Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Environ Res ; 258: 119476, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38909949

RESUMO

The present study aims to analyze the effects of developmental exposure to phthalates at environmentally relevant doses on the neural control of male and female reproduction. For this purpose, C57Bl/6J mice were exposed to di-(2-ethylexyl) phthalate (DEHP) alone (5 or 50 µg/kg/d), or DEHP (5 µg/kg/d) in a phthalate mixture. Exposure through diet started 6 weeks before the first mating and lasted until weaning of litters from the second gestation (multiparous dams). Analyses of offspring born from multiparous dams exposed to DEHP alone or in a phthalate mixture showed that females experienced a delayed pubertal onset, and as adults they had prolonged estrous cyclicity and reduced Kiss1 expression in the preoptic area and mediobasal hypothalamus. Male littermates showed a reduced anogenital distance and delayed pubertal onset compared with controls. However, in adulthood the weight of androgen-sensitive organs and hypothalamic Kiss1 expression were unaffected, suggesting normal functioning of the male gonadotropic axis. Developmental exposure to DEHP alone or in a phthalate mixture reduced the ability of intact males and ovariectomized and hormonally primed females to attract a sexual partner and to express copulatory behaviors. In addition, females were unable to discriminate between male and female stimuli in the olfactory preference test. Social interaction was also impaired in females, while locomotor activity and anxiety-like behavior in both sexes were unaffected by the treatment. The sexual deficiencies were associated with reduced expression of the androgen receptor in the preoptic area and progesterone receptor in the mediobasal hypothalamus, the key regions involved in male and female sexual behavior, respectively. Thus, the neural structures controlling reproduction are vulnerable to developmental exposure to phthalates at environmentally relevant doses in male and female mice. Adult females had an impaired gonadotropic axis and showed more affected behaviors than adult males.

2.
Neuroendocrinology ; 113(12): 1215-1231, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37903467

RESUMO

BACKGROUND: The nervous system is a sensitive target for exposure to environmental endocrine-disrupting compounds (EDC). This vulnerability is particularly important during the critical windows of development and puberty and lasts even at later stages of life. Among these environmental EDC, phthalates have largely been described for their neurotoxic effects. These effects have been reported for a large majority of studies using high to very high doses of these substances, which are not relevant for environmental exposure. SUMMARY: The aim of this review was to analyze specifically the male rodent studies using low doses of phthalates. This analysis focuses on reproductive and cognitive behaviors, given the described antiandrogenic effects of phthalates and the known regulation of these behaviors by sex steroids. We also analyze the other neural effects in the hypothalamus and hippocampus/cortex, the brain regions governing these behaviors. A particular focus is on the neurovascular unit, which is newly investigated in the field of endocrine disruption. KEY MESSAGES: Exposure to low doses of phthalates can induce modifications in reproductive and cognitive behaviors. Whether these changes are triggered by common initiating cellular and molecular mechanisms in the brain areas controlling these behaviors still needs to be extensively investigated. In this context, given the high sensitivity of the neurovascular unit to sex steroid regulation and its impairment by low doses of phthalates, it could represent a possible initiating trigger for behavioral alterations to assess for phthalate exposure.


Assuntos
Disruptores Endócrinos , Ácidos Ftálicos , Animais , Masculino , Roedores , Maturidade Sexual , Ácidos Ftálicos/toxicidade , Disruptores Endócrinos/toxicidade , Cognição
3.
Environ Res ; 222: 115330, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36693459

RESUMO

Severe hypothyroidism has been reported in humans during resorcinol therapeutic use. However, available data highlight differences in the severity of resorcinol-induced thyroid effects between humans and rodents, leading to a debate on the relevance of human data for its classification as a thyroid disruptor. The aim of this review is to illustrate some of the limitations of the evaluation framework for thyroid disrupters using resorcinol as a case study of a chemical with clear thyroid-disrupting properties in humans that could not have been identified solely from regulatory studies on animals. The reliability of human data has been called into question due to the specific exposure patterns in humans and the paucity of robust toxicokinetic data. In humans, therapeutic use of resorcinol induces severe hypothyroidism, but in rodents, thyroid disruption is limited to decreased thyroxine concentrations and histological changes in the thyroid. The adverse effects of thyroid disruption, such as impaired neurodevelopment, have not been sufficiently investigated, and experimental neurobehavioral data for resorcinol remain scarce and inconclusive. Although regulatory toxicological evaluations have not included in-depth investigations of thyroid regulation and related adverse effects, they have been used to challenge the relevance of human data. Resorcinol is an emblematic example of how the framework for regulatory evaluations of thyroid disruptors relies almost exclusively on animal studies which may not be suitable for assessing thyroid disruption. This review highlights the need to revise regulatory guidelines and to adopt strategies based on up-to-date, scientifically sound approaches to identify thyroid disruptors. The limits of the current regulatory framework for identifying thyroid disruptors can lead to opposing positions between regulatory bodies. The French Agency for Food, Environmental and Occupational Health & Safety (ANSES)'proposal to identify resorcinol as a "substance of very high concern" due to its ED properties has not been adopted by the European instances.


Assuntos
Disruptores Endócrinos , Hipotireoidismo , Animais , Humanos , Reprodutibilidade dos Testes , Hipotireoidismo/induzido quimicamente , Resorcinóis/toxicidade
4.
Front Neuroendocrinol ; 63: 100949, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34687674

RESUMO

Male sexual behavior is subject to learning, resulting in increased efficiency of experienced males compared to naive ones. The improvement in behavioral parameters is underpinned by cellular and molecular changes in the neural circuit controlling sexual behavior, particularly in the hypothalamic medial preoptic area. This review provides an update on the mechanisms related to the sexual experience in male rodents, emphasizing the differences between rats and mice.


Assuntos
Área Pré-Óptica , Comportamento Sexual Animal , Animais , Hipotálamo , Masculino , Camundongos , Ratos
5.
Environ Res ; 207: 112235, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34678253

RESUMO

We have previously shown that adult male mice exposure to low doses of di (2-ethylhexyl)phthalate (DEHP) impacts the blood-brain barrier (BBB) integrity and surrounding parenchyma in the medial preoptic area (mPOA), a key hypothalamic area involved in the male sexual behavior. BBB leakage was associated with a decrease in the endothelial tight junction accessory protein, zona occludens-1, and caveolae protein Cav-1, added to an inflammatory profile including glial activation accompanied by enhanced expression of inducible nitric oxide synthase. As this failure of BBB functionality in the mPOA could participate, at least in part, in reported alteration of sexual behavior following DEHP exposure, we explored the cellular pathway connecting cerebral capillaries and neurons. Two-month-old C57BL/6J male mice were orally exposed for 6 weeks to DEHP alone (5 and 50 µg/kg/day) or to DEHP (5 µg/kg/day) in an environmental phthalate mixture. The presence of androgen receptor (AR) and estrogen receptor-α (ERα) were first evidenced in brain capillaries. Protein levels of AR but not of ERα were reduced in cerebral capillaries after phthalate exposure. The amounts of basement membrane and cell-matrix interaction components were decreased, while matrix metalloprotease MMP-2 and MMP-9 activities were increased. Fluorojade® labelling suggested that exposure to phthalates also lead to a neurodegenerative process in the mPOA. Altogether, the data suggest that environmental exposure to endocrine disruptors such as phthalates, could alter AR/Cav-1 interaction, impacting a Cav-1/nitric oxide/MMP pathway. This would lead to disruption of the glio-neurovascular coupling which is essential to neuronal functioning.


Assuntos
Dietilexilftalato , Disruptores Endócrinos , Ácidos Ftálicos , Animais , Dietilexilftalato/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
Environ Res ; 205: 112495, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34883077

RESUMO

Pharmaceutical drugs have become consumer products, with a daily use for some of them. The volume of production and consumption of drugs is such that they have become environmental pollutants. Their transfer to wastewater through urine, feces or rinsing in case of skin use, associated with partial elimination by wastewater treatment plants generalize pollution in the hydrosphere, including drinking water, sediments, soils, the food chain and plants. Here, we review the potential effects of environmental exposure to three classes of pharmaceutical drugs, i.e. antibiotics, antidepressants and non-steroidal anti-inflammatory drugs, on neurodevelopment. Experimental studies analyzing their underlying modes of action including those related to endocrine disruption, and molecular mechanisms including epigenetic modifications are presented. In addition, the contribution of brain imaging to the assessment of adverse effects of these three classes of pharmaceuticals is approached.


Assuntos
Poluentes Ambientais , Poluentes Químicos da Água , Monitoramento Ambiental/métodos , Poluentes Ambientais/toxicidade , Preparações Farmacêuticas , Águas Residuárias , Poluentes Químicos da Água/análise , Poluentes Químicos da Água/toxicidade
7.
Environ Res ; 204(Pt C): 112233, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34688643

RESUMO

The close structural analogy of bisphenol (BP) S with BPA, a recognized endocrine-disrupting chemical and a substance of very high concern in the European Union, highlights the need to assess the extent of similarities between the two compounds and carefully scrutinize BPS potential toxicity for human health. This analysis aimed to investigate human health toxicity data regarding BPS, to find a point of departure for the derivation of human guidance values. A systematic and transparent methodology was applied to determine whether European or international reference values have been established for BPS. In the absence of such values, the scientific literature on human health effects was evaluated by focusing on human epidemiological and animal experimental studies. The results were analyzed by target organ/system: male and female reproduction, mammary gland, neurobehavior, and metabolism/obesity. Academic experimental studies were analyzed and compared to regulatory data including subchronic studies and an extended one-generation and reproduction study. In contrast to the regulatory studies, which were performed at dose levels in the mg/kg bw/day range, the academic dataset on specific target organs or systems showed adverse effects for BPS at much lower doses (0.5-10 µg/kg bw/day). A large disparity between the lowest-observed-adverse-effect levels (LOAELs) derived from regulatory and academic studies was observed for BPS, as for BPA. Toxicokinetic data on BPS from animal and human studies were also analyzed and showed a 100-fold higher oral bioavailability compared to BPA in a pig model. The similarities and differences between the two bisphenols, in particular the higher bioavailability of BPS in its active (non-conjugated) form and its potential impact on human health, are discussed. Based on the available experimental data, and for a better human protection, we propose to derive human reference values for exposure to BPS from the N(L)OAELs determined in academic studies.


Assuntos
Disruptores Endócrinos , Sulfonas , Animais , Compostos Benzidrílicos/toxicidade , Disponibilidade Biológica , Disruptores Endócrinos/toxicidade , Feminino , Humanos , Masculino , Fenóis , Valores de Referência , Sulfonas/toxicidade , Suínos
8.
Int J Mol Sci ; 23(5)2022 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-35269705

RESUMO

Phthalates have been widely studied for their reprotoxic effects in male rodents and in particular on testosterone production, for which reference doses were established. The female rodent brain can also represent a target for exposure to these environmental endocrine disruptors. Indeed, a large range of behaviors including reproductive behaviors, mood-related behaviors, and learning and memory are regulated by sex steroid hormones. Here we review the experimental studies addressing the effects and mechanisms of phthalate exposure on these behaviors in female rodents, paying particular attention to the experimental conditions (period of exposure, doses, estrous stage of analyses etc.). The objective of this review is to provide a clear picture of the consistent effects that can occur in female rodents and the gaps that still need to be filled in terms of effects and mode(s) of action for a better risk assessment for human health.


Assuntos
Disruptores Endócrinos , Ácidos Ftálicos , Animais , Disruptores Endócrinos/toxicidade , Exposição Ambiental , Feminino , Masculino , Ácidos Ftálicos/toxicidade , Roedores , Testosterona
9.
Int J Mol Sci ; 23(6)2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35328575

RESUMO

This review provides an overview of the assessment of the endocrine disrupting (ED) properties of carbon disulfide (CS2), following the methodology used at the European level to identify endocrine disruptors. Relevant in vitro, in vivo studies and human data are analyzed. The assessment presented here focuses on one endocrine activity, i.e., thyroid disruption, and two main adverse effects, neurotoxicity and cardiotoxicity. The data available on the different ED or non-ED modes of action (MoA), known to trigger these adverse effects, are described and the strength of evidence of the different MoA is weighted. We conclude that the adverse effects could be due to systemic toxicity rather than endocrine-mediated toxicity. This assessment illustrates the scientific and regulatory challenges in differentiating a specific endocrine disruption from an indirect endocrine effect resulting from a non-ED mediated systemic toxicity. This issue of evaluating the ED properties of highly toxic and reactive substances has been insufficiently developed by European guidance so far and needs to be further addressed. Finally, this example also raises questions about the capacity of the technics available in toxicology to address such a complex issue with certainty.


Assuntos
Dissulfeto de Carbono , Disruptores Endócrinos , Dissulfeto de Carbono/toxicidade , Disruptores Endócrinos/toxicidade , Sistema Endócrino , Humanos , Medição de Risco/métodos , Glândula Tireoide
10.
Int J Mol Sci ; 23(22)2022 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-36430167

RESUMO

Mini-puberty of infancy is a short developmental phase occurring in humans and other mammals after birth. In females, it corresponds to transient and robust activation of the hypothalamo-pituitary-ovarian (HPO) axis revealed by high levels of gonadotropin hormones, follicular growth, and increased estradiol production by the ovary. The roles of estradiol signaling during this intriguing developmental phase are not yet well known, but accumulating data support the idea that it aids in the implementation of reproductive function. This review aims to provide in-depth information on HPO activity during this particular developmental phase in several mammal species, including humans, and to propose emerging hypotheses on the putative effect of estradiol signaling on the development and function of organs involved in female reproduction.


Assuntos
Estradiol , Maturidade Sexual , Camundongos , Humanos , Animais , Feminino , Estradiol/farmacologia , Ovário , Hipófise , Transdução de Sinais , Mamíferos
11.
Proc Natl Acad Sci U S A ; 113(51): 14829-14834, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27930320

RESUMO

Lost myelin can be replaced after injury or during demyelinating diseases in a regenerative process called remyelination. In the central nervous system (CNS), the myelin sheaths, which protect axons and allow the fast propagation of electrical impulses, are produced by oligodendrocytes. The abundance and widespread distribution of oligodendrocyte progenitors (OPs) within the adult CNS account for this remarkable regenerative potential. Here, we report a key role for the male gonad, testosterone, and androgen receptor (AR) in CNS remyelination. After lysolecithin-induced demyelination of the male mouse ventral spinal cord white matter, the recruitment of glial fibrillary acidic protein-expressing astrocytes was compromised in the absence of testes and testosterone signaling via AR. Concomitantly, the differentiation of OPs into oligodendrocytes forming myelin basic protein (MBP)+ and proteolipid protein-positive myelin was impaired. Instead, in the absence of astrocytes, axons were remyelinated by protein zero (P0)+ and peripheral myelin protein 22-kDa (PMP22)+ myelin, normally only produced by Schwann cells in the peripheral nervous system. Thus, testosterone favors astrocyte recruitment and spontaneous oligodendrocyte-mediated remyelination. This finding may have important implications for demyelinating diseases, psychiatric disorders, and cognitive aging. The testosterone dependency of CNS oligodendrocyte remyelination may have roots in the evolutionary history of the AR, because the receptor has evolved from an ancestral 3-ketosteroid receptor through gene duplication at the time when myelin appeared in jawed vertebrates.


Assuntos
Bainha de Mielina/metabolismo , Receptores Androgênicos/metabolismo , Remielinização , Androgênios/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Neuroglia/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Receptores de Esteroides/metabolismo , Células de Schwann/metabolismo , Transdução de Sinais , Testículo/metabolismo , Testosterona/metabolismo
12.
Neuroendocrinology ; 107(4): 400-416, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30326485

RESUMO

Sexual behavior constitutes a chain of behavioral responses beginning with courtship and leading to copulation. These responses, which are exhibited in a sexually dimorphic manner by the two partners, are tightly regulated by sex steroid hormones as early as the perinatal period. Hormonal changes or exposure to exogenous factors exhibiting hormone-mimetic activities, such as endocrine disrupting compounds (EDC), can therefore interfere with their expression. Here we review the experimental studies in rodents performed to address the potential effects of exposure to EDC on sexual behavior and underlying mechanisms, with particular attention to molecules with estrogenic and/or anti-androgenic activities.


Assuntos
Disruptores Endócrinos/farmacologia , Hormônios Esteroides Gonadais/farmacologia , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Feminino , Masculino , Camundongos , Ratos , Roedores , Caracteres Sexuais
13.
Hum Mol Genet ; 24(25): 7326-38, 2015 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-26464488

RESUMO

Ovarian oestradiol is essential for pubertal maturation and adult physiology of the female reproductive axis. It acts at central and peripheral sites through two main oestrogen receptors (ER) α and ß. Here we investigate the role of ERß on central effects of oestradiol, by generating a mouse line specifically lacking the ERß gene in neuronal and glial cells. Central ERß deletion delays the age at vaginal opening and first oestrous and reduces uterine weight without affecting body growth. Analysis of factors necessary for pubertal progression shows reduced levels of Kiss1 transcripts at postnatal (P) day 25 in the preoptic area, but not in the mediobasal hypothalamus (MBH) of mutant females. In agreement with these data, the number of kisspeptin-immunoreactive neurons was decreased by 57-72% in the three subdivisions of the rostral periventricular area of the third ventricle (RP3V), whereas the density of kisspeptin-immunoreactive fibres was unchanged in the arcuate nucleus of mutant mice. These alterations do not involve changes in ERα mRNAs in the preoptic area and protein levels in the RP3V. The number and distribution of GnRH-immunoreactive cells were unaffected, but gonadotropin-releasing hormone (GnRH) transcript levels were higher in the P25 preoptic area of mutants. At adulthood, mutant females have normal oestrous cyclicity, kisspeptin system and exhibit unaltered sexual behaviour. They display, however, reduced ovary weight and increased anxiety-related behaviour during the follicular phase. This argues for the specific involvement of central ERß in the regulation of pubertal onset in female reproduction, possibly through prepubertal induction of kisspeptin expression in the RP3V.


Assuntos
Receptor beta de Estrogênio/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Animais , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Ciclo Estral/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Camundongos , Puberdade/genética , Puberdade/metabolismo
14.
Horm Behav ; 80: 1-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26836767

RESUMO

Estradiol derived from neural aromatization of gonadal testosterone plays a key role in the perinatal organization of the neural circuitry underlying male sexual behavior. The aim of this study was to investigate the contribution of neural estrogen receptor (ER) ß in estradiol-induced effects without interfering with its peripheral functions. For this purpose, male mice lacking ERß in the nervous system were generated. Analyses of males in two consecutive tests with a time interval of two weeks showed an effect of experience, but not of genotype, on the latencies to the first mount, intromission, pelvic thrusting and ejaculation. Similarly, there was an effect of experience, but not of genotype, on the number of thrusts and mating length. Neural ERß deletion had no effect on the ability of males to adopt a lordosis posture in response to male mounts, after castration and priming with estradiol and progesterone. Indeed, only low percentages of both genotypes exhibited a low lordosis quotient. It also did not affect their olfactory preference. Quantification of tyrosine hydroxylase- and kisspeptin-immunoreactive neurons in the preoptic area showed unaffected sexual dimorphism of both populations in mutants. By contrast, the number of androgen receptor- and ERα-immunoreactive cells was significantly increased in the bed nucleus of stria terminalis of mutant males. These data show that neural ERß does not play a crucial role in the organization and activation of the neural circuitry underlying male sexual behavior. These discrepancies with the phenotype of global ERß knockout models are discussed.


Assuntos
Receptor beta de Estrogênio/genética , Camundongos , Mutagênese/genética , Gravidez , Comportamento Sexual Animal/fisiologia , Animais , Deleção Cromossômica , Feminino , Fertilidade/genética , Hipotálamo Anterior/metabolismo , Masculino , Camundongos Knockout , Neuroglia/metabolismo , Neurônios/metabolismo , Área Pré-Óptica/fisiologia , Núcleos Septais/metabolismo
15.
Brain ; 136(Pt 1): 132-46, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23365095

RESUMO

Myelin regeneration is a major therapeutic goal in demyelinating diseases, and the failure to remyelinate rapidly has profound consequences for the health of axons and for brain function. However, there is no efficient treatment for stimulating myelin repair, and current therapies are limited to anti-inflammatory agents. Males are less likely to develop multiple sclerosis than females, but often have a more severe disease course and reach disability milestones at an earlier age than females, and these observations have spurred interest in the potential protective effects of androgens. Here, we demonstrate that testosterone treatment efficiently stimulates the formation of new myelin and reverses myelin damage in chronic demyelinated brain lesions, resulting from the long-term administration of cuprizone, which is toxic for oligodendrocytes. In addition to the strong effect of testosterone on myelin repair, the number of activated astrocytes and microglial cells returned to low control levels, indicating a reduction of neuroinflammatory responses. We also identify the neural androgen receptor as a novel therapeutic target for myelin recovery. After the acute demyelination of cerebellar slices in organotypic culture, the remyelinating actions of testosterone could be mimicked by 5α-dihydrotestosterone, a metabolite that is not converted to oestrogens, and blocked by the androgen receptor antagonist flutamide. Testosterone treatment also failed to promote remyelination after chronic cuprizone-induced demyelination in mice with a non-functional androgen receptor. Importantly, testosterone did not stimulate the formation of new myelin sheaths after specific knockout of the androgen receptor in neurons and macroglial cells. Thus, the neural brain androgen receptor is required for the remyelination effect of testosterone, whereas the presence of the receptor in microglia and in peripheral tissues is not sufficient to enhance remyelination. The potent synthetic testosterone analogue 7α-methyl-19-nortestosterone, which has been developed for long-term male contraception and androgen replacement therapy in hypogonadal males and does not stimulate prostate growth, also efficiently promoted myelin repair. These data establish the efficacy of androgens as remyelinating agents and qualify the brain androgen receptor as a promising drug target for remyelination therapy, thus providing the preclinical rationale for a novel therapeutic use of androgens in males with multiple sclerosis.


Assuntos
Encéfalo/metabolismo , Doenças Desmielinizantes/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Receptores Androgênicos/metabolismo , Androgênios/farmacologia , Androgênios/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/patologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/patologia , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia , Receptores Androgênicos/genética , Testosterona/farmacologia , Testosterona/uso terapêutico
16.
Front Endocrinol (Lausanne) ; 15: 1408677, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38978624

RESUMO

Infertility is becoming a major public health problem, with increasing frequency due to medical, environmental and societal causes. The increasingly late age of childbearing, growing exposure to endocrine disruptors and other reprotoxic products, and increasing number of medical reproductive dysfunctions (endometriosis, polycystic ovary syndrome, etc.) are among the most common causes. Fertility relies on fine-tuned control of both neuroendocrine function and reproductive behaviors, those are critically regulated by sex steroid hormones. Testosterone and estradiol exert organizational and activational effects throughout life to establish and activate the neural circuits underlying reproductive function. This regulation is mediated through estrogen receptors (ERs) and androgen receptor (AR). Estradiol acts mainly via nuclear estrogen receptors ERα and ERß. The aim of this review is to summarize the genetic studies that have been undertaken to comprehend the specific contribution of ERα and ERß in the neural circuits underlying the regulation of the hypothalamic-pituitary-gonadal axis and the expression of reproductive behaviors, including sexual and parental behavior. Particular emphasis will be placed on the neural role of these receptors and the underlying sex differences.


Assuntos
Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Reprodução , Humanos , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Animais , Reprodução/fisiologia , Feminino , Comportamento Reprodutivo/fisiologia , Masculino
17.
Environ Int ; : 108861, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38991890

RESUMO

Lithium is a key medication for the treatment of psychiatric disorders and is also used in various industrial applications (including battery production and recycling). Here, we review published data on the endocrine-disrupting potential of lithium, with a particular focus on the thyroid hormone system. To this end, we used PubMed and Scopus databases to search for, select and review primary research addressing human and animal health endpoints during or after lithium exposure at non-teratogenic doses. Given the key role of thyroid hormones in neurodevelopmental processes, we focused at studies of the neural effects of developmental exposure to lithium in humans and animals. Our results show that lithium meets the World Health Organization's definition of a thyroid hormone system disruptor - particularly when used at therapeutic doses. When combined with knowledge of adverse outcome pathways linking molecular initiating events targeting thyroid function and neurodevelopmental outcomes, the neurodevelopmental data reported in animal experiments prompt us to suggest that lithium influences neurodevelopment. However, we cannot rule out the involvement of additional modes of action (i.e. unrelated to the thyroid hormone system) in the described neural effects. Given the increasing use of lithium salts in new technologies, attention must be paid to this emerging pollutant - particularly with regard to its potential effects at environmental doses on the thyroid hormone system and potential consequences on the developing nervous system.

18.
Gen Comp Endocrinol ; 188: 218-25, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23583766

RESUMO

In the present study, we investigated the role of the androgen receptor (AR) in the nervous system in the regulation of aggressive behavior and arginine vasopressin and galanin systems by testosterone. For this purpose, we used a conditional mouse line selectively lacking AR gene in the nervous system, backcrossed onto the C57BL/6J strain. Adult males were gonadectomized and supplemented with similar amounts of testosterone. When tested on two consecutive days in the resident intruder paradigm, fewer males of the mutant group exhibited aggressive behavior compared to their control littermates. In addition, a high latency to the first offensive attack was observed for the few animals that exhibited fighting behavior. This alteration was associated with a normal anogenital chemoinvestigation of intruder males. In olfactory discrimination tasks, sexual experience enhanced preference towards female-soiled bedding rather than male-soiled bedding and estrus females rather than intact males, regardless of genotype. This indicated that the behavioral alteration induced by neural AR mutation occurs in brain areas located downstream from the olfactory bulb. Quantification of the sexually dimorphic cell populations expressing preprovasopressin and galanin mRNAs in the bed nucleus of stria terminalis (BNST) and vasopressin-neurophysin 2 and galanin immunoreactivity in the lateral septum showed no significant differences between the two genotypes. The present findings indicate that the neural AR is required in the expression of aggressive behavior but not in the sexual differentiation of AVP and galanin cell number in the BNST and fiber immunoreactivity in the lateral septum. They also suggest that AR in the nervous system could mediate activational effects of testosterone in the regulation of aggressive behavior during adulthood.


Assuntos
Arginina Vasopressina/metabolismo , Galanina/metabolismo , Receptores Androgênicos/metabolismo , Agressão/fisiologia , Animais , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Nervoso/metabolismo , RNA Mensageiro/genética , Receptores Androgênicos/genética , Testosterona/metabolismo
19.
Environ Pollut ; 323: 121341, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-36828353

RESUMO

We recently showed that chronic exposure of adult male mice to environmental doses of DEHP alone or in a phthalate mixture altered blood brain barrier integrity and induced an inflammatory profile in the hippocampus. Here, we investigate whether such exposure alters hippocampus-dependent behavior and underlying cellular mechanisms. Adult C57BL/6 J male mice were continuously exposed orally to the vehicle or DEHP alone (5 or 50 µg/kg/d) or to DEHP (5 µg/kg/d) in a phthalate mixture. In the Morris water maze, males showed reduced latencies across days to find the platform in the cue and spatial reference memory tasks, regardless of their treatment group. In the probe test, DEHP-50 exposed males displayed a higher latency to find the platform quadrant. In the temporal order memory test, males exposed to DEHP alone or in a phthalate mixture were unable to discriminate between the most recently and previously seen objects. They also displayed reduced ability to show a preference for the new object in the novel object recognition test. These behavioral alterations were associated with a lowered dendritic spine density and protein levels of glutamate receptors and postsynaptic markers, and increased protein levels of the presynaptic synaptophysin in the hippocampus. Metabolomic analysis of the hippocampus indicated changes in amino acid levels including reduced tryptophan and L-kynurenine and elevated NAD + levels, respectively, a precursor, intermediate and endproduct of the kynurenine pathway of tryptophan metabolism. Interestingly, the protein amounts of the xenobiotic aryl hydrocarbon receptor, a target of this metabolic pathway, were elevated in the CA1 area. These data indicate that chronic exposure of adult male mice to environmental doses of DEHP alone or in a phthalate mixture impacted hippocampal function and structure, associated with modifications in amino acid metabolites with a potential involvement of the kynurenine pathway of tryptophan metabolism.


Assuntos
Dietilexilftalato , Disruptores Endócrinos , Ácidos Ftálicos , Camundongos , Animais , Masculino , Dietilexilftalato/toxicidade , Cinurenina/farmacologia , Triptofano , Camundongos Endogâmicos C57BL , Ácidos Ftálicos/farmacologia , Hipocampo , Cognição , Disruptores Endócrinos/farmacologia
20.
Environ Health Perspect ; 131(7): 77008, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37458746

RESUMO

BACKGROUND: We have previously shown that chronic exposure of adult male mice to low doses of di(2-ethylhexyl) phthalate (DEHP) altered male sexual behavior and induced down-regulation of the androgen receptor (AR) in the neural circuitry controlling this behavior. OBJECTIVES: The cellular mechanisms induced by chronic exposure of adult male mice to low doses of DEHP alone or in an environmental phthalate mixture were studied. METHODS: Two-month-old C57BL/6J males were exposed orally for 8 wk to DEHP alone (0, 5, or 50µg/kg/d) or to DEHP (50µg/kg/d) in a phthalate mixture. Behavior, dendritic density per 50-µm length, pre-/postsynaptic markers, synapse ultrastructure, and bioenergetic activity were analyzed. RESULTS: Mice exposed to DEHP either alone or in a phthalate mixture differed in mating, emission of ultrasonic vocalizations, and the ability to attract receptive females in urinary preference tests from control mice. Analyses in the medial preoptic area, the key hypothalamic region involved in male sexual behavior, showed lower dendritic spine density and protein levels of glutamate receptors and differences in other postsynaptic components and presynaptic markers between the treated groups. Ultrastructural observation of dendritic synapses by electron microscopy showed comparable morphology between the treated groups. Metabolic analyses highlighted differences in hypothalamic metabolites of males exposed to DEHP alone or in a phthalate mixture compared to control mice. These differences included lower tryptophan and higher NAD+ levels, respectively, a precursor and end product of the kynurenine pathway of tryptophan metabolism. The protein amounts of the xenobiotic aryl hydrocarbon receptor, one of the targets of this metabolic pathway and known negative regulator of the AR, were higher in the medial preoptic area of exposed male mice. DISCUSSION: Differences in behavior of male mice exposed to environmental doses of phthalates were associated with differences in neural structure and metabolism, with possibly a key role of the kynurenine pathway of tryptophan metabolism in the effects mediated by these substances. https://doi.org/10.1289/EHP11514.


Assuntos
Dietilexilftalato , Ácidos Ftálicos , Feminino , Camundongos , Animais , Masculino , Dietilexilftalato/toxicidade , Triptofano , Cinurenina , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA