Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Oper Neurosurg (Hagerstown) ; 26(3): 347-348, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37962341

RESUMEN

INDICATIONS CORRIDOR AND LIMITS OF EXPOSURE: Cavernous malformations of the third ventricle arise from the medial thalamus and/or periaqueductal midbrain. Microsurgical resection is indicated when the lifetime risk of hemorrhage outweighs the surgical risks. ANATOMIC ESSENTIALS NEED FOR PREOPERATIVE PLANNING AND ASSESSMENT: superior sagittal sinus, callosomarginal and pericallosal arteries, corpus callosum, foramen of Monro, choroidal fissure, fornix, thalamostriate veins, internal cerebral veins (ICVs), velum interpositum, and thalamus. ESSENTIAL STEPS OF THE PROCEDURE: The patient consents to the procedure. With the patient supine, the head is turned 90° and laterally flexed 45°. A bifrontal craniotomy positioned two-thirds anterior and one-third posterior to the coronal suture is performed. The interhemispheric fissure is opened, and a 2-cm corpus callosotomy is performed. Choroid plexus cauterization exposes the choroidal fissure. Sharp division of the taenia fornicea opens the velum interpositum, where the thalamostriate vein can be followed around the venous angle to the ICV. The anterior septal vein may be divided to communicate between the foramen of Monro and choroidal fissure. Dissection between the ICVs opens the velum interpositum into the third ventricle. PITFALLS/AVOIDANCE OF COMPLICATIONS: Frontal or deep vein occlusion causes venous infarction, and dissection on the nondominant hemisphere is preferred. Other complications include arterial infarction, fornix injury from choroidal fissure dissection or forniceal retraction, and thalamic or midbrain injury during lesion resection. VARIANTS AND INDICATIONS FOR THEIR USE: The contralateral choroidal fissure is used for low-lying medial thalamic and midbrain lesions. The ipsilateral choroidal fissure is used for high-lying or large lesions extending laterally. Transchoroidal approaches are not needed for superior (transcallosal only) or anterior (contralateral transcallosal-contralateral transforaminal) thalamic lesions. Used with permission from Barrow Neurological Institute, Phoenix, Arizona.


Asunto(s)
Plexo Coroideo , Tercer Ventrículo , Humanos , Plexo Coroideo/cirugía , Tercer Ventrículo/cirugía , Tálamo/diagnóstico por imagen , Tálamo/cirugía , Mesencéfalo/diagnóstico por imagen , Mesencéfalo/cirugía , Infarto
2.
Cell Rep ; 42(8): 112903, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37515772

RESUMEN

The choroid plexus (CP) is a source of trophic factors for the developing and mature brain. Insulin is produced in epithelial cells of the CP (EChPs), and its secretion is stimulated by Htr2c-mediated signaling. We modulated insulin expression in EChPs with intracerebroventricular injections of AAV5. Insulin overexpression in CP decelerates food intake, whereas its knockdown has the opposite effect. Insulin overexpression also results in reduced anxious behavior. Transcriptomic changes in the hypothalamus, especially in synapse-related processes, are also seen in mice overexpressing insulin in CP. Last, activation of Gq signaling in CP leads to acute Akt phosphorylation in neurons of the arcuate nucleus, indicating a direct action of CP-derived insulin on the hypothalamus. Taken together, our findings signify that CP is a relevant source of insulin in the central nervous system and that CP-derived insulin should be taken into consideration in future work pertaining to insulin actions in the brain.


Asunto(s)
Plexo Coroideo , Insulina , Ratones , Animales , Insulina/metabolismo , Plexo Coroideo/metabolismo , Encéfalo , Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo
3.
Sci Rep ; 13(1): 8510, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37231062

RESUMEN

Manganese dioxide nanoparticles (MnO2-NPs) have a wide range of applications in biomedicine. Given this widespread usage, it is worth noting that MnO2-NPs are definitely toxic, especially to the brain. However, the damage caused by MnO2-NPs to the choroid plexus (CP) and to the brain after crossing CP epithelial cells has not been elucidated. Therefore, this study aims to investigate these effects and elucidate potential underlying mechanisms through transcriptomics analysis. To achieve this objective, eighteen SD rats were randomly divided into three groups: the control group (control), low-dose exposure group (low-dose) and high-dose exposure group (high-dose). Animals in the two treated groups were administered with two concentrations of MnO2-NPs (200 mg kg-1 BW and 400 mg kg-1 BW) using a noninvasive intratracheal injection method once a week for three months. Finally, the neural behavior of all the animals was tested using a hot plate tester, open-field test and Y-type electric maze. The morphological characteristics of the CP and hippocampus were observed by H&E stain, and the transcriptome of CP tissues was analysed by transcriptome sequencing. The representative differentially expressed genes were quantified by qRT-PCR. We found that treatment with MnO2-NPs could induce learning capacity and memory faculty decline and destroy the structure of hippocampal and CP cells in rats. High doses of MnO2-NPs had a more obvious destructive capacity. For transcriptomic analysis, we found that there were significant differences in the numbers and types of differential genes in CP between the low- and high-dose groups compared to the control. Through GO terms and KEGG analysis, high-dose MnO2-NPs significantly affected the expression of transporters, ion channel proteins, and ribosomal proteins. There were 17 common differentially expressed genes. Most of them were transporter and binding genes on the cell membrane, and some of them had kinase activity. Three genes, Brinp, Synpr and Crmp1, were selected for qRT-PCR to confirm their expression differences among the three groups. In conclusion, high-dose MnO2-NPs exposure induced abnormal neurobehaviour, impaired memory function, destroyed the structure of the CP and changed its transcriptome in rats. The most significant DEGs in the CP were within the transport system.


Asunto(s)
Nanopartículas , Óxidos , Ratas , Animales , Óxidos/toxicidad , Óxidos/química , Compuestos de Manganeso/química , Plexo Coroideo , Transcriptoma , Ratas Sprague-Dawley , Nanopartículas/toxicidad
4.
PLoS Genet ; 19(1): e1010558, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36626371

RESUMEN

Copper (Cu) has a multifaceted role in brain development, function, and metabolism. Two homologous Cu transporters, Atp7a (Menkes disease protein) and Atp7b (Wilson disease protein), maintain Cu homeostasis in the tissue. Atp7a mediates Cu entry into the brain and activates Cu-dependent enzymes, whereas the role of Atp7b is less clear. We show that during postnatal development Atp7b is necessary for normal morphology and function of choroid plexus (ChPl). Inactivation of Atp7b causes reorganization of ChPl' cytoskeleton and cell-cell contacts, loss of Slc31a1 from the apical membrane, and a decrease in the length and number of microvilli and cilia. In ChPl lacking Atp7b, Atp7a is upregulated but remains intracellular, which limits Cu transport into the brain and results in significant Cu deficit, which is reversed only in older animals. Cu deficiency is associated with down-regulation of Atp7a in locus coeruleus and catecholamine imbalance, despite normal expression of dopamine-ß-hydroxylase. In addition, there are notable changes in the brain lipidome, which can be attributed to inhibition of diacylglyceride-to-phosphatidylethanolamine conversion. These results identify the new role for Atp7b in developing brain and identify metabolic changes that could be exacerbated by Cu chelation therapy.


Asunto(s)
Cobre , Síndrome del Pelo Ensortijado , Ratones , Animales , ATPasas Transportadoras de Cobre , Cobre/metabolismo , Plexo Coroideo/metabolismo , Síndrome del Pelo Ensortijado/metabolismo , Encéfalo/metabolismo
5.
Inflammation ; 46(2): 556-572, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36269513

RESUMEN

Neuropsychiatric systemic lupus erythematosus (NPSLE) is a serious complication of systemic lupus erythematosus (SLE) involving the nervous system with high morbidity and mortality. A key hypothesis in NPSLE is that a disrupted barrier allows autoantibodies and immune components of peripheral blood to penetrate into the central nervous system (CNS), resulting in inflammation and damage. The blood cerebrospinal fluid barrier (BCSFB), which consists of the choroid plexus and the hypothalamic tanycytes, has long been regarded as an immunological sanctuary site. 1,25-Dihydroxyvitamin D3 [1,25-(OH)2D3] is the active form of vitamin D, which plays multiple roles in inflammation and immunoregulation. In this study, we investigated the possible protective effects of 1,25-dihydroxyvitamin D3 against BCSFB dysfunction in NPSLE in MRL/lpr mice and explored the mechanism by which 1,25-dihydroxyvitamin D3 inhibits the progression of NPSLE. In this study, we found that supplementation with 1,25-dihydroxyvitamin D3 markedly improved serological and immunological indices, delayed inflammatory infiltration, delayed neuronal deformation, and upregulated the expression of brain-derived neurotrophic factor (BDNF) proteins in the brain. Furthermore, 1,25-dihydroxyvitamin D3 downregulated proinflammatory cytokines such as nuclear factor kappa-B (NF-κB) and tumor necrosis factor-α (TNF-α) by activating peroxisome proliferator-activated receptor γ (PPARγ), and it reduced the expression of the TGF-ß/Smad signaling pathway. Our findings demonstrate that 1,25-dihydroxyvitamin D3 delayed cell infiltration into the choroid plexus and decreased markers suggestive of cognitive decline in MRL/lpr mice, and the mechanism may be related to protection against BCSFB disruption through activation of the anti-inflammatory PPARγ/NF-κB/TNF-α pathway as well as upregulation of BDNF and inhibition of the TGF-ß/Smad signaling pathway. These findings provide a novel direction for the study of NPSLE.


Asunto(s)
Lupus Eritematoso Sistémico , Factor de Necrosis Tumoral alfa , Animales , Ratones , Factor Neurotrófico Derivado del Encéfalo , Calcitriol/farmacología , Calcitriol/uso terapéutico , Plexo Coroideo , Inflamación/tratamiento farmacológico , Inflamación/complicaciones , Lupus Eritematoso Sistémico/terapia , Ratones Endogámicos MRL lpr , FN-kappa B , PPAR gamma , Factor de Crecimiento Transformador beta , Proteínas Smad/metabolismo
6.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34948033

RESUMEN

This study was designed to determine the effect of acute caffeine (CAF) administration, which exerts a broad spectrum of anti-inflammatory activity, on the synthesis of pro-inflammatory cytokines and their receptors in the hypothalamus and choroid plexus (ChP) during acute inflammation caused by the injection of bacterial endotoxin-lipopolysaccharide (LPS). The experiment was performed on 24 female sheep randomly divided into four groups: control; LPS treated (iv.; 400 ng/kg of body mass (bm.)); CAF treated (iv.; 30 mg/kg of bm.); and LPS and CAF treated. The animals were euthanized 3 h after the treatment. It was found that acute administration of CAF suppressed the synthesis of interleukin (IL-1ß) and tumor necrosis factor (TNF)α, but did not influence IL-6, in the hypothalamus during LPS-induced inflammation. The injection of CAF reduced the LPS-induced expression of TNF mRNA in the ChP. CAF lowered the gene expression of IL-6 cytokine family signal transducer (IL6ST) and TNF receptor superfamily member 1A (TNFRSF1) in the hypothalamus and IL-1 type II receptor (IL1R2) in the ChP. Our study on the sheep model suggests that CAF may attenuate the inflammatory response at the hypothalamic level and partly influence the inflammatory signal generated by the ChP cells. This suggests the potential of CAF to suppress neuroinflammatory processes induced by peripheral immune/inflammatory challenges.


Asunto(s)
Cafeína/administración & dosificación , Plexo Coroideo/inmunología , Citocinas/genética , Encefalitis/tratamiento farmacológico , Hipotálamo/inmunología , Lipopolisacáridos/efectos adversos , Administración Intravenosa , Animales , Cafeína/farmacología , Plexo Coroideo/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Encefalitis/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-6/metabolismo , Ovinos , Factor de Necrosis Tumoral alfa/genética
7.
Int J Mol Sci ; 22(16)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34445563

RESUMEN

Choroid plexus (CP) sequesters cadmium and other metals, protecting the brain from these neurotoxins. These metals can induce cellular stress and modulate homeostatic functions of CP, such as solute transport. We previously showed in primary cultured neonatal rat CP epithelial cells (CPECs) that cadmium induced cellular stress and stimulated choline uptake at the apical membrane, which interfaces with cerebrospinal fluid in situ. Here, in CPECs, we characterized the roles of glutathione (GSH) and Zinc supplementation in the adaptive stress response to cadmium. Cadmium increased GSH and decreased the reduced GSH-to-oxidized GSH (GSSG) ratio. Heat shock protein-70 (Hsp70), heme oxygenase (HO-1), and metallothionein (Mt-1) were induced along with the catalytic and modifier subunits of glutamate cysteine ligase (GCL), the rate-limiting enzyme in GSH synthesis. Inhibition of GCL by l-buthionine sulfoximine (BSO) enhanced stress protein induction and stimulation of choline uptake by cadmium. Zinc alone did not induce Hsp70, HO-1, or GCL subunits, or modulate choline uptake. Zinc supplementation during cadmium exposure attenuated stress protein induction and stimulation of choline uptake; this effect persisted despite inhibition of GSH synthesis. These data indicated up-regulation of GSH synthesis promotes adaptation to cadmium-induced cellular stress in CP, but Zinc may confer cytoprotection independent of GSH.


Asunto(s)
Cadmio/toxicidad , Colina/metabolismo , Plexo Coroideo/efectos de los fármacos , Epitelio/efectos de los fármacos , Glutatión/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Zinc/administración & dosificación , Animales , Animales Recién Nacidos , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Suplementos Dietéticos , Epitelio/metabolismo , Epitelio/patología , Ratas , Ratas Sprague-Dawley
8.
Commun Biol ; 4(1): 260, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637884

RESUMEN

Neuroinflammation is a key component of virtually all neurodegenerative diseases, preceding neuronal loss and associating directly with cognitive impairment. Neuroinflammatory signals can originate and be amplified at barrier tissues such as brain vasculature, surrounding meninges and the choroid plexus. We designed a high content screening system to target inflammation in human brain-derived cells of the blood-brain barrier (pericytes and endothelial cells) to identify inflammatory modifiers. Screening an FDA-approved drug library we identify digoxin and lanatoside C, members of the cardiac glycoside family, as inflammatory-modulating drugs that work in blood-brain barrier cells. An ex vivo assay of leptomeningeal and choroid plexus explants confirm that these drugs maintain their function in 3D cultures of brain border tissues. These results suggest that cardiac glycosides may be useful in targeting inflammation at border regions of the brain and offer new options for drug discovery approaches for neuroinflammatory driven degeneration.


Asunto(s)
Antiinflamatorios/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Plexo Coroideo/efectos de los fármacos , Digoxina/farmacología , Células Endoteliales/efectos de los fármacos , Inflamación/tratamiento farmacológico , Lanatosidos/farmacología , Meninges/efectos de los fármacos , Pericitos/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Células Cultivadas , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Evaluación Preclínica de Medicamentos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Meninges/metabolismo , Meninges/patología , Pericitos/metabolismo , Pericitos/patología , Técnicas de Cultivo de Tejidos
9.
Biol Reprod ; 104(3): 578-588, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33331645

RESUMEN

The objective of the study was to characterize the anatomical framework and sites of action of the nerve growth factor (NGF)-mediated ovulation-inducing system of llamas. The expression patterns of NGF and its receptors in the hypothalamus of llamas (n = 5) were examined using single and double immunohistochemistry/immunofluorescence. We also compare the expression pattern of the P75 receptor in the hypothalamus of llama and a spontaneous ovulator species (sheep, n = 5). Both NGF receptors (TrkA and P75) were highly expressed in the medial septum and diagonal band of Broca, and populations of TrkA cells were observed in the periventricular and dorsal hypothalamus. Unexpectedly, we found NGF immunoreactive cell bodies with widespread distribution in the hypothalamus but not in areas endowed with NGF receptors. The organum vasculosum of the lamina terminalis (OVLT) and the median eminence displayed immunoreactivity for P75. Double immunofluorescence using vimentin, a marker of tanycytes, confirmed that tanycytes were immunoreactive to P75 in the median eminence and in the OVLT. Additionally, tanycytes were in close association with GnRH and kisspeptin in the arcuate nucleus and median eminence of llamas. The choroid plexus of llamas contained TrkA and NGF immunoreactivity but no P75 immunoreactivity. Results of the present study demonstrate sites of action of NGF in the llama hypothalamus, providing support for the hypothesis of a central effect of NGF in the ovulation-inducing mechanism in llamas.


Asunto(s)
Hipotálamo/fisiología , Factor de Crecimiento Nervioso/metabolismo , Ovulación/fisiología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Animales , Camélidos del Nuevo Mundo , Plexo Coroideo , Femenino , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Inmunohistoquímica , Kisspeptinas/genética , Kisspeptinas/metabolismo , Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/genética , Receptor trkA/genética , Receptor trkA/metabolismo , Ovinos/fisiología , Vimentina/genética , Vimentina/metabolismo
10.
Schizophr Bull ; 46(3): 722-731, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-31603232

RESUMEN

Although schizophrenia is a brain disorder, increasing evidence suggests that there may be body-wide involvement in this illness. However, direct evidence of brain structures involved in the presumed peripheral-central interaction in schizophrenia is still unclear. Seventy-nine previously treatment-naïve first-episode schizophrenia patients who were within 2-week antipsychotics initial stabilization, and 41 age- and sex-matched healthy controls were enrolled in the study. Group differences in subcortical brain regional structures measured by MRI and the subclinical cardiovascular, metabolic, immune, and neuroendocrine biomarkers as indexed by allostatic load, and their associations were explored. Compared with controls, patients with schizophrenia had significantly higher allostatic load (P = .001). Lateral ventricle (P < .001), choroid plexus (P < .001), and thalamus volumes (P < .001) were significantly larger, whereas amygdala volume (P = .001) was significantly smaller in patients. The choroid plexus alone was significantly correlated with higher allostatic load after age, sex, education level, and the total intracranial volume were taken into account (t = 3.60, P < .001). Allostatic load was also significantly correlated with PANSS positive (r = 0.28, P = .016) and negative (r = -0.31, P = .008) symptoms, but in opposite directions. The peripheral multisystemic and central nervous system abnormalities in schizophrenia may interact through the choroid plexus during the early stage of the illness. The choroid plexus might provide a sensitive structural biomarker to study the treatment and prevention of brain-periphery interaction abnormalities in schizophrenia.


Asunto(s)
Alostasis , Plexo Coroideo/patología , Esquizofrenia , Estrés Psicológico , Adulto , Alostasis/fisiología , Amígdala del Cerebelo/diagnóstico por imagen , Amígdala del Cerebelo/patología , Biomarcadores , Plexo Coroideo/diagnóstico por imagen , Femenino , Humanos , Ventrículos Laterales/diagnóstico por imagen , Ventrículos Laterales/patología , Imagen por Resonancia Magnética , Masculino , Esquizofrenia/inmunología , Esquizofrenia/metabolismo , Esquizofrenia/patología , Esquizofrenia/fisiopatología , Estrés Psicológico/inmunología , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología , Tálamo/diagnóstico por imagen , Tálamo/patología , Adulto Joven
11.
J Pharmacol Exp Ther ; 371(1): 25-35, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31375638

RESUMEN

Methylcellulose (MC; 0.5% concentration) is commonly used when evaluating investigational agents for efficacy in preclinical models of disease. When administered by the oral (PO) route, MC is considered a Food and Drug Administration "generally recognized as safe" compound. Yet, there is limited data pertaining to the tolerability and impact on model fidelity of repeated intraperitoneal administration of 0.5% MC. Chronic administration of high-concentration MC (2%-2.5%) has been used to induce anemia, splenomegaly, and lesions in multiple organ systems in several preclinical species. Histopathological findings from a diagnostic pathologic analysis of a single mouse from our laboratory with experimentally induced chronic seizures that had received repeated intraperitoneal administration of antiseizure drugs delivered in MC revealed similar widespread lesions. This study thus tested the hypothesis that chronic administration of intraperitoneal, but not PO, MC incites histologic lesions without effects on preclinical phenotype. Male CF-1 mice (n = 2-14/group) were randomized to receive either 6 weeks of twice weekly 0.5% MC or saline (intraperitoneal or PO) following induction of chronic seizures. Histology of a subset of mice revealed lesions in kidney, liver, mediastinal lymph nodes, mesentery, aorta, and choroid plexus only in intraperitoneal MC-treated mice (n = 7/7). Kindled mice that received MC PO (n = 5) or saline (intraperitoneal n = 6, PO n = 3) had no lesions. There were no effects of intraperitoneal MC treatment on body weight, appearance, seizure stability, or behavior. Nonetheless, our findings suggest that repeated intraperitoneal, but not PO, MC elicits systemic organ damage without impacting the model phenotype, which may confound interpretation of investigational drug-induced histologic lesions. SIGNIFICANCE STATEMENT: Methylcellulose (0.5% concentration) is commonly used when evaluating investigational agents for efficacy in preclinical models of disease. Herein, we demonstrate that repeated administration of 0.5% methylcellulose by the intraperitoneal, but not oral, route results in systemic inflammation and presence of foam-laden macrophages but does not impact the behavioral phenotype of a rodent model of neurological disease.


Asunto(s)
Inyecciones Intraperitoneales/efectos adversos , Metilcelulosa/efectos adversos , Fenotipo , Convulsiones/inducido químicamente , Animales , Aorta/efectos de los fármacos , Plexo Coroideo/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Femenino , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Masculino , Metilcelulosa/administración & dosificación , Metilcelulosa/toxicidad , Ratones , Ratones Endogámicos C57BL
12.
Mediators Inflamm ; 2018: 9150207, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30402044

RESUMEN

The study was designed to examine whether the administration of neostigmine (0.5 mg/animal), a peripheral inhibitor of acetylcholinesterase (AChE), during an immune/inflammatory challenge provoked by intravenous injection of bacterial endotoxin-lipopolysaccharide (LPS; 400 ng/kg)-attenuates the synthesis of proinflammatory cytokines in the ovine preoptic area (POA), the hypothalamic structure playing an essential role in the control of the reproduction process, and in the choroid plexus (CP), a multifunctional organ sited at the interface between the blood and cerebrospinal fluid in the ewe. Neostigmine suppressed (p < 0.05) LPS-stimulated synthesis of cytokines such as interleukin- (IL-) 1ß, IL-6, and tumor necrosis factor (TNF) α in the POA, and this effect was similar to that induced by the treatment with systemic AChE inhibitor-donepezil (2.5 mg/animal). On the other hand, both AChE inhibitors did not influence the gene expression of these cytokines and their corresponding receptors in the CP. It was found that this structure seems to not express the neuronal acetylcholine (ACh) receptor subunit alpha-7, required for anti-inflammatory action of ACh. The mechanism of action involves inhibition of the proinflammatory cytokine synthesis on the periphery as well as inhibition of their de novo synthesis rather in brain microvessels and not in the CP. In conclusion, it is suggested that the AChE inhibitors incapable of reaching brain parenchyma might be used in the treatment of neuroinflammatory processes induced by peripheral inflammation.


Asunto(s)
Plexo Coroideo/metabolismo , Citocinas/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Neostigmina/uso terapéutico , Área Preóptica/metabolismo , Animales , Inhibidores de la Colinesterasa/uso terapéutico , Plexo Coroideo/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Interleucina-1beta/sangre , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Área Preóptica/efectos de los fármacos , Ovinos , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Chem Neuroanat ; 94: 1-7, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30118754

RESUMEN

The eusocial Damaraland mole-rat (Fukomys damarensis) represents an extreme example of reproductive skew, in that reproduction is completely blocked in female subordinate group members. It is thought that in these animals normal GnRH secretion from the hypothalamus is disrupted. Prolactin, a peptide hormone secreted from the anterior pituitary gland, has been implicated in a wide variety of functions. Well documented in rodents is its role in mediating lactational infertility. Elevated circulating prolactin levels, such as during lactation, are associated with reduced GnRH release into the portal blood and with a reduction in the frequency and amplitude of LH pulses. The present study aimed at investigating whether such a mechanism could act in reproductively suppressed female Damaraland mole-rats. By means of in situ hybridisation we studied the distribution and gene expression of the prolactin receptor (Prlr) in wild-caught female Damaraland mole-rats with different reproductive status. Substantial Prlr expression was found in several brain regions, with highest levels in the choroid plexus and moderate expression in the preoptic and tuberal hypothalamus. While in reproductive and non-reproductive females plasma prolactin levels were very low and not significantly different, quantification of the Prlr hybridisation signal revealed significant differences in relation to reproductive status. Reproductively suppressed females had increased expression of Prlr in the choroid plexus and in the arcuate nucleus (ARC) when compared to reproductive females. This suggests higher local prolactin levels in the brain of suppressed females. Together with previous findings, it could indicate that prolactin inhibits ARC kisspeptin neurons, which then would lead to reduced activation of GnRH neurons in such females.


Asunto(s)
Plexo Coroideo/metabolismo , Hipotálamo/metabolismo , Prolactina/sangre , Receptores de Prolactina/metabolismo , Reproducción/fisiología , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Ratas Topo , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Prolactina/genética
14.
Chronobiol Int ; 35(2): 270-279, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29172740

RESUMEN

The suprachiasmatic nucleus (SCN) of the hypothalamus is considered the master circadian oscillator in mammals. However, extra-SCN structures in the brain also display daily rhythms. Recently, we have demonstrated that the choroid plexus (CP) expresses core clock genes that are subjected to circadian regulation in a sex-dependent manner. By using CP explants cultured from female knock-in mice carrying the Period-luciferase transgene, we show that CP exhibits endogenous circadian rhythms of PERIOD2::LUCIFERASE expression. Furthermore, we demonstrate that estrogen declines following ovariectomy modulates the daily rhythm expression of Bmal1, Per1 and Per2 in female rat CP, corroborating data obtained in experiments where rat CP epithelial cell (CPEC) cultures were incubated with 17ß-estradiol (E2). The molecular mechanism underlying these effects was also investigated, and we provide evidence that the estrogen receptor (ER) mediates the response of clock genes to E2. In conclusion, our study proves that the CP harbors a circadian oscillator that is modulated by estrogens and demonstrates that E2 regulation occurs through an estrogen-receptor-dependent mechanism.


Asunto(s)
Plexo Coroideo/metabolismo , Ritmo Circadiano/fisiología , Estrógenos/metabolismo , Proteínas Circadianas Period/metabolismo , Animales , Femenino , Hipotálamo/metabolismo , Luciferasas/metabolismo , Ratones Transgénicos , Neuronas/metabolismo , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiología
15.
Turk Neurosurg ; 27(5): 823-826, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28820225

RESUMEN

Bilateral thalamic dysfunction secondary to venous congestion may result from either venous sinus thrombosis or high flow arteriovenous malformations or a combination of both. We present a case of bilateral thalamic edema resulting from concomitant choroid plexus arteriovenous malformation (AVM) and straight sinus thrombosis and describe our treatment approach. The patient presented with several weeks of progressive confusion and memory deficits. Magnetic resonance imaging and venography (MRI/ MRV) showed bilateral thalamic T2 hyperintensities and straight sinus thrombosis. Subsequent cerebral angiography revealed a choroid plexus AVM within the right lateral ventricle. The patient underwent surgical resection of the AVM resulting in postoperative resolution of bilateral thalamic edema on MRI and improvement of his confusion and memory deficits. This case demonstrates a rare example of reversible bilateral thalamic edema secondary to venous hypertension from both an AVM and sinus occlusion after appropriate treatment of the AVM.


Asunto(s)
Plexo Coroideo/anomalías , Edema/etiología , Malformaciones Arteriovenosas Intracraneales/complicaciones , Trombosis de los Senos Intracraneales/complicaciones , Tálamo/cirugía , Angiografía Cerebral , Plexo Coroideo/diagnóstico por imagen , Plexo Coroideo/cirugía , Edema/diagnóstico por imagen , Edema/cirugía , Humanos , Malformaciones Arteriovenosas Intracraneales/diagnóstico por imagen , Malformaciones Arteriovenosas Intracraneales/cirugía , Angiografía por Resonancia Magnética , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Trombosis de los Senos Intracraneales/diagnóstico por imagen , Trombosis de los Senos Intracraneales/cirugía , Tálamo/diagnóstico por imagen , Resultado del Tratamiento
16.
Turk Neurosurg ; 27(5): 823-826, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27509460

RESUMEN

Bilateral thalamic dysfunction secondary to venous congestion may result from either venous sinus thrombosis or high flow arteriovenous malformations or a combination of both. We present a case of bilateral thalamic edema resulting from concomitant choroid plexus arteriovenous malformation (AVM) and straight sinus thrombosis and describe our treatment approach. The patient presented with several weeks of progressive confusion and memory deficits. Magnetic resonance imaging and venography (MRI/ MRV) showed bilateral thalamic T2 hyperintensities and straight sinus thrombosis. Subsequent cerebral angiography revealed a choroid plexus AVM within the right lateral ventricle. The patient underwent surgical resection of the AVM resulting in postoperative resolution of bilateral thalamic edema on MRI and improvement of his confusion and memory deficits. This case demonstrates a rare example of reversible bilateral thalamic edema secondary to venous hypertension from both an AVM and sinus occlusion after appropriate treatment of the AVM.


Asunto(s)
Plexo Coroideo/patología , Edema/cirugía , Malformaciones Arteriovenosas Intracraneales/cirugía , Trombosis de los Senos Intracraneales/cirugía , Tálamo/patología , Adulto , Edema/complicaciones , Humanos , Malformaciones Arteriovenosas Intracraneales/complicaciones , Malformaciones Arteriovenosas Intracraneales/diagnóstico por imagen , Masculino , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/cirugía , Trombosis de los Senos Intracraneales/inducido químicamente
17.
PLoS One ; 11(11): e0166416, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27893788

RESUMEN

Prolactin (PRL) is well characterized for its roles in initiation and maintenance of lactation, and it also suppresses stress-induced responses. Feeding a high-fat diet (HFD) disrupts activity of the hypothalamic-pituitary-adrenal (HPA) axis. Whether PRL regulates HPA axis activation under HFD feeding is not clear. Male and female wildtype (WT) and PRL knockout (KO) mice were fed either a standard low-fat diet (LFD) or HFD for 12 weeks. Circulating corticosterone (CORT) levels were measured before, during, and after mice were subjected to an acute restraint stress or remained in their home cages as no stress controls. HFD feeding increased leptin levels, but the increase was lower in KO than in WT mice. All stressed female groups and only LFD-fed stressed males had elevated CORT levels compared to their no stress same-sex counterparts regardless of genotype. These results indicated that HFD consumption blunted the HPA axis response to acute stress in males but not females. Additionally, basal hypothalamic CRH content was lower in HFD than LFD males, but was similar among female groups. Furthermore, although basal CORT levels were similar among KO and WT groups, CORT levels were higher in KO mice than their WT counterparts during stress, suggesting that loss of PRL led to greater HPA axis activation. Basal PRL receptor mRNA levels in the choroid plexus were higher in HFD than LFD same-sex counterparts, suggesting activation of central PRL's action by HFD feeding in both males and females. Current results confirmed PRL's roles in suppression of the stress-induced HPA axis activation. Although HFD feeding activated central PRL's action in both sexes, only the male HPA axis was dampened by HFD feeding.


Asunto(s)
Dieta Alta en Grasa , Prolactina/genética , Estrés Fisiológico , Animales , Composición Corporal , Peso Corporal , Plexo Coroideo/metabolismo , Corticosterona/sangre , Dieta con Restricción de Grasas , Ingestión de Energía , Femenino , Hipotálamo/metabolismo , Leptina/sangre , Masculino , Ratones , Ratones Noqueados , Prolactina/deficiencia , ARN Mensajero/metabolismo , Receptores de Prolactina/genética , Receptores de Prolactina/metabolismo
18.
Artículo en Chino | MEDLINE | ID: mdl-27514264

RESUMEN

OBJECTIVE: To investigate the impairment in primary cultured rat choroid plexus epithelial cells (CPECs)induced by aluminum. METHODS: The choroid plexus isolated from Sprague-Dawley rats 14 days old was cut into pieces and digested by trypsin in the sterile area. The obtained single cells were cultured in DMEM with 1% epidermal growth factor and 20% fetal calf serum. Five days later, immunohistochemistry with anti-transthyretin antibody was used to identify the purity of cultured cells. The well-grown cells were treated with aluminum lactate at different concentrations (0, 100, 400, and 1 600 µmol/L for control, lowdose, mediumdose, and highdose groups). Fortyeight hours later, the cell viability, apoptotic rate, level of reactive oxygen species (ROS), and activity of superoxide dismutase (SOD)were measured in each group to evaluate the impairment in primary cultured rat CPECs by aluminum. RESULTS: More than 95% of the cultured cells were identified as CPECs. The medium-and high-dose groups had significantly lower cell viability than the control group(86.74%±4.03% vs 100%, P<0.01; 81.90%±9.17% vs 100%, P<0.01). The high-dose group had significantly lower cell viability than the lowdose group (81.90%±9.17% vs 92.92%±8.81%, P<0.01). The medium-and high-dose groups had significantly higher apoptotic rates than the control group (7.26%±0.99% vs 1.29%±0.03%, P<0.01; 22.25%±1.55% vs 1.29%±0.03%, P<0.01)and the low-dose group (7.26%±0.99% vs 1.68%±0.27%, P<0.01; 22.25%±1.55% vs 1.68%±0.27%, P<0.01). The high-dose group had a significantly higher apoptotic rate than the medium-dose group (22.25%±1.55% vs 7.26%±0.99%, P<0.01). The mediumand high-dose groups had significantly higher fluorescence intensity of ROS than the control group (22.23%±0.41% vs 17.24%±0.09%, P<0.05; 25.10%±1.13% vs 17.24%±0.09%, P<0.05)and the lowdose group (22.23%±0.41% vs 18.31%±0.21%, P<0.05; 25.10%±1.13% vs 18.31%±0.21%, P<0.05). The highdose group had significantly higher fluorescence intensity of ROS than the mediumdose group (25.10%±1.13% vs 22.23%±0.41%, P< 0.05). The low-, medium-and high-dose groups had significantly lower SOD activity than the control group[(28.65±0.74)U/g Hb vs (37.35±1.05)U/g Hb, P<0.05; (22.75±1.94)U/g Hb vs (37.35±1.05)U/g Hb, P<0.05; (13.29±0.64)U/g Hb vs(37.35±1.05)U/g Hb, P<0.05]. The medium-and high-dose groups had significantly lower SOD activity than the low-dose group[(22.75±1.94)U/g Hb vs(28.65±0.74)U/g Hb, P<0.05; (13.29±0.64)U/g Hb vs (28.65±0.74)U/g Hb, P<0.05], while the high-dose group had had significantly lower SOD activity than the medium-dose group[(13.29±0.64)U/g Hb vs (22.75±1.94)U/g Hb, P<0.05]. There were no significant differences in cell viability, apoptotic rate, level of ROS, or activity of SOD between any other two groups (P>0.05). CONCLUSION: Aluminum lactate may induce impairment in primary cultured rat CPECs. It reduces the cell viability, elevates the apoptotic rate, and causes oxidative stress.


Asunto(s)
Plexo Coroideo , Células Epiteliales , Aluminio , Animales , Supervivencia Celular , Células Cultivadas , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Superóxido Dismutasa
19.
PLoS One ; 10(3): e0119493, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25774914

RESUMEN

In the current study, we tested the in vivo effects of Yy1 gene dosage on the Peg3 imprinted domain with various breeding schemes utilizing two sets of mutant alleles. The results indicated that a half dosage of Yy1 coincides with the up-regulation of Peg3 and Zim1, suggesting a repressor role of Yy1 in this imprinted domain. This repressor role of Yy1 is consistent with the observations derived from previous in vitro studies. The current study also provided an unexpected observation that the maternal allele of Peg3 is also normally expressed, and thus the expression of Peg3 is bi-allelic in the specific areas of the brain, including the choroid plexus, the PVN (Paraventricular Nucleus) and the SON (Supraoptic Nucleus) of the hypothalamus. The exact roles of the maternal allele of Peg3 in these cell types are currently unknown, but this new finding confirms the previous prediction that the maternal allele may be functional in specific cell types based on the lethality associated with the homozygotes for several mutant alleles of the Peg3 locus. Overall, these results confirm the repressor role of Yy1 in the Peg3 domain and also provide a new insight regarding the bi-allelic expression of Peg3 in mouse brain.


Asunto(s)
Encéfalo/metabolismo , Proteínas Portadoras/genética , Dosificación de Gen , Factores de Transcripción de Tipo Kruppel/genética , Proteínas del Tejido Nervioso/genética , Factor de Transcripción YY1/genética , Alelos , Animales , Plexo Coroideo/metabolismo , Proteínas de Unión al ADN , Impresión Genómica , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Mutación , Especificidad de Órganos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Supraóptico/metabolismo , Regulación hacia Arriba
20.
Amino Acids ; 47(5): 1053-63, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25691144

RESUMEN

TFF3 is a member of the trefoil factor family (TFF) predominantly secreted by mucous epithelia. Minute amounts are also expressed in the immune system and the brain. In the latter, particularly the hypothalamo-pituitary axis has been investigated in detail in the past. Functionally, cerebral TFF3 has been reported to be involved in several processes such as fear, depression, learning and object recognition, and opiate addiction. Furthermore, TFF3 has been linked with neurodegenerative and neuropsychiatric disorders (e.g., Alzheimer's disease, schizophrenia, and alcoholism). Here, using immunohistochemistry, a systematic survey of the TFF3 localization in the adult human brain is presented focusing on extrahypothalamic brain areas. In addition, the distribution of TFF3 in the developing human brain is described. Taken together, neurons were identified as the predominant cell type to express TFF3, but to different extent; TFF3 was particularly enriched in various midbrain and brain stem nuclei. Besides, TFF3 immunostaining staining was observed in oligodendroglia and the choroid plexus epithelium. The wide cerebral distribution should help to explain its multiple effects in the CNS.


Asunto(s)
Plexo Coroideo/metabolismo , Mesencéfalo/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Péptidos/genética , Aborto Espontáneo , Adulto , Amígdala del Cerebelo/química , Amígdala del Cerebelo/metabolismo , Mapeo Encefálico , Cerebelo/química , Cerebelo/metabolismo , Corteza Cerebral/química , Corteza Cerebral/metabolismo , Plexo Coroideo/química , Femenino , Feto , Expresión Génica , Hipocampo/química , Hipocampo/metabolismo , Humanos , Hipotálamo/química , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Mesencéfalo/química , Persona de Mediana Edad , Neuronas/química , Oligodendroglía/química , Especificidad de Órganos , Péptidos/metabolismo , Hipófisis/química , Hipófisis/metabolismo , Neurohipófisis/química , Neurohipófisis/metabolismo , Factor Trefoil-3 , Sustancia Blanca/química , Sustancia Blanca/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA