Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurochem Res ; 44(6): 1410-1424, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30661228

RESUMEN

Traumatic brain injury triggers neuroinflammation that may contribute to progressive neurodegeneration. We investigated patterns of recruitment of astrocytes and microglia to inflammation after brain trauma by firstly characterising expression profiles over time of marker genes following TBI, and secondly by monitoring glial morphologies reflecting inflammatory responses in a rat model of traumatic brain injury (i.e. the lateral fluid percussion injury). Gene expression profiles revealed early elevation of expression of astrocytic marker glial fibrillary acidic protein relative to microglial marker allograft inflammatory factor 1 (also known as ionized calcium-binding adapter molecule 1). Adult rat brains collected at day 7 after injury were processed for immunohistochemistry with allograft inflammatory factor 1, glial fibrillary acidic protein and complement C3 (marker of bad/disruptive astrocytic A1 phenotype). Astrocytes positive for glial fibrillary acidic protein and complement C3 were significant increased in the injured cortex and displayed more complex patterns of arbourisation with significantly increased bifurcations. Our observations suggested that traumatic brain injury changed the phenotype of microglia from a ramified appearance with long, thin, highly branched processes to a swollen amoeboid shape in the injured cortex. These findings suggest differential glial activation with astrocytes likely undergoing strategic changes in morphology and function. Whilst a detailed analysis is needed of temporal patterns of glial activation, ours is the first evidence of a role for the bad/disruptive astrocytic A1 phenotype in an open head model of traumatic brain injury.


Asunto(s)
Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Animales , Astrocitos/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Complemento C3/metabolismo , Equidae , Proteína Ácida Fibrilar de la Glía/metabolismo , Cabras , Masculino , Ratones , Microglía/patología , Conejos , Ratas Sprague-Dawley
2.
J Mol Cell Cardiol ; 111: 96-101, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28822806

RESUMEN

A correlation exists between the extent of pericardial adipose and atrial fibrillation (AF) risk, though the underlying mechanisms remain unclear. Selected adipose depots express high levels of aromatase, capable of converting androgens to estrogens - no studies have investigated aromatase occurrence/expression regulation in pericardial adipose. The Women's Health Initiative reported that estrogen-only therapy in women elevated AF incidence, indicating augmented estrogenic influence may exacerbate cardiac vulnerability. The aim of this study was to identify the occurrence of pericardial adipose aromatase, evaluate the age- and sex-dependency of local cardiac steroid synthesis capacity and seek preliminary experimental evidence of a link between pericardial adipose aromatase capacity and arrhythmogenic vulnerability. Both human atrial appendage and epicardial adipose exhibited immunoblot aromatase expression. In rodents, myocardium and pericardial adipose aromatase expression increased >20-fold relative to young controls. Comparing young, aged and aged-high fat diet animals, a significant positive correlation was determined between the total aromatase content of pericardial adipose and the occurrence/duration of triggered atrial arrhythmias. Incidence and duration of arrhythmias were increased in hearts perfused with 17ß-estradiol. This study provides novel report of pericardial adipose aromatase expression. We show that aromatase expression is remarkably upregulated with aging, and aromatase estrogen conversion capacity significantly elevated with obesity-related cardiac adiposity. Our studies suggest an association between adiposity, aromatase estrogenic capacity and atrial arrhythmogenicity - additional investigation is required to establish causality. The potential impact of these findings may be considerable, and suggests that focus on local cardiac steroid conversion (rather than systemic levels) may yield translational outcomes.


Asunto(s)
Tejido Adiposo/metabolismo , Envejecimiento/patología , Aromatasa/metabolismo , Arritmias Cardíacas/terapia , Obesidad/terapia , Pericardio/patología , Investigación Biomédica Traslacional , Animales , Arritmias Cardíacas/enzimología , Arritmias Cardíacas/patología , Estradiol/farmacología , Estrógenos/biosíntesis , Femenino , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/patología , Humanos , Masculino , Ratones , Obesidad/enzimología , Obesidad/patología , Ratas
3.
Am J Physiol Heart Circ Physiol ; 306(9): H1265-74, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24610919

RESUMEN

Estrogen in females is conventionally considered a cardioprotective influence, but a role for estrogen in male cardioprotection has yet to be defined. Estrogen biosynthesis from testosterone is regulated by aromatase. Aromatase has recently been shown to be expressed in the adult heart, although little is known about its involvement in the regulation of myocardial function and stress responses. The goal of this study was to determine whether upregulation of tissue aromatase expression could improve ischemic resilience in male hearts. Isolated hearts from male transgenic aromatase-overexpressing (AROM(+); high estrogen, low testosterone) mice and wild-type (WT) mice (12 wk) were Langendorff perfused and subjected to ischemia-reperfusion (25 min ischemia and 60 min of reperfusion). Basal systolic function was lower in AROM(+) hearts (dP/dtmax: 4,121 ± 255 vs. 4,992 ± 283 mmHg/s, P < 0.05) and associated with augmented Akt phosphorylation, consistent with a suppressor action of estrogen on contractility. Ischemic contracture was attenuated in AROM(+) hearts (43 ± 3 vs. 55 ± 4 mmHg, P < 0.05), yet AROM(+) hearts were more arrhythmic in early reperfusion. At the end of 60 min of reperfusion, AROM(+) systolic functional recovery was lower (left ventricular developed pressure: 39 ± 6 vs. 56 ± 5 %basal, P < 0.05) and diastolic dysfunction was accentuated (36 ± 4 vs. 24 ± 2 mmHg, P < 0.05). This is the first study to show that in vivo aromatase upregulation modulates basal cardiac performance and the response to ischemic stress. These data suggest that while chronic exposure to enhanced estrogenic influence may have benefits in limiting ischemic contracture severity, acute functional recovery in reperfusion is compromised. A temporally targeted, tissue-specific intervention combining aromatase treatment with inotropic support may offer therapeutic potential for men and women.


Asunto(s)
Aromatasa/metabolismo , Contracción Miocárdica , Daño por Reperfusión Miocárdica/fisiopatología , Estrés Fisiológico , Regulación hacia Arriba , Animales , Aromatasa/genética , Estrógenos/metabolismo , Femenino , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/metabolismo , Factores Sexuales , Testosterona/metabolismo
4.
EBioMedicine ; 100: 104949, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199043

RESUMEN

BACKGROUND: Attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) are neurodevelopmental conditions with early life origins. Alterations in blood lipids have been linked to ADHD and ASD; however, prospective early life data are limited. This study examined (i) associations between the cord blood lipidome and ADHD/ASD symptoms at 2 years of age, (ii) associations between prenatal and perinatal predictors of ADHD/ASD symptoms and cord blood lipidome, and (iii) mediation by the cord blood lipidome. METHODS: From the Barwon Infant Study cohort (1074 mother-child pairs, 52.3% male children), child circulating lipid levels at birth were analysed using ultra-high-performance liquid chromatography-tandem mass spectrometry. These were clustered into lipid network modules via Weighted Gene Correlation Network Analysis. Associations between lipid modules and ADHD/ASD symptoms at 2 years, assessed with the Child Behavior Checklist, were explored via linear regression analyses. Mediation analysis identified indirect effects of prenatal and perinatal risk factors on ADHD/ASD symptoms through lipid modules. FINDINGS: The acylcarnitine lipid module is associated with both ADHD and ASD symptoms at 2 years of age. Risk factors of these outcomes such as low income, Apgar score, and maternal inflammation were partly mediated by higher birth acylcarnitine levels. Other cord blood lipid profiles were also associated with ADHD and ASD symptoms. INTERPRETATION: This study highlights that elevated cord blood birth acylcarnitine levels, either directly or as a possible marker of disrupted cell energy metabolism, are on the causal pathway of prenatal and perinatal risk factors for ADHD and ASD symptoms in early life. FUNDING: The foundational work and infrastructure for the BIS was sponsored by the Murdoch Children's Research Institute, Deakin University, and Barwon Health. Subsequent funding was secured from the Minderoo Foundation, the European Union's Horizon 2020 research and innovation programme (ENDpoiNTs: No 825759), National Health and Medical Research Council of Australia (NHMRC) and Agency for Science, Technology and Research Singapore [APP1149047], The William and Vera Ellen Houston Memorial Trust Fund (via HOMER Hack), The Shepherd Foundation, The Jack Brockhoff Foundation, the Scobie & Claire McKinnon Trust, the Shane O'Brien Memorial Asthma Foundation, the Our Women Our Children's Fund Raising Committee Barwon Health, the Rotary Club of Geelong, the Ilhan Food Allergy Foundation, Geelong Medical and Hospital Benefits Association, Vanguard Investments Australia Ltd, the Percy Baxter Charitable Trust, and Perpetual Trustees.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Trastorno del Espectro Autista , Carnitina/análogos & derivados , Lactante , Recién Nacido , Humanos , Masculino , Femenino , Trastorno del Espectro Autista/epidemiología , Trastorno del Espectro Autista/etiología , Estudios de Cohortes , Trastorno por Déficit de Atención con Hiperactividad/epidemiología , Trastorno por Déficit de Atención con Hiperactividad/etiología , Sangre Fetal , Estudios Prospectivos , Lípidos
5.
Transgenic Res ; 21(2): 415-28, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21614586

RESUMEN

Aromatase converts androgens to estrogens and it is expressed in gonads and non-reproductive tissues (e.g. brain and adipose tissues). As circulating levels of estrogens in males are low, we hypothesize that local estrogen production is important for the regulation of physiological functions (e.g. metabolism) and pathological development (e.g. breast and prostate cancers) by acting in a paracrine and/or intracrine manner. We generated a tissue-specific doxycycline-inducible, aromatase transgenic mouse to test this hypothesis. The transgene construct (pTetOAROM) consists of a full-length human aromatase cDNA (hAROM) and a luciferase gene under the control of a bi-directional tetracycline-responsive promoter (pTetO), which is regulated by transactivators (rtTA or tTA) and doxycycline. Our in vitro studies using MBA-MB-231tet cells stably expressing rtTA, showed that doxycycline treatment induced transgene expression of hAROM transcripts by 17-fold (P = 0.01), aromatase activity by 26-fold, (P = 0.0008) and luciferase activity by 9.6-fold (P = 0.0006). Pronuclear microinjection of the transgene generated four pTetOAROM founder mice. A male founder was bred with a female mammary gland-specific rtTA mouse (MMTVrtTA) to produce MMTVrtTA-pTetOAROM double-transgenic mice. Upon doxycycline treatment via drinking water, human aromatase expression was detected by RT-PCR, specifically in mammary glands, salivary glands and seminal vesicles of double-stransgenic mice. Luciferase expression and activity was detected in these tissues by in vivo bioluminescence imaging, in vitro luciferase assay and RT-PCR. In summary, we generated a transgenic mouse model that expresses the human aromatase transgene in a temporal- and spatial-specific manner, which will be a useful model to study the physiological importance of local estrogen production.


Asunto(s)
Aromatasa/metabolismo , Doxiciclina/farmacología , Regulación Enzimológica de la Expresión Génica , Animales , Aromatasa/genética , Línea Celular Tumoral , Clonación Molecular , ADN Complementario/genética , ADN Complementario/metabolismo , Doxiciclina/administración & dosificación , Activación Enzimática , Pruebas de Enzimas , Femenino , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Mediciones Luminiscentes/métodos , Masculino , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Ratones , Ratones Transgénicos , Microinyecciones , Plásmidos/genética , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Glándulas Salivales/citología , Glándulas Salivales/metabolismo , Vesículas Seminales/citología , Vesículas Seminales/metabolismo , Transgenes
6.
Behav Genet ; 42(4): 509-27, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22426781

RESUMEN

Inspired by the localization, on 15q21.2 of the CYP19A1 gene in the linkage region of speech and language disorders, and a rare translocation in a dyslexic individual that was brought to our attention, we conducted a series of studies on the properties of CYP19A1 as a candidate gene for dyslexia and related conditions. The aromatase enzyme is a member of the cytochrome P450 super family, and it serves several key functions: it catalyzes the conversion of androgens into estrogens; during early mammalian development it controls the differentiation of specific brain areas (e.g. local estrogen synthesis in the hippocampus regulates synaptic plasticity and axonal growth); it is involved in sexual differentiation of the brain; and in songbirds and teleost fishes, it regulates vocalization. Our results suggest that variations in CYP19A1 are associated with dyslexia as a categorical trait and with quantitative measures of language and speech, such as reading, vocabulary, phonological processing and oral motor skills. Variations near the vicinity of its brain promoter region altered transcription factor binding, suggesting a regulatory role in CYP19A1 expression. CYP19A1 expression in human brain correlated with the expression of dyslexia susceptibility genes such as DYX1C1 and ROBO1. Aromatase-deficient mice displayed increased cortical neuronal density and occasional cortical heterotopias, also observed in Robo1-/- mice and human dyslexic brains, respectively. An aromatase inhibitor reduced dendritic growth in cultured rat neurons. From this broad set of evidence, we propose CYP19A1 as a candidate gene for human cognitive functions implicated in reading, speech and language.


Asunto(s)
Aromatasa/genética , Encéfalo/crecimiento & desarrollo , Dislexia/genética , Trastornos del Lenguaje/genética , ARN Mensajero/análisis , Trastornos del Habla/genética , Animales , Aromatasa/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Cohortes , Proteínas del Citoesqueleto , Dislexia/metabolismo , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Trastornos del Lenguaje/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Sitios de Carácter Cuantitativo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Trastornos del Habla/metabolismo , Translocación Genética , Proteínas Roundabout
7.
Front Endocrinol (Lausanne) ; 13: 808479, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35273564

RESUMEN

Sex steroid hormones have been implicated as disease modifiers in the neurodegenerative disorder amyotrophic lateral sclerosis (ALS). Androgens, signalling via the androgen receptor (AR), predominate in males, and have widespread actions in the periphery and the central nervous system (CNS). AR translocates to the cell nucleus when activated upon binding androgens, whereby it regulates transcription of target genes via the classical genomic signalling pathway. We previously reported that AR protein is decreased in the lumbar spinal cord tissue of symptomatic male SOD1G93A mice. Here, we further explored the changes in AR within motor neurons (MN) of the CNS, assessing their nuclear AR content and propensity to degenerate by endstage disease in male SOD1G93A mice. We observed that almost all motor neuron populations had undergone significant loss in nuclear AR in SOD1G93A mice. Interestingly, loss of nuclear AR was evident in lumbar spinal MNs as early as the pre-symptomatic age of 60 days. Several MN populations with high AR content were identified which did not degenerate in SOD1G93A mice. These included the brainstem ambiguus and vagus nuclei, and the sexually dimorphic spinal MNs: cremaster, dorsolateral nucleus (DLN) and spinal nucleus of bulbocavernosus (SNB). In conclusion, we demonstrate that AR loss directly associates with MN vulnerability and disease progression in the SOD1G93A mouse model of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Andrógenos/metabolismo , Animales , Masculino , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
8.
Neurochem Int ; 149: 105143, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34311029

RESUMEN

Lipids are essential in maintaining brain function, and lipid profiles have been reported to be altered in aged and Alzheimer's disease (AD) brains as compared to healthy mature brains. Both age and AD share common metabolic hallmarks such as increased oxidative stress and perturbed metabolic function, and age remains the most strongly correlated risk factor for AD, a neurodegenerative disease. A major accompanying pathological symptom of these conditions is cognitive impairment, which is linked with changes in lipid metabolism. Thus, nutraceuticals that affect brain lipid metabolism or lipid levels as a whole have the potential to ameliorate cognitive decline. Lipid analyses and lipidomic studies reveal changes in specific lipid types with aging and AD, which can identify potential lipid-based nutraceuticals to restore the brain to a healthy lipid phenotype. The brain lipid profile can be influenced directly with dietary administration of lipids themselves, although because of synergistic effects of nutrients it may be more useful to consider a multi-component diet rather than single nutrient supplementation. Gut microbiota also serve as a source of beneficial lipids, and the value of treatments that manipulate the composition of gut microbiome should not be ignored. Lastly, instead of direct supplementation, compounds that affect pathways involved with lipid metabolism should also be considered as a way of manipulating lipid levels to improve cognition. In this review, we briefly discuss the role of lipids in the brain, the changing lipid profile in AD, current research on lipid-based nutraceuticals and their therapeutic potential to combat cognitive impairment.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Cognición/fisiología , Suplementos Dietéticos , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/fisiología , Anciano , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Disfunción Cognitiva/terapia , Ácidos Grasos/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
9.
Mol Cell Neurosci ; 41(1): 1-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19185610

RESUMEN

The aromatase knockout (ArKO) mouse is completely estrogen deficient. We previously detected apoptosis in the hypothalamus of 1 year-old male ArKO mice. This study shows that 12 week-old female ArKO mice display spontaneous apoptosis of pyramidal neurons in the frontal cortex while wild-type (WT) littermates show no signs of apoptosis. Concomitantly, bcl-2 related anti-apoptotic genes are down-regulated whereas the pro-apoptotic gene TRADD is up-regulated in the female ArKO frontal cortex. This phenotype can be rescued by 3-week replacement of 17beta-estradiol. Furthermore, the apoptosis phenotype is exacerbated in 12-15 month-old female ArKO mice, which have 30% less neurons in the frontal cortex and lower brain weights than WT counterparts. These data show that estrogens are essential for the survival of female cortical neurons even in the absence of pathological conditions or external assaults. Our observations also demonstrate the sexually dimorphic susceptibility of neurons to estrogen deficiency.


Asunto(s)
Apoptosis/fisiología , Aromatasa , Estrógenos/deficiencia , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Animales , Aromatasa/genética , Aromatasa/metabolismo , Caspasa 3/metabolismo , Supervivencia Celular , Proteínas de Unión al ADN , Estradiol/administración & dosificación , Femenino , Lóbulo Frontal/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Proteínas Nucleares/metabolismo , Tamaño de los Órganos
10.
Compr Physiol ; 9(2): 457-475, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30873596

RESUMEN

As adipose tissue depots are active endocrine organs, they secrete a variety of hormones (including estrogens from white adipose) and inflammatory mediators, which have important implications in numerous obesity-associated diseases. Adipose tissues are broadly characterized as consisting of white, beige, and brown depot types. The endocrine, metabolic, and inflammatory profiles of adipose are depot dependent and influenced by the estrogenic and androgenic status of the adipose tissue. Estrogen receptors mediate both the genomic and nongenomic actions of estrogens and are expressed in the brain, heart, and other peripheral tissues. All three known estrogen receptor α (ERα) and estrogen receptor ß (ERß), and the G-protein coupled estrogen receptor (GPER/GPR30) are expressed in white adipose and can modulate adipose mass. Expression of each receptor is dependent on depot location, adipose cell type, and estrogen levels. Estrogen receptor expression profiles in beige and brown adipocytes are less well established. This review will discuss the effects of estrogens on the differential deposition of the major adipose tissues and the impact of estrogens within white adipose depots. © 2019 American Physiological Society. Compr Physiol 9:457-475, 2019.


Asunto(s)
Tejido Adiposo/metabolismo , Estrógenos/metabolismo , Adiposidad , Animales , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Glándulas Mamarias Humanas/metabolismo
11.
Biol Psychiatry ; 61(3): 359-66, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16566897

RESUMEN

BACKGROUND: Aromatase converts androgen to estrogen. Thus, the aromatase knockout (ArKO) mouse is estrogen deficient. We investigated the compulsive behaviors of these animals and the protein levels of catechol-O-methyltransferase (COMT) in frontal cortex, hypothalamus and liver. METHODS: Grooming was analyzed during the 20-min period immediately following a water-mist spray. Running wheel activity over two consecutive nights and barbering were analyzed. COMT protein levels were measured by Western analysis. RESULTS: Six-month old male but not female ArKO mice develop compulsive behaviors such as excessive barbering, grooming and wheel-running. Excessive activities were reversed by 3 weeks of 17beta-estradiol replacement. Interestingly, the presentation of compulsive behaviors is accompanied by concomitant decreases (p < .05) in hypothalamic COMT protein levels in male ArKO mice. These values returned to normal upon 17beta-estradiol treatment. In contrast, hepatic and frontal cortex COMT levels were not affected by the estrogen status, indicating region- and tissue-specific regulation of COMT levels by estrogen. No differences in COMT levels were detectable between female animals of both genotypes. CONCLUSIONS: This study describes the novel observation of a possible link between estrogen, COMT and development of compulsive behaviors in male animals which may have therapeutic implications in obsessive compulsive disorder (OCD) patients.


Asunto(s)
Conducta Compulsiva/fisiopatología , Conducta Compulsiva/psicología , Estrógenos/deficiencia , Animales , Aromatasa/genética , Aromatasa/fisiología , Western Blotting , Encéfalo/enzimología , Catecol O-Metiltransferasa/genética , Catecol O-Metiltransferasa/metabolismo , Conducta Compulsiva/genética , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Aseo Animal/fisiología , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología
12.
Nat Clin Pract Endocrinol Metab ; 3(5): 414-21, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17452968

RESUMEN

Aromatase deficiency is rare in humans. Affected individuals cannot synthesize endogenous estrogens. Aromatase is the enzyme that catalyzes conversion of androgens into estrogens, and if aromatase is nonfunctional because of an inactivating mutation, estrogen synthesis cannot occur. If the fetus lacks aromatase activity, dehydroepiandrosterone sulfate produced by the fetal adrenal glands cannot be converted to estrogen by the placenta, so is converted to testosterone peripherally and results in virilization of both fetus and mother. Virilization manifests as pseudohermaphroditism in female infants, with hirsutism and acne in the mother; the maternal indicators resolve following delivery. To date, only seven males and seven females with aromatase deficiency have been reported. Affected females are typically diagnosed at birth because of the pseudohermaphroditism. Cystic ovaries and delayed bone maturation can occur during childhood and adolescence in these girls, who present at puberty with primary amenorrhea, failure of breast development, virilization, and hypergonadotrophic hypogonadism. Affected males, on the other hand, do not present with obvious defects at birth, so are diagnosed much later in life, presenting with clinical symptoms, which include tall stature, delayed skeletal maturation, delayed epiphyseal closure, bone pain, eunuchoid body proportions and excess adiposity. Estrogen replacement therapy reverses the symptoms in male and female patients.


Asunto(s)
Aromatasa/deficiencia , Adiposidad/fisiología , Adolescente , Adulto , Aromatasa/genética , Huesos/metabolismo , Terapia de Reemplazo de Estrógeno , Estrógenos/uso terapéutico , Femenino , Hormonas/uso terapéutico , Humanos , Infertilidad Masculina/metabolismo , Resistencia a la Insulina/fisiología , Masculino , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/terapia , Mutación , Embarazo , Factores Sexuales
13.
Trends Endocrinol Metab ; 17(2): 55-64, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16480891

RESUMEN

We are rapidly becoming aware of the importance of estrogen in maintaining virtually all facets of male health. In order for estrogens to be synthesized endogenously, the enzyme responsible for their synthesis from androgens, aromatase, must be functional. The seven known men in whom aromatase is nonfunctional all have a mutation in either exon V or IX of the CYP19 gene, which encodes aromatase. Collectively, these men are reported to have undetectable estrogen; normal to high levels of testosterone and gonadotropins; tall stature with delayed skeletal maturation and epiphyseal closure; osteoporosis; impaired lipid and insulin metabolism; and impaired reproductive function. The aromatase knockout mouse presents with a phenotype that is similar in many aspects and provides a valuable tool with which to examine and manipulate the actions of estrogen. By studying the naturally occurring aromatase-deficient humans, together with studies of the aromatase-knockout mouse, we are expanding our understanding of the essential role of estrogen in male physiology.


Asunto(s)
Aromatasa/deficiencia , Andrógenos/fisiología , Animales , Aromatasa/fisiología , Conducta/fisiología , Desarrollo Óseo/fisiología , Fenómenos Fisiológicos Cardiovasculares , Metabolismo Energético/fisiología , Estrógenos/deficiencia , Estrógenos/fisiología , Humanos , Sistema Inmunológico/fisiología , Síndrome Metabólico/fisiopatología , Ratones , Fenotipo
14.
Behav Brain Res ; 319: 48-62, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27829127

RESUMEN

Chronic social behavior problems after pediatric traumatic brain injury (TBI) significantly contribute to poor quality of life for survivors. Using a well-characterized mouse model of early childhood TBI, we have previously demonstrated that young brain-injured mice develop social deficits by adulthood. As biological sex may influence both normal and aberrant social development, we here evaluated potential sex differences in post-TBI psychosocial deficits by comparing the behavior of male and female mice at adulthood (8 weeks post-injury). Secondly, we hypothesized that pediatric TBI would influence neuronal morphology identified by Golgi-Cox staining in the hippocampus and prefrontal cortex, regions involved in social cognition and behavior, before the onset of social problems (3 weeks post-injury). Morphological analysis of pyramidal neurons in the ipsilateral prefrontal cortex and granule cells of the hippocampal dentate gyrus revealed a reduction in dendritic complexity after pediatric TBI. This was most apparent in TBI males, whereas neurons from females were less affected. At adulthood, consistent with previous studies, TBI males showed deficits in sociability and social recognition. TBI females also showed a reduction in sociability, but intact social recognition and increased sociosexual avoidance. Together, these findings indicate that sex is a determinant of regional neuroplasticity and social outcomes after pediatric TBI. Reduced neuronal complexity in the prefrontal cortex and hippocampus, several weeks after injury in male mice, appears to precede the subsequent emergence of social deficits. Sex-specific alterations in the social brain network are thus implicated as an underlying mechanism of social dysfunction after pediatric TBI.


Asunto(s)
Lesiones Encefálicas/patología , Lesiones Encefálicas/psicología , Hipocampo/patología , Neuronas/patología , Caracteres Sexuales , Conducta Social , Análisis de Varianza , Animales , Dendritas/ultraestructura , Conducta Exploratoria , Femenino , Hipocampo/ultraestructura , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/ultraestructura , Tinción con Nitrato de Plata , Natación/psicología
15.
J Steroid Biochem Mol Biol ; 170: 39-48, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27353462

RESUMEN

We investigated the effects of estrogens on glucose homeostasis using the Aromatase Knockout (ArKO) mouse, which is unable to convert androgens into estrogens. The ArKO mouse is a model of total estrogen ablation which develops symptoms of metabolic syndrome. To determine the development and progression of whole body state of insulin resistance of ArKO mice, comprehensive whole body tolerance tests were performed on WT, ArKO and estrogen administrated mice at 3 and 12 months of age. The absence of estrogens in the male ArKO mice leads to hepatic insulin resistance, glucose and pyruvate intolerance from 3 to 12 months with consistent improvement upon estrogen treatment. Estrogen absence in the female ArKO mice leads to glucose intolerance without pyruvate intolerance or insulin resistance. The replacement of estrogens in the female WT and ArKO mice exhibited both insulin sensitizing and resistance effects depending on age and dosage. In conclusion, this study presents information on the sexually dimorphic roles of estrogens on glucose homeostasis regulation.


Asunto(s)
Aromatasa/deficiencia , Aromatasa/genética , Estrógenos/metabolismo , Glucosa/metabolismo , Homeostasis , Animales , Aromatasa/metabolismo , Índice de Masa Corporal , Femenino , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Caracteres Sexuales
16.
Stem Cell Reports ; 8(2): 417-431, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28132885

RESUMEN

Estrogen stimulates breast development during puberty and mammary tumors in adulthood through estrogen receptor-α (ERα). These effects are proposed to occur via ERα+ luminal cells and not the mammary stem cells (MaSCs) that are ERαneg. Since ERα+ luminal cells express stem cell antigen-1 (SCA-1), we sought to determine if SCA-1 could define an ERα+ subset of EpCAM+/CD24+/CD49fhi MaSCs. We show that the MaSC population has a distinct SCA-1+ population that is abundant in pre-pubertal mammary glands. The SCA-1+ MaSCs have less stem cell markers and less in vivo repopulating activity than their SCA-1neg counterparts. However, they express ERα and specifically enter the cell cycle at puberty. Using estrogen-deficient aromatase knockouts (ArKO), we showed that the SCA-1+ MaSC could be directly modulated by estrogen supplementation. Thus, SCA-1 enriches for an ERα+, estrogen-sensitive subpopulation within the CD24+/CD49fhi MaSC population that may be responsible for the hormonal sensitivity of the developing mammary gland.


Asunto(s)
Antígenos Ly/metabolismo , Estrógenos/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Proteínas de la Membrana/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Antígeno CD24/metabolismo , Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Inmunofenotipificación , Integrina alfa6/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Transgénicos , Fenotipo , Trasplante de Células Madre , Células Madre/efectos de los fármacos
17.
Neurochem Int ; 95: 75-84, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26939763

RESUMEN

There is little doubt that we are what we eat. Fatty acid supplementation and diets rich in fatty acids are being promoted as ways to a healthier brain. Short chain fatty acids are a product of intestinal microbiota metabolism of dietary fibre; and their derivatives are used as an anti-convulstant. They demonstrated therapeutic potential in neurodegenerative conditions as HDAC inhibitors; and while the mechanism is not well understood, have been shown to lower amyloid ß in Alzheimer's Disease in preclinical studies. Medium chain fatty acids consumed as a mixture in dietary oils can induce ketogenesis without the need for a ketogentic diet. Hence, this has the potential to provide an alternative energy source to prevent neuronal cell death due to lack of glucose. Long chain fatty acids are commonly found in the diet as omega fatty acids. They act as an anti-oxidant protecting neuronal cell membranes from oxidative damage and as an anti-inflammatory mediator in the brain. We review which agents, from each fatty acid class, have the most therapeutic potential for neurological disorders (primarily Alzheimer's disease, Parkinson's disease, Autism Spectrum Disorder as well as possible applications to traumatic brain injury), by discussing what is known about their biological mechanisms from preclinical studies.


Asunto(s)
Encéfalo/efectos de los fármacos , Suplementos Dietéticos , Ácidos Grasos/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Ensayos Clínicos como Asunto/métodos , Ácidos Grasos/metabolismo , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-3/uso terapéutico , Humanos , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología
18.
Life Sci ; 158: 130-6, 2016 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-27373424

RESUMEN

AIMS: Controversy surrounds the gender basis of progression in chronic kidney disease. Unfortunately, most experimental studies addressing this question do not distinguish between direct effects of estrogen and indirect activation of estrogen receptors through conversion of testosterone to 17ß-estradiol by aromatase. We examined the pathogenesis of renal fibrosis in female aromatase knockout (ArKO) mice, which lack circulating and stored estrogens, while having normal levels of testosterone. MAIN METHODS: ArKO mice and their wild-type (ArWT) counterparts were subjected to unilateral ureteric obstruction (UUO), with kidney tissue collected at day(D) 0, 3 and 9 post-UUO. Effects of 5α-dihydrotestosterone (DHT) administration on each genotype were also studied. Tissue was assessed biochemically and histochemically for fibrosis. Western blot analysis was used to measure α-smooth muscle actin (α-SMA) expression and TGF-ß1 signalling. Matrix metalloproteinase-2 (MMP-2) activity was measured by zymography. KEY FINDINGS: UUO increased collagen content over time (p<0.05 (D3) and p<0.01 (D9) vs day 0), with no difference between genotypes in qualitative (collagen IV staining) and quantitative (hydroxyproline concentration) analyses. Systemic administration of non-aromatizable DHT increased collagen content after 3days of UUO in both genotypes. This was not paralleled by any change in α-SMA (myofibroblast burden) or TGF-ß1 signalling but was commensurate with DHT reducing MMP2 activity in both genotypes (p<0.05 vs genotype controls). SIGNIFICANCE: Physiological concentrations of estrogens do not protect the injured kidney from fibrosis progression. Androgens rather than estrogens are the relevant factor involved in regulating disease-related renal scarring in this model.


Asunto(s)
Andrógenos/fisiología , Colágeno/metabolismo , Estrógenos/fisiología , Riñón/metabolismo , Obstrucción Ureteral/metabolismo , Animales , Femenino , Fibrosis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obstrucción Ureteral/patología
19.
Brain Res Mol Brain Res ; 140(1-2): 127-32, 2005 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-16083992

RESUMEN

The aromatase knockout (ArKO) mouse is estrogen deficient. Using reverse-transcription and real-time PCR, we showed that transcript levels of the N-methyl-d-aspartate (NMDA) receptor subunit NR2B are significantly higher in the hippocampus of female ArKO mice compared to wild-type (WT) littermates. Expression levels of NR1, NR2A, but not NR2C, also tended to be higher in ArKO mice. In the Morris watermaze test for spatial memory, both genotypes displayed equal significant improvement in the latency in locating the invisible platform over the 5-day training period. These findings show that selective loss of estrogen synthesis is associated with changes in NMDA receptor subunit expression in the hippocampus but little change in spatial learning ability.


Asunto(s)
Estrógenos/deficiencia , Hipocampo/fisiología , Aprendizaje por Laberinto/fisiología , Receptores de N-Metil-D-Aspartato/genética , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa
20.
Menopause ; 12(2): 210-5, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15772569

RESUMEN

OBJECTIVE: Although natural menopause is associated with loss of ovarian estrogen production, this life phase is followed by a significant increase in estrogen-related cancers, namely breast and endometrial cancer. These tissues, as well as adipose, skeletal, and vascular tissues and the brain are important sites of postmenopausal estrogen production. Circulating C19 steroid precursors are essential substrates for extragonadal estrogen synthesis; however, the levels of these androgenic precursors decline markedly with advancing age. This implies an increase in capacity for extragonadal tissues to produce estrogen with age. DESIGN: To explore this, and the effects of the menopause transition and postmenopausal estrogen therapy on extragonadal estrogen biosynthesis, we have compared the expression of the aromatase gene and estrogen (ER) and androgen receptors (AR) in subcutaneous abdominal and gluteal fat taken from premenopausal (group 1: n = 11), postmenopausal (group 2: n = 10), and postmenopausal women taking estrogen therapy (group 3: n = 10). All subjects were of normal body mass index, euglycemic, and normolipemic. RESULTS: The postmenopausal women were older (group 1, 43.1 +/- 5.0 vs groups 2 and 3, 57.9 +/- 7.4 years, P < 0.001 and 56.1 +/- 4.5 years, P < 0.001, respectively) and had lower serum estradiol levels (group 2, 22.2 +/- 3.2 vs group 1, 442.5 +/- 248.2 pmol/L, P < 0.05), which were restored to premenopausal levels with estrogen therapy. Expression analysis revealed that levels of transcripts encoding aromatase were greater in gluteal than abdominal depots in each group in postmenopausal versus premenopausal women (P < 0.05). Use of hormone therapy did not influence aromatase gene expression in either depot. No differences were detected in the expression of ER or AR between groups of between tissue depots. CONCLUSION: Thus, the capacity of adipose tissue to produce estrogen seems to increase significantly with age at the time of menopause and to be unaltered by exogenous estrogen therapy. This difference in extragonadal estrogen production with age may play a pivotal role in the increase in estrogen-dependent malignancies in the postmenopausal years.


Asunto(s)
Tejido Adiposo/metabolismo , Aromatasa/genética , Terapia de Reemplazo de Estrógeno , Posmenopausia , Tejido Adiposo/patología , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Expresión Génica , Humanos , Persona de Mediana Edad , ARN/análisis , Receptores Androgénicos/genética , Receptores de Estrógenos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA