Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38328219

ABSTRACT

The strongest risk factors for Alzheimer's disease (AD) include the χ4 allele of apolipoprotein E (APOE), the R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2), and female sex. Here, we combine APOE4 and TREM2R47H ( R47H ) in female P301S tauopathy mice to identify the pathways activated when AD risk is the strongest, thereby highlighting disease-causing mechanisms. We find that the R47H variant induces neurodegeneration in female APOE4 mice without impacting hippocampal tau load. The combination of APOE4 and R47H amplified tauopathy-induced cell-autonomous microglial cGAS-STING signaling and type-I interferon response, and interferon signaling converged across glial cell types in the hippocampus. APOE4-R47H microglia displayed cGAS- and BAX-dependent upregulation of senescence, showing association between neurotoxic signatures and implicating mitochondrial permeabilization in pathogenesis. By uncovering pathways enhanced by the strongest AD risk factors, our study points to cGAS-STING signaling and associated microglial senescence as potential drivers of AD risk.

2.
Front Aging Neurosci ; 15: 1280218, 2023.
Article in English | MEDLINE | ID: mdl-38035277

ABSTRACT

Dementia is often characterized by age-dependent cerebrovascular pathology, neuroinflammation, and cognitive deficits with notable sex differences in risk, disease onset, progression and severity. Women bear a disproportionate burden of dementia, and the onset of menopause (i.e., perimenopause) may be a critical period conferring increased susceptibility. However, the contribution of early ovarian decline to the neuroinflammatory processes associated with cerebrovascular dementia risks, particularly at the initial stages of pathology that may be more amenable to proactive intervention, is unknown. To better understand the influence of early ovarian failure on dementia-associated neuroinflammation we developed a model of perimenopausal cerebral amyloid angiopathy (CAA), an important contributor to dementia. For this, accelerated ovarian failure (AOF) was induced by 4-vinylcyclohexene diepoxide (VCD) treatment to isolate early-stage ovarian failure comparable to human perimenopause (termed "peri-AOF") in transgenic SWDI mice expressing human vasculotropic mutant amyloid beta (Aß) precursor protein, that were also tested at an early stage of amyloidosis. We found that peri-AOF SWDI mice showed increased astrocyte activation accompanied by elevated Aß in select regions of the hippocampus, a brain system involved in learning and memory that is severely impacted during dementia. However, although SWDI mice showed signs of increased hippocampal microglial activation and impaired cognitive function, this was not further affected by peri-AOF. In sum, these results suggest that elevated dysfunction of key elements of the neurovascular unit in select hippocampal regions characterizes the brain pathology of mice at early stages of both CAA and AOF. However, neurovascular unit pathology may not yet have passed a threshold that leads to further behavioral compromise at these early periods of cerebral amyloidosis and ovarian failure. These results are consistent with the hypothesis that the hormonal dysregulation associated with perimenopause onset represents a stage of emerging vulnerability to dementia-associated neuropathology, thus providing a selective window of opportunity for therapeutic intervention prior to the development of advanced pathology that has proven difficult to repair or reverse.

3.
bioRxiv ; 2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37961594

ABSTRACT

Pathogenic tau accumulation fuels neurodegeneration in Alzheimer's disease (AD). Enhancing aging brain's resilience to tau pathology would lead to novel therapeutic strategies. DAP12 (DNAX-activation protein 12) is critically involved in microglial immune responses. Previous studies have showed that mice lacking DAP12 in tauopathy mice exhibit higher tau pathology but are protected from tau-induced cognitive deficits. However, the exact mechanism remains elusive. Our current study uncovers a novel resilience mechanism via microglial interaction with oligodendrocytes. Despite higher tau inclusions, Dap12 deletion curbs tau-induced brain inflammation and ameliorates myelin and synapse loss. Specifically, removal of Dap12 abolished tau-induced disease-associated clusters in microglia (MG) and intermediate oligodendrocytes (iOli), which are spatially correlated with tau pathology in AD brains. Our study highlights the critical role of interactions between microglia and oligodendrocytes in tau toxicity and DAP12 signaling as a promising target for enhancing resilience in AD.

4.
Res Sq ; 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37961627

ABSTRACT

Pathogenic tau accumulation fuels neurodegeneration in Alzheimer's disease (AD). Enhancing aging brain's resilience to tau pathology would lead to novel therapeutic strategies. DAP12 (DNAX-activation protein 12) is critically involved in microglial immune responses. Previous studies have showed that mice lacking DAP12 in tauopathy mice exhibit higher tau pathology but are protected from tau-induced cognitive deficits. However, the exact mechanism remains elusive. Our current study uncovers a novel resilience mechanism via microglial interaction with oligodendrocytes. Despite higher tau inclusions, Dap12 deletion curbs tau-induced brain inflammation and ameliorates myelin and synapse loss. Specifically, removal of Dap12 abolished tau-induced disease-associated clusters in microglia (MG) and intermediate oligodendrocytes (iOli), which are spatially correlated with tau pathology in AD brains. Our study highlights the critical role of interactions between microglia and oligodendrocytes in tau toxicity and DAP12 signaling as a promising target for enhancing resilience in AD.

5.
bioRxiv ; 2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37781600

ABSTRACT

Demyelination occurs in aging and associated diseases, including Alzheimer's disease. Several of these diseases exhibit sex differences in prevalence and severity. Biological sex primarily stems from sex chromosomes and gonads releasing sex hormones. To dissect mechanisms underlying sex differences in demyelination of aging brains, we constructed a transcriptomic atlas of cell type-specific responses to illustrate how sex chromosomes, gonads, and their interaction shape responses to demyelination. We found that sex-biased oligodendrocyte and microglial responses are driven by interaction of sex chromosomes and gonads prior to myelin loss. Post demyelination, sex chromosomes mainly guide microglial responses, while gonadal composition influences oligodendrocyte signaling. Significantly, ablation of the X-linked gene Toll-like receptor 7 (Tlr7), which exhibited sex-biased expression during demyelination, abolished the sex-biased responses and protected against demyelination.

6.
J Neurosci ; 41(24): 5190-5205, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33941651

ABSTRACT

Hypertension susceptibility in women increases at the transition to menopause, termed perimenopause, a state characterized by erratic estrogen fluctuation and extended hormone cycles. Elucidating the role of estrogen signaling in the emergence of hypertension during perimenopause has been hindered by animal models that are confounded by abrupt estrogen cessation or effects of aging. In the present study, accelerated ovarian failure (AOF) in estrogen receptor ß (ERß) reporter mice was induced by 4-vinylcyclohexene diepoxide in young mice to model early-stage ovarian failure (peri-AOF) characteristic of peri-menopause. It was found that administering ERß agonists suppressed elevated blood pressure in a model of neurogenic hypertension induced by angiotensin II (AngII) in peri-AOF, but not in age-matched male mice. It was also found that ERß agonist administration in peri-AOF females, but not males, suppressed the heightened NMDAR signaling and reactive oxygen production in ERß neurons in the hypothalamic paraventricular nucleus (PVN), a critical neural regulator of blood pressure. It was further shown that deleting ERß in the PVN of gonadally intact females produced a phenotype marked by a sensitivity to AngII hypertension. These results suggest that ERß signaling in the PVN plays an important role in blood pressure regulation in female mice and contributes to hypertension susceptibility in females at an early stage of ovarian failure comparable to human perimenopause.


Subject(s)
Estrogen Receptor beta/metabolism , Hypertension/metabolism , Neuronal Plasticity/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Perimenopause/metabolism , Animals , Disease Models, Animal , Female , Hypertension/etiology , Mice , Mice, Inbred C57BL
7.
Neuroscience ; 410: 274-292, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31071414

ABSTRACT

Prescription opioid abuse is a serious public health issue. Recently, we showed that female and male Sprague-Dawley rats acquire conditioned place preference (CPP) to the mu opioid receptor agonist oxycodone. Anatomical analysis of the hippocampus from these rats unveiled sex differences in the opioid system in a way that would support excitation and opiate associative learning processes especially in females. In this study, we examined the expression and protein densities of opioid, plasticity, stress and related kinase and signaling molecules in the hippocampus of female and male rats following oxycodone CPP. Oxycodone CPP females have: a) increases in ARC (activity regulated cytoskeletal-associated protein)-immunoreactivity (ir) in CA3 pyramidal cells; b) decreases in Npy (neuropeptide Y) gene expression in the medial hippocampus but higher numbers of NPY-containing hilar interneurons compared to males; c) increases in Crhr2 (corticotropin releasing factor receptor 2) expression in CA2/3; d) increases in Akt1 (AKT serine/threonine kinase 1) expression in medial hippocampus; and e) decreases in phosphorylated MAPK (mitogen activated protein kinase)-ir in CA1 and dentate gyrus. Oxycodone CPP males have: a) increases in Bdnf (brain derived-neurotrophic factor) expression, which is known to be produced in granule cells, relative to females; b) elevated Mapk1 expression and pMAPK-ir in the dentate hilus which harbors newly generated granule cells; and c) increases in CRHR1-ir in CA3 pyramidal cell soma. These sex-specific changes in plasticity, stress and kinase markers in hippocampal circuitry parallel previously observed sex differences in the opioid system after oxycodone CPP.


Subject(s)
Analgesics, Opioid/pharmacology , Conditioning, Psychological/physiology , Neuronal Plasticity/physiology , Oxycodone/pharmacology , Sex Characteristics , Stress, Psychological/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Conditioning, Psychological/drug effects , Female , Gene Expression , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Stress, Psychological/genetics
8.
J Neurosci ; 36(49): 12425-12435, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27927959

ABSTRACT

Passive immunization with anti-tau monoclonal antibodies has been shown by several laboratories to reduce age-dependent tau pathology and neurodegeneration in mutant tau transgenic mice. These studies have used repeated high weekly doses of various tau antibodies administered systemically for several months and have reported reduced tau pathology of ∼40-50% in various brain regions. Here we show that direct intrahippocampal administration of the adeno-associated virus (AAV)-vectored anti-phospho-tau antibody PHF1 to P301S tau transgenic mice results in high and durable antibody expression, primarily in neurons. Hippocampal antibody levels achieved after AAV delivery were ∼50-fold more than those reported following repeated systemic administration. In contrast to systemic passive immunization, we observed markedly reduced (≥80-90%) hippocampal insoluble pathological tau species and neurofibrillary tangles following a single dose of AAV-vectored PHF1 compared with mice treated with an AAV-IgG control vector. Moreover, the hippocampal atrophy observed in untreated P301S mice was fully rescued by treatment with the AAV-vectored PHF1 antibody. Vectored passive immunotherapy with an anti-tau monoclonal antibody may represent a viable therapeutic strategy for treating or preventing such tauopathies as frontotemporal dementia, progressive supranuclear palsy, or Alzheimer's disease. SIGNIFICANCE STATEMENT: We have used an adeno-associated viral (AAV) vector to deliver the genes encoding an anti-phospho-tau monoclonal antibody, PHF1, directly to the brain of mice that develop neurodegeneration due to a tau mutation that causes frontotemporal dementia (FTD). When administered systemically, PHF1 has been shown to modestly reduce tau pathology and neurodegeneration. Since such antibodies do not readily cross the blood-brain barrier, we used an AAV vector to deliver antibody directly to the hippocampus and observed much higher antibody levels and a much greater reduction in tau pathology. Using AAV vectors to deliver antibodies like PHF1 directly to brain may constitute a novel approach to treating various neurodegenerative disorders, such as FTD and Alzheimer's disease.


Subject(s)
Antibodies, Monoclonal/immunology , Immunization, Passive/methods , Tauopathies/immunology , Tauopathies/prevention & control , Transcription Factors/immunology , tau Proteins/genetics , Animals , Antibodies, Monoclonal/administration & dosage , DNA-Binding Proteins , Dependovirus/immunology , Female , Hippocampus , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microinjections , Mutation/genetics , Neurofibrillary Tangles/pathology , Polycomb-Group Proteins , Tissue Distribution
9.
Am J Pathol ; 179(5): 2551-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21925470

ABSTRACT

ß-Amyloid (Aß) accumulation and aggregation are hallmarks of Alzheimer's disease (AD). High-resolution three-dimensional (HR-3D) volumetric imaging allows for better analysis of fluorescence confocal microscopy and 3D visualization of Aß pathology in brain. Early intraneuronal Aß pathology was studied in AD transgenic mouse brains by HR-3D volumetric imaging. To better visualize and analyze the development of Aß pathology, thioflavin S staining and immunofluorescence using antibodies against Aß, fibrillar Aß, and structural and synaptic neuronal proteins were performed in the brain tissue of Tg19959, wild-type, and Tg19959-YFP mice at different ages. Images obtained by confocal microscopy were reconstructed into three-dimensional volumetric datasets. Such volumetric imaging of CA1 hippocampus of AD transgenic mice showed intraneuronal onset of Aß42 accumulation and fibrillization within cell bodies, neurites, and synapses before plaque formation. Notably, early fibrillar Aß was evident within individual synaptic compartments, where it was associated with abnormal morphology. In dendrites, increasing intraneuronal thioflavin S correlated with decreases in neurofilament marker SMI32. Fibrillar Aß aggregates could be seen piercing the cell membrane. These data support that Aß fibrillization begins within AD vulnerable neurons, leading to disruption of cytoarchitecture and degeneration of spines and neurites. Thus, HR-3D volumetric image analysis allows for better visualization of intraneuronal Aß pathology and provides new insights into plaque formation in AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , CA1 Region, Hippocampal/pathology , Plaque, Amyloid/pathology , Synapses/metabolism , Animals , Antibodies, Monoclonal/metabolism , Cell Membrane/metabolism , Disease Progression , Female , Imaging, Three-Dimensional , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Neurites/pathology , Neurons/pathology , Synapses/pathology
10.
J Alzheimers Dis ; 27(1): 211-23, 2011.
Article in English | MEDLINE | ID: mdl-21799249

ABSTRACT

Increased oxidative stress is implicated in the pathogenesis of Alzheimer's disease (AD). A large body of evidence suggests that mitochondrial dysfunction and increased reactive oxygen species occur prior to amyloid-ß (Aß) deposition. Coenzyme Q10 (CoQ10), a component of the mitochondrial electron transport chain, is well characterized as a neuroprotective antioxidant in animal models and human trials of Huntington's disease and Parkinson's disease, and reduces plaque burden in AßPP/PS1 mice. We now show that CoQ10 reduces oxidative stress and amyloid pathology and improves behavioral performance in the Tg19959 mouse model of AD. CoQ10 treatment decreased brain levels of protein carbonyls, a marker of oxidative stress. CoQ10 treatment resulted in decreased plaque area and number in hippocampus and in overlying cortex immunostained with an Aß42-specific antibody. Brain Aß42 levels were also decreased by CoQ10 supplementation. Levels of amyloid-ß protein precursor (AßPP) ß-carboxyterminal fragments were decreased. Importantly, CoQ10-treated mice showed improved cognitive performance during Morris water maze testing. Our results show decreased pathology and improved behavior in transgenic AD mice treated with the naturally occurring antioxidant compound CoQ10. CoQ10 is well tolerated in humans and may be promising for therapeutic trials in AD.


Subject(s)
Alzheimer Disease/diet therapy , Amyloid beta-Peptides/metabolism , Behavioral Symptoms/drug therapy , Ubiquinone/analogs & derivatives , Vitamins/therapeutic use , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid/drug effects , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/metabolism , Behavioral Symptoms/etiology , Cognition Disorders/diet therapy , Cognition Disorders/etiology , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Motor Skills/drug effects , Motor Skills/physiology , Mutation/genetics , Neuroblastoma/pathology , Peptide Fragments/metabolism , Protein Carbonylation/drug effects , Protein Carbonylation/genetics , Time Factors , Ubiquinone/therapeutic use
11.
J Neurosci ; 30(43): 14299-304, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20980585

ABSTRACT

Accumulation of ß-amyloid (Aß) and loss of synapses are hallmarks of Alzheimer's disease (AD). How synaptic activity relates to Aß accumulation and loss of synapses is a current topic of major interest. Synaptic activation promotes Aß secretion, and chronic reduction of synaptic activity reduced Aß plaques in an AD transgenic mouse model. This suggested beneficial effects of reducing synaptic activity in AD. We now show that reduced synaptic activity causes detrimental effects on synapses and memory despite reducing plaques using two different models of chronic synaptic inhibition: deafferentation of the barrel cortex and administration of benzodiazepine. An interval of prolonged synaptic inhibition exacerbated loss of synaptophysin compared with synaptically more active brain in AD transgenic but not wild-type mice. Furthermore, an interval of benzodiazepine treatment, followed by a washout period, exacerbated memory impairment in AD transgenic mice. Exacerbation of synaptic and behavioral abnormalities occurred in the setting of reduced Aß plaques but elevated intraneuronal Aß immunoreactivity. These data support beneficial effects of synaptic activation on Aß-related synaptic and behavioral impairment in AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Memory/physiology , Synapses/physiology , Synaptophysin/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Blotting, Western , Cerebral Cortex/pathology , Diazepam/pharmacology , Female , Hippocampus/pathology , Hypnotics and Sedatives/pharmacology , Immunohistochemistry , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Motor Cortex/pathology , Plaque, Amyloid/pathology , Synapses/drug effects , Synapses/pathology , Vibrissae/innervation , Vibrissae/physiology
12.
PLoS One ; 5(9)2010 Sep 20.
Article in English | MEDLINE | ID: mdl-20862226

ABSTRACT

BACKGROUND: The mammalian target of rapamycin (mTOR) is an evolutionarily conserved Ser/Thr protein kinase that plays a pivotal role in multiple fundamental biological processes, including synaptic plasticity. We explored the relationship between the mTOR pathway and ß-amyloid (Aß)-induced synaptic dysfunction, which is considered to be critical in the pathogenesis of Alzheimer's disease (AD). METHODOLOGY/PRINCIPAL FINDINGS: We provide evidence that inhibition of mTOR signaling correlates with impairment in synaptic plasticity in hippocampal slices from an AD mouse model and in wild-type slices exposed to exogenous Aß1-42. Importantly, by up-regulating mTOR signaling, glycogen synthase kinase 3 (GSK3) inhibitors rescued LTP in the AD mouse model, and genetic deletion of FK506-binding protein 12 (FKBP12) prevented Aß-induced impairment in long-term potentiation (LTP). In addition, confocal microscopy demonstrated co-localization of intraneuronal Aß42 with mTOR. CONCLUSIONS/SIGNIFICANCE: These data support the notion that the mTOR pathway modulates Aß-related synaptic dysfunction in AD.


Subject(s)
Alzheimer Disease/metabolism , Neuronal Plasticity , Signal Transduction , Synapses/metabolism , TOR Serine-Threonine Kinases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Peptide Fragments/metabolism , TOR Serine-Threonine Kinases/genetics
13.
Cardiovasc Toxicol ; 7(3): 192-201, 2007.
Article in English | MEDLINE | ID: mdl-17901562

ABSTRACT

The tobacco industry markets potentially reduced exposure products (PREPs) as less harmful or addictive alternatives to conventional cigarettes. This study compared the effects of mainstream smoke from Quest, Eclipse, and 2R4F reference cigarettes on the development of atherosclerosis in apolipoprotein E-deficient (apoE -/-) mice. Mice were exposed to smoke from four cigarette types for 12 weeks beginning at age of 12 weeks, and in a separate study for 8 weeks, beginning at age of 8 weeks. In both studies, mice exposed to smoke from high-nicotine, high-tar Quest 1, and 2R4F cigarettes developed greater areas of lipid-rich aortic lesions than did non-smoking controls. Exposure to smoke from the lower-nicotine products, Eclipse, and Quest 3, was associated with smaller lesion areas, but animals exposed to smoke from all of the tested types of cigarette had larger lesions than did control animals not exposed to smoke. Urinary levels of isoprostane F2 alpha VI, increased proportionally to cigarette nicotine yield, whereas induction of pulmonary cytochrome P4501A1 was proportional to tar yield. Lesion area was associated with both nicotine and tar yields, although in multiple regression analysis only nicotine was a significant predictor of lesion area. Smoke exposure did not alter systolic blood pressure (SBP), heart rate (HR), blood cholesterol, or leukocyte count. Taken together, these observations suggest that smoking may accelerate atherosclerosis by increasing oxidative stress mediated at least in part via the actions of nicotine.


Subject(s)
Coronary Artery Disease/etiology , Nicotine/toxicity , Smoking/adverse effects , Administration, Inhalation , Animals , Aorta/drug effects , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Azo Compounds/chemistry , Coloring Agents/chemistry , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Cotinine/urine , Cytochrome P-450 CYP1A1/metabolism , Gene Silencing , Lung/drug effects , Lung/enzymology , Male , Mice , Mice, Knockout , Nicotine/urine , Oxidative Stress , Tars/analysis , Nicotiana/chemistry
14.
J Am Soc Hypertens ; 1(6): 423-32, 2007.
Article in English | MEDLINE | ID: mdl-20409875

ABSTRACT

Clinical and animal experimental studies suggest that combination therapy using angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors provides superior blood pressure (BP) lowering and target organ protection than either agent alone. We tested combination therapy with telmisartan and ramipril in lowering BP and protecting against stroke and target-organ damage in salt-fed stroke prone spontaneously hypertensive rats. Twenty-five rats were assigned to each of five groups: control (C), telmisartan (T), ramipril (R), and telmisartan + ramipril at full (TR) and at half-dose ((1/2)TR). Full dose telmisartan was 1 mg/kg/day and ramipril .4 mg/kg/day. Rats were fed a stroke prone diet for 8 weeks starting at age 7.5 weeks. Eighty-three percent C and 56% R showed behavioral signs of stroke. There were no strokes in other groups. BP was lower than control in all groups and lowest in TR. Urinary protein excretion, renal damage scores, and left ventricle cardiac collagen areas were lower than controls in all telmisartan treatment groups and lowest in TR. Telmisartan was superior to ramipril in preventing strokes, and telmisartan/ramipril combination therapy provided better BP control and greater cardio-renal protection than telmisartan alone.

15.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 23(2): 235-8, 2006 Apr.
Article in Chinese | MEDLINE | ID: mdl-16706337

ABSTRACT

In this study ECG signal of unstrained rat was recorded by telemetry device, and heart rate variability (HRV) was analyzed in order to evaluate 24h autonomic nervous activity. The results demonstrated an obvious circadian rhythm in the autonomic nervous activity: sympathetic activity being dominant during wake phase, and parasympathetic activity, dominant during sleep phase. The ratio of the low frequency to high frequency components in HRV power spectrum (LF/HF) fluctuates with the change in the sleep stages. It is concluded that 24h HRV analyses may reveal plentiful information about the behavior of autonomic nervous system and thus facilitate the investigation of its regulating role in physiological and pathological processes.


Subject(s)
Autonomic Nervous System/physiology , Circadian Rhythm , Heart Rate/physiology , Telemetry , Animals , Electrocardiography , Male , Rats , Rats, Sprague-Dawley
16.
Conf Proc IEEE Eng Med Biol Soc ; 2005: 6654-7, 2005.
Article in English | MEDLINE | ID: mdl-17281798

ABSTRACT

Telemetry technology offers possibility to observe physiological activity of animals in a nature state. In this paper, 24h physiologic signals of rats under different conditions were recorded. New indices for blood pressure evaluation were developed. Meanwhile, approaches to heart rate variability spectrum analysis, linear and non-linear analysis of electroencephalogram (EEG) were developed to estimate the activity of autonomic nervous system and central nervous system, respectively. The results demonstrate that the new parameters extracted from telemetry data may reveal more efficient information of physiological activity, which is impossible by conventional methods. The results also provide the evidence that the activity of central nervous system relates closely to the sympathovagal balance and the sympathetic activity.

17.
J Neurochem ; 89(5): 1308-12, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15147524

ABSTRACT

A growing body of evidence suggests a relationship between oxidative stress and beta-amyloid (Abeta) peptide accumulation, a hallmark in the pathogenesis of Alzheimer's disease (AD). However, a direct causal relationship between oxidative stress and Abeta pathology has not been established in vivo. Therefore, we crossed mice with a knockout of one allele of manganese superoxide dismutase (MnSOD), a critical antioxidant enzyme, with Tg19959 mice, which overexpress a doubly mutated human beta-amyloid precursor protein (APP). Partial deficiency of MnSOD, which is well established to cause elevated oxidative stress, significantly increased brain Abeta levels and Abeta plaque burden in Tg19959 mice. These results indicate that oxidative stress can promote the pathogenesis of AD and further support the feasibility of antioxidant approaches for AD therapy.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Brain/metabolism , Plaque, Amyloid/genetics , Superoxide Dismutase/deficiency , Amyloid beta-Protein Precursor/biosynthesis , Animals , Brain/pathology , Crosses, Genetic , Disease Models, Animal , Disease Progression , Heterozygote , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Oxidative Stress/genetics , Plaque, Amyloid/pathology , Superoxide Dismutase/genetics
18.
J Neurosci ; 24(14): 3592-9, 2004 Apr 07.
Article in English | MEDLINE | ID: mdl-15071107

ABSTRACT

Multiple lines of evidence implicate beta-amyloid (Abeta) in the pathogenesis of Alzheimer's disease (AD), but the mechanisms whereby Abeta is involved remain unclear. Addition of Abeta to the extracellular space can be neurotoxic. Intraneuronal Abeta42 accumulation is also associated with neurodegeneration. We reported previously that in Tg2576 amyloid precursor protein mutant transgenic mice, brain Abeta42 localized by immunoelectron microscopy to, and accumulated with aging in, the outer membranes of multivesicular bodies, especially in neuronal processes and synaptic compartments. We now demonstrate that primary neurons from Tg2576 mice recapitulate the in vivo localization and accumulation of Abeta42 with time in culture. Furthermore, we demonstrate that Abeta42 aggregates into oligomers within endosomal vesicles and along microtubules of neuronal processes, both in Tg2576 neurons with time in culture and in Tg2576 and human AD brain. These Abeta42 oligomer accumulations are associated with pathological alterations within processes and synaptic compartments in Tg2576 mouse and human AD brains.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Synapses/metabolism , Adult , Aging/metabolism , Aging/pathology , Alzheimer Disease/pathology , Animals , Brain/pathology , Cells, Cultured , Disease Models, Animal , Endosomes/metabolism , Endosomes/pathology , Fluorescent Antibody Technique , Humans , Macromolecular Substances , Mice , Mice, Transgenic , Microtubules/metabolism , Microtubules/pathology , Middle Aged , Neurons/cytology , Neurons/pathology , Organelles/metabolism , Organelles/pathology , Synapses/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...