative levels of N-Myc compared to Max appears to be crucial in stimulating neuroblastoma growth or differentiation, and may contribute to explain the remarkable clinical behaviour of neuroblastomas.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Genes myc , Neuroblastoma/genética , Fatores de Transcrição , Tretinoína/farmacologia , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fatores de Transcrição de Zíper de Leucina Básica , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/fisiologia , Humanos , Dados de Sequência Molecular , Neuroblastoma/patologia , Células Tumorais Cultivadas
9.
Oncogene ; 14(15): 1805-10, 1997 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-9150386

RESUMO

The NFKB-2 (Lyt-10) gene codes for an NF-kappaB-related transcription factor containing rel-polyG-ankyrin domains. Rearrangements of the NFKB-2 locus leading to the production of 3' truncated NFKB-2 proteins are recurrently found in lymphoid neoplasms, particularly cutaneous lymphomas. Such mutant NFKB-2 proteins have lost the ability to repress transcription that is typical of NFKB-2 subunit p52, and function as constitutive transcriptional activators. To verify whether the expression of abnormal NFKB-2 proteins can lead to malignant transformations in mammalian cells, we transfected human lymphoblastoid cell lines and murine fibroblasts (Balb/3T3) with expression vectors carrying the cDNAs coding for normal NFKB-2p52, Lyt-10C alpha or LB40 proteins, which are representative of the abnormal types found in lymphoma cases. The expression of both normal and mutant NFKB-2 proteins has a lethal effect on lymphoblastoid cells and a cytotoxic effect was also observed in murine fibroblasts. The fibroblast cell lines expressing Lyt-10C alpha or LB40, but not those expressing normal NFKB-2p52, were capable of forming colonies in soft agar. The analysis of individual clones revealed that cloning efficiency correlated with the expression levels of the abnormal proteins. Injection of the Lyt-10C alpha-transfected Balb cells in SCID mice led to tumor formation in all of the animals, whereas no tumors were observed in the mice injected with control or NFKB-2p52-transfected cells, thus indicating that abnormal NFKB-2 protein expression is tumorigenic in vivo. Our results show that mutant NFKB-2 proteins can lead to the transformed phenotype, and support the hypothesis that alterations in NFKB-2 genes may play a role in lymphomagenesis.


Assuntos
Células 3T3/efeitos dos fármacos , Transformação Celular Neoplásica , Linfoma/metabolismo , NF-kappa B/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Células 3T3/citologia , Células 3T3/fisiologia , Animais , Transformação Celular Neoplásica/efeitos dos fármacos , Feminino , Humanos , Linfoma/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Mutação , NF-kappa B/biossíntese , NF-kappa B/genética , Subunidade p52 de NF-kappa B , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética
10.
J Neurosci ; 21(6): 1809-18, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11245665

RESUMO

After neuronal injury and in several neurodegenerative diseases, activated microglia secrete proinflammatory molecules that can contribute to the progressive neural damage. The recent demonstration of a protective role of estrogen in neurodegenerative disorders in humans and experimental animal models led us to investigate whether this hormone regulates the inflammatory response in the CNS. We here show that estrogen exerts an anti-inflammatory activity on primary cultures of rat microglia, as suggested by the blockage of the phenotypic conversion associated with activation and by the prevention of lipopolysaccharide-induced production of inflammatory mediators: inducible form of NO synthase (iNOS), prostaglandin-E(2) (PGE(2)), and metalloproteinase-9 (MMP-9). These effects are dose-dependent, maximal at 1 nm 17beta-estradiol, and can be blocked by the estrogen receptor (ER) antagonist ICI 182,780. The demonstration of ERalpha and ERbeta expression in microglia and macrophages and the observation of estrogen blockade of MMP-9 mRNA accumulation and MMP-9 promoter induction further support the hypothesis of a genomic activity of estrogen via intracellular receptors. This is the first report showing an anti-inflammatory activity of estrogen in microglia. Our study proposes a novel explanation for the protective effects of estrogen in neurodegenerative and inflammatory diseases and provides new molecular and cellular targets for the screening of ER ligands acting in the CNS.


Assuntos
Estradiol/análogos & derivados , Estrogênios/farmacologia , Inflamação/prevenção & controle , Lipopolissacarídeos/farmacologia , Microglia/fisiologia , Animais , Células Cultivadas , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Fulvestranto , Humanos , Inflamação/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Microglia/citologia , Microglia/efeitos dos fármacos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética
11.
Brain Res Bull ; 65(3): 241-7, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15811587

RESUMO

The insecticide dichlorodiphenyltrichloroethane (DDT) interferes with physiological endocrine processes modulating estrogens receptor activity. Most of the data describing the DDT mechanism of action have been collected in vitro or in reproductive tissues in vivo. Here we use a new transgenic mouse model to investigate the DDT effects on estrogens receptor activation in vivo in non-reproductive tissues. In particular, we demonstrate that DDT is able to activate estrogen receptors in the brain and the liver of adult mice after acute administration, and it is active in lactating mice when accumulated in the mother's milk. Furthermore, we demonstrate that the acute administration of DDT activates estrogen receptors with a different kinetics with respect to 17beta-estradiol. Experiments with a breast cancer cell line engineered to express luciferase under the transcriptional control of activated estrogen receptors reveal that the microsomal metabolization of DDT is required for its full activity on estrogen receptors. Taken together these data lead to hypothesize that the delayed DDT time course on estrogen receptor activation in vivo might be due to a necessary step of metabolism of the compound.


Assuntos
Encéfalo/efeitos dos fármacos , DDT/toxicidade , Lactação/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Receptores de Estrogênio/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting/métodos , Carcinoma , Linhagem Celular Tumoral , Estradiol/farmacologia , Feminino , Fígado/efeitos dos fármacos , Fígado/metabolismo , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Ensaio Radioligante/métodos , Receptores de Estrogênio/genética , Fatores de Tempo , Transfecção/métodos
12.
Mol Endocrinol ; 15(7): 1104-13, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11435611

RESUMO

In addition to their well known control of reproductive functions, estrogens modulate important physiological processes. The identification of compounds with tissue-selective activity will lead to new drugs mimicking the beneficial effects of estrogen on the prevention of osteoporosis and cardiovascular or neurodegenerative diseases, while avoiding its detrimental proliferative effects. As an innovative model for the in vivo identification of new selective estrogen receptor modulators (SERMs), we engineered a mouse genome to express a luciferase reporter gene ubiquitously. The constructs for transgenesis consist of the reporter gene driven by a dimerized estrogen-responsive element (ERE) and a minimal promoter. Insulator sequences, either matrix attachment region (MAR) or beta-globin hypersensitive site 4 (HS4), flank the construct to achieve a generalized, hormoneresponsive luciferase expression. In the mouse we generated, the reporter expression is detectable in all 26 tissues examined, but is induced by 17beta-estradiol (E2) only in 15 of them, all expressing estrogen receptors (ERs). Immunohistochemical studies show that in the mouse uterus, luciferase and ERs colocalize. In primary cultures of bone marrow cells explanted from the transgenic mice and in vivo, luciferase activity accumulates with increasing E(2) concentration. E2 activity is blocked by the ER full antagonist ICI 182,780. Tamoxifen shows partial agonist activity in liver and bone when administered to the animals. In the mouse system here illustrated, by biochemical, immunohistochemical, and pharmacological criteria, luciferase content reflects ER transcriptional activity and thus represents a novel system for the study of ER dynamics during physiological fluctuations of estrogen and for the identification of SERMs or endocrine disruptors.


Assuntos
Engenharia Genética , Receptores de Estrogênio/genética , Animais , Células da Medula Óssea/metabolismo , Neoplasias da Mama , Dimerização , Estradiol/administração & dosagem , Estradiol/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Genes Reporter/genética , Células HeLa , Humanos , Imuno-Histoquímica , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Neuroblastoma , Ovariectomia , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Elementos de Resposta/genética , Moduladores Seletivos de Receptor Estrogênico/análise , Transcrição Gênica/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas , Útero/metabolismo
13.
Endocrinology ; 143(12): 4544-51, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12446581

RESUMO

We investigated the tissue-specific effects of dichlorodyphenyltrichloroethane (DDT) isomers in adult and suckling newborn mice, using a novel mouse line engineered to express a reporter of estrogen receptor transcriptional activity (ERE-tkLUC mouse). The DDT isomers p,p'-DDT [1,1,1-trichloro2,2-bis(p-chlorophenyl) ethane] and o,p'-DDT [1,1,1-trichloro-2(p-chlorophenyl)-2-(o-chlorophenyl) ethane] were specifically selected as a weak and a strong estrogen, respectively. In adult male mice, p,p'-DDT induced luciferase activity in liver, brain, thymus, and prostate but not in heart and lung. The effect of p,p'-DDT was dose-dependent, maximal at 16 h after sc treatment, and completely blocked by the estrogen receptor antagonist ICI-182,780. In all the organs analyzed, except the liver, administration of o,p'-DDT showed a pattern of luciferase induction superimposable to that of its isomer p,p'-DDT. In liver, o,p'-DDT significantly decreased basal luciferase activity and blocked the reporter induction by 17beta-estradiol. These data lead us to hypothesize that a modulation of ER activity may be involved in the toxic effects of DDT demonstrated by epidemiological and experimental studies. Luciferase activity was also studied in 4-d-old mice lactating from a mother injected with either p,p'-DDT or o,p'-DDT. Both isomers induced a 2-fold increase in the newborn brain. An opposite effect was observed in liver, where p,p'-DDT increased and o,p'-DDT decreased luciferase, thus indicating that these compounds modulate ER activity in adult and newborn tissues by use of a similar mechanism. The ERE-tkLUC mouse proves to be a suitable tool to functionally assess the tissue specificity of estrogenic/antiestrogenic compounds in adult (as well as in suckling) mice.


Assuntos
DDT/química , DDT/farmacologia , Estradiol/análogos & derivados , Genes Reporter , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/genética , Envelhecimento , Animais , Animais Recém-Nascidos , Animais Lactentes , Encéfalo/metabolismo , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Fulvestranto , Cromatografia Gasosa-Espectrometria de Massas , Expressão Gênica/efeitos dos fármacos , Cinética , Fígado/química , Fígado/metabolismo , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Próstata/metabolismo , Receptores de Estrogênio/fisiologia , Receptores de Progesterona/efeitos dos fármacos , Receptores de Progesterona/genética , Elementos de Resposta , Relação Estrutura-Atividade , Timidina Quinase/genética
14.
Exp Gerontol ; 35(9-10): 1309-16, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11113609

RESUMO

Estrogens are thought to play a protective role against neurodegeneration through a variety of mechanisms including the activation of growth factors and neurotransmitter synthesis, the control of synaptic plasticity and functions, and the blockade of oxidative reactions. We here propose a novel mechanism to explain the neuroprotective effects of estradiol by showing that estrogens may antagonize nitric oxide synthase activity and reduce the accumulation of nitrites and nitrates consequent to various inflammatory stimuli. The potential anti-inflammatory activity of estradiol is analyzed in vitro in cells in culture including primary cultures of microglia and in vivo in a well-known model of inflammation.


Assuntos
Estrogênios/farmacologia , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Óxido Nítrico Sintase/antagonistas & inibidores , Animais , Carragenina , Células Cultivadas , Citocinas/farmacologia , Estradiol/farmacologia , Exsudatos e Transudatos/química , Exsudatos e Transudatos/efeitos dos fármacos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Masculino , Microglia/citologia , Microglia/enzimologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Nitritos/metabolismo , Pleurisia/induzido quimicamente , Pleurisia/metabolismo , Ratos , Ratos Sprague-Dawley
15.
J Neuroendocrinol ; 12(11): 1051-9, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11069120

RESUMO

We have recently identified nip-2 as a gene target for 17beta-oestradiol activity in the neuroblastoma SK-ER3 cells expressing the oestrogen receptor (ER) alpha. Here we show 17beta-oestradiol treatment of neuroblastoma and rat embryo neurones in culture blocks the increase in nip-2 mRNA induced by apoptotic stimuli and prevents cell death as indicated by cell counting, 3,(4,5-dimethylthiazol-2-yl)2,5-diphenil-tetrazoliumbromi de and DNA fragmentation assays. Neither of these effects are observed in the presence of the specific ER antagonist ICI 182,780, and are absent in neuroblastoma cells not expressing ER. We propose that nip-2 plays a relevant role in neural cell apoptosis and that a decrease in its expression is instrumental for the oestrogen anti-apoptotic effect described here. The experimental evidence presented supports the recent hypothesis of a protective role of oestrogens in neurodegenerative diseases such as Alzheimer's disease and highlights the importance of the development of new ER ligands for the prevention of neural cell damage.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/genética , Proteínas de Transporte , Estradiol/análogos & derivados , Estradiol/farmacologia , Expressão Gênica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Proteínas de Ligação ao Cálcio/fisiologia , Contagem de Células , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Fragmentação do DNA , Embrião de Mamíferos , Fulvestranto , Glucose/administração & dosagem , Neuroblastoma/patologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , RNA Mensageiro/análise , Ratos , Receptores de Estrogênio/antagonistas & inibidores , Transfecção , Células Tumorais Cultivadas
16.
J Steroid Biochem Mol Biol ; 74(5): 319-25, 2000 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11162940

RESUMO

In mammals, estrogens have a multiplicity of effects ranging from control of differentiation of selected brain nuclei, reproductive functions, sexual behavior. In addition, these hormones influence the manifestation of disorders like depression and Alzheimer's. Study of the cells target for the hormone has shown that estrogen receptors (ERs) are expressed in all known neural cells, including microglia. In view of the potential interest in the use of estrogens in the therapy of several pathologies of the nervous system, it would be of interest to fully understand the mechanism of estrogen activity in the various neural target cells and get an insight on the molecular means allowing the hormone to display such a variety of effects. We have proposed the use of a reductionist approach for the systematic understanding of the estrogen activities in each specific type of target cell. Thus, we have generated a model system in which to study the activation of one of the known (ERs), estrogen receptor alpha. This system allowed us to identify a number of novel genes which expression may be influenced following the activation of this receptor subtype by estradiol (E(2)). We here report on data recently obtained by the study of one of these target genes, nip2, which encodes a proapoptotic protein product. We hypothesize that nip2 might be an important molecular determinant for estrogen anti-apoptotic activity in cells of neural origin and represents a potential target for drugs aimed at mimicking the E(2) beneficial effects in neural cells.


Assuntos
Proteínas de Transporte , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Elementos de Resposta/genética , Apoptose/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Estradiol/metabolismo , Humanos , Ligantes , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/citologia , Neurônios/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Toxicology ; 205(1-2): 65-73, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15458791

RESUMO

The present work tested the estrogenic activity of three weak environmental estrogens p,p'DDT [1,1,1-trichloro-2,2-bis(p-chlorophenyl) ethane], p,p'DDE [1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene] and betaBHC [beta-benzene-hexachloride] in the transgenic estrogen-reporter mouse model (ERE-tK-LUC). By a time dependent analysis of the transgenic reporter expression (luciferase), we showed that all these chemicals modulated the estrogen receptors (ERs) in the whole body, although with a different efficacy and depending upon the tissue analyzed. Peak activity was registered at 16 h of treatment with 5000 microg/kg of each compound. Organochlorines are lipophylic molecules that accumulate in fat. During weight loss they are mobilized and their concentration increases in blood. We tested whether after experimental accumulation in fat tissue, followed by a 48 h period of fasting, these compounds could be modulated to reach sufficient levels to activate the ERs in target tissues. This experimental setting produced results that were different from those obtained following acute treatments. In loaded mice, fasting induced betaBHC mobilization resulted in strong ER activation in the liver, lung, eye, cerebellum, hypothalamus and cortex. p,p'DDT mobilization had no effect in these tissues, but efficiently acted in the testis, where, on the contrary, betaBHC inhibited reporter expression. During fasting, betaBHC, p,p'DDT and the metabolite p,p'DDE increased in blood concentration, from 2.7 +/- 0.36, 0.65 +/- 0.01 and 0.48 +/- 0.06 microg/ml to 9.51 +/- 1.1, 4.98 +/- 0.77 and 6.0 +/- 0.71 microg/ml, respectively. We conclude that these organochlorines modulate differently the expression of estrogen regulated genes in a tissue- and compound-specific manner and that their action is dependent on the energy balance. Moreover, we show that this mouse model is suitable to detect the estrogenic activity of chemicals with variable structures such as alkyl phenols and polychlorobiphenyls.


Assuntos
DDT/toxicidade , Diclorodifenil Dicloroetileno/toxicidade , Estrogênios/toxicidade , Hexaclorocicloexano/toxicidade , Receptores de Estrogênio/efeitos dos fármacos , Animais , DDT/sangue , Diclorodifenil Dicloroetileno/sangue , Regulação da Expressão Gênica/efeitos dos fármacos , Hexaclorocicloexano/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Receptores de Estrogênio/fisiologia
19.
Ann N Y Acad Sci ; 1163: 475-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19456391

RESUMO

Here we show that genistein, through an estrogen receptor-mediated action, modulates gene expression in the mouse testis throughout development. Genistein passed from the lactating mother to the suckling offspring at levels sufficient to activate gene expression in the testis of the pups. Testis are already responsive to genistein as well as to estradiol at day 14.5 of fetal development. Activation of luciferase correlates with an activation of cell proliferation. In conclusion, our results show that genistein affects reproductive organs of male mice at all developmental ages.


Assuntos
Genisteína/farmacologia , Receptores de Estrogênio/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Animais , Masculino , Camundongos , Camundongos Transgênicos , Receptores de Estrogênio/genética , Técnicas de Cultura de Tecidos
20.
Q J Nucl Med Mol Imaging ; 51(2): 127-38, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17420714

RESUMO

The development of novel drugs is a lengthy process requiring years of preclinical research and many steps indispensable to ensure that the molecule of interest can be administered to humans with a minimal risk of toxic effects. Even a minimal reduction in the initial stages of drug development would result in a tremendous saving in time; therefore, pharmaceutical companies are eager to apply novel methodologies that shorten the time required for pharmacodynamic, pharmacokinetic and toxicological studies to be carried out in vitro and in animal systems. Currently, quantitative analysis of molecular events in living organisms is done with the combined application of imaging and genetic engineering technologies. In vivo imaging provides surrogate endpoints that can improve the identification of new drug candidates and speed up their research at preclinical stages. The integration of reporter systems in animal models of human diseases represents a reachable frontier that will dramatically advance drug development in terms of costs, time and efficacy. The present review outlines the applicability of imaging technologies for drug development and presents a panorama on the state of the art of currently available imaging technologies suitable for preclinical studies, with particular focus on bioluminescence and fluorescence as the methodologies of election.


Assuntos
Diagnóstico por Imagem/tendências , Sistemas de Liberação de Medicamentos/tendências , Desenho de Fármacos , Genes Reporter , Técnicas de Sonda Molecular/tendências , Medicina Nuclear/tendências , Animais , Previsões , Humanos , Aumento da Imagem/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA