Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Pharmacol Res ; : 107296, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38971269

ABSTRACT

The activity of sirtuin 1 (SIRT1, a member of the NAD+-dependent deacetylases family) decreases during aging as NAD+ levels naturally decline, thus increasing the risk of several age-associated diseases. Several sirtuin-activating compounds (STACs) have been developed to counteract the age-associated reduction in SIRT1 activity, and some of them are currently under development in clinical trials. STACs induce SIRT1 activation, either through allosteric activation of the enzyme in the presence of NAD+, or by increasing NAD+ levels by inhibiting its degradation or by supplying a key precursor in biosynthesis. In this study, we have identified (E)-2'-des-methyl sulindac analogues as a novel class of STACs that act also in the absence of NAD+, a peculiar behavior demonstrated through enzymatic and mass spectrometry experiments, both in vitro and in cell lines. The activation of the SIRT1 pathway was confirmed in vivo through gene expression and metabolomics analysis. Our data suggest that these compounds could serve as candidate leads for a novel therapeutic strategy aimed at addressing a key metabolic deficiency that may contribute to metabolic and age-associated diseases.

2.
J Control Release ; 294: 165-175, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30557650

ABSTRACT

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.


Subject(s)
Adenoviridae , Antineoplastic Agents/administration & dosage , Extracellular Vesicles , Oncolytic Viruses , Paclitaxel/administration & dosage , Animals , Cell Line, Tumor , Cell Survival , Combined Modality Therapy , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice, Transgenic , Neoplasms/immunology , Neoplasms/therapy , Tissue Distribution
3.
J Control Release ; 283: 223-234, 2018 08 10.
Article in English | MEDLINE | ID: mdl-29864473

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Extracellular Vesicles , Lung Neoplasms/therapy , Oncolytic Viruses , Paclitaxel/administration & dosage , Animals , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Liver/drug effects , Liver/pathology , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Spleen/drug effects , Spleen/pathology
4.
J Neuroendocrinol ; 30(2)2018 02.
Article in English | MEDLINE | ID: mdl-28650102

ABSTRACT

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.


Subject(s)
Brain/metabolism , Encephalitis/metabolism , Animals , Cytokines/metabolism , Luciferases/metabolism , Luminescent Measurements , Mice , NF-kappa B/metabolism
5.
Toxicol Appl Pharmacol ; 255(1): 65-75, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21683088

ABSTRACT

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.


Subject(s)
Adipose Tissue/drug effects , Environmental Pollutants/toxicity , Receptors, Estrogen/drug effects , Trialkyltin Compounds/toxicity , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Diethylstilbestrol/pharmacology , Dose-Response Relationship, Drug , Estradiol/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , PPAR gamma/physiology
6.
Ann N Y Acad Sci ; 1163: 475-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19456391

ABSTRACT

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.


Subject(s)
Genistein/pharmacology , Receptors, Estrogen/metabolism , Testis/drug effects , Testis/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Animals , Male , Mice , Mice, Transgenic , Receptors, Estrogen/genetics , Tissue Culture Techniques
7.
Q J Nucl Med Mol Imaging ; 51(2): 127-38, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17420714

ABSTRACT

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.


Subject(s)
Diagnostic Imaging/trends , Drug Delivery Systems/trends , Drug Design , Genes, Reporter , Molecular Probe Techniques/trends , Nuclear Medicine/trends , Animals , Forecasting , Humans , Image Enhancement/methods
8.
Phytomedicine ; 14(2-3): 147-52, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16860978

ABSTRACT

Isolation and biological characterization of pure compounds was used to identify and characterize estrogenic activity and estrogen receptors (ER) preference in chemical components of Achillea millefolium. This medicinal plant is used in folk medicine as an emmenagogue. In vitro assay, based on recombinant MCF-7 cells, showed estrogenic activity in a crude extract of the aerial parts of A. millefolium. After fractionation of the crude extract with increasing polar solvents, estrogenic activity was found in the methanol/water fraction. Nine compounds were isolated and characterized by HR-MS spectra and 1D- and 2D-NMR techniques. In particular, dihydrodehydrodiconiferyl alcohol 9-O-beta-D-glucopyranoside - a glycosyl-neolignan - was isolated for the first time from the genus Achillea in addition to six flavone derivatives, apigenin, apigenin-7-O-beta-D-glucopyranoside, luteolin, luteolin-7-O-beta-D-glucopyranoside, luteolin-4'-O-beta-D-glucopyranoside, rutin, and two caffeic acid derivatives, 3,5-dicaffeoylquinic acid and chlorogenic acid. Apigenin and luteolin, the most important estrogenic compounds among those tested, were studied for their ability to activate alpha or beta estrogen receptors (ERalpha, ERbeta) using transiently transfected cells. Our results suggest that isolation and biological characterization of estrogenic compounds in traditionally used medicinal plants could be a first step in better assessing further (e.g. in vivo) tests of nutraceutical and pharmacological strategies based on phytoestrogens.


Subject(s)
Achillea , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Phytoestrogens/pharmacology , Phytotherapy , Plant Extracts/pharmacology , Cells, Cultured/drug effects , Female , Humans , In Vitro Techniques , Phytoestrogens/administration & dosage , Phytoestrogens/chemistry , Phytoestrogens/therapeutic use , Plant Components, Aerial , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/therapeutic use
9.
Maturitas ; 54(4): 315-20, 2006 Jul 20.
Article in English | MEDLINE | ID: mdl-16753274

ABSTRACT

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.


Subject(s)
Receptors, Estrogen/metabolism , Animals , Female , Genes, Reporter , Luciferases/metabolism , Mice , Models, Animal
10.
Mol Cell Endocrinol ; 246(1-2): 69-75, 2006 Feb 26.
Article in English | MEDLINE | ID: mdl-16388894

ABSTRACT

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.


Subject(s)
Diagnostic Imaging/methods , Diagnostic Imaging/trends , Models, Animal , Molecular Biology/methods , Molecular Biology/trends , Animals , Genes, Reporter/genetics , Genetic Vectors , Mice , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
11.
Brain Res Bull ; 65(3): 241-7, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15811587

ABSTRACT

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.


Subject(s)
Brain/drug effects , DDT/toxicity , Lactation/drug effects , Prenatal Exposure Delayed Effects , Receptors, Estrogen/metabolism , Animals , Animals, Newborn , Blotting, Western/methods , Carcinoma , Cell Line, Tumor , Estradiol/pharmacology , Female , Liver/drug effects , Liver/metabolism , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Radioligand Assay/methods , Receptors, Estrogen/genetics , Time Factors , Transfection/methods
12.
Toxicol Appl Pharmacol ; 201(2): 137-48, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15541753

ABSTRACT

Organochlorines are lipophylic molecules that accumulate in the fat where they remain for years. During weight loss, they are mobilized and their concentration increases in blood. The present work tests, in transgenic estrogen-reporter mice (ERE-tK-LUC), whether this increase is sufficient to modulate the estrogen receptors (ERs) in the whole body. Three weak estrogens were studied: p,p'DDT [1,1,1-trichloro2,2-bis(p-chlorophenyl) ethane], p,p'DDE [1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene], and betaBHC [beta-benzene-hexachloride]. Dose-dependent analysis of reporter expression (luciferase) were performed in tissues of acutely treated mice. A body map of ER activation was obtained. All these chemicals modulated the reporter, although with a different efficiency and depending upon the tissue analyzed. Induction was confirmed in the liver by determining the expression of the endogenous progesterone receptor (PR) gene, at the dose and time point at which the luciferase gene was maximally induced. After experimental accumulation in the fat tissue, followed by a 48-h period of fasting, we tested whether these compounds could be mobilized to reach sufficient levels to activate the ERs in selected reproductive and nonreproductive tissues (testicle, prostate, liver, and lung). This experimental setting produced results that were different than those obtained following acute treatments. In loaded mice, fasting induced betaBHC mobilization resulted in strong ER activation in the liver and the lung, which was blocked by ICI-182780. p,p'DDT mobilization had no effect in these tissues, but it acted efficiently in the prostate and testis. betaBHC inhibited the ERE-mediated reporter in the testicle and induced the reporter in the prostate. In this tissue, betaBHC action was not inhibited by the anti-estrogen ICI-182780. During fasting, betaBHC, p,p'DDT, and metabolite p,p'DDE increased in blood concentration, from 2.25 +/- 0.25, 0.51 +/- 0.09, and 0.38 +/- 0.06 microg/ml to 8.24 +/- 0.95, 4.52 +/- 0.68, and 5.06 +/- 0.57 microg/ml, respectively. The effect produced by these organochlorines in the liver correlates with the modulation of the ERalpha protein. We conclude that these organochlorines modulate differently the expression of estrogen-regulated genes in male mice. Their effect is tissue- and compound-specific and is dependent on the energetic balance.


Subject(s)
Estrogens/genetics , Genes, Reporter/genetics , Genitalia, Male/drug effects , Hydrocarbons, Chlorinated/toxicity , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blotting, Western , Cell Line, Tumor , DDT/metabolism , DDT/toxicity , Dichlorodiphenyl Dichloroethylene/metabolism , Dichlorodiphenyl Dichloroethylene/toxicity , Estrogens, Non-Steroidal/toxicity , Female , Gas Chromatography-Mass Spectrometry , Hexachlorocyclohexane/metabolism , Hexachlorocyclohexane/toxicity , Humans , Hydrocarbons, Chlorinated/pharmacokinetics , Luminescent Measurements , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
13.
Toxicology ; 205(1-2): 65-73, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15458791

ABSTRACT

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.


Subject(s)
DDT/toxicity , Dichlorodiphenyl Dichloroethylene/toxicity , Estrogens/toxicity , Hexachlorocyclohexane/toxicity , Receptors, Estrogen/drug effects , Animals , DDT/blood , Dichlorodiphenyl Dichloroethylene/blood , Gene Expression Regulation/drug effects , Hexachlorocyclohexane/blood , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Receptors, Estrogen/physiology
15.
Endocrinology ; 143(12): 4544-51, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12446581

ABSTRACT

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.


Subject(s)
DDT/chemistry , DDT/pharmacology , Estradiol/analogs & derivatives , Genes, Reporter , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Aging , Animals , Animals, Newborn , Animals, Suckling , Brain/metabolism , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Fulvestrant , Gas Chromatography-Mass Spectrometry , Gene Expression/drug effects , Kinetics , Liver/chemistry , Liver/metabolism , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Transgenic , Prostate/metabolism , Receptors, Estrogen/physiology , Receptors, Progesterone/drug effects , Receptors, Progesterone/genetics , Response Elements , Structure-Activity Relationship , Thymidine Kinase/genetics
17.
Brain Res Brain Res Rev ; 37(1-3): 335-42, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11744098

ABSTRACT

To identify genes selectively induced by estrogens in cells of neural origin we have treated with a low concentration of 17 beta-estradiol (E2) the estrogen-receptor positive SK-ER3 neuroblastoma cells and we have isolated messages modulated by the hormonal treatment at short (1 h) and longer (17 h) times. By using the ddPCR approach we identified numerous messages which content was significantly and reproducibly altered by the hormonal treatment. Among these messages we focused our attention on bnip2, which expression was inhibited by estradiol. bnip2 was found to be a member of the BNIP family of genes of unknown physiological activity at the time. Investigations carried out in our laboratory proved a strong correlation between the increased expression of bnip2 gene and cell death induced by toxic stimuli. Furthermore, we showed that transfection of the bnip2 cDNA results in massive cell death and Bcl-2 overexpression counteracts the toxic effect of bnip2. These findings suggest that the proteins encoded by these two genes either interact or act in an opposite manner on the same mechanisms triggering the apoptotic cascade of events. Time-course experiments carried out in different cell systems and with a variety of neurotoxic agents proved a strong correlation between estrogen-induced decrease in bnip2 expression and the time required for estrogen to exert its protective effect. These observations led us to hypothesize an involvement of bnip2 in estrogen effects on cell survival. The finding that bnip2 is developmentally regulated may suggest a role of this gene in those brain areas where the differentiation is orchestrated by estradiol. Investigations in non-neural cells show that bnip2 is the mediator of the anti-apoptotic activity of estrogens in a variety of cells and thus might represent an important target for the evaluation of the activity of novel synthetic ligands for the estrogen receptor.


Subject(s)
Carrier Proteins/metabolism , Estrogens/pharmacology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/toxicity , Cell Death/drug effects , Cell Death/physiology , Cell Line , Gene Expression/drug effects , Gene Expression Profiling , Humans , Neuroblastoma , Neurons/cytology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/pharmacology , RNA, Messenger/metabolism , Rats
19.
Mol Endocrinol ; 15(7): 1104-13, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11435611

ABSTRACT

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.


Subject(s)
Genetic Engineering , Receptors, Estrogen/genetics , Animals , Bone Marrow Cells/metabolism , Breast Neoplasms , Dimerization , Estradiol/administration & dosage , Estradiol/pharmacology , Female , Gene Expression/drug effects , Genes, Reporter/genetics , HeLa Cells , Humans , Immunohistochemistry , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Neuroblastoma , Ovariectomy , Promoter Regions, Genetic , Receptors, Estrogen/metabolism , Response Elements/genetics , Selective Estrogen Receptor Modulators/analysis , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured , Uterus/metabolism
20.
Oncogene ; 20(7): 775-87, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11314012

ABSTRACT

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.


Subject(s)
Carbonic Anhydrases/genetics , Enhancer Elements, Genetic , Leukemia, Erythroblastic, Acute/genetics , Oncogene Proteins v-erbA/genetics , Receptors, Thyroid Hormone/genetics , Animals , Base Sequence , Binding Sites , Chickens , Erythroid Precursor Cells , Gene Expression Regulation, Leukemic , Humans , Mice , Models, Genetic , Molecular Sequence Data , Protein Binding , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...