Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Acta Neuropathol Commun ; 7(1): 82, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31113487

ABSTRACT

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.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Brain/pathology , Microglia/pathology , Plaque, Amyloid/pathology , Animals , Female , Genotype , Male , Membrane Glycoproteins/metabolism , Mice, Transgenic , Receptors, Immunologic/metabolism , Sex Characteristics
2.
Transl Psychiatry ; 8(1): 261, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30498214

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Behavior, Animal , Hippocampus/physiopathology , Neurogenesis , Prenatal Exposure Delayed Effects/chemically induced , Vehicle Emissions/toxicity , Animals , Astrocytes/physiology , Blood-Brain Barrier/metabolism , Depression/chemically induced , Feeding Behavior , Female , Male , Memory , Microglia/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Rats, Sprague-Dawley
3.
Transl Psychiatry ; 7(1): e1022, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28140404

ABSTRACT

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.


Subject(s)
Cognitive Dysfunction/epidemiology , Dementia/epidemiology , Environmental Exposure/statistics & numerical data , Gene-Environment Interaction , Particulate Matter , Aged , Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/metabolism , Animals , Apolipoprotein E4/genetics , Atrophy , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Cell Line, Tumor , Cerebrum/drug effects , Cerebrum/metabolism , Cognitive Dysfunction/genetics , Dementia/genetics , Female , Humans , In Vitro Techniques , Mice , Mice, Transgenic , Neurites/drug effects , Neurites/pathology , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism
4.
Article in English | MEDLINE | ID: mdl-28042517

ABSTRACT

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.

5.
Front Neurosci ; 7: 157, 2013.
Article in English | MEDLINE | ID: mdl-24027494

ABSTRACT

Pgrmc1 (progesterone receptor membrane component 1) is a multifunctional 22 kDa protein with heme-binding and P450-activating capacity which was recognized under different names for roles in cell motility during neural development and in cancer, and apoptosis. Pgrmc1 expression in microglia was recently shown by the present authors to mediate estrogen-progesterone interactions during axonal sprouting and to mediate microglial activation itself. We also discuss other functions of Pgramc1 in the nervous system and its possible relationship to the 18 kDa sigma-2 receptor (S2R).

6.
Endocrinology ; 154(7): 2468-80, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23653459

ABSTRACT

Neuronal plasticity is regulated by the ovarian steroids estradiol (E2) and progesterone (P4) in many normal brain functions, as well as in acute response to injury and chronic neurodegenerative disease. In a female rat model of axotomy, the E2-dependent compensatory neuronal sprouting is antagonized by P4. To resolve complex glial-neuronal cell interactions, we used the "wounding-in-a-dish" model of neurons cocultured with astrocytes or mixed glia (microglia to astrocytes, 1:3). Although both astrocytes and mixed glia supported E2-enhanced neurite outgrowth, P4 antagonized E2-induced neurite outgrowth only with mixed glia, but not astrocytes alone. We now show that P4-E2 antagonism of neurite outgrowth is mediated by microglial expression of progesterone receptor (Pgr) membrane component 1 (Pgrmc1)/S2R, a putative nonclassical Pgr mediator with multiple functions. The P4-E2 antagonism of neurite outgrowth was restored by add-back of microglia to astrocyte-neuron cocultures. Because microglia do not express the classical Pgr, we examined the role of Pgrmc1, which is expressed in microglia in vitro and in vivo. Knockdown by siRNA-Pgrmc1 in microglia before add-back to astrocyte-neuron cocultures suppressed the P4-E2 antagonism of neurite outgrowth. Conditioned media from microglia restored the P4-E2 activity, but only if microglia were activated by lipopolysaccharide or by wounding. Moreover, the microglial activation was blocked by Pgmrc1-siRNA knockdown. These findings explain why nonwounded cultures without microglial activation lack P4 antagonism of E2-induced neurite outgrowth. We suggest that microglial activation may influence brain responses to exogenous P4, which is a prospective therapy in traumatic brain injury.


Subject(s)
Estradiol/pharmacology , Microglia/drug effects , Microglia/metabolism , Neurites/drug effects , Neurites/metabolism , Progesterone/pharmacology , Receptors, Progesterone/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cells, Cultured , Immunohistochemistry , Lipopolysaccharides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Interdiscip Top Gerontol ; 35: 83-97, 2007.
Article in English | MEDLINE | ID: mdl-17063034

ABSTRACT

Dietary restriction (DR) remains the most powerful and general environmental manipulation of aging processes in laboratory animals with strong beneficial effects on most age-related degenerative changes throughout the body. Underlying the beneficial effects of DR is the attenuation of system-wide inflammatory processes including those occurring within the central nervous system. During normal aging a progressive neuroinflammatory state builds in the brain involving astrocytes and microglia, the primary cellular components of neuroinflammation. DR attenuates the age-related activation of astrocytes and microglia with concomitant beneficial effects on neurodegeneration and cognition. Increasing evidence suggests that common pathways are emerging that link many normal aging inflammatory processes with age-related diseases such as Alzheimer, cancer, diabetes and cardiovascular disease.


Subject(s)
Aging/metabolism , Anti-Inflammatory Agents , Caloric Restriction , Inflammation , Longevity/physiology , Aging/physiology , Animals , Glucocorticoids , Glucose , Humans , Models, Animal , Peroxisome Proliferator-Activated Receptors
8.
J Neurochem ; 93(4): 1038-46, 2005 May.
Article in English | MEDLINE | ID: mdl-15857407

ABSTRACT

Apolipoprotein J (apoJ; also known as clusterin and sulfated glycoprotein (SGP)-2) is associated with senile plaques in degenerating regions of Alzheimer's disease brains, where activated microglia are also prominent. We show a functional link between apoJ and activated microglia by demonstrating that exogenous apoJ activates rodent microglia in vivo and in vitro. Intracerebroventricular infusion of purified human plasma apoJ ( approximately 4 microg over 28 days) activated parenchymal microglia to a phenotype characterized by enlarged cell bodies and processes (phosphotyrosine immunostaining). In vitro, primary rat microglia were also activated by apoJ, with changes in morphology and induction of major histocompatibility complex class II (MHCII) antigen. ApoJ increased the secretion of reactive nitrogen intermediates in a dose-dependent manner (EC(50) 112 nm), which was completely blocked by aminoguanidine (AG), a nitric oxide synthase inhibitor. However, AG did not block the increased secretion of tumor necrosis factor-alpha by apoJ (EC(50) 55 nm). Microglial activation by apoJ was also blocked by an anti-apoJ monoclonal antibody (G7), and by chemical cleavage of apoJ with 2-nitro-5-thiocyanobenzoate. The mitogen-activated protein kinase kinase and protein kinase C inhibitors PD98059 and H7 inhibited apoJ-mediated induction of reactive nitrogen intermediate secretion from cultured microglia. As a functional measure, apoJ-activated microglia secreted neurotoxic agents in a microglia-neuron co-culture model. We hypothesize that ApoJ contributes to chronic inflammation and neurotoxicity through direct effects on microglia.


Subject(s)
Cerebral Cortex/cytology , Complement Inactivator Proteins/pharmacology , Glycoproteins/pharmacology , Microglia/drug effects , Molecular Chaperones/pharmacology , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Clusterin , Complement Hemolytic Activity Assay/methods , Complement Inactivator Proteins/isolation & purification , Diagnostic Imaging/methods , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Flavonoids/pharmacology , Glycoproteins/isolation & purification , Humans , Immunohistochemistry/methods , In Vitro Techniques , Interferons/pharmacology , Microglia/metabolism , Molecular Chaperones/isolation & purification , Neurons/drug effects , Neurons/metabolism , Nitrites/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Rats , Rats, Inbred F344 , Thiocyanates/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
9.
J Neurochem ; 79(3): 595-605, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11701763

ABSTRACT

In recent studies of transgenic models of Alzheimer's disease (AD), it has been reported that antibodies to aged beta amyloid peptide 1-42 (Abeta(1-42)) solutions (mixtures of Abeta monomers, oligomers and amyloid fibrils) cause conspicuous reduction of amyloid plaques and neurological improvement. In some cases, however, neurological improvement has been independent of obvious plaque reduction, and it has been suggested that immunization might neutralize soluble, non-fibrillar forms of Abeta. It is now known that Abeta toxicity resides not only in fibrils, but also in soluble protofibrils and oligomers. The current study has investigated the immune response to low doses of Abeta(1-42) oligomers and the characteristics of the antibodies they induce. Rabbits that were injected with Abeta(1-42) solutions containing only monomers and oligomers produced antibodies that preferentially bound to assembled forms of Abeta in immunoblots and in physiological solutions. The antibodies have proven useful for assays that can detect inhibitors of oligomer formation, for immunofluorescence localization of cell-attached oligomers to receptor-like puncta, and for immunoblots that show the presence of SDS-stable oligomers in Alzheimer's brain tissue. The antibodies, moreover, were found to neutralize the toxicity of soluble oligomers in cell culture. Results support the hypothesis that immunizations of transgenic mice derive therapeutic benefit from the immuno-neutralization of soluble Abeta-derived toxins. Analogous immuno-neutralization of oligomers in humans may be a key in AD vaccines.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/immunology , Antibody Specificity , Peptide Fragments/immunology , Vaccination , Amyloid beta-Peptides/toxicity , Animals , Epitopes , Fluorescent Antibody Technique , Hippocampus/cytology , Humans , Neurons/cytology , Neurons/immunology , Neuroprotective Agents/immunology , PC12 Cells , Peptide Fragments/toxicity , Rats , Solubility
10.
J Biol Chem ; 276(42): 38787-94, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11483593

ABSTRACT

The DSCR1 (Adapt78) gene was independently discovered as a resident of the "Down syndrome candidate region"and as an "adaptive response"shock or stress gene that is transiently induced during oxidative stress. Recently the DSCR1 (Adapt78) gene product was discovered to be an inhibitor of the serine/threonine phosphatase, calcineurin, and its signaling pathways. We hypothesized that DSCR1 (Adapt78) might also be involved in the development of Alzheimer's disease. To address this question we first studied DSCR1 (Adapt78) in multiple human tissues and found significant expression in brain, spinal cord, kidney, liver, mammary gland, skeletal muscle, and heart. Within the brain DSCR1 (Adapt78) is predominantly expressed in neurons within the cerebral cortex, hippocampus, substantia nigra, thalamus, and medulla oblongata. When we compared DSCR1 (Adapt78) mRNA expression in post-mortem brain samples from Alzheimer's disease patients and individuals who had died with no Alzheimer's diagnosis, we found that DSCR1 (Adapt78) mRNA levels were about twice as high in age-matched Alzheimer's patients as in controls. DSCR1 (Adapt78) mRNA levels were actually three times higher in patients with extensive neurofibrillary tangles (a hallmark of Alzheimer's disease) than in controls. In comparison, post-mortem brain samples from Down syndrome patients (who suffer Alzheimer's symptoms) also exhibited DSCR1 (Adapt78) mRNA levels two to three times higher than controls. Using a cell culture model we discovered that the amyloid beta(1-42) peptide, which is a major component of senile plaques in Alzheimer's, can directly induce increased expression of DSCR1 (Adapt78). Our findings associate DSCR1 (Adapt78) with such major hallmarks of Alzheimer's disease as amyloid protein, senile plaques, and neurofibrillary tangles.


Subject(s)
Alzheimer Disease/metabolism , Muscle Proteins/biosynthesis , RNA, Messenger/biosynthesis , Aged , Aged, 80 and over , Amyloid beta-Peptides/biosynthesis , Animals , Blotting, Northern , Brain/metabolism , Breast/metabolism , Case-Control Studies , Cells, Cultured , DNA-Binding Proteins , Down Syndrome/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Kidney/metabolism , Liver/metabolism , Male , Middle Aged , Muscle, Skeletal/metabolism , Myocardium/metabolism , Neurons/cytology , Peptide Fragments/biosynthesis , Peptides/chemistry , Protein Binding , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spinal Cord/metabolism , Tissue Distribution
11.
J Clin Exp Neuropsychol ; 23(2): 240-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309677

ABSTRACT

A comprehensive neuropsychological battery was administered to 48 veterans with Gulf War Illness (GWI) characterized by severe fatigue (GV-F) and 39 healthy veterans (GV-H). Subjects were matched on intelligence and did not differ on age, gender, race, and alcohol consumption. Compared to GVs-H, GVs-F were significantly impaired on four tasks: three attention, concentration, information processing tasks and one measure of abstraction and conceptualization. After considering the presence of post-war Axis I psychopathology, GWI remained a significant predictor of cognitive performance on one of the attention, concentration, and information processing tasks and one abstraction and conceptualization measure. Performance on the remaining two attention, concentration, and information processing tasks was only significantly predicted by Axis I psychopathology with post-war onset. The results suggest that Gulf War Illness is associated with some aspects of cognitive dysfunction in Gulf Veterans, over and above the contribution of psychopathology.


Subject(s)
Attention , Cognition Disorders/diagnosis , Cognition , Fatigue Syndrome, Chronic/psychology , Persian Gulf Syndrome/psychology , Adult , Case-Control Studies , Cognition Disorders/etiology , Fatigue Syndrome, Chronic/etiology , Female , Humans , Male , Models, Psychological , Neuropsychological Tests , Persian Gulf Syndrome/complications , Persian Gulf Syndrome/diagnosis
12.
Neuroscience ; 101(2): 313-21, 2000.
Article in English | MEDLINE | ID: mdl-11074155

ABSTRACT

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.


Subject(s)
Brain/drug effects , Complement C1q/drug effects , Microglia/drug effects , Receptors, Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Complement C1q/genetics , Complement C1q/metabolism , Denervation/adverse effects , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Encephalitis/physiopathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Homeostasis/drug effects , Homeostasis/physiology , Male , Microglia/cytology , Microglia/metabolism , Perforant Pathway/pathology , Perforant Pathway/physiopathology , Perforant Pathway/surgery , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
13.
Exp Neurol ; 165(1): 46-57, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964484

ABSTRACT

Age and estrogen treatment influenced fiber outgrowth and compensatory neuronal sprouting after unilateral entorhinal cortex lesions (ECL) which model Alzheimer disease-like deafferentation in the dentate gyrus of the hippocampus. In young F344 rats (3 months old), ovariectomy (OVX) decreased reactive fiber outgrowth by 60%. Sprouting in middle-aged rats (18 months old) was reduced in intact females; no further reduction was caused by OVX. Several astrocyte mRNAs were measured in the dentate gyrus of young and middle-aged female rats in three different estrogen states (sham OVX, OVX, or OVX + estradiol) 1 week after ECL. Glial fibrillary acidic protein (GFAP) mRNA was twofold greater in middle-aged rats than young, although both ages showed threefold increases in response to ECL. In prior studies GFAP was found to be decreased by estradiol treatment 3-4 days after ECL; in this study GFAP mRNA had returned to sham OVX levels in young rats by 7 days post-ECL. Surprisingly, estradiol treatment increased GFAP mRNA levels by 25% above OVX in middle-aged rats. Apolipoprotein E (apoE) mRNA was decreased 20% by age in the dentate, although both age groups showed a 25% increase in apoE mRNA in response to ECL. Apolipoprotein J (apoJ) mRNA was increased 20% in the dentate gyrus of middle-aged rats, and both age groups responded to ECL with a 65% increase in apoJ mRNA. The estrogen state did not alter levels of either apolipoprotein mRNA in the deafferented dentate. The data suggest that the estrogen-induced decrease of GFAP in response to lesions does not persist at 7 days post-ECL during sprouting. Overall effects of age on the dentate gyrus include elevated GFAP mRNA and decreased apoE mRNA. The cortical wound site showed consistent enhancement of GFAP mRNA in both age groups by estradiol above sham OVX and greater responses in middle-aged rats.


Subject(s)
Aging/physiology , Estradiol/pharmacology , Gene Expression/physiology , Molecular Chaperones , Nerve Regeneration/physiology , Synapses/drug effects , Synapses/physiology , Afferent Pathways/physiology , Animals , Apolipoproteins E/genetics , Clusterin , Dentate Gyrus/metabolism , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Female , Glial Fibrillary Acidic Protein/genetics , Glycoproteins/genetics , Neuroglia/drug effects , Neuroglia/physiology , Ovariectomy , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Time Factors
14.
Exp Neurol ; 163(1): 220-30, 2000 May.
Article in English | MEDLINE | ID: mdl-10785461

ABSTRACT

Transforming growth factor-beta1 (TGFbeta1) is a cytokine/growth factor found within the pathological central nervous system. TGFbeta1 has been shown to inhibit the release of cytotoxic molecules from microglia and macrophages, decrease astrocyte proliferation, and promote neuron survival. Because of the relevance of these actions to spinal cord injury, we examined TGFbeta1 and its receptors betaRI and betaRII mRNA levels and localization within the contused rat spinal cord using in situ hybridization. At the lesion site, TGFbeta1 mRNA peaked at 7 days postinjury and declined thereafter. Temporal and spatial localization of the betaRI and betaRII receptor mRNA closely mimicked that for TGFbeta1 in the epicenter. TGFbeta1, betaRI, and betaRII mRNAs also were elevated rostral and caudal to the injury, especially in regions known to contain activated microglia and degenerating axon profiles. Immunohistochemical staining of nearby sections confirmed that the highest levels of TGFbeta1 and receptor mRNA corresponded to regions filled with activated microglia and macrophages. The similar expression pattern of TGFbeta1, betaRI, and betaRII mRNA within the injured spinal cord suggests a local site of action. Since TGFbeta1 can act as an immunosuppressant as well as a stimulant for growth factors and neurite sprouting, it likely plays an important role, both temporally and spatially, in orchestrating postinjury events within the spinal cord.


Subject(s)
Contusions/metabolism , RNA, Messenger/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Transforming Growth Factor beta/metabolism , Animals , Axons/metabolism , Axons/pathology , Female , Immunohistochemistry , Macrophages/metabolism , Macrophages/pathology , Microglia/metabolism , Microglia/pathology , Rats , Rats, Inbred Lew , Receptors, Transforming Growth Factor beta/genetics , Spinal Cord/pathology , Transforming Growth Factor beta/genetics , Up-Regulation/genetics
15.
Neurosci Lett ; 271(1): 65-7, 1999 Aug 13.
Article in English | MEDLINE | ID: mdl-10471215

ABSTRACT

We analyzed postmortem tissues of sporadic amyotrophic lateral sclerosis (SALS) for mRNA levels of two inflammatory proteins, complement C1qB and clusterin (apoJ). By Northern blot hybridization, SALS was associated with increased mRNA for C1qB and clusterin in the motor cortex (Brodmann area A4), but not in superior temporal cortex (A17), relative to neurologically normal controls. By in situ hybridization, SALS spinal cords showed increased C1qB and clusterin mRNA in areas undergoing neurodegeneration. This evidence implicates inflammatory mechanisms during neurodegenerative processes in SALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cerebral Cortex/metabolism , Complement C1q/genetics , Complement Inactivator Proteins/genetics , Gene Expression Regulation , Glycoproteins/genetics , Molecular Chaperones , Nerve Degeneration/genetics , Spinal Cord/metabolism , Transcription, Genetic , Adolescent , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/pathology , Cerebral Cortex/pathology , Clusterin , Humans , Nerve Degeneration/etiology , Nerve Degeneration/pathology , RNA, Messenger/genetics , Reference Values , Somatosensory Cortex/metabolism , Spinal Cord/pathology , Temporal Lobe/metabolism
16.
Neuroscience ; 89(3): 687-99, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10199605

ABSTRACT

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.


Subject(s)
Aging/pathology , Food Deprivation , Gliosis/prevention & control , Animals , Astrocytes/pathology , Frozen Sections , Glial Fibrillary Acidic Protein/genetics , Gliosis/etiology , Hippocampus/pathology , In Situ Hybridization , Male , Microglia/pathology , Oxidative Stress , RNA, Messenger/analysis , Rats , Rats, Inbred BN , Rats, Inbred F344 , Transcription, Genetic
17.
J Neurosci ; 18(9): 3180-5, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9547226

ABSTRACT

Estrogen replacement therapy appears to delay the onset of Alzheimer's disease (AD), but the mechanisms for this action are incompletely known. We show how the enhancement of synaptic sprouting by estradiol (E2) in response to an entorhinal cortex (EC) lesion model of AD may operate via an apolipoprotein E (apoE)-dependent mechanism. In wild-type (WT) mice, ovariectomy decreased commissural/associational sprouting to the inner molecular layer of the dentate gyrus, with synaptophysin (SYN) as a marker. E2 replacement returned SYN in the inner layer to levels of EC-lesioned, ovary-bearing controls and increased the area of compensatory synaptogenesis in the outer molecular layer. In EC-lesioned apoE-knock-out (KO) mice, however, E2 did not enhance sprouting. We also examined apoJ (clusterin) mRNA, which is implicated in AD by its presence in senile plaques, its transport of Abeta across the blood-brain barrier, and its induction by neurodegenerative lesioning. ApoJ mRNA levels were increased by E2 replacement in EC-lesioned WT mice but not in apoE-KO mice. These data suggest a mechanism for the protective effects of estrogens on AD and provide a link between two important risk factors in the etiology of AD, the apoE epsilon4 genotype and an estrogen-deficient state. This is also the first evidence that SYN, a presynaptic protein involved in neurotransmitter release, is regulated by E2 in the adult brain, and that apoE is necessary for the induction of apoJ mRNA by E2 in brain injury.


Subject(s)
Alzheimer Disease/prevention & control , Apolipoproteins E/physiology , Estrogen Replacement Therapy , Molecular Chaperones , Synapses/drug effects , Alzheimer Disease/physiopathology , Animals , Clusterin , Female , Glycoproteins/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , Synaptophysin/analysis
18.
Proc Natl Acad Sci U S A ; 95(11): 6448-53, 1998 May 26.
Article in English | MEDLINE | ID: mdl-9600986

ABSTRACT

Abeta1-42 is a self-associating peptide whose neurotoxic derivatives are thought to play a role in Alzheimer's pathogenesis. Neurotoxicity of amyloid beta protein (Abeta) has been attributed to its fibrillar forms, but experiments presented here characterize neurotoxins that assemble when fibril formation is inhibited. These neurotoxins comprise small diffusible Abeta oligomers (referred to as ADDLs, for Abeta-derived diffusible ligands), which were found to kill mature neurons in organotypic central nervous system cultures at nanomolar concentrations. At cell surfaces, ADDLs bound to trypsin-sensitive sites and surface-derived tryptic peptides blocked binding and afforded neuroprotection. Germ-line knockout of Fyn, a protein tyrosine kinase linked to apoptosis and elevated in Alzheimer's disease, also was neuroprotective. Remarkably, neurological dysfunction evoked by ADDLs occurred well in advance of cellular degeneration. Without lag, and despite retention of evoked action potentials, ADDLs inhibited hippocampal long-term potentiation, indicating an immediate impact on signal transduction. We hypothesize that impaired synaptic plasticity and associated memory dysfunction during early stage Alzheimer's disease and severe cellular degeneration and dementia during end stage could be caused by the biphasic impact of Abeta-derived diffusible ligands acting upon particular neural signal transduction pathways.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/drug effects , Brain/metabolism , Neurotoxins/toxicity , Alzheimer Disease , Amyloid beta-Peptides/metabolism , Animals , Cell Death/drug effects , Cell Membrane/metabolism , Cells, Cultured , Ligands , Mice
19.
Neurobiol Aging ; 19(1): 97-103, 1998.
Article in English | MEDLINE | ID: mdl-9562510

ABSTRACT

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.


Subject(s)
Aging/physiology , Astrocytes/physiology , Down-Regulation/physiology , Microglia/physiology , Animals , Cell Division/physiology , Cells, Cultured , Glial Fibrillary Acidic Protein/biosynthesis , Immunohistochemistry , Male , Nitrogen/metabolism , Phenotype , Rats , Rats, Inbred F344 , Thymidine/metabolism , Transforming Growth Factor beta/metabolism
20.
J Neurochem ; 68(5): 2046-52, 1997 May.
Article in English | MEDLINE | ID: mdl-9109531

ABSTRACT

The first component of the classic complement cascade, C1q, was increased in whole rat brain after lesioning by intraperitoneally injected kainic acid (KA) (20-fold, 3 days after KA) and in the striatum ipsilateral to unilateral decortication (fivefold, 10 days after decortication). C1q was measured after purification by chromatography and electrophoresis. De novo biosynthesis of C1q 3 days after KA was increased >10-fold, as measured by the incorporation of [35S]methionine into C1q after incubation of brain slices from KA-treated rats for 2 h. In parallel with these responses, KA induced fivefold increase of C1q bioactivity, as evaluated with C1q-dependent hemolysis. The contribution of C1q from entrapped cerebrovascular blood was evaluated by the effects of perfusion and was minor relative to the increases of C1q in response to KA lesioning. These findings support the hypothesis that the C1q protein detected by immunocytochemistry in senile plaques of Alzheimer brains and in the hippocampus after deafferenting lesions is synthesized by resident brain cells.


Subject(s)
Brain/drug effects , Brain/metabolism , Complement C1q/metabolism , Corpus Striatum/physiology , Kainic Acid/pharmacology , Animals , Blotting, Western , Cerebrovascular Circulation , Chromatography , Complement Hemolytic Activity Assay , Decerebrate State , Denervation , Humans , Injections, Intraperitoneal , Male , Methionine/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...