Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(11)2022 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-35683029

RESUMO

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.


Assuntos
Neuropeptídeo Y , Receptores de Neuropeptídeo Y , Animais , Feminino , Hormônios Gonadais , Masculino , Camundongos , Doenças Neuroinflamatórias , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Obesidade/genética , Obesidade/metabolismo , Prosencéfalo/metabolismo , Receptores de Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/metabolismo
2.
J Neuroinflammation ; 18(1): 220, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34551802

RESUMO

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.


Assuntos
Encéfalo/enzimologia , Glucosilceramidase/antagonistas & inibidores , Microglia/enzimologia , Neurônios/metabolismo , Neuroproteção/fisiologia , Animais , Encéfalo/patologia , Comunicação Celular/fisiologia , Camundongos , Microglia/patologia , Neurônios/patologia
3.
Front Neuroendocrinol ; 52: 156-164, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30481522

RESUMO

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.


Assuntos
Encefalopatias , Encéfalo/crescimento & desenvolvimento , Microglia/fisiologia , Caracteres Sexuais , Diferenciação Sexual/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Encefalopatias/epidemiologia , Encefalopatias/metabolismo , Encefalopatias/fisiopatologia , Humanos
4.
Br J Cancer ; 120(5): 537-546, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30739913

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Anticarcinógenos/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Sirtuína 1/efeitos dos fármacos , Sulindaco/análogos & derivados , Proteína Supressora de Tumor p53/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anticarcinógenos/efeitos adversos , Linhagem Celular Tumoral , Simulação por Computador , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidores de Ciclo-Oxigenase/efeitos adversos , Humanos , Cetorolaco/efeitos adversos , Cetorolaco/uso terapêutico , Camundongos , Modelos Moleculares , Sirtuína 1/metabolismo , Sulindaco/farmacologia , Proteína Supressora de Tumor p53/metabolismo
5.
Nucleic Acids Res ; 45(6): e37, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-27899606

RESUMO

Deciphering the etiology of complex pathologies at molecular level requires longitudinal studies encompassing multiple biochemical pathways (apoptosis, proliferation, inflammation, oxidative stress). In vivo imaging of current reporter animals enabled the spatio-temporal analysis of specific molecular events, however, the lack of a multiplicity of loci for the generalized and regulated expression of the integrated transgenes hampers the creation of systems for the simultaneous analysis of more than a biochemical pathways at the time. We here developed and tested an in vivo-based methodology for the identification of multiple insertional loci suitable for the generation of reliable reporter mice. The validity of the methodology was tested with the generation of novel mice useful to report on inflammation and oxidative stress.


Assuntos
Regulação da Expressão Gênica , Genes Reporter , Loci Gênicos , Camundongos Transgênicos , Animais , Linhagem Celular , Eletroporação , Células-Tronco Embrionárias/metabolismo , Feminino , Humanos , Luciferases/biossíntese , Luciferases/genética , Substâncias Luminescentes , Medições Luminescentes , Masculino , Camundongos , Estresse Oxidativo , Regiões Promotoras Genéticas , Transgenes , Imagem Corporal Total
6.
Neurobiol Dis ; 114: 74-84, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29486298

RESUMO

The Parkinson's disease (PD) evolves over an extended period of time with the onset occurring long before clinical signs begin to manifest. Characterization of the molecular events underlying the PD onset is instrumental for the development of diagnostic markers and preventive treatments, progress in this field is hindered by technical limitations. We applied an imaging approach to demonstrate the activation of Nrf2 transcription factor as a hallmark of neurodegeneration in neurotoxin-driven models of PD. In dopaminergic SK-N-BE neuroblastoma cells, Nrf2 activation was detected in cells committed to die as proven by time lapse microscopy; in the substantia nigra pars compacta area of the mouse brain, the Nrf2 activation preceded dopaminergic neurodegeneration as demonstrated by in vivo and ex vivo optical imaging, a finding confirmed by co-localization experiments carried out by immunohistochemistry. Collectively, our results identify the Nrf2 signaling as an early marker of neurodegeneration, anticipating dopaminergic neurodegeneration and motor deficits.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Medições Luminescentes/métodos , Fator 2 Relacionado a NF-E2/metabolismo , Imagem Óptica/métodos , Transtornos Parkinsonianos/diagnóstico por imagem , Transtornos Parkinsonianos/metabolismo , Animais , Morte Celular/fisiologia , Modelos Animais de Doenças , Neurônios Dopaminérgicos/patologia , Humanos , Células MCF-7 , Masculino , Camundongos , Camundongos Transgênicos , Células NIH 3T3
7.
J Neuroinflammation ; 14(1): 236, 2017 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-29202771

RESUMO

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.


Assuntos
Ativação de Macrófagos/efeitos dos fármacos , Microglia/citologia , Microglia/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Estradiol/farmacologia , Feminino , Interleucina-4/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
8.
Adv Exp Med Biol ; 1043: 141-151, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29224094

RESUMO

Recent studies have demonstrated that in mice, the estrogen receptor alpha (ERα) is expressed in the liver and has a direct effect on the regulation of the hepatic genes relevant for energy metabolism and drug metabolism. The sex-related differential expression of the hepatic ERα raises the questions as to whether this receptor is responsible for the sexual differences observed in the physiopathology of the liver.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Fígado/metabolismo , Caracteres Sexuais , Ativação Metabólica/genética , Animais , Metabolismo Energético/genética , Receptor alfa de Estrogênio/genética , Feminino , Regulação da Expressão Gênica , Inativação Metabólica/genética , Masculino , Camundongos , Reprodução/genética , Fatores Sexuais , Transdução de Sinais , Transcrição Gênica
9.
Proc Natl Acad Sci U S A ; 111(26): 9561-6, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24979764

RESUMO

Decades of studies provided a detailed view of the mechanism of estrogen receptor-α (ERα) regulated gene transcription and the physio-pathological relevance of the genetic programs controlled by this receptor in a variety of tissues. However, still limited is our knowledge on the regulation of ERα synthesis. Preliminary observations showed that the expression of ERα is cell cycle regulated. Here, we have demonstrated that a well described polymorphic sequence in the first intron of ERα (PvuII and XbaI) has a key role in regulating the ERα content in cycling cells. We have shown that the RNA Pol II (Pol II) elongation is blocked at the polymorphic site and that the proto-oncogene c-MYB modulates the release of the pausing polymerase. It is well known that the two SNPs are associated to an increased risk, progression, survival and mortality of endocrine-related cancers, here we have demonstrated that the c-MYB-dependent release of Pol II at a specific phase of the cell cycle is facilitated by the px haplotype, thus leading to a higher ERα mitogenic signal. In breast cancer, this mechanism is disrupted when the hormone refractory phenotype is established; therefore, we propose this oscillator as a novel target for the development of therapies aimed at sensitizing breast cancer resistant to hormonal treatments. Because PvuII and XbaI were associated to a broad range physio-pathological conditions beside neoplastic transformation, we expect that the ERα oscillator contributes to the regulation of the estrogen signal in several tissues.


Assuntos
Ciclo Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica/fisiologia , RNA Polimerase II/metabolismo , Análise de Variância , Imunoprecipitação da Cromatina , Primers do DNA/genética , Receptor alfa de Estrogênio/genética , Genótipo , Humanos , Células MCF-7 , Plasmídeos/genética , Polimorfismo de Nucleotídeo Único , Proto-Oncogene Mas , Reação em Cadeia da Polimerase em Tempo Real
10.
Alzheimers Dement ; 12(9): 964-976, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27107518

RESUMO

INTRODUCTION: In Alzheimer's disease (AD), pathologic amyloid-beta (Aß) is synaptotoxic and impairs neuronal function at the microscale, influencing brain networks at the macroscale before Aß deposition. The latter can be detected noninvasively, in vivo, using resting-state functional MRI (rsfMRI), a technique used to assess brain functional connectivity (FC). METHODS: RsfMRI was performed longitudinally in TG2576 and PDAPP mice, starting before Aß deposition to determine the earliest FC changes. Additionally, the role of pathologic Aß on early FC alterations was investigated by treating TG2576 mice with the 3D6 anti-Aß-antibody. RESULTS: Both transgenic models showed hypersynchronized FC before Aß deposition and hyposynchronized FC at later stages. Early anti-Aß treatment in TG2576 mice prevented hypersynchronous FC and the associated synaptic impairments and excitatory/inhibitory disbalances. DISCUSSION: Hypersynchrony of FC may be used as a new noninvasive read out of early AD and can be recovered by anti-Aß treatment, encouraging preventive treatment strategies in familial AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Doença de Alzheimer/diagnóstico por imagem , Animais , Autoanticorpos/farmacologia , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Circulação Cerebrovascular/fisiologia , Sincronização Cortical/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Estudos Longitudinais , Imageamento por Ressonância Magnética , Camundongos Transgênicos , Vias Neurais/diagnóstico por imagem , Vias Neurais/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Oxigênio/sangue , Placa Amiloide/diagnóstico por imagem , Placa Amiloide/fisiopatologia , Placa Amiloide/prevenção & controle , Sintomas Prodrômicos , Descanso
11.
J Biol Chem ; 289(10): 6908-6920, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24451380

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors regulating lipid and glucose metabolism. Ongoing drug discovery programs aim to develop dual PPARα/γ agonists devoid of the side effects of the marketed antidiabetic agents thiazolidinediones and the dual agonists glitazars. Recently, we described a new dual PPARα/γ ligand, LT175, with a partial agonist profile against PPARγ and interacting with a newly identified region of the PPARγ-ligand binding domain (1). Here we show that LT175 differentially activated PPARγ target genes involved in fatty acid esterification and storage in 3T3-L1-derived adipocytes. This resulted in a less severe lipid accumulation compared with that triggered by rosiglitazone, suggesting that LT175 may have a lower adipogenic activity. Consistent with this hypothesis, in vivo administration of LT175 to mice fed a high-fat diet decreased body weight, adipocyte size, and white adipose tissue mass, as assessed by magnetic resonance imaging. Furthermore, LT175 significantly reduced plasma glucose, insulin, non-esterified fatty acids, triglycerides, and cholesterol and increased circulating adiponectin and fibroblast growth factor 21 levels. Oral glucose and insulin tolerance tests showed that the compound improves glucose homeostasis and insulin sensitivity. Moreover, we demonstrate that the peculiar interaction of LT175 with PPARγ affected the recruitment of the coregulators cyclic-AMP response element-binding protein-binding protein and nuclear corepressor 1 (NCoR1), fundamentals for the PPARγ-mediated adipogenic program. In conclusion, our results describe a new PPAR ligand, modulating lipid and glucose metabolism with reduced adipogenic activity, that may be used as a model for a series of novel molecules with an improved pharmacological profile for the treatment of dyslipidemia and type 2 diabetes.


Assuntos
Adipogenia/efeitos dos fármacos , Compostos de Bifenilo/administração & dosagem , Hipoglicemiantes/farmacologia , Resistência à Insulina , Insulina/farmacologia , PPAR alfa/agonistas , PPAR gama/agonistas , Fenilpropionatos/administração & dosagem , Células 3T3-L1 , Animais , Compostos de Bifenilo/metabolismo , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dislipidemias/tratamento farmacológico , Glucose/metabolismo , Teste de Tolerância a Glucose , Hipoglicemiantes/metabolismo , Insulina/sangue , Ligantes , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Correpressor 1 de Receptor Nuclear/metabolismo , PPAR alfa/metabolismo , PPAR gama/metabolismo , Fenilpropionatos/metabolismo
12.
Proc Natl Acad Sci U S A ; 109(29): 11806-11, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22761311

RESUMO

In the liver of female mice, the transcriptional activity of estrogen receptor (ER) α oscillates in phase with the 4-d-long estrous cycle. Here systemic, genome-wide analysis demonstrates that ER tetradian oscillation is necessary to generate pulses of expression in genes for fatty acid and cholesterol synthesis. This ER-dependent metabolic programming changes with pregnancy and after cessation of ovarian function due to age or surgical menopause, suggesting that ER signaling is optimized to coordinate liver functions with the energetic requirements of each reproductive stage. Alterations of amplitude and frequency of the tetradian cycle, as observed after surgical menopause, age, or specific ablation of the hepatic Igf-1 gene, are associated with liver fat deposition. Appropriate hormone replacement therapy reinstating the oscillatory activity of liver ER prevents the effect of surgical menopause on fat deposition in liver.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Ciclo Estral/fisiologia , Regulação da Expressão Gênica/fisiologia , Fígado/metabolismo , Periodicidade , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Animais , Colesterol/metabolismo , Primers do DNA/genética , Estradiol/sangue , Ciclo Estral/metabolismo , Ácidos Graxos/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Histocitoquímica , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Radioimunoensaio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Moduladores Seletivos de Receptor Estrogênico/classificação
13.
J Biol Chem ; 287(43): 35899-911, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22910911

RESUMO

Recent evidence highlights the peroxisome proliferator-activated receptors (PPARs) as critical neuroprotective factors in several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). To gain new mechanistic insights into the role of these receptors in the context of ALS, here we investigated how PPAR transcriptional activity varies in hSOD1(G93A) ALS transgenic mice. We demonstrate that PPARγ-driven transcription selectively increases in the spinal cord of symptomatic hSOD1(G93A) mice. This phenomenon correlates with the up-regulation of target genes, such as lipoprotein lipase and glutathione S-transferase α-2, which are implicated in scavenging lipid peroxidation by-products. Such events are associated with enhanced PPARγ immunoreactivity within motor neuronal nuclei. This observation, and the fact that PPARγ displays increased responsiveness in cultured hSOD1(G93A) motor neurons, points to a role for this receptor in neutralizing deleterious lipoperoxidation derivatives within the motor cells. Consistently, in both motor neuron-like cultures and animal models, we report that PPARγ is activated by lipid peroxidation end products, such as 4-hydroxynonenal, whose levels are elevated in the cerebrospinal fluid and spinal cord from ALS patients. We propose that the accumulation of critical concentrations of lipid peroxidation adducts during ALS progression leads to the activation of PPARγ in motor neurons. This in turn triggers self-protective mechanisms that involve the up-regulation of lipid detoxification enzymes, such as lipoprotein lipase and glutathione S-transferase α-2. Our findings indicate that anticipating natural protective reactions by pharmacologically modulating PPARγ transcriptional activity may attenuate neurodegeneration by limiting the damage induced by lipid peroxidation derivatives.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Peroxidação de Lipídeos , Neurônios Motores/metabolismo , PPAR gama/metabolismo , Superóxido Dismutase/metabolismo , Substituição de Aminoácidos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica/genética , Glutationa Transferase/biossíntese , Glutationa Transferase/genética , Humanos , Isoenzimas/biossíntese , Isoenzimas/genética , Lipase Lipoproteica/biossíntese , Lipase Lipoproteica/genética , Camundongos , Camundongos Transgênicos , Neurônios Motores/patologia , Mutação de Sentido Incorreto , PPAR gama/genética , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Transcrição Gênica/genética , Regulação para Cima/genética
14.
Cells ; 12(3)2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36766684

RESUMO

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.


Assuntos
Doença de Parkinson , Animais , Feminino , Masculino , Camundongos , Glucosilceramidase/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo
15.
Biochim Biophys Acta ; 1812(8): 1054-60, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21605666

RESUMO

Over the past two decades, our understanding of estrogen receptor physiology in mammals widened considerably as we acquired a deeper appreciation of the roles of estrogen receptor alpha and beta (ERα and ERß) in reproduction as well as in bone and metabolic homeostasis, depression, vascular disorders, neurodegenerative diseases and cancer. In addition, our insights on ER transcriptional functions in cells increased considerably with the demonstration that ER activity is not strictly dependent on ligand availability. Indeed, unliganded ERs may be transcriptionally active and post-translational modifications play a major role in this context. The finding that several intracellular transduction molecules may regulate ER transcriptional programs indicates that ERs may act as a hub where several molecular pathways converge: this allows to maintain ER transcriptional activity in tune with all cell functions. Likely, the biological relevant role of ER was favored by evolution as a mean of integration between reproductive and metabolic functions. We here review the post-translational modifications modulating ER transcriptional activity in the presence or in the absence of estrogens and underline their potential role for ER tissue-specific activities. In our opinion, a better comprehension of the variety of molecular events that control ER activity in reproductive and non-reproductive organs is the foundation for the design of safer and more efficacious hormone-based therapies, particularly for menopause. This article is part of a Special Issue entitled: Translating Nuclear receptors from health to disease.


Assuntos
Terapia de Reposição de Estrogênios , Receptores de Estrogênio/metabolismo , Animais , Estrogênios/metabolismo , Humanos , Ligantes , Processamento de Proteína Pós-Traducional , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/genética , Receptores de Estrogênio/fisiologia , Ativação Transcricional/fisiologia
16.
Pharmacol Res ; 65(5): 531-6, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22391453

RESUMO

Detailed knowledge of drug metabolism is relevant information provided by preclinical drug development research. Oxidative enzymes such as those belonging to P450 family of cytochromes (CYP) play a prominent role in drug metabolism. Here, we propose an innovative method based on bioluminescence in vivo imaging which has the potential to simplify the in vivo measurement of CYP activity also providing a dynamic measure of the effects of a drug on a specific P450 enzyme complex in a living mouse. The method is based on a pro-luciferin which can be converted into the active luciferase substrate by a specific P450 activity. The pro-luciferin is administered to a luciferase reporter mouse which produces luminescent signals in relation to the cytochrome activity present in each tissue. The photon emission generated can be easily localized and quantified by optical imaging. To demonstrate the validity of the system, we pharmacologically induced hepatic Cyp3a in the reporter mouse and proved that pro-luciferin administration generates a Cyp3a selective signal in the chest area that can be efficiently detected by optical imaging. The kind of tool generated has the potential to be exploited for the study of additional CYPs.


Assuntos
Citocromo P-450 CYP3A/metabolismo , Acetais/metabolismo , Animais , Dexametasona/farmacologia , Luciferina de Vaga-Lumes/análogos & derivados , Luciferina de Vaga-Lumes/metabolismo , Genes Reporter , Fígado/efeitos dos fármacos , Fígado/metabolismo , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Substâncias Luminescentes/metabolismo , Medições Luminescentes , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Distribuição Tecidual
18.
Endocrinology ; 163(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34758075

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Hormônios Esteroides Gonadais/metabolismo , Hepatopatias/metabolismo , Doenças Metabólicas/metabolismo , Animais , Evolução Biológica , Feminino , Humanos , Hepatopatias/genética , Masculino , Doenças Metabólicas/genética , Fatores Sexuais
19.
Nutrients ; 14(16)2022 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-36014766

RESUMO

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.


Assuntos
Dieta Hiperlipídica , Receptor alfa de Estrogênio , Peptídeos e Proteínas de Sinalização Intercelular , Fígado , Hepatopatia Gordurosa não Alcoólica , Animais , Dieta Hiperlipídica/efeitos adversos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/genética , Estrogênios/metabolismo , Feminino , Homeostase/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo
20.
Nat Med ; 9(1): 82-6, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12483206

RESUMO

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.


Assuntos
Estradiol/metabolismo , Receptores de Estrogênio/metabolismo , Transcrição Gênica , Animais , Diagnóstico por Imagem , Ciclo Estral/fisiologia , Feminino , Genes Reporter , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovariectomia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa