Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Autophagy ; : 1-16, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38522425

ABSTRACT

TFEB and TFE3 belong to the MiT/TFE family of transcription factors that bind identical DNA responsive elements in the regulatory regions of target genes. They are involved in regulating lysosomal biogenesis, function, exocytosis, autophagy, and lipid catabolism. Precise control of TFEB and TFE3 activity is crucial for processes such as senescence, stress response, energy metabolism, and cellular catabolism. Dysregulation of these factors is implicated in various diseases, thus researchers have explored pharmacological approaches to modulate MiT/TFE activity, considering these transcription factors as potential therapeutic targets. However, the physiological complexity of their functions and the lack of suitable in vivo tools have limited the development of selective MiT/TFE modulating agents. Here, we have created a reporter-based biosensor, named CLEARoptimized, facilitating the pharmacological profiling of TFEB- and TFE3-mediated transcription. This innovative tool enables the measurement of TFEB and TFE3 activity in living cells and mice through imaging and biochemical techniques. CLEARoptimized consists of a promoter with six coordinated lysosomal expression and regulation motifs identified through an in-depth bioinformatic analysis of the promoters of 128 TFEB-target genes. The biosensor drives the expression of luciferase and tdTomato reporter genes, allowing the quantification of TFEB and TFE3 activity in cells and in animals through optical imaging and biochemical assays. The biosensor's validity was confirmed by modulating MiT/TFE activity in both cell culture and reporter mice using physiological and pharmacological stimuli. Overall, this study introduces an innovative tool for studying autophagy and lysosomal pathway modulation at various biological levels, from individual cells to the entire organism.Abbreviations: CLEAR: coordinated lysosomal expression and regulation; MAR: matrix attachment regions; MiT: microphthalmia-associated transcription factor; ROI: region of interest; TBS: tris-buffered saline; TF: transcription factor; TFE3: transcription factor binding to IGHM enhancer 3; TFEB: transcription factor EB; TH: tyrosine hydroxylase; TK: thymidine kinase; TSS: transcription start site.

3.
Nat Med ; 29(10): 2643-2655, 2023 10.
Article in English | MEDLINE | ID: mdl-37749332

ABSTRACT

Fatty liver disease (FLD) caused by metabolic dysfunction is the leading cause of liver disease and the prevalence is rising, especially in women. Although during reproductive age women are protected against FLD, for still unknown and understudied reasons some develop rapidly progressive disease at the menopause. The patatin-like phospholipase domain-containing 3 (PNPLA3) p.I148M variant accounts for the largest fraction of inherited FLD variability. In the present study, we show that there is a specific multiplicative interaction between female sex and PNPLA3 p.I148M in determining FLD in at-risk individuals (steatosis and fibrosis, P < 10-10; advanced fibrosis/hepatocellular carcinoma, P = 0.034) and in the general population (P < 10-7 for alanine transaminase levels). In individuals with obesity, hepatic PNPLA3 expression was higher in women than in men (P = 0.007) and in mice correlated with estrogen levels. In human hepatocytes and liver organoids, PNPLA3 was induced by estrogen receptor-α (ER-α) agonists. By chromatin immunoprecipitation and luciferase assays, we identified and characterized an ER-α-binding site within a PNPLA3 enhancer and demonstrated via CRISPR-Cas9 genome editing that this sequence drives PNPLA3 p.I148M upregulation, leading to lipid droplet accumulation and fibrogenesis in three-dimensional multilineage spheroids with stellate cells. These data suggest that a functional interaction between ER-α and PNPLA3 p.I148M variant contributes to FLD in women.


Subject(s)
Acyltransferases , Non-alcoholic Fatty Liver Disease , Phospholipases A2, Calcium-Independent , Receptors, Estrogen , Animals , Female , Humans , Male , Mice , Acyltransferases/genetics , Acyltransferases/metabolism , Carcinoma, Hepatocellular , Fibrosis , Genetic Predisposition to Disease , Liver/metabolism , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Phospholipases A2, Calcium-Independent/genetics , Phospholipases A2, Calcium-Independent/metabolism , Receptors, Estrogen/metabolism
4.
Cells ; 12(3)2023 01 17.
Article in English | MEDLINE | ID: mdl-36766684

ABSTRACT

Microglia are heterogenous cells characterized by distinct populations each contributing to specific biological processes in the nervous system, including neuroprotection. To elucidate the impact of sex-specific microglia heterogenicity to the susceptibility of neuronal stress, we video-recorded with time-lapse microscopy the changes in shape and motility occurring in primary cells derived from mice of both sexes in response to pro-inflammatory or neurotoxic stimulations. With this morpho-functional analysis, we documented distinct microglia subpopulations eliciting sex-specific responses to stimulation: male microglia tended to have a more pro-inflammatory phenotype, while female microglia showed increased sensitivity to conduritol-B-epoxide (CBE), a small molecule inhibitor of glucocerebrosidase, the enzyme encoded by the GBA1 gene, mutations of which are the major risk factor for Parkinson's Disease (PD). Interestingly, glucocerebrosidase inhibition particularly impaired the ability of female microglia to enhance the Nrf2-dependent detoxification pathway in neurons, attenuating the sex differences observed in this neuroprotective function. This finding is consistent with the clinical impact of GBA1 mutations, in which the 1.5-2-fold reduced risk of developing idiopathic PD observed in female individuals is lost in the GBA1 carrier population, thus suggesting a sex-specific role for microglia in the etiopathogenesis of PD-GBA1.


Subject(s)
Parkinson Disease , Animals , Female , Male , Mice , Glucosylceramidase/metabolism , Microglia/metabolism , Neurons/metabolism , Parkinson Disease/metabolism
5.
Nutrients ; 14(16)2022 Aug 10.
Article in English | MEDLINE | ID: mdl-36014766

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) represents a public health issue, due to its prevalence and association with other cardiometabolic diseases. Growing evidence suggests that NAFLD alters the production of hepatokines, which, in turn, influence several metabolic processes. Despite accumulating evidence on the major role of estrogen signaling in the sexually dimorphic nature of NAFLD, dependency of hepatokine expression on sex and estrogens has been poorly investigated. Through in vitro and in vivo analysis, we determined the extent to which hepatokines, known to be altered in NAFLD, can be regulated, in a sex-specific fashion, under different hormonal and nutritional conditions. Our study identified four hepatokines that better recapitulate sex and estrogen dependency. Among them, adropin resulted as one that displays a sex-specific and estrogen receptor alpha (ERα)-dependent regulation in the liver of mice under an excess of dietary lipids (high-fat diet, HFD). Under HFD conditions, the hepatic induction of adropin negatively correlates with the expression of lipogenic genes and with fatty liver in female mice, an effect that depends upon hepatic ERα. Our findings support the idea that ERα-mediated induction of adropin might represent a potential approach to limit or prevent NAFLD.


Subject(s)
Diet, High-Fat , Estrogen Receptor alpha , Intercellular Signaling Peptides and Proteins , Liver , Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat/adverse effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/genetics , Estrogens/metabolism , Female , Homeostasis/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism
6.
Int J Mol Sci ; 23(11)2022 Jun 06.
Article in English | MEDLINE | ID: mdl-35683029

ABSTRACT

We previously demonstrated that Npy1rrfb mice, which carry the conditional inactivation of the Npy1r gene in forebrain principal neurons, display a sexually dimorphic phenotype, with male mice showing metabolic, hormonal and behavioral effects and females being only marginally affected. Moreover, exposure of Npy1rrfb male mice to a high-fat diet (HFD) increased body weight growth, adipose tissue, blood glucose levels and caloric intake compared to Npy1r2lox male controls. We used conditional knockout Npy1rrfb and Npy1r2lox control mice to examine whether forebrain disruption of the Npy1r gene affects susceptibility to obesity and associated disorders of cycling and ovariectomized (ovx) female mice in a standard diet (SD) regimen or exposed to an HFD for 3 months. The conditional deletion of the Npy1r gene increased body weight and subcutaneous white adipose tissue weight in both SD- and HFD-fed ovx females but not in cycling females. Moreover, compared with ovx control females on the same diet regimen, Npy1rrfb females displayed increased microglia number and activation, increased expression of Neuropeptide Y (NPY)-immunoreactivity (IR) and decreased expression of proopiomelanocortin-IR in the hypothalamic arcuate nucleus (ARC). These results suggest that in the ARC NPY-Y1R reduces the susceptibility to obesity of female mice with low levels of gonadal hormones and that this effect may be mediated via NPY-Y1R ability to protect the brain against neuroinflammation.


Subject(s)
Neuropeptide Y , Receptors, Neuropeptide Y , Animals , Female , Gonadal Hormones , Male , Mice , Neuroinflammatory Diseases , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Obesity/genetics , Obesity/metabolism , Prosencephalon/metabolism , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism
8.
Pharmacol Res ; 175: 105982, 2022 01.
Article in English | MEDLINE | ID: mdl-34798263

ABSTRACT

All the different coronavirus SARS-CoV-2 variants isolated so far share the same mechanism of infection mediated by the interaction of their spike (S) glycoprotein with specific residues on their cellular receptor: the angiotensin converting enzyme 2 (ACE2). Therefore, the steric hindrance on this cellular receptor created by a bulk macromolecule may represent an effective strategy for the prevention of the viral spreading and the onset of severe forms of Corona Virus disease 19 (COVID-19). Here, we applied a systematic evolution of ligands by exponential enrichment (SELEX) procedure to identify two single strand DNA molecules (aptamers) binding specifically to the region surrounding the K353, the key residue in human ACE2 interacting with the N501 amino acid of the SARS-CoV-2 S. 3D docking in silico experiments and biochemical assays demonstrated that these aptamers bind to this region, efficiently prevent the SARS-CoV-2 S/human ACE2 interaction and the viral infection in the nanomolar range, regardless of the viral variant, thus suggesting the possible clinical development of these aptamers as SARS-CoV-2 infection inhibitors. Our approach brings a significant innovation to the therapeutic paradigm of the SARS-CoV-2 pandemic by protecting the target cell instead of focusing on the virus; this is particularly attractive in light of the increasing number of viral mutants that may potentially escape the currently developed immune-mediated neutralization strategies.


Subject(s)
Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Aptamers, Nucleotide/pharmacology , COVID-19 Drug Treatment , Receptors, Virus/antagonists & inhibitors , SARS-CoV-2/pathogenicity , Virus Internalization/drug effects , A549 Cells , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , COVID-19/enzymology , COVID-19/genetics , COVID-19/virology , HEK293 Cells , Host-Pathogen Interactions , Humans , Mutation , Receptors, Virus/genetics , Receptors, Virus/metabolism , SARS-CoV-2/genetics , SELEX Aptamer Technique
9.
Nat Commun ; 12(1): 6883, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824281

ABSTRACT

In female mammals, the cessation of ovarian functions is associated with significant metabolic alterations, weight gain, and increased susceptibility to a number of pathologies associated with ageing. The molecular mechanisms triggering these systemic events are unknown because most tissues are responsive to lowered circulating sex steroids. As it has been demonstrated that isoform alpha of the estrogen receptor (ERα) may be activated by both estrogens and amino acids, we test the metabolic effects of a diet enriched in specific amino acids in ovariectomized (OVX) mice. This diet is able to block the OVX-induced weight gain and fat deposition in the liver. The use of liver-specific ERα KO mice demonstrates that the hepatic ERα, through the control of liver lipid metabolism, has a key role in the systemic response to OVX. The study suggests that the liver ERα might be a valuable target for dietary treatments for the post-menopause.


Subject(s)
Amino Acids, Essential/pharmacology , Estrogen Receptor alpha/metabolism , Liver/drug effects , Ovariectomy/adverse effects , Amino Acids, Branched-Chain/pharmacology , Amino Acids, Branched-Chain/therapeutic use , Amino Acids, Essential/therapeutic use , Animals , Diet Therapy , Estrogen Receptor alpha/deficiency , Female , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Sex Characteristics , Transcriptome/drug effects , Weight Gain/drug effects
10.
Mol Metab ; 32: 97-108, 2020 02.
Article in English | MEDLINE | ID: mdl-32029233

ABSTRACT

OBJECTIVE: Among obesity-associated metabolic diseases, non-alcoholic fatty liver disease (NAFLD) represents an increasing public health issue due to its emerging association with atherogenic dyslipidemia and cardiovascular diseases (CVDs). The lower prevalence of NAFLD in pre-menopausal women compared with men or post-menopausal women led us to hypothesize that the female-inherent ability to counteract this pathology might strongly rely on estrogen signaling. In female mammals, estrogen receptor alpha (ERα) is highly expressed in the liver, where it acts as a sensor of the nutritional status and adapts the metabolism to the reproductive needs. As in the male liver this receptor is little expressed, we here hypothesize that hepatic ERα might account for sex differences in the ability of males and females to cope with an excess of dietary lipids and counteract the accumulation of lipids in the liver. METHODS: Through liver metabolomics and transcriptomics we analyzed the relevance of hepatic ERα in the metabolic response of males and females to a diet highly enriched in fats (HFD) as a model of diet-induced obesity. RESULTS: The study shows that the hepatic ERα strongly contributes to the sex-specific response to an HFD and its action accounts for opposite consequences for hepatic health in males and females. CONCLUSION: This study identified hepatic ERα as a novel target for the design of sex-specific therapies against fatty liver and its cardio-metabolic consequences.


Subject(s)
Diet, High-Fat , Estrogen Receptor alpha/metabolism , Lipids/administration & dosage , Liver/metabolism , Sex Characteristics , Animals , Estrogen Receptor alpha/deficiency , Female , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism
11.
J Endocrinol ; 238(3): 165-176, 2018 09.
Article in English | MEDLINE | ID: mdl-30012715

ABSTRACT

Oestrogens are well-known proliferation and differentiation factors that play an essential role in the correct development of sex-related organs and behaviour in mammals. With the use of the ERE-Luc reporter mouse model, we show herein that throughout mouse development, oestrogen receptors (ERs) are active starting from day 12 post conception. Most interestingly, we show that prenatal luciferase expression in each organ is proportionally different in relation to the germ layer of the origin. The luciferase content is highest in ectoderm-derived organs (such as brain and skin) and is lowest in endoderm-derived organs (such as liver, lung, thymus and intestine). Consistent with the testosterone surge occurring in male mice at the end of pregnancy, in the first 2 days after birth, we observed a significant increase in the luciferase content in several organs, including the liver, bone, gonads and hindbrain. The results of the present study show a widespread transcriptional activity of ERs in developing embryos, pointing to the potential contribution of these receptors in the development of non-reproductive as well as reproductive organs. Consequently, the findings reported here might be relevant in explaining the significant differences in male and female physiopathology reported by a growing number of studies and may underline the necessity for more systematic analyses aimed at the identification of the prenatal effects of drugs interfering with ER signalling, such as aromatase inhibitors or endocrine disrupter chemicals.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , Receptors, Estrogen/physiology , Animals , Embryo, Mammalian , Embryonic Development/drug effects , Estrogens/pharmacology , Female , Fulvestrant/pharmacology , Gene Expression Regulation, Developmental/drug effects , Genes, Reporter/drug effects , Luciferases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Response Elements/drug effects , Response Elements/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
12.
Cell Metab ; 28(2): 256-267.e5, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29909969

ABSTRACT

Sex impacts on liver physiology with severe consequences for energy metabolism and response to xenobiotic, hepatic, and extra-hepatic diseases. The comprehension of the biology subtending sex-related hepatic differences is therefore very relevant in the medical, pharmacological, and dietary perspective. The extensive application of metabolomics paired to transcriptomics here shows that, in the case of short-term fasting, the decision to maintain lipid synthesis using amino acids (aa) as a source of fuel is the key discriminant for the hepatic metabolism of male and female mice. Pharmacological and genetic interventions indicate that the hepatic estrogen receptor (ERα) has a key role in this sex-related strategy that is primed around birth by the aromatase-dependent conversion of testosterone into estradiol. This energy partition strategy, possibly the result of an evolutionary pressure enabling mammals to tailor their reproductive capacities to nutritional status, is most important to direct future sex-specific dietary and medical interventions.


Subject(s)
Amino Acids/metabolism , Estrogen Receptor alpha/physiology , Fasting/metabolism , Lipogenesis/physiology , Liver/metabolism , Sex Characteristics , Animals , Aromatase/metabolism , Energy Metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Male , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Factors
13.
Sci Rep ; 7(1): 2510, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28566747

ABSTRACT

Estrogen deprivation is associated with delayed healing, while estrogen replacement therapy (ERT) accelerates acute wound healing and protects against development of chronic wounds. However, current estrogenic molecules have undesired systemic effects, thus the aim of our studies is to generate new molecules for topic administration that are devoid of systemic effects. Following a preliminary study, the new 17ß-estradiol derivatives 1 were synthesized. The estrogenic activity of these novel compounds was evaluated in vitro using the cell line ERE-Luc B17 stably transfected with an ERE-Luc reporter. Among the 17ß-estradiol derivatives synthesized, compounds 1e and 1f showed the highest transactivation potency and were therefore selected for the study of their systemic estrogenic activity. The study of these compounds in the ERE-Luc mouse model demonstrated that both compounds lack systemic effects when administered in the wound area. Furthermore, wound-healing experiments showed that 1e displays a significant regenerative and anti-inflammatory activity. It is therefore confirmed that this class of compounds are suitable for topical administration and have a clear beneficial effect on wound healing.


Subject(s)
Estradiol/administration & dosage , Estrogen Replacement Therapy , Skin/drug effects , Wound Healing/drug effects , Administration, Topical , Animals , Cells, Cultured , Estradiol/analogs & derivatives , Estradiol/chemical synthesis , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Estrone/metabolism , Female , Humans , Mice , Ovariectomy , Skin/injuries , Skin/pathology , Wound Healing/physiology
14.
Sci Rep ; 7(1): 1194, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28446774

ABSTRACT

Recent work revealed the major role played by liver Estrogen Receptor α (ERα) in the regulation of metabolic and reproductive functions. By using mutant mice with liver-specific ablation of Erα, we here demonstrate that the hepatic ERα is essential for the modulation of the activity of Agouti Related Protein (AgRP) neurons in relation to the reproductive cycle and diet. Our results suggest that the alterations of hepatic lipid metabolism due to the lack of liver ERα activity are responsible for a neuroinflammatory status that induces refractoriness of AgRP neurons to reproductive and dietary stimuli. The study therefore points to the liver ERα as a necessary sensor for the coordination of systemic energy metabolism and reproductive functions.


Subject(s)
Agouti-Related Protein/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Estrogen Receptor alpha/metabolism , Liver/metabolism , Neurons/drug effects , Animals , Feeding Behavior , Female , Mice , Sexual Behavior, Animal
15.
Cell Rep ; 15(2): 360-71, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27050513

ABSTRACT

Lipoprotein synthesis is controlled by estrogens, but the exact mechanisms underpinning this regulation and the role of the hepatic estrogen receptor α (ERα) in cholesterol physiology are unclear. Utilizing a mouse model involving selective ablation of ERα in the liver, we demonstrate that hepatic ERα couples lipid metabolism to the reproductive cycle. We show that this receptor regulates the synthesis of cholesterol transport proteins, enzymes for lipoprotein remodeling, and receptors for cholesterol uptake. Additionally, ERα is indispensable during proestrus for the generation of high-density lipoproteins efficient in eliciting cholesterol efflux from macrophages. We propose that a specific interaction with liver X receptor α (LXRα) mediates the broad effects of ERα on the hepatic lipid metabolism.


Subject(s)
Estrogen Receptor alpha/metabolism , Liver/metabolism , Reproduction , Adiposity , Animals , Cholesterol/metabolism , Collagen/metabolism , Estrous Cycle , Female , Gene Deletion , Lipoproteins/metabolism , Lipoproteins, HDL/metabolism , Liver X Receptors/metabolism , Mice, Knockout , PPAR alpha/metabolism , Protein Binding , Transcription, Genetic
16.
Oncotarget ; 6(13): 10801-11, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25719423

ABSTRACT

This study shows that lack of ovarian activity has a negative impact on the life span of female mice. The extent to which this phenomenon could be associated with the anti-inflammatory effect of estrogens was analyzed in metabolic organs and aorta, by quantitative analysis of mRNAs encoding proteins in the inflammatory cascade. We demonstrate that the TNFα, IL-1ß, MCP-1, MIP-2 and IL-6 mRNA contents are increased in the liver, adipose tissue and aorta 7 months after ovariectomy (ovx) and this increased basal inflammation is maintained as the mice aged. In contrast, the extent of inflammatory gene expression is directly proportional to age in sham-operated mice. As a consequence, at 22 months, most of the inflammatory parameters examined were higher in the sham-operated group compared with the ovx group. These observations led us to propose that the decreased longevity of ovx mice may be due to an acceleration of the basal state of inflammation in metabolic organs, which is likely driven by the combination of a lack of estrogen-mediated anti-inflammatory activity and the loss of gonadal control of energy metabolism.


Subject(s)
Aging/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Life Expectancy , Ovariectomy/adverse effects , Adipose Tissue/metabolism , Age Factors , Aging/genetics , Animals , Aorta/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Female , Forkhead Transcription Factors/metabolism , Inflammation/etiology , Inflammation/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/metabolism , Mice, Inbred C57BL , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/metabolism , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
Chemosphere ; 112: 496-502, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25048945

ABSTRACT

A positive effect of estrogen treatment has been observed in neurodegenerative diseases such as Parkinson's disease. Since 17ß-estradiol can modulate positively dopaminergic system, here we sought to evaluate the effect of 17ß-estradiol supplementation on an animal model developing dopaminergic alterations on nucleus of striatum after neonatal exposure to permethrin pesticide. The goal of the study was to verify if the co-treatment with 17ß-estradiol could protect against the damage induced by pesticide exposure in early life. Permethrin treated rats showed a decrease of dopamine and Nurr1 gene expression in striatum, while a more pronounced decrease of dopamine was observed in rats co-administered with permethrin+17ß-estradiol. No difference between control and permethrin treated rats was observed in both mRNA of ERα and ERß, whereas the rats co-administered with permethrin+17ß-estradiol showed a down-regulation of ERα expression. The in vitro studies showed that permethrin, at high concentration may have an antagonist effect on ERα and even more pronounced in ERß, thus suggesting that permethrin may block the estrogen neuroprotective effects. In conclusion, in male rats, the administration of estrogen further enhanced the impairment of dopaminergic transmission due to exposure to permethrin.


Subject(s)
Dopamine/metabolism , Environmental Pollutants/toxicity , Estradiol/pharmacology , Estrogens/pharmacology , Neostriatum/drug effects , Neuroprotective Agents/pharmacology , Permethrin/toxicity , Synaptic Transmission/drug effects , Animals , Down-Regulation/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , MCF-7 Cells , Male , Neostriatum/cytology , Neostriatum/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors
18.
Endocrinology ; 155(6): 2213-21, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24635349

ABSTRACT

Estrogens play an important role in the regulation of energy homeostasis in female mammals and a reduced ovarian function, due to natural aging or surgery, is associated with body weight increase and fat redistribution. This disruption of energy homeostasis may constitute a trigger for several pathologies known to be associated with climacterium; however, so far, limited attention has been devoted to the ability of estrogen replacement therapies (ERT) to reinstate the balanced energy metabolism characteristic of cycling female mammals. The purpose of the present study was to compare the efficacy of selected ERTs in reversing the ovariectomy-induced gain in body weight. To this aim female ERE-Luc mice were ovariectomized and, after 3 weeks, treated per os for 21 days with: conjugated estrogens, two selective estrogen receptor modulators (bazedoxifene and raloxifene), and the combination of bazedoxifene plus conjugated estrogens (tissue-selective estrogen complex, TSEC). The study shows that the therapy based on TSEC was the most efficacious in reducing the body weight accrued by ovariectomy (OVX). In addition, by means of in vivo imaging, the TSEC treatment was shown to increase estrogen receptor (ER) transcriptional activity selectively in the arcuate nucleus, which is a key area for the control of energy homeostasis. Finally, quantitative analysis of the mRNAs encoding orexigenic and anorexigenic peptides indicated that following ERT with TSEC there was a significant change in Agrp, NPY, and Kiss-1 mRNA accumulation in the whole hypothalamus. Considering that prior studies showed that ERT with TSEC was able to mimic the rhythm of ER oscillatory activity during the reproductive cycle and that such fluctuations were relevant for energy metabolism, the present observations further point to the ER tetradian oscillation as an important component of the ER signaling necessary for the full hormone action and therefore for an efficacious ERT.


Subject(s)
Energy Metabolism/drug effects , Estrogen Replacement Therapy , Hypothalamus/drug effects , Hypothalamus/metabolism , Indoles/pharmacology , Raloxifene Hydrochloride/pharmacology , Animals , Body Weight/drug effects , Estrogen Receptor Modulators/pharmacology , Estrogens/pharmacology , Female , Mice
19.
Mol Cell Endocrinol ; 375(1-2): 149-56, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23727624

ABSTRACT

Estrogen deprivation is associated with delayed healing, while Hormone Replacement Therapy (HRT) accelerates acute wound healing and protects against development of chronic wounds. Estrogen exerts its effects on healing via numerous cell types by signalling through the receptors ERα and ERß, which bind to the Estrogen Responsive Element (ERE) and initiate gene transcription. The ERE-luciferase transgenic mouse model has been influential in assessing real-time in vivo estrogen receptor activation across a range of tissues and pathologies. Using this model we demonstrate novel temporally regulated peri-wound activation of estrogen signalling in female mice. Using histological methods we reveal that this signal is specifically localised to keratinocytes of the neoepidermis and wound margin dermal cells. Moreover using pharmacological agonists we reveal that ERß induces ERE-mediated signal in both epidermal and dermal cells while ERα induces ERE-mediated signal in dermal cells alone. Collectively these novel data demonstrate rapid and regional activation of estrogen signalling in wounded skin. A more complete understanding of local hormonal signalling during repair is essential for the focussed development of new therapies for wound healing.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Signal Transduction , Transcriptional Activation , Animals , Cells, Cultured , Estradiol/pharmacology , Estradiol/physiology , Estrogens/pharmacology , Estrogens/physiology , Female , Keratinocytes/metabolism , Mice , Mice, Transgenic , Response Elements , Skin/physiopathology , Wound Healing
20.
Endocrinology ; 153(6): 2777-88, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22492304

ABSTRACT

Although several lines of evidence have indicated that menopause is associated with increased susceptibility to neurological disorders, the mechanisms involved in this phenomenon remain to be elucidated. Because neuroinflammation is a common feature of a number of brain diseases, we hypothesized that the cessation of ovarian functions and the consequent decrease in estrogen receptor (ER)-mediated antiinflammatory activity may represent a trigger for postmenopausal brain dysfunctions. The aim of the present study was to investigate the effects of aging and surgical menopause on the activity of ER in neuroinflammation. The present study shows that ER genes are expressed in the hippocampus, but ER transcriptional activity decreases significantly beginning at 12 months of age in intact and ovariectomized mice. With ovariectomy, we observe an age-dependent accumulation of mRNA encoding inflammatory mediators (e.g. TNFα, IL1ß, and macrophage inflammatory protein-2) and changes in the morphology of astroglia and microglia. In addition, we show that aging itself is coupled with an exaggerated response to acute inflammatory stimuli with a major accumulation of TNFα, IL1ß, macrophage inflammatory protein-2, and macrophage chemoattractant protein-1 mRNA in response to lipopolysaccharide administration. The response to acute inflammatory stimuli appears to be differentially modulated by the duration of hormone deprivation in 12-month-old mice. Taken together, the present results show that aging is associated with decreased ER activity, despite continuous ER synthesis, and that age-dependent neuroinflammation is strongly influenced by hormone deprivation.


Subject(s)
Aging , Hippocampus/metabolism , Inflammation Mediators/metabolism , Ovariectomy , Receptors, Estrogen/genetics , Age Factors , Animals , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Hippocampus/drug effects , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , Injections, Intraventricular , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Male , Mice , Mice, Inbred C57BL , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...