Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Cells ; 12(3)2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36766684

RESUMEN

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.


Asunto(s)
Enfermedad de Parkinson , Animales , Femenino , Masculino , Ratones , Glucosilceramidasa/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo
2.
Nutrients ; 14(16)2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-36014766

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa , Receptor alfa de Estrógeno , Péptidos y Proteínas de Señalización Intercelular , Hígado , Enfermedad del Hígado Graso no Alcohólico , Animales , Dieta Alta en Grasa/efectos adversos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/genética , Estrógenos/metabolismo , Femenino , Homeostasis/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo
3.
Int J Mol Sci ; 23(11)2022 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-35683029

RESUMEN

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.


Asunto(s)
Neuropéptido Y , Receptores de Neuropéptido Y , Animales , Femenino , Hormonas Gonadales , Masculino , Ratones , Enfermedades Neuroinflamatorias , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Obesidad/genética , Obesidad/metabolismo , Prosencéfalo/metabolismo , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo
4.
Endocrinology ; 163(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34758075

RESUMEN

The number of studies illuminating major sex differences in liver metabolic activities is growing, but we still lack a theory to explain the origin of the functional differences we are identifying. In the animal kingdom, energy metabolism is tightly associated with reproduction; conceivably, the major evolutionary step that occurred about 200 million years ago with placentation determined a significant change in female physiology, as females had to create new energy strategies to allow the growth of the embryo in the womb and the lactation of the newborn. In vertebrates the liver is the metabolic organ most tuned to gonadal functions because the liver synthesizes and transports of all the components necessary for the maturation of the egg upon estrogenic stimulation. Thus, in mammals, evolution must have worked on the already strict gonad-liver relationship fostering these novel reproductive needs. As a consequence, the functions of mammalian liver in females diverged from that in males to acquire the flexibility necessary to tailor metabolism according to reproductive status and to ensure the parsimonious exploitation and storage of energy for the continuation of gestation in case of food scarcity. Indeed, several studies show that male and female livers adopt very different strategies when confronted with nutritional stress of varied origins. Considering the role of liver and energy metabolism in most pathologies, a better focus on liver functions in the 2 sexes might be of considerable help in personalizing medicine and pharmacology for male and female needs.


Asunto(s)
Regulación de la Expresión Génica , Hormonas Esteroides Gonadales/metabolismo , Hepatopatías/metabolismo , Enfermedades Metabólicas/metabolismo , Animales , Evolución Biológica , Femenino , Humanos , Hepatopatías/genética , Masculino , Enfermedades Metabólicas/genética , Factores Sexuales
5.
Nat Commun ; 12(1): 6883, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34824281

RESUMEN

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.


Asunto(s)
Aminoácidos Esenciales/farmacología , Receptor alfa de Estrógeno/metabolismo , Hígado/efectos de los fármacos , Ovariectomía/efectos adversos , Aminoácidos de Cadena Ramificada/farmacología , Aminoácidos de Cadena Ramificada/uso terapéutico , Aminoácidos Esenciales/uso terapéutico , Animales , Dietoterapia , Receptor alfa de Estrógeno/deficiencia , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Caracteres Sexuales , Transcriptoma/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
6.
Biomed Pharmacother ; 144: 112274, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34653752

RESUMEN

Sex differences in immune-mediated diseases are linked to the activity of estrogens on innate immunity cells, including macrophages. Tamoxifen (TAM) is a selective estrogen receptor modulator (SERM) used in estrogen receptor-alpha (ERα)-dependent breast cancers and off-target indications such as infections, although the immune activity of TAM and its active metabolite, 4-OH tamoxifen (4HT), is poorly characterized. Here, we aimed at investigating the endocrine and immune activity of these SERMs in macrophages. Using primary cultures of female mouse macrophages, we analyzed the expression of immune mediators and activation of effector functions in competition experiments with SERMs and 17ß-estradiol (E2) or the bacterial endotoxin LPS. We observed that 4HT and TAM induce estrogen antagonist effects when used at nanomolar concentrations, while pharmacological concentrations that are reached by TAM in clinical settings regulate the expression of VEGFα and other immune activation genes by ERα- and G protein-coupled receptor 1 (GPER1)-independent mechanisms that involve NRF2 through PI3K/Akt-dependent mechanisms. Importantly, we observed that SERMs potentiate cell phagocytosis and modify the effects of LPS on the expression of inflammatory cytokines, such as TNFα and IL1ß, with an overall increase in cell inflammatory phenotype, further sustained by potentiation of IL1ß secretion through caspase-1 activation. Altogether, our data unravel a novel molecular mechanism and immune functions for TAM and 4HT, sustaining their repurposing in infective and other estrogen receptors-unrelated pathologies.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Agentes Inmunomoduladores/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/análogos & derivados , Animales , Células Cultivadas , Receptor alfa de Estrógeno/genética , Femenino , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Fenotipo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Tamoxifeno/farmacología
7.
J Neuroinflammation ; 18(1): 220, 2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34551802

RESUMEN

BACKGROUND: Homozygotic mutations in the GBA gene cause Gaucher's disease; moreover, both patients and heterozygotic carriers have been associated with 20- to 30-fold increased risk of developing Parkinson's disease. In homozygosis, these mutations impair the activity of ß-glucocerebrosidase, the enzyme encoded by GBA, and generate a lysosomal disorder in macrophages, which changes morphology towards an engorged phenotype, considered the hallmark of Gaucher's disease. Notwithstanding the key role of macrophages in this disease, most of the effects in the brain have been attributed to the ß-glucocerebrosidase deficit in neurons, while a microglial phenotype for these mutations has never been reported. METHODS: We applied the bioluminescence imaging technology, immunohistochemistry and gene expression analysis to investigate the consequences of microglial ß-glucocerebrosidase inhibition in the brain of reporter mice, in primary neuron/microglia cocultures and in cell lines. The use of primary cells from reporter mice allowed for the first time, to discriminate in cocultures neuronal from microglial responses consequent to the ß-glucocerebrosidase inhibition; results were finally confirmed by pharmacological depletion of microglia from the brain of mice. RESULTS: Our data demonstrate the existence of a novel neuroprotective mechanism mediated by a direct microglia-to-neuron contact supported by functional actin structures. This cellular contact stimulates the nuclear factor erythroid 2-related factor 2 activity in neurons, a key signal involved in drug detoxification, redox balance, metabolism, autophagy, lysosomal biogenesis, mitochondrial dysfunctions, and neuroinflammation. The central role played by microglia in this neuronal response in vivo was proven by depletion of the lineage in the brain of reporter mice. Pharmacological inhibition of microglial ß-glucocerebrosidase was proven to induce morphological changes, to turn on an anti-inflammatory/repairing pathway, and to hinder the microglia ability to activate the nuclear factor erythroid 2-related factor 2 response, thus increasing the neuronal susceptibility to neurotoxins. CONCLUSION: This mechanism provides a possible explanation for the increased risk of neurodegeneration observed in carriers of GBA mutations and suggest novel therapeutic strategies designed to revert the microglial phenotype associated with ß-glucocerebrosidase inhibition, aimed at resetting the protective microglia-to-neuron communication.


Asunto(s)
Encéfalo/enzimología , Glucosilceramidasa/antagonistas & inhibidores , Microglía/enzimología , Neuronas/metabolismo , Neuroprotección/fisiología , Animales , Encéfalo/patología , Comunicación Celular/fisiología , Ratones , Microglía/patología , Neuronas/patología
8.
Prog Neurobiol ; 197: 101895, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32781107

RESUMEN

Women are more prone than men to develop age-related dementia, such as Alzheimer's disease (AD). This has been linked to the marked decrease in circulating estrogens during menopause. This review proposes to change this perspective and consider women's vulnerability to developing AD as a consequence of sex differences in the neurobiology of memory, focusing on the hippocampus. The hippocampus of cognitively impaired subjects tends to shrink with age; however, in many cases, this can be prevented by exercise or cognitive training, suggesting that if you do not use the hippocampus you lose it. We will review the developmental trajectory of sex steroids-regulated differences on the hippocampus, proposing that the overall shaping action of sex-steroids results in a lower usage of the hippocampus in females, which in turn makes them more vulnerable to the effects of ageing, the "network fragility hypothesis". To explain why women rely less on hippocampus-dependent strategies, we propose a "computational hypothesis" that is based on experimental evidence suggesting that the direct effects of estrogens on hippocampal synaptic and structural plasticity during the estrous-cycle confers instability to the memory-dependent hippocampal network. Finally, we propose to counteract AD with training and/or treatments, such as orienteering, which specifically favour the use of the hippocampus.


Asunto(s)
Hipocampo , Envejecimiento , Enfermedad de Alzheimer , Demencia , Estrógenos , Femenino , Humanos , Masculino , Factores de Riesgo
9.
Pharmacol Res Perspect ; 8(4): e00638, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32794353

RESUMEN

The metabolic and immune adaptation to extracellular signals allows macrophages to carry out specialized functions involved in immune protection and tissue homeostasis. Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor that coordinates cell redox and metabolic responses to stressors. However, the individual and concomitant activation of NRF2 and inflammatory pathways have been poorly investigated in isolated macrophages. We here took advantage of reporter mice for the transcriptional activities of NRF2 and nuclear factor-kB (NFκB), a key transcription factor in inflammation, and observe a persisting reciprocal interference in the response of peritoneal macrophages to the respective activators, tert-Butylhydroquinone (tBHQ) and lipopolysaccharide (LPS). When analyzed separately by gene expression studies, these pathways trigger macrophage-specific metabolic and proliferative target genes that are associated with tBHQ-induced pentose phosphate pathway (PPP) with no proliferative response, and with opposite effects observed with LPS. Importantly, the simultaneous administration of tBHQ + LPS alters the effects of each individual pathway in a target gene-specific manner. In fact, this co-treatment potentiates the effects of tBHQ on the antioxidant enzyme, HMOX1, and the antibacterial enzyme, IRG1, respectively; moreover, the combined treatment reduces tBHQ activity on the glycolytic enzymes, TALDO1 and TKT, and decreases LPS effects on the metabolic enzyme IDH1, the proliferation-related proteins KI67 and PPAT, and the inflammatory cytokines IL-1ß, IL-6, and TNFα. Altogether, our results show that the activation of NRF2 redirects the metabolic, immune, and proliferative response of peritoneal macrophages to inflammatory signals, with relevant consequences for the pharmacological treatment of diseases that are associated with unopposed inflammatory responses.


Asunto(s)
Inflamación/inmunología , Macrófagos Peritoneales/inmunología , FN-kappa B/genética , Transducción de Señal/inmunología , Animales , Proliferación Celular/fisiología , Citocinas/inmunología , Femenino , Genes Reporteros , Hidroquinonas/toxicidad , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética
10.
Mol Metab ; 32: 97-108, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32029233

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa , Receptor alfa de Estrógeno/metabolismo , Lípidos/administración & dosificación , Hígado/metabolismo , Caracteres Sexuales , Animales , Receptor alfa de Estrógeno/deficiencia , Femenino , Masculino , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/metabolismo
11.
Endocr Rev ; 41(2)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31544208

RESUMEN

Neurodegenerative diseases (NDs) are a wide class of disorders of the central nervous system (CNS) with unknown etiology. Several factors were hypothesized to be involved in the pathogenesis of these diseases, including genetic and environmental factors. Many of these diseases show a sex prevalence and sex steroids were shown to have a role in the progression of specific forms of neurodegeneration. Estrogens were reported to be neuroprotective through their action on cognate nuclear and membrane receptors, while adverse effects of male hormones have been described on neuronal cells, although some data also suggest neuroprotective activities. The response of the CNS to sex steroids is a complex and integrated process that depends on (i) the type and amount of the cognate steroid receptor and (ii) the target cell type-either neurons, glia, or microglia. Moreover, the levels of sex steroids in the CNS fluctuate due to gonadal activities and to local metabolism and synthesis. Importantly, biochemical processes involved in the pathogenesis of NDs are increasingly being recognized as different between the two sexes and as influenced by sex steroids. The aim of this review is to present current state-of-the-art understanding on the potential role of sex steroids and their receptors on the onset and progression of major neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, and the peculiar motoneuron disease spinal and bulbar muscular atrophy, in which hormonal therapy is potentially useful as disease modifier.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Receptores de Esteroides/metabolismo , Caracteres Sexuales , Femenino , Humanos , Masculino , Enfermedades Neurodegenerativas/tratamiento farmacológico
12.
Br J Cancer ; 120(5): 537-546, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30739913

RESUMEN

BACKGROUND: Nonsteroidal anti-inflammatory drugs (NSAIDs) have been proposed as chemopreventive agents for many tumours; however, the mechanism responsible for their anti-neoplastic activity remains elusive and the side effects due to cyclooxygenase (COX) inhibition prevent this clinical application. METHODS: Molecular biology, in silico, cellular and in vivo tools, including innovative in vivo imaging and classical biochemical assays, were applied to identify and characterise the COX-independent anti-cancer mechanism of NSAIDs. RESULTS: Here, we show that tumour-protective functions of NSAIDs and exisulind (a sulindac metabolite lacking anti-inflammatory activity) occur through a COX-independent mechanism. We demonstrate these NSAIDs counteract carcinogen-induced proliferation by inhibiting the sirtuin 1 (SIRT1) deacetylase activity, augmenting acetylation and activity of the tumour suppressor p53 and increasing the expression of the antiproliferative gene p21. These properties are shared by all NSAIDs except for ketoprofen lacking anti-cancer properties. The clinical interest of the mechanism identified is underlined by our finding that p53 is activated in mastectomy patients undergoing intraoperative ketorolac, a treatment associated with decreased relapse risk and increased survival. CONCLUSION: Our study, for the first-time, links NSAID chemopreventive activity with direct SIRT1 inhibition and activation of the p53/p21 anti-oncogenic pathway, suggesting a novel strategy for the design of tumour-protective drugs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Anticarcinógenos/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Sirtuina 1/efectos de los fármacos , Sulindac/análogos & derivados , Proteína p53 Supresora de Tumor/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Anticarcinógenos/efectos adversos , Línea Celular Tumoral , Simulación por Computador , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidores de la Ciclooxigenasa/efectos adversos , Humanos , Ketorolaco/efectos adversos , Ketorolaco/uso terapéutico , Ratones , Modelos Moleculares , Sirtuina 1/metabolismo , Sulindac/farmacología , Proteína p53 Supresora de Tumor/metabolismo
13.
Front Neuroendocrinol ; 52: 156-164, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30481522

RESUMEN

Sex plays a role in the incidence and outcome of neurological illnesses, also influencing the response to treatments. Despite sexual differentiation of the brain has been extensively investigated, the study of sex differences in microglia, the brain's resident immune cells, has been largely neglected until recently. To fulfill this gap, our laboratory developed several tools, including cellular and animal models, which bolstered in-depth studies on sexual differentiation of microglia and its impact on brain physiology, as well as on the onset and progression of neurological disorders. Here, we summarize the current status of knowledge on the sex-dependent function of microglia, and report recent evidence linking these cells to the sexual bias in the susceptibility to neurological brain diseases.


Asunto(s)
Encefalopatías , Encéfalo/crecimiento & desarrollo , Microglía/fisiología , Caracteres Sexuales , Diferenciación Sexual/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Encefalopatías/epidemiología , Encefalopatías/metabolismo , Encefalopatías/fisiopatología , Humanos
14.
Theranostics ; 8(19): 5400-5418, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555554

RESUMEN

Microglia are potential targets for therapeutic intervention in neurological and neurodegenerative diseases affecting the central nervous system. In order to assess the efficacy of therapies aimed to reduce the tissue damaging activities of microglia and/or to promote the protective potential of these cells, suitable pre-clinical and clinical tools for the in vivo analysis of microglia activities and dynamics are required. The aim of this work was to identify new translational markers of the anti-inflammatory / protective state of microglia for the development of novel PET tracers. Methods: New translational markers of the anti-inflammatory/protective activation state of microglia were selected by bioinformatic approaches and were in vitro and ex vivo validated by qPCR and immunohistochemistry in rodent and human samples. Once a viable marker was identified, a novel PET tracer was developed. This tracer was subsequently confirmed by autoradiography experiments in murine and human brain tissues. Results: Here we provide evidence that P2RY12 expression increases in murine and human microglia following exposure to anti-inflammatory stimuli, and that its expression is modulated in the reparative phase of experimental and clinical stroke. We then synthesized a novel carbon-11 labeled tracer targeting P2RY12, showing increased binding in brain sections of mice treated with IL4, and low binding to brain sections of a murine stroke model and of a stroke patient. Conclusion: This study provides new translational targets for PET tracers for the anti-inflammatory/protective activation state of microglia and shows the potential of a rationale-based approach. It therefore paves the way for the development of novel non-invasive methodologies aimed to monitor the success of therapeutic approaches in various neurological diseases.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/inmunología , Microglía/inmunología , Imagen Molecular/métodos , Tomografía de Emisión de Positrones/métodos , Animales , Antiinflamatorios/administración & dosificación , Radioisótopos de Carbono/administración & dosificación , Biología Computacional , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Interleucina-4/administración & dosificación , Ratones , Trazadores Radiactivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Purinérgicos P2Y12/análisis , Roedores , Accidente Cerebrovascular/patología
15.
Hum Reprod Update ; 24(6): 652-672, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30256960

RESUMEN

BACKGROUND: Estrogens are known to orchestrate reproductive events and to regulate the immune system during infections and following tissue damage. Recent findings suggest that, in the absence of any danger signal, estrogens trigger the physiological expansion and functional specialization of macrophages, which are immune cells that populate the female reproductive tract (FRT) and are increasingly being recognized to participate in tissue homeostasis beyond their immune activity against infections. Although estrogens are the only female gonadal hormones that directly target macrophages, a comprehensive view of this endocrine-immune communication and its involvement in the FRT is still missing. OBJECTIVE AND RATIONALE: Recent accomplishments encourage a revision of the literature on the ability of macrophages to respond to estrogens and induce tissue-specific functions required for reproductive events, with the aim to envision macrophages as key players in FRT homeostasis and mediators of the regenerative and trophic actions of estrogens. SEARCH METHODS: We conducted a systematic search using PubMed and Ovid for human, animal (rodents) and cellular studies published until 2018 on estrogen action in macrophages and the activity of these cells in the FRT. OUTCOMES: Our search identified the remarkable ability of macrophages to activate biochemical processes in response to estrogens in cell culture experiments. The distribution at specific locations, interaction with selected cells and acquisition of distinct phenotypes of macrophages in the FRT, as well as the cyclic renewal of these properties at each ovarian cycle, demonstrate the involvement of these cells in the homeostasis of reproductive events. Moreover, current evidence suggests an association between estrogen-macrophage signaling and the generation of a tolerant and regenerative environment in the FRT, although a causative link is still missing. WIDER IMPLICATIONS: Dysregulation of the functions and estrogen responsiveness of FRT macrophages may be involved in infertility and estrogen- and macrophage-dependent gynecological diseases, such as ovarian cancer and endometriosis. Thus, more research is needed on the physiology and pharmacological control of this endocrine-immune interplay.


Asunto(s)
Estrógenos/fisiología , Macrófagos/fisiología , Reproducción/fisiología , Animales , Endometriosis/metabolismo , Endometriosis/patología , Femenino , Genitales Femeninos/citología , Genitales Femeninos/metabolismo , Homeostasis/fisiología , Humanos , Infertilidad/metabolismo , Infertilidad/patología , Ciclo Menstrual/fisiología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Transducción de Señal/fisiología
16.
J Endocrinol ; 238(3): 165-176, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30012715

RESUMEN

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.


Asunto(s)
Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Receptores de Estrógenos/fisiología , Animales , Embrión de Mamíferos , Desarrollo Embrionario/efectos de los fármacos , Estrógenos/farmacología , Femenino , Fulvestrant/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Luciferasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo , Elementos de Respuesta/efectos de los fármacos , Elementos de Respuesta/genética , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
17.
Cell Rep ; 23(12): 3501-3511, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29924994

RESUMEN

Sex has a role in the incidence and outcome of neurological illnesses, also influencing the response to treatments. Neuroinflammation is involved in the onset and progression of several neurological diseases, and the fact that estrogens have anti-inflammatory activity suggests that these hormones may be a determinant in the sex-dependent manifestation of brain pathologies. We describe significant differences in the transcriptome of adult male and female microglia, possibly originating from perinatal exposure to sex steroids. Microglia isolated from adult brains maintain the sex-specific features when put in culture or transplanted in the brain of the opposite sex. Female microglia are neuroprotective because they restrict the damage caused by acute focal cerebral ischemia. This study therefore provides insight into a distinct perspective on the mechanisms underscoring a sexual bias in the susceptibility to brain diseases.


Asunto(s)
Envejecimiento/fisiología , Microglía/fisiología , Caracteres Sexuales , Animales , Encéfalo/metabolismo , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Progresión de la Enfermedad , Estradiol/sangre , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Microglía/trasplante , Fenotipo , Ratas Sprague-Dawley , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/patología , Transcriptoma/genética
18.
Cell Metab ; 28(2): 256-267.e5, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-29909969

RESUMEN

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.


Asunto(s)
Aminoácidos/metabolismo , Receptor alfa de Estrógeno/fisiología , Ayuno/metabolismo , Lipogénesis/fisiología , Hígado/metabolismo , Caracteres Sexuales , Animales , Aromatasa/metabolismo , Metabolismo Energético , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Femenino , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores Sexuales
19.
Sci Rep ; 8(1): 6580, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29700413

RESUMEN

The P2X7 receptor plays a significant role in microglial activation, and as a potential drug target, the P2X7 receptor is also an interesting target in positron emission tomography. The current study aimed at the development and evaluation of a potent tracer targeting the P2X7 receptor, to which end four adamantanyl benzamide analogues with high affinity for the human P2X7 receptor were labelled with carbon-11. All four analogues could be obtained in excellent radiochemical yield and high radiochemical purity and molar activity, and all analogues entered the rat brain. [11C]SMW139 showed the highest metabolic stability in rat plasma, and showed high binding to the hP2X7 receptor in vivo in a hP2X7 receptor overexpressing rat model. Although no significant difference in binding of [11C]SMW139 was observed between post mortem brain tissue of Alzheimer's disease patients and that of healthy controls in in vitro autoradiography experiments, [11C]SMW139 could be a promising tracer for P2X7 receptor imaging using positron emission tomography, due to high receptor binding in vivo in the hP2X7 receptor overexpressing rat model. However, further investigation of both P2X7 receptor expression and binding of [11C]SMW139 in other neurological diseases involving microglial activation is warranted.


Asunto(s)
Microglía/metabolismo , Imagen Molecular , Tomografía de Emisión de Positrones , Antagonistas del Receptor Purinérgico P2X/química , Radiofármacos/química , Receptores Purinérgicos P2X7/química , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Humanos , Microglía/efectos de los fármacos , Estructura Molecular , Tomografía de Emisión de Positrones/métodos , Unión Proteica , Antagonistas del Receptor Purinérgico P2X/farmacocinética , Radioquímica , Radiofármacos/farmacocinética , Ratas , Coloración y Etiquetado , Distribución Tisular
20.
Mol Metab ; 15: 3-7, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29550349

RESUMEN

BACKGROUND: Epidemiological and clinical studies have largely demonstrated major differences in the prevalence of metabolic disorders in males and females, but the biological cause of these dissimilarities remain to be elucidated. Mammals are characterized by a major change in reproductive strategies and it is conceivable that these changes subjected females to a significant evolutionary pressure that perfected the coupling between energy metabolism and reproduction. SCOPE OF REVIEW: This review will address the plausibility that female liver functions diverged significantly from males given the role of liver in the control of metabolism. Indeed, it is well known that the liver is sexually dimorphic, and this might be relevant to explain the lower susceptibility to hepatic diseases and liver-derived metabolic disturbances (such as the cardiovascular diseases) characteristic of females during their fertile period. Furthermore, estrogens and the hepatic ERα play a significant role in liver sexual-specific functions and in the control of metabolic functions. CONCLUSIONS: A better grasp of the role of male and female sex steroids in the liver of the two sexes may therefore represent an important element to conceive novel treatments aimed at preventing metabolic diseases particularly in ageing women or limiting undesired side effect in the treatment of gender dysphoria.


Asunto(s)
Hepatopatías/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Caracteres Sexuales , Animales , Femenino , Hormonas Gonadales/metabolismo , Humanos , Hígado/fisiología , Hepatopatías/epidemiología , Masculino , Enfermedades Metabólicas/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...