Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
J Cell Biol ; 122(3): 623-33, 1993 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7687605

RESUMEN

Actin depolymerizing factor (ADF) is an 18.5-kD protein with pH-dependent reciprocal F-actin binding and severing/depolymerizing activities. We previously showed developing muscle down-regulates ADF (J. R. Bamburg and D. Bray. 1987. J. Cell Biol. 105: 2817-2825). To further study this process, we examined ADF expression in chick myocytes cultured in vitro. Surprisingly, ADF immunoreactivity increases during the first 7-10 d in culture. This increase is due to the presence of a new ADF species with higher relative molecular weight which reacts identically to brain ADF with antisera raised against either brain ADF or recombinant ADF. We have purified both ADF isoforms from myocytes and have shown by peptide mapping and partial sequence analysis that the new isoform is structurally related to ADF. Immunoprecipitation of both isoforms from extracts of cells prelabeled with [32P]orthophosphate showed that the new isoform is radiolabeled, predominantly on a serine residue, and hence is called pADF. pADF can be converted into a form which comigrates with ADF on 1-D and 2-D gels by treatment with alkaline phosphatase. pADF has been quantified in a number of cells and tissues where it is present from approximately 18% to 150% of the amount of unphosphorylated ADF. pADF, unlike ADF, does not bind to G-actin, or affect the rate or extent of actin assembly. Four ubiquitous protein kinases failed to phosphorylate ADF in vitro suggesting that ADF phosphorylation in vivo is catalyzed by a more specific kinase. We conclude that the ability to regulate ADF activity is important to muscle development since myocytes have both pre- and posttranslational mechanisms for regulating ADF activity. The latter mechanism is apparently a general one for cell regulation of ADF activity.


Asunto(s)
Actinas/metabolismo , Proteínas de Microfilamentos/aislamiento & purificación , Proteínas de Microfilamentos/metabolismo , Músculos/metabolismo , Factores Despolimerizantes de la Actina , Fosfatasa Alcalina/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Embrión de Pollo , Pollos , Destrina , Electroforesis en Gel de Poliacrilamida , Epítopos , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/inmunología , Datos de Secuencia Molecular , Músculos/química , Músculos/citología , Mapeo Peptídico , Fosforilación , Fosfoserina/análisis , Proteínas Quinasas/metabolismo
2.
Acta Neuropathol Commun ; 7(1): 82, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31113487

RESUMEN

Microglia affect Alzheimer's disease (AD) pathogenesis in opposing manners, by protecting against amyloid accumulation in early phases of the disease and promoting neuropathology in advanced stages. Recent research has identified specific microglial interactions with amyloid plaques that exert important protective functions including attenuation of early pathology. It is unknown how these protective microglial interactions with plaques are affected by apolipoprotein E (APOE) genotype and sex, two well-established AD risk factors that modulate microglial function. We investigated this question using quantitative confocal microscopy to compare microglial interactions with amyloid plaques in male and female EFAD mice across APOE3 and APOE4 genotypes at 6 months of age. We observed that microglial coverage of plaques is highest in male APOE3 mice with significant reductions in coverage observed with both APOE4 genotype and female sex. Plaque compaction, a beneficial consequence of microglial interactions with plaques, showed a similar pattern in which APOE4 genotype and female sex were associated with significantly lower values. Within the plaque environment, microglial expression of triggering receptor expressed on myeloid cells 2 (TREM2), a known regulator of microglial plaque coverage, was highest in male APOE3 mice and reduced by APOE4 genotype and female sex. These differences in plaque interactions were unrelated to the number of microglial processes in the plaque environment across groups. Interestingly, the pattern of amyloid burden across groups was opposite to that of microglial plaque coverage, with APOE4 genotype and female sex showing the highest amyloid levels. These findings suggest a possible mechanism by which microglia may contribute to the increased AD risk associated with APOE4 genotype and female sex.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Encéfalo/patología , Microglía/patología , Placa Amiloide/patología , Animales , Femenino , Genotipo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones Transgénicos , Receptores Inmunológicos/metabolismo , Caracteres Sexuales
3.
Transl Psychiatry ; 8(1): 261, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30498214

RESUMEN

Exposure to traffic-related air pollution (TRAP) is associated with a range of neurodevelopmental disorders in human populations. In rodent models, prenatal TRAP exposure increased depressive behaviors and increased brain microglial activity. To identify cellular mechanisms, we examined adult neurogenesis and the blood-brain barrier (BBB) in relation to cognition and motivated behaviors in rats that were exposed to a nano-sized TRAP subfraction from gestation into adulthood. At age 5 months, exposed male rats had 70% fewer newly generated neurons in the dentate gyrus (DG) of the hippocampus. Microglia were activated in DG and CA1 subfields (35% more Iba1). The BBB was altered, with a 75% decrease of the tight junction protein ZO-1 in the CA1 layer, and twofold more iron deposits, a marker of microhemorrhages. The exposed rats had impaired contextual memory (novel object in context), reduced food-seeking behavior, and increased depressive behaviors (forced swim). Deficits of de novo neurogenesis were inversely correlated with depressive behavior, whereas increased microbleeds were inversely correlated with deficits in contextual memory. These findings give the first evidence that prenatal and early life exposure to TRAP impairs adult hippocampal neurogenesis and increases microbleeds in association with behavioral deficits.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Conducta Animal , Hipocampo/fisiopatología , Neurogénesis , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Emisiones de Vehículos/toxicidad , Animales , Astrocitos/fisiología , Barrera Hematoencefálica/metabolismo , Depresión/inducido químicamente , Conducta Alimentaria , Femenino , Masculino , Memoria , Microglía/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas Sprague-Dawley
4.
Transl Psychiatry ; 7(1): e1022, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28140404

RESUMEN

Exposure to particulate matter (PM) in the ambient air and its interactions with APOE alleles may contribute to the acceleration of brain aging and the pathogenesis of Alzheimer's disease (AD). Neurodegenerative effects of particulate air pollutants were examined in a US-wide cohort of older women from the Women's Health Initiative Memory Study (WHIMS) and in experimental mouse models. Residing in places with fine PM exceeding EPA standards increased the risks for global cognitive decline and all-cause dementia respectively by 81 and 92%, with stronger adverse effects in APOE ɛ4/4 carriers. Female EFAD transgenic mice (5xFAD+/-/human APOE ɛ3 or ɛ4+/+) with 225 h exposure to urban nanosized PM (nPM) over 15 weeks showed increased cerebral ß-amyloid by thioflavin S for fibrillary amyloid and by immunocytochemistry for Aß deposits, both exacerbated by APOE ɛ4. Moreover, nPM exposure increased Aß oligomers, caused selective atrophy of hippocampal CA1 neurites, and decreased the glutamate GluR1 subunit. Wildtype C57BL/6 female mice also showed nPM-induced CA1 atrophy and GluR1 decrease. In vitro nPM exposure of neuroblastoma cells (N2a-APP/swe) increased the pro-amyloidogenic processing of the amyloid precursor protein (APP). We suggest that airborne PM exposure promotes pathological brain aging in older women, with potentially a greater impact in ɛ4 carriers. The underlying mechanisms may involve increased cerebral Aß production and selective changes in hippocampal CA1 neurons and glutamate receptor subunits.


Asunto(s)
Disfunción Cognitiva/epidemiología , Demencia/epidemiología , Exposición a Riesgos Ambientales/estadística & datos numéricos , Interacción Gen-Ambiente , Material Particulado , Anciano , Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteína E4/genética , Atrofia , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Línea Celular Tumoral , Cerebro/efectos de los fármacos , Cerebro/metabolismo , Disfunción Cognitiva/genética , Demencia/genética , Femenino , Humanos , Técnicas In Vitro , Ratones , Ratones Transgénicos , Neuritas/efectos de los fármacos , Neuritas/patología , Receptores AMPA/efectos de los fármacos , Receptores AMPA/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-28042517

RESUMEN

Cerebral microbleeds (MB) and small vessel disease (SVD) with congophilic arterial angiopathy (CAA) are increasingly recognized as a variable factor in AD cognitive impairments. This commentary on our recent report on sex-ApoE interactions in MBs published this February, briefly explores three aspects of MBs that could not be fully discussed therein: I, A possible gap between the prevalence of MBs as detected by MRI and post mortem analysis; II, The role of hemoglobin-degradation products in amyloid-attributed neurodegenerative changes; and III, Possible assessment of MB by cerebrospinal fluid (CSF) assays for iron-related markers to better screen patient subgroups for AD interventions.

6.
Neurobiol Aging ; 19(1): 97-103, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9562510

RESUMEN

Astrocytes and microglia from cerebral cortex of 3-, 6-, 12-, and 24-month-old F344 male rat donors showed progressively greater proliferation during primary culture. Microglia from aging donor brains exhibited an amoeboid-like morphology and express antigens characteristic of an activated state (e.g., major histocompatibility complex class II). Moreover, microglia from aging donors were less sensitive to several types of regulators. Granulocyte-macrophage colony stimulating factor stimulated proliferation in microglia from young, but not aging brains. Transforming growth factor (TGF)-beta1 inhibited astrocytic and microglial proliferation in cultures from young, but not aging donors. Similarly, the inhibition of lipopolysaccharide-induced NO production by TGF-beta1 in microglia was impaired in cultures from 12-month (middle-age) brains. Another aging change detected by middle age, increased glial fibrillary acidic protein (GFAP) expression, also persisted in astrocytes from 12- to 24-month-old brains, as evaluated by increased activity of a 5'-upstream GFAP promoter construct. Thus, both microglia and astrocytes originated from aging cerebral cortex maintain in vitro at least some of the activated phenotypes of aging glia that are observed in vivo. This new in vitro cell model may allow efficient analysis of glial age changes.


Asunto(s)
Envejecimiento/fisiología , Astrocitos/fisiología , Regulación hacia Abajo/fisiología , Microglía/fisiología , Animales , División Celular/fisiología , Células Cultivadas , Proteína Ácida Fibrilar de la Glía/biosíntesis , Inmunohistoquímica , Masculino , Nitrógeno/metabolismo , Fenotipo , Ratas , Ratas Endogámicas F344 , Timidina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
7.
Free Radic Biol Med ; 23(3): 524-8, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9214592

RESUMEN

Glial fibrillary acidic protein (GFAP), an intermediate filament of astrocytes, shows increased expression during aging. Because we found that chronic food restriction retards the increase of GFAP mRNA in aging rats and because food restriction decreases the load of oxidized proteins and lipids in association with increased life span, we investigated the regulation of GFAP during oxidative stress and aging. First, we showed that food restriction decreased the transcription of GFAP in aging rats. This result generalizes effects of food restriction on age changes of transcription; whether transcription decreases during aging as in hepatic genes, or increases during aging as in astrocytic GFAP, food restriction attenuates the age change. Moreover, food restriction decreased microglial activation during aging, which suggested the hypothesis that GFAP expression is sensitive to oxidative stress. Because GFAP transcription in cultured glia is increased by oxidative stress in response to hydrogen peroxide and cysteamine whether or not microglia were present, we conclude that responses of GFAP to oxidative stress in astrocytes do not depend on microglial activation. The results implicate oxidative stress in the increased expression of GFAP during aging, but also in responses to brain injury.


Asunto(s)
Envejecimiento/metabolismo , Dieta , Regulación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Hipocampo/metabolismo , Estrés Oxidativo , Animales , Astrocitos/metabolismo , Células Cultivadas , Cisteamina/farmacología , Proteína Ácida Fibrilar de la Glía/análisis , Peróxido de Hidrógeno/farmacología , Inmunohistoquímica , Lipopolisacáridos/farmacología , Complejo Mayor de Histocompatibilidad/genética , Masculino , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas , Transcripción Genética/genética
8.
Am J Surg Pathol ; 2(4): 339-55, 1978 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-736209

RESUMEN

In reviewing all proliferative serous tumors of the ovary seen at Barnes Hospital from 1950 to 1974, we quantitated histologic characteristics and defined criteria for diagnosis in 55 borderline tumors, 13 well-differentiated cystadenocarcinomas, and 15 cystadenomas with unusual proliferative areas. This last type with focal proliferation behaved in a benign fashion and should be considered a variant of a simple cystadenoma. Stromal invasion was the only histologic feature which consistently distinguished carcinomas from the borderline tumors. The presence in some borderline tumors of severe cellular atypia, marked epithelial disorganization, frequent mitoses, and cribriform glands in the stroma neither signified carcinoma nor indicated poor prognosis. No patient with a Stage I borderline lesion died of tumor. Although the mortality of patients with Stage IIb or Stage III borderline tumors is high, tumor-related deaths rarely occurred before 5 years, and three patients lived more than 10 years. Borderline serous tumors are low-grade malignant neoplasms which differ from overt carcinomas in the excellent prognosis of Stage I lesions and in long survival even with widespread abdominal disease.


Asunto(s)
Cistadenocarcinoma/patología , Cistoadenoma/patología , Neoplasias Ováricas/patología , Femenino , Humanos , Pronóstico
9.
J Neuroimmunol ; 56(1): 53-63, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7822482

RESUMEN

Entorhinal cortex lesions (ECL) that damage the perforant path to the dentate gyrus of the hippocampal formation were used to model the regulation of vimentin (VIM) mRNA. ECL increased VIM mRNA in the ipsilateral hippocampus and in the ipsilateral cortex including the wound cavity within 1 day. By in situ hybridization, at 4 days post-ECL, VIM mRNA increased two-fold in the molecular layer of the dentate gyrus. VIM protein was co-localized by immunocytochemistry to astrocytes and microglia/macrophages. Transforming growth factor-beta 1 (TGF-beta 1), which was previously shown to increase in microglia/macrophages of the molecular layer after hippocampal deafferentation by ECL, was investigated as a regulator of VIM expression. Infusions of TGF-beta 1 into the lateral ventricle induced VIM mRNA with dose-dependence, e.g. infusion of 100 ng TGF-beta 1 increased VIM mRNA three-fold. The increase in VIM mRNA was localized by in situ hybridization to astrocytes and microglia in the molecular layer of the dentate gyrus. These findings further implicate TGF-beta 1 as a regulator of cytoskeletal proteins during synaptic reorganization.


Asunto(s)
Corteza Cerebral/metabolismo , Corteza Entorrinal/fisiología , Hipocampo/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Vimentina/metabolismo , Animales , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas de Neurofilamentos/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Distribución Tisular , Vimentina/genética
10.
J Neuroimmunol ; 58(1): 101-10, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7730444

RESUMEN

This study characterizes the effect of transforming growth factor (TGF) beta 1 on clusterin expression in rat brain cells. 24 h after an acute unilateral intracerebroventricular infusion of TGF-beta 1, clusterin mRNA prevalence was increased in astrocytes that contained immunoreactive (IR) glial fibrillary acidic protein (GFAP). TGF-beta 1 selectively induced clusterin mRNA in astrocytes, as no clusterin mRNA was detected in neurons, oligodendrocytes, or microglia. TGF-beta 1 induced a bilateral increase in clusterin mRNA per astrocyte. Astrocyte hypertrophy (GFAP-IR area) was only increased on the ipsilateral side. In pure astrocyte cultures, TGF-beta 1 (200 pM) decreased clusterin mRNA levels and the rate of clusterin RNA transcription. However, in cultures of astrocytes that contained microglia and oligodendrocytes (mixed glia cultures), TGF-beta 1 caused a dose-dependent increase in astrocytic clusterin mRNA levels. The astrocytes that responded to TGF-beta 1 included two GFAP-IR subtypes, type 1 and 2. TGF-beta 1 increased clusterin protein in the conditioned medium from cultured glia, in either monotypic or mixed glial cultures. Thus, TGF-beta 1 and heterotypic cell interactions influence clusterin expression by astrocytes and may be important to the role of clusterin in multiple sclerosis, AIDS, and Alzheimer's disease.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Ventrículos Cerebrales/fisiología , Cuerpo Estriado/metabolismo , Expresión Génica , Glicoproteínas/biosíntesis , Chaperonas Moleculares , Factor de Crecimiento Transformador beta/farmacología , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Ventrículos Cerebrales/efectos de los fármacos , Clusterina , Cuerpo Estriado/citología , Proteína Ácida Fibrilar de la Glía/análisis , Infusiones Parenterales , Masculino , Microglía/citología , Microglía/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas F344 , Transcripción Genética , Factor de Crecimiento Transformador beta/administración & dosificación
11.
Neuroscience ; 58(3): 563-72, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8170537

RESUMEN

Transforming growth factor-beta 1 was studied as a possible regulator of messenger RNAs in astrocytes and neurons that increase after hippocampal deafferentation by perforant path transection: tubulin alpha 1, clusterin and glial fibrillary acidic protein messenger RNA. Because transforming growth factor-beta 1 messenger RNA is increased after this lesion, we examined which messenger RNA lesion responses could be induced by transforming growth factor-beta 1 alone. Porcine transforming growth factor-beta 1 infused into the lateral ventricle elevated the messenger RNAs for tubulin alpha 1, clusterin and glial fibrillary acidic protein 24 h after infusion in the ipsilateral hippocampus. As assayed by nuclear run-on, the transcription of glial fibrillary acidic protein RNA was increased in the ipsilateral hippocampus after perforant path transection and in primary rat astrocyte cultures by transforming growth factor-beta 1. In contrast, transforming growth factor-beta 1 did not change apolipoprotein-E messenger RNA or transcription, or growth associated protein-43 messenger RNA levels. We conclude that transforming growth factor-beta 1 increases subsets of neuronal and astrocyte messenger RNAs coding for cytoskeletal proteins that are also elevated in response to experimental lesions and Alzheimer's disease. This suggests that transforming growth factor-beta 1 might be a local organizing factor of neuronal and astrocyte responses to brain injury.


Asunto(s)
Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/biosíntesis , Glicoproteínas/biosíntesis , Chaperonas Moleculares , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Tubulina (Proteína)/biosíntesis , Animales , Apolipoproteínas E/biosíntesis , Apolipoproteínas E/genética , Astrocitos/efectos de los fármacos , Secuencia de Bases , Northern Blotting , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Clusterina , Proteína Ácida Fibrilar de la Glía/genética , Glicoproteínas/genética , Hibridación in Situ , Masculino , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas F344 , Sinapsis/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Tubulina (Proteína)/genética
12.
Neuroscience ; 101(2): 313-21, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11074155

RESUMEN

Transforming growth factor-beta1 is a multifunctional peptide with increased expression during Alzheimer's disease and other neurodegenerative conditions which involve inflammatory mechanisms. We examined the autoregulation of transforming growth factor-beta1 and transforming growth factor-beta receptors and the effects of transforming growth factor-beta1 on complement C1q in brains of adult Fischer 344 male rats and in primary glial cultures. Perforant path transection by entorhinal cortex lesioning was used as a model for the hippocampal deafferentation of Alzheimer's disease. In the hippocampus ipsilateral to the lesion, transforming growth factor-beta1 peptide was increased >100-fold; the messenger RNAs encoding transforming growth factor-beta1, transforming growth factor-beta type I and type II receptors were also increased, but to a smaller degree. In this acute lesion paradigm, microglia are the main cell type containing transforming growth factor-beta1, transforming growth factor-beta type I and II receptor messenger RNAs, shown by immunocytochemistry in combination with in situ hybridization. Autoregulation of the transforming growth factor-beta1 system was examined by intraventricular infusion of transforming growth factor-beta1 peptide, which increased hippocampal transforming growth factor-beta1 messenger RNA levels in a dose-dependent fashion. Similarly, transforming growth factor-beta1 increased levels of transforming growth factor-beta1 messenger RNA and transforming growth factor-beta type II receptor messenger RNA (IC(50), 5pM) and increased release of transforming growth factor-beta1 peptide from primary microglia cultures. Interactions of transforming growth factor-beta1 with complement system gene expression are also indicated, because transforming growth factor-beta1 decreased C1qB messenger RNA in the cortex and hippocampus, after intraventricular infusion, and in cultured glia. These indications of autocrine regulation of transforming growth factor-beta1 in the rodent brain support a major role of microglia in neural activities of transforming growth factor-beta1 and give a new link between transforming growth factor-beta1 and the complement system. The auto-induction of the transforming growth factor-beta1 system has implications for transgenic mice that overexpress transforming growth factor-beta1 in brain cells and for its potential role in amyloidogenesis.


Asunto(s)
Encéfalo/efectos de los fármacos , Complemento C1q/efectos de los fármacos , Microglía/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Complemento C1q/genética , Complemento C1q/metabolismo , Desnervación/efectos adversos , Modelos Animales de Enfermedad , Encefalitis/metabolismo , Encefalitis/patología , Encefalitis/fisiopatología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Masculino , Microglía/citología , Microglía/metabolismo , Vía Perforante/patología , Vía Perforante/fisiopatología , Vía Perforante/cirugía , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1
13.
Neuroscience ; 62(3): 741-58, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7870303

RESUMEN

This study concerns expression of the genes encoding three multifunctional proteins: clusterin and two complement cascade components, C1q and C4. Previous work from this and other laboratories has established that clusterin, Clq and C4 messenger RNAs are elevated during Alzheimer's disease, and in response to deafferenting and excitotoxic brain lesion. This study addresses hippocampal clusterin, ClqB and C4 expression in response to neurotoxins that caused selective neuron death. Kainate, which preferentially kills hippocampal CA3 pyramidal neurons but not dentate gyrus granule neurons induced clusterin immunoreactivity in CA1 and CA3 pyramidal neurons and adjacent astrocytes, but not in dentate gyrus granule neurons. In contrast, colchicine, which preferentially kills the dentate gyrus granule neurons, induced clusterin immunoreactivity in the local neuropil as punctate deposits, but not in the surviving or degenerating dentate gyrus granule neurons. Clusterin messenger RNA was increased in astrocytes. ClqB and C4 messenger RNAs increased within 48 h after kainate injections, particularly in the CA3 pyramidal layer, less in the dentate gyrus-CA4, and less in CA1. Clq immunoreactivity was detected in CA1 pyramidal neurons and also as small punctate deposits in the CA1 region at eight and 14 days after kainate. The increase of both clusterin and ClqB messenger RNAs after kainate injections was blocked by barbiturates that prevented seizures and neurodegeneration. In primary hippocampal neuronal cultures treated with glutamate, a subpopulation of cultured neurons that survived glutamate toxicity also had parallel elevations of clusterin and ClqB messenger RNA. In conclusion, cytotoxins that target selective hippocampal neurons increase the expression of both clusterin and ClqB in vivo and in vitro. These results show that elevations of clusterin messenger RNA or protein can be dissociated from each other and from cell death. These increased messenger RNAs were associated with immunoreactive deposits that differed by cell type and intra- versus extracellular locations. These results suggest that the complement system is involved in brain responses to injury.


Asunto(s)
Astrocitos/efectos de los fármacos , Complemento C1q/biosíntesis , Complemento C4/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Glutámico/toxicidad , Glicoproteínas/biosíntesis , Hipocampo/efectos de los fármacos , Ácido Kaínico/toxicidad , Chaperonas Moleculares , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/efectos de los fármacos , Convulsiones/inducido químicamente , Animales , Astrocitos/metabolismo , Muerte Celular , Clusterina , Complemento C1q/genética , Complemento C4/genética , Ácido Glutámico/farmacología , Glicoproteínas/genética , Glicoproteínas/fisiología , Hipocampo/metabolismo , Hipocampo/patología , Hibridación in Situ , Ácido Kaínico/farmacología , Masculino , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/patología
14.
Neuroscience ; 89(3): 687-99, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10199605

RESUMEN

Food restriction of adult rodents increases lifespan, with commensurate attenuation of age-related pathological lesions in many organs, as well as attenuation of normal ageing changes that are distinct from gross lesions. Previous work showed that chronic food restriction attenuated age-associated astrocyte and microglial hyperactivity in the hippocampal hilus, as measured by expression of glial fibrillary acidic protein and major histocompatibility complex II antigen (OX6). Here, we examined other markers of astrocyte and microglial activation in gray and white matter regions of ad libitum-fed (Brown Norway x Fischer 344) F1 male rats aged three and 24 months and chronic food-restricted rats aged 24 months. In situ hybridization and immunohistochemical techniques evaluated glial expression of glial fibrillary acidic protein, apolipoprotein E, apolipoprotein J (clusterin), heme oxygenase-1, complement 3 receptor (OX42), OX6 and transforming growth factor-beta1. All markers were elevated in the corpus callosum during ageing and were attenuated by food restriction, but other regions showed marked dissociation of the extent and direction of changes. Astrocytic activation, as measured with glial fibrillary acidic protein expression (coding and intron-containing RNA, immunoreactivity), increased with age in the corpus callosum, basal ganglia and hippocampus. Generally, food restriction attenuated the age-related increase in glial fibrillary acidic protein messenger RNA and immunoreactivity. Food restriction also reduced the age-related increase in apolipoprotein J and E messenger RNA and heme oxygenase-1 immunoreactivity in the basal ganglia and corpus callosum. However, astrocytes in the hilus of the hippocampus showed an age-related decrease in apolipoprotein J and E messenger RNA, which was further intensified by food restriction. The age-associated microglial activation measured by OX6 and OX42 immunoreactivity was reduced by food restriction in most subregions. The localized subsets of glial age changes and effects of food restriction comprise a mosaic of ageing consistent with the regional heterogeneity of ageing changes reported by others. In particular, age has a differential effect on astrocytic and microglial hyperactivity in gray versus white matter areas. The evident mosaic of glial ageing and responses to food restriction suggests that multiple mechanisms are at work during ageing.


Asunto(s)
Envejecimiento/patología , Privación de Alimentos , Gliosis/prevención & control , Animales , Astrocitos/patología , Secciones por Congelación , Proteína Ácida Fibrilar de la Glía/genética , Gliosis/etiología , Hipocampo/patología , Hibridación in Situ , Masculino , Microglía/patología , Estrés Oxidativo , ARN Mensajero/análisis , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344 , Transcripción Genética
15.
Am J Cardiol ; 67(16): 1354-9, 1991 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-1904189

RESUMEN

To assess the long-term effect of thrombolytic therapy on left ventricular (LV) systolic function, 222 patients with acute myocardial infarction treated with intravenous tissue plasminogen activator within 4 hours of symptom onset underwent assessment of LV ejection fraction (EF) by radionuclide equilibrium angiography at hospital discharge and 1 year later. Mean EF at hospital discharge (46 +/- 12) was similar to that at 1 year (45 +/- 13). Stepwise multivariate linear regression analysis identified EF at discharge and patency of the infarct-related artery before discharge as independent predictors of EF change at 1 year (p = 0.0002 and 0.003, respectively). Random assignments to invasive versus conservative treatment strategies or to early versus delayed beta-blocker therapy did not affect EF change during follow-up. No significant deterioration of EF was observed in patients with larger infarcts. However, EF decreased from 45 +/- 10 at hospital discharge to 39 +/- 12 (p = 0.005) at 1-year follow-up in a subgroup of patients with history of prior infarction. Thus, patients with acute myocardial infarction, treated with intravenous tissue plasminogen activator early after onset of symptoms, appear to have stable LV function between hospital discharge and 1 year follow-up. The change in EF between hospital discharge and 1 year can be predicted from the EF value at discharge, patency of the infarct-related artery before discharge and history of previous myocardial infarction.


Asunto(s)
Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/fisiopatología , Volumen Sistólico/efectos de los fármacos , Terapia Trombolítica , Activador de Tejido Plasminógeno/uso terapéutico , Función Ventricular Izquierda/fisiología , Anciano , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/diagnóstico por imagen , Angiografía por Radionúclidos , Recurrencia
16.
J Endocrinol ; 149(1): 19-27, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8676051

RESUMEN

Transforming growth factor beta 1 (TGF beta 1) has recently been shown to be produced in the prolactin (PRL)-secreting lactotropes of the pituitary gland. TGF beta 1 inhibits lactotropic secretion and proliferation, and the production of TGF beta 1 in lactotropes is reduced during lactotropic growth following estrogen treatment in ovariectomized rats. In many estrogen-responsive tissues, TGF beta 1 has been shown to exert its effect by binding to TGF beta 1 type II receptors (T beta R II) at the cell surface. In this study, we sought to ascertain whether T beta R II is involved in TGF beta 1 action on lactotropes by determining the changes of T beta R II mRNA and protein levels and specific 125I-TGF beta 1 binding sites on the lactotropes during estrogen-induced proliferation of lactotropes in Fischer 344 rats. Double immunohistochemical procedures were employed to identify immunoreactive T beta R II in PRL-reactive cells. The majority of T beta R II-reactive cells in the anterior pituitary were observed to be lactotropes. Dual immunohistochemistry and in situ hybridization procedures also indicated that lactotropes were the major cell types containing T beta R II mRNA hybrids. Both the levels of immunoreactive T beta R II protein and in situ T beta R II mRNA hybrids in the pituitary were significantly decreased in ovariectomized rats after 15 days of estrogen treatment. Determination of 125I-TGF beta 1 binding sites in lactotropes by double immunohistochemistry and receptor autoradiography also revealed specific binding sites of 125I-TGF beta 1 in lactotropes in the anterior pituitary. 125I-TGF beta 1 binding in the anterior pituitary was also reduced following estrogen treatment in ovariectomized rats. These data suggest that down-regulation of T beta R II may be an important mechanism of estrogen action on lactotropic cell growth and PRL secretion, and further support the notion that TGF beta 1 controls lactotropic function by autocrine/paracrine mechanisms.


Asunto(s)
Adenohipófisis/metabolismo , Prolactina/metabolismo , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Sitios de Unión , Estradiol/farmacología , Femenino , Inmunohistoquímica , Hibridación in Situ , Ovariectomía , Adenohipófisis/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Ratas , Ratas Endogámicas F344 , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
17.
18.
Neuroreport ; 8(5): 1077-81, 1997 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-9175088

RESUMEN

The expression and localization of two distinct mRNAs from the macrophage scavenger receptor gene family were studied in rat brain cells in vivo and in vitro. In general, brains of control male rats showed low level signals by in situ hybridization for the macrophage scavenger receptor (MSR) and murine adherent macrophage (MAMA) receptor. In contrast, the reticular thalamic nucleus had a subpopulation of intensely labeled cells. Kainic acid (KA) treatment induced MSR and MAMA mRNA levels on different schedules in brain regions that are susceptible to KA, including hippocampal areas CA1 and CA3. The combination of immunocytochemistry and in situ hybridization localized the MSR and MAMA mRNA to microglia of KA-treated rats. Northern blot hybridization detected both MSR and MAMA mRNAs in primary cultures of mixed glia that contained microglia. Both MSR and MAMA mRNA were induced by treatment of primary mixed glia with lipopolysaccharide and interferon-gamma, but not TGF beta 1. MSR, but not MAMA, mRNA levels were increased after treatment with interleukin-1 alpha. These results demonstrate the differential regulation of scavenger receptor mRNAs in microglia that is consistent with distinct roles for scavenger receptors in responses to neurodegeneration.


Asunto(s)
Encéfalo/efectos de los fármacos , Moléculas de Adhesión Celular/genética , Citocinas/farmacología , Microglía/efectos de los fármacos , ARN Mensajero/biosíntesis , Receptores Inmunológicos/genética , Animales , Encéfalo/metabolismo , Células Cultivadas , Hibridación in Situ , Ácido Kaínico , Masculino , Ratones , Microglía/metabolismo , Familia de Multigenes , Neurotoxinas , Ratas , Ratas Endogámicas F344 , Receptores Depuradores
19.
Science ; 201(4355): 482, 1978 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-17790421
20.
Urology ; 5(5): 658-61, 1975 May.
Artículo en Inglés | MEDLINE | ID: mdl-1168963

RESUMEN

A case of choriocarcinoma presenting with metastases to both kidneys is reported. The epidemiologic and pathologic aspects of gestational choriocarcinoma are briefly discussed. Prior reports of renal metastases with choriocarcinoma are reviewed, and the fairly typical clinical presentation for this entity is emphasized.


Asunto(s)
Coriocarcinoma/diagnóstico , Neoplasias Renales/diagnóstico , Adulto , Coriocarcinoma/patología , Diagnóstico Diferencial , Enfermedad de Hodgkin/diagnóstico por imagen , Humanos , Neoplasias Renales/diagnóstico por imagen , Neoplasias Renales/patología , Masculino , Metástasis de la Neoplasia , Urografía
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda