Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharmacol Res ; 197: 106956, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37820857

RESUMEN

Several immunopharmacological agents are effective in the treatment of cancer and immune-mediated conditions, with a favorable impact on life expectancy and clinical outcomes for a large number of patients. Nevertheless, response variation and undesirable effects of these drugs represent major issues, and overall efficacy remains unpredictable. Males and females show a distinct difference in immune system responses, with females generally mounting stronger responses to a variety of stimuli. Therefore, exploring sex differences in the efficacy and safety of immunopharmacological agents would strengthen the practice of precision medicine. As a pharmacological target highlight, programmed cell death 1 ligand 1 (PD-L1) is the first functionally characterized ligand of the coinhibitory programmed death receptor 1 (PD-1). The PD-L1/PD-1 crosstalk plays an important role in the immune response and is relevant in cancer, infectious and autoimmune disease. Sex differences in the response to immune checkpoint inhibitors are well documented, with male patients responding better than female patients. Similarly, higher efficacy of and adherence to tumor necrosis factor inhibitors in chronic inflammatory conditions including rheumatoid arthritis and Crohn's disease have been reported in male patients. The pharmacological basis of sex-specific responses to immune system modulating drugs is actively investigated in other settings such as stroke and type 1 diabetes. Advances in therapeutics targeting the endothelium could soon be wielded against autoimmunity and metabolic disorders. Based on the established sexual dimorphism in immune-related pathophysiology and disease presentation, sex-specific immunopharmacological protocols should be integrated into clinical guidelines.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Humanos , Masculino , Femenino , Receptor de Muerte Celular Programada 1 , Neoplasias/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Autoinmunidad
2.
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
3.
J Neuroinflammation ; 14(1): 236, 2017 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-29202771

RESUMEN

BACKGROUND: Microglia are resident myeloid cells of the central nervous system (CNS) that are maintained by self-renewal and actively participate in tissue homeostasis and immune defense. Under the influence of endogenous or pathological signals, microglia undertake biochemical transformations that are schematically classified as the pro-inflammatory M1 phenotype and the alternatively activated M2 state. Dysregulated proliferation of M1-activated microglia has detrimental effects, while an increased number of microglia with the alternative, pro-resolving phenotype might be beneficial in brain pathologies; however, the proliferative response of microglia to M2 signals is not yet known. We thus evaluated the ability of interleukin-4 (IL-4), a typical M2 and proliferative signal for peripheral macrophages, to induce microglia proliferation and compared it with other proliferative and M2 polarizing stimuli for macrophages, namely colony-stimulating factor-1 (CSF-1) and the estrogen hormone, 17ß-estradiol (E2). METHODS: Recombinant IL-4 was delivered to the brain of adult mice by intracerebroventricular (i.c.v.) injection; whole brain areas or ex vivo-sorted microglia were analyzed by real-time PCR for assessing the mRNA levels of genes related with cell proliferation (Ki67, CDK-1, and CcnB2) and M2 polarization (Arg1, Fizz1, Ym-1) or by FACS analyses of in vivo BrdU incorporation in microglia. Primary cultures of microglia and astrocytes were also tested for proliferative effects. RESULTS: Our results show that IL-4 only slightly modified the expression of cell cycle-related genes in some brain areas but not in microglia, where it strongly enhanced M2 gene expression; on the contrary, brain delivery of CSF-1 triggered proliferation as well as M2 polarization of microglia both in vivo and in vitro. Similar to IL-4, the systemic E2 administration failed to induce microglia proliferation while it increased M2 gene expression. CONCLUSIONS: Our data show that, in contrast to the wider responsiveness of peripheral macrophages, microglia proliferation is stimulated by selected M2 polarizing stimuli suggesting a role for the local microenvironment and developmental origin of tissue macrophages in regulating self-renewal following alternative activating stimuli.


Asunto(s)
Activación de Macrófagos/efectos de los fármacos , Microglía/citología , Microglía/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Estradiol/farmacología , Femenino , Interleucina-4/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Ratones Endogámicos C57BL , Fenotipo
4.
J Neuroinflammation ; 11: 211, 2014 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-25551794

RESUMEN

BACKGROUND: Acquisition of the M1 or M2 phenotypes by microglia has been shown to occur during the development of pathological conditions, with M1 activation being widely involved in neurotoxicity in relation with the anatomical localization and the reactivity of subtypes of microglia cells. On the contrary, little is known on the ability of microglia to undergo M2 polarization by interleukin-4 (IL4), the typical M2a polarization signal for peripheral macrophages. METHODS: Recombinant mouse IL4 was injected in the third cerebral ventricle of mice to induce brain alternative polarization. The mRNA levels of Fizz1, Arg1, and Ym1 genes, known to be up-regulated by IL4 in peripheral macrophages, together with additional polarization markers, were evaluated in the striatum and frontal cortex at different time intervals after central administration of IL4; in parallel, M2a protein expression was evaluated in tissue extracts and at the cellular level. RESULTS: Our results show that the potency and temporal profile of IL4-mediated M2a gene induction vary depending on the gene analyzed and according to the specific brain area analyzed, with the striatum showing a reduced M2a response compared with the frontal cortex, as further substantiated by assays of polarization protein levels. Of notice, Fizz1 mRNA induction reached 100-fold level, underscoring the potency of this specific IL4 signaling pathway in the brain. In addition, immunochemistry assays demonstrated the localization of the M2 response specifically to microglia cells and, more interestingly, the existence of a subpopulation of microglia cells amenable to undergoing M2a polarization in the healthy mouse brain. CONCLUSIONS: These results show that the responsiveness of brain macrophages to centrally administered IL4 may vary depending on the gene and brain area analyzed, and that M2a polarization can be ascribed to a subpopulation of IL4-responsive microglia cells. The biochemical pathways that enable microglia to undergo M2a activation represent key aspects for understanding the physiopathology of neuroinflammation and for developing novel therapeutic and diagnostic agents.


Asunto(s)
Lóbulo Frontal/metabolismo , Interleucina-4/farmacología , Microglía/metabolismo , Neostriado/metabolismo , Animales , Polaridad Celular/efectos de los fármacos , Polaridad Celular/fisiología , Lóbulo Frontal/efectos de los fármacos , Inyecciones Intraventriculares , Interleucina-4/administración & dosificación , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Neostriado/efectos de los fármacos , Fenotipo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo
5.
Biomed Pharmacother ; 165: 115008, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37442065

RESUMEN

Raloxifene belongs to the family of Selective Estrogen Receptor Modulators (SERMs), which are drugs widely prescribed for Estrogen Receptor alpha (ERα)-related pathologies. Recently, SERMs are being tested in repurposing strategies for ERα-independent clinical indications, including a wide range of microbial infections. Macrophages are central in the fight against pathogen invasion. Despite estrogens have been shown to regulate macrophage phenotype, SERMs activity in these cells is still poorly defined. We investigated the activity of Raloxifene in comparison with another widely used SERM, Tamoxifen, on immune gene expression in macrophages obtained from mouse and human tissues, including mouse peritoneal macrophages, bone marrow-derived macrophages, microglia or human blood-derived macrophages, assaying for the involvement of the ERα, PI3K and NRF2 pathways also under inflammatory conditions. Our data demonstrate that Raloxifene acts by a dual mechanism, which entails ERα antagonism and off-target mediators. Moreover, micromolar concentrations of Raloxifene increase the expression of immune metabolic genes, such as Vegfa and Hmox1, through PI3K and NRF2 activation selectively in peritoneal macrophages. Conversely, Il1b mRNA down-regulation by SERMs is consistently observed in all macrophage subtypes and unrelated to the PI3K/NRF2 system. Importantly, the production of the inflammatory cytokine TNFα induced by the bacterial endotoxin, LPS, is potentiated by SERMs and paralleled by the cell subtype-specific increase in IL1ß secretion. This work extends our knowledge on the biological and molecular mechanisms of SERMs immune activity and indicate macrophages as a pharmacological target for the exploitation of the antimicrobial potential of these drugs.


Asunto(s)
Clorhidrato de Raloxifeno , Moduladores Selectivos de los Receptores de Estrógeno , Ratones , Humanos , Animales , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Clorhidrato de Raloxifeno/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Regulación hacia Abajo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Tamoxifeno/farmacología , Macrófagos/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo
6.
Proteomics ; 12(4-5): 691-707, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22247026

RESUMEN

More than a decade ago our groups pioneered the analysis of serum proteins of laboratory animals with up-to-date proteomic techniques. We were, and still are, convinced that conforming animal procedures to the minimally invasive approaches typical of clinical biochemistry focuses attention on the actual conditions under which any finding arrived at on animal models of disease may eventually be applied to human patients for screening/diagnosis. We are also convinced that, besides the proteins present in trace level as a result of tissue leakage during disorders affecting specific peripheral organs, changes in the concentration of some of the major serum proteins as part of an acute-phase response may be taken as biological end-points during a number of experimental procedures. When reviewing literature data about proteomic investigations on plasma or serum of mice, we realized that not much work has been done in the direction we favor. In addition, we noticed that sometimes information about serum proteome has been coarsely treated and in a few cases even misunderstood/misused. In the following, we present current findings on serum/plasma proteome of the laboratory mouse not only under control conditions and during an experimentally induced acute-phase reaction, but also in a number of models of disease, mainly related to cancer and to metabolic disorders.


Asunto(s)
Biomarcadores/sangre , Proteínas Sanguíneas/análisis , Proteómica/métodos , Animales , Modelos Animales de Enfermedad , Espectrometría de Masas , Ratones , Ratones Transgénicos , Neoplasias/sangre , Neoplasias/patología , Proteoma/análisis
7.
Methods Mol Biol ; 2418: 153-172, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35119665

RESUMEN

In spite of the fact that women spend 1/3 of their lives in postmenopause, the search for appropriate therapies able to counteract the derangements associated with the menopause still represents a sort of sought after the "Holy Grail."Nowadays, the combination of estrogens and selective estrogen receptor modulators (SERMs), a class of compounds with a mixed agonist/antagonistic activity on the estrogen receptor (ER) in various tissues, represents the most promising approach to improve postmenopausal women's health, by preserving the benefits while avoiding the side effects of estrogen-based therapy.Given their complex mechanisms of action, the evaluation of SERM activity in combination with conjugated estrogens (CE) requires a multifactorial analysis that takes into account the multifaceted and dynamic effects of these compounds in target tissues, even in relation to the physiological/pathological status.To accomplish such a goal, we took advantage of the ERE-Luc model, a reporter mouse that allows the monitoring of ER transcriptional activity in a spatio-temporal dimension. Cluster analyses performed on in vivo/ex vivo bioluminescence (BLI) data and ex vivo luciferase activity enabled to sustain the combination of CE plus bazedoxifene (TSEC, tissue-selective estrogen complex) as a valuable option for the pharmacological treatment of the postmenopause.


Asunto(s)
Estrógenos Conjugados (USP) , Receptores de Estrógenos , Animales , Estrógenos/farmacología , Estrógenos Conjugados (USP)/efectos adversos , Femenino , Humanos , Menopausia , Ratones , Receptores de Estrógenos/genética , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico
8.
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
9.
Front Pharmacol ; 13: 879020, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35431927

RESUMEN

Beyond the wide use of tamoxifen in breast cancer chemotherapy due to its estrogen receptor antagonist activity, this drug is being assayed in repurposing strategies against a number of microbial infections. We conducted a literature search on the evidence related with tamoxifen activity in macrophages, since these immune cells participate as a first line-defense against pathogen invasion. Consistent data indicate the existence of estrogen receptor-independent targets of tamoxifen in macrophages that include lipid mediators and signaling pathways, such as NRF2 and caspase-1, which allow these cells to undergo phenotypic adaptation and potentiate the inflammatory response, without the induction of cell death. Thus, these lines of evidence suggest that the widespread antimicrobial activity of this drug can be ascribed, at least in part, to the potentiation of the host innate immunity. This widens our understanding of the pharmacological activity of tamoxifen with relevant therapeutic implications for infections and other clinical indications that may benefit from the immunomodulatory effects of this drug.

10.
FASEB J ; 24(7): 2262-72, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20203089

RESUMEN

The beneficial effects of estrogens on the cardiovascular system are associated with adverse effects on reproductive tissues. On the basis of previous work indicating a major role for estrogen receptor (ER)-alpha in maintaining cardiovascular health, we evaluated the tissue selectivity of the ER alpha-selective agonist propyl pyrazole triol (PPT) compared with 17beta-estradiol (E2) in vivo. Four weeks postovariectomy, equimolar doses of PPT and E2 were administered to rats in subcutaneous implants for 5 d. Both treatments restored rapid vasorelaxation of aortic tissue to estrogenic agents and prevented coronary hyperresponsiveness to angiotensin II in isolated heart preparations. Accordingly, multiple endpoints of myocardial ischemia-reperfusion injury exacerbated by ovariectomy returned to baseline following treatment. These protective effects were linked to increased in vivo levels of endothelial progenitor cells (EPCs). Human EPC function was enhanced in vitro after PPT treatment. In sharp contrast to E2, PPT treatment had no effect on uterine weight and histomorphology except for vessel density, and failed to up-regulate classic estrogen target genes. Dissection of the effects on vascular reactivity and uterine morphology was also observed following increased exposure to PPT at a higher dose for longer time. These data provide the first in vivo evidence for tissue-specific ER alpha activation. By conferring cardiovascular protection dissected from unwanted uterotrophic effects, ER alpha-selective agonists may represent a potential safer alternative to natural hormones.


Asunto(s)
Cardiotónicos/farmacología , Sistema Cardiovascular/efectos de los fármacos , Endotelio Vascular/citología , Receptor alfa de Estrógeno/agonistas , Miocardio/citología , Células Madre/citología , Angiotensina II/farmacología , Animales , Aorta/fisiología , Sistema Cardiovascular/citología , Sistema Cardiovascular/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Estradiol/farmacología , Femenino , Miocardio/metabolismo , Fenoles , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Vasodilatación/efectos de los fármacos
11.
Nat Med ; 9(1): 82-6, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12483206

RESUMEN

Through intracellular receptors, estrogens control growth, differentiation and function of not only reproductive tissues, but also other systems. Estrogen receptors are ligand-dependent transcription factors whose activity is modulated either by estrogens, or by alternative intracellular signaling pathways downstream of growth factors and neurotransmitters. To determine the dynamics of estrogen receptor activity and the dependence of estrogen receptor on 17beta-estradiol in vivo, we generated a transgenic mouse that expresses a luciferase reporter gene under the control of activated estrogen receptors. As expected, luciferase activity, monitored with a cooled charged coupled device camera, paralleled circulating estrogen levels in reproductive tissues and in liver, indicating that the peak transcriptional activity of the estrogen receptor occurred at proestrus. In contrast, in tissues such as bone and brain, the peak activity of estrogen receptors was observed at diestrus. These tissue-specific responses are masked when mice undergo conventional hormone treatment. We also demonstrate that estrogen receptors are active in immature mice before gonadal production of sex hormones as well as in ovariectomized adult mice. These findings emphasize the importance of hormone-independent activation of the estrogen receptor, and have implications for the therapeutic use of estrogens, such as hormone replacement therapy.


Asunto(s)
Estradiol/metabolismo , Receptores de Estrógenos/metabolismo , Transcripción Genética , Animales , Diagnóstico por Imagen , Ciclo Estral/fisiología , Femenino , Genes Reporteros , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovariectomía , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Distribución Tisular
12.
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
13.
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
14.
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
15.
Front Neuroendocrinol ; 29(4): 507-19, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18522863

RESUMEN

Recent studies highlight the prominent role played by estrogens in protecting the central nervous system (CNS) against the noxious consequences of a chronic inflammatory reaction. The neurodegenerative process of several CNS diseases, including Multiple Sclerosis, Alzheimer's and Parkinson's Diseases, is associated with the activation of microglia cells, which drive the resident inflammatory response. Chronically stimulated during neurodegeneration, microglia cells are thought to provide detrimental effects on surrounding neurons. The inhibitory activity of estrogens on neuroinflammation and specifically on microglia might thus be considered as a beneficial therapeutic opportunity for delaying the onset or progression of neurodegenerative diseases; in addition, understanding the peculiar activity of this female hormone on inflammatory signalling pathways will possibly lead to the development of selected anti-inflammatory molecules. This review summarises the evidence for the involvement of microglia in neuroinflammation and the anti-inflammatory activity played by estrogens specifically in microglia.


Asunto(s)
Antiinflamatorios , Encéfalo/efectos de los fármacos , Estrógenos , Menopausia/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Estrógenos/farmacología , Estrógenos/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Microglía/efectos de los fármacos , Microglía/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Enfermedades Neurodegenerativas/inmunología , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/patología
16.
J Pharmacol Exp Ther ; 328(1): 174-82, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18832649

RESUMEN

Estrogen is known to affect vascular function and diabetes development, but the relative contribution of estrogen receptor (ER) isoforms is unclear. The aim of this study was to determine how individual ER isoforms modulate inflammatory enzymes in the vascular wall of control and streptozotocin (STZ)-injected rodents. Primary cultures of rat aortic smooth muscle cells (SMCs) were stimulated with inflammatory agents in the presence or absence of increasing concentrations of the ER alpha and ER beta-selective agonists 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) and diarylpropionitrile (DPN), respectively. The production of inducible nitric-oxide synthase (iNOS), a classical indicator of vascular inflammation, was significantly reduced by PPT in control but not diabetic SMCs, whereas it was further enhanced by DPN treatment in both groups. This distinct action profile was not related to changes in ER transcriptional activity. However, extracellular signal-regulated kinase 1/2 signaling was activated by DPN but not by PPT in cytokine-treated SMCs. In cultured aortic rings from both normoglycemic and STZ-diabetic mice, pharmacological activation of ER alpha attenuated cytokine-driven iNOS induction by 30 to 50%. Vascular iNOS levels were decreased consistently when adding 1 nM 17beta-estradiol to aortic tissues from ER beta- but not ER alpha-knockout mice. These findings suggest a possible role for ER alpha-selective ligands in reducing vascular inflammatory responses under normo- and hyperglycemic conditions.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/deficiencia , Receptor beta de Estrógeno/fisiología , Músculo Liso Vascular/fisiopatología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Aorta/enzimología , Aorta/fisiopatología , Diabetes Mellitus Experimental/enzimología , Inducción Enzimática , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Inflamación/enzimología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/enzimología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Nitrilos/farmacología , Nitritos/metabolismo , Fenoles , Propionatos/farmacología , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Transfección
17.
Mol Cell Biol ; 25(8): 2957-68, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15798185

RESUMEN

Estrogen is an immunoregulatory agent, in that hormone deprivation increases while 17beta-estradiol (E2) administration blocks the inflammatory response; however, the underlying mechanism is still unknown. The transcription factor p65/relA, a member of the nuclear factor kappaB (NF-kappaB) family, plays a major role in inflammation and drives the expression of proinflammatory mediators. Here we report a novel mechanism of action of E2 in inflammation. We observe that in macrophages E2 blocks lipopolysaccharide-induced DNA binding and transcriptional activity of p65 by preventing its nuclear translocation. This effect is selectively activated in macrophages to prevent p65 activation by inflammatory agents and extends to other members of the NF-kappaB family, including c-Rel and p50. We observe that E2 activates a rapid and persistent response that involves the activation of phosphatidylinositol 3-kinase, without requiring de novo protein synthesis or modifying Ikappa-Balpha degradation and mitogen-activated protein kinase activation. Using a time course experiment and the microtubule-disrupting agent nocodazole, we observe that the hormone inhibits p65 intracellular transport to the nucleus. This activity is selectively mediated by estrogen receptor alpha (ERalpha) and not ERbeta and is not shared by conventional anti-inflammatory drugs. These results unravel a novel and unique mechanism for E2 anti-inflammatory activity, which may be useful for identifying more selective ligands for the prevention of the inflammatory response.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Estradiol/fisiología , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Línea Celular , Núcleo Celular/química , Citoplasma/química , Regulación hacia Abajo , Estradiol/farmacología , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/fisiología , Humanos , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , FN-kappa B/análisis , FN-kappa B/antagonistas & inhibidores , Subunidad p50 de NF-kappa B , Nocodazol/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , Factor de Transcripción ReIA , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA