Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Comp Neurol ; 532(4): e25610, 2024 04.
Article in English | MEDLINE | ID: mdl-38605461

ABSTRACT

The cerebellum is involved in higher order cognitive function and is susceptible to age-related atrophy. However, limited evidence has directly examined the cerebellum's role in cognitive aging. To interrogate potential substrates of the relationship between cerebellar structure and memory in aging, here we target the Purkinje cells (PCs). The sole output neurons of the cerebellum, PC loss and/or degeneration underlie a variety of behavioral abnormalities. Using a rat model of normal cognitive aging, we immunostained sections through the cerebellum for the PC-specific protein, calbindin-D28k. Although morphometric quantification revealed no significant difference in total PC number as a function of age or cognitive status, regional cell number was a more robust correlate of memory performance in the young cerebellum than in aged animals. Parallel biochemical analysis of PC-specific protein levels in whole cerebellum additionally revealed that calbindin-D28k and Purkinje cell protein-2 (pcp-2) levels were lower selectively in aged rats with spatial memory impairment compared to both young animals and aged rats with intact memory. These results suggest that cognitive aging is associated with cerebellum vulnerability, potentially reflecting disruption of the cerebellum-medial temporal lobe network.


Subject(s)
Purkinje Cells , S100 Calcium Binding Protein G , Rats , Animals , Purkinje Cells/metabolism , Calbindin 1/metabolism , S100 Calcium Binding Protein G/chemistry , S100 Calcium Binding Protein G/metabolism , Cerebellum , Neurons/metabolism
2.
J Neurosci ; 43(49): 8425-8441, 2023 12 06.
Article in English | MEDLINE | ID: mdl-37798131

ABSTRACT

Basal forebrain (BF) projections to the hippocampus and cortex are anatomically positioned to influence a broad range of cognitive capacities that are known to decline in normal aging, including executive function and memory. Although a long history of research on neurocognitive aging has focused on the role of the cholinergic basal forebrain system, intermingled GABAergic cells are numerically as prominent and well positioned to regulate the activity of their cortical projection targets, including the hippocampus and prefrontal cortex. The effects of aging on noncholinergic BF neurons in primates, however, are largely unknown. In this study, we conducted quantitative morphometric analyses in brains from young adult (6 females, 2 males) and aged (11 females, 5 males) rhesus monkeys (Macaca mulatta) that displayed significant impairment on standard tests that require the prefrontal cortex and hippocampus. Cholinergic (ChAT+) and GABAergic (GAD67+) neurons were quantified through the full rostrocaudal extent of the BF. Total BF immunopositive neuron number (ChAT+ plus GAD67+) was significantly lower in aged monkeys compared with young, largely because of fewer GAD67+ cells. Additionally, GAD67+ neuron volume was greater selectively in aged monkeys without cognitive impairment compared with young monkeys. These findings indicate that the GABAergic component of the primate BF is disproportionally vulnerable to aging, implying a loss of inhibitory drive to cortical circuitry. Moreover, adaptive reorganization of the GABAergic circuitry may contribute to successful neurocognitive outcomes.SIGNIFICANCE STATEMENT A long history of research has confirmed the role of the basal forebrain in cognitive aging. The majority of that work has focused on BF cholinergic neurons that innervate the cortical mantle. Codistributed BF GABAergic populations are also well positioned to influence cognitive function, yet little is known about this prominent neuronal population in the aged brain. In this unprecedented quantitative comparison of both cholinergic and GABAergic BF neurons in young and aged rhesus macaques, we found that neuron number is significantly reduced in the aged BF compared with young, and that this reduction is disproportionately because of a loss of GABAergic neurons. Together, our findings encourage a new perspective on the functional organization of the primate BF in neurocognitive aging.


Subject(s)
Basal Forebrain , Cognitive Aging , Animals , Male , Female , Basal Forebrain/physiology , Macaca mulatta , Cholinergic Neurons , Aging/physiology , Cholinergic Agents
3.
Neurobiol Aging ; 87: 132-137, 2020 03.
Article in English | MEDLINE | ID: mdl-31952867

ABSTRACT

The glycoprotein reelin has been implicated in both memory-related synaptic plasticity and Alzheimer's disease pathogenesis. Aged rats with memory impairment display decreased reelin expression in layer II of the entorhinal cortex (EC) relative to memory-intact subjects, and here we tested whether this effect extends to the primate brain. Seven young adult (8-10 years) and 14 aged (27-38 years) rhesus monkeys (Macaca mulatta) were examined, including 7 old animals classified as impaired based on their scores from a delayed nonmatching-to-sample recognition memory test. Histological sections spanning the rostrocaudal extent of the intermediate and caudal divisions of EC were processed by immunohistochemistry and the total number of reelin-positive neurons in layer II was estimated using design-based stereological techniques. The main finding was that the number of reelin-expressing neurons in EC layer II is decreased selectively in aged monkeys with memory deficits relative to young adult and aged subjects with intact memory. The results add to evidence implicating EC-hippocampal integrity in neurocognitive aging, and they suggest that disrupted reelin signaling may be among the mechanisms that mediate the associated vulnerability of this circuitry in Alzheimer's disease.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Extracellular Matrix Proteins/metabolism , Memory Disorders/metabolism , Memory Disorders/pathology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Serine Endopeptidases/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Animals , Cognitive Aging , Macaca mulatta , Memory Disorders/etiology , Rats , Reelin Protein , Signal Transduction/genetics , Signal Transduction/physiology
4.
Hippocampus ; 29(12): 1165-1177, 2019 12.
Article in English | MEDLINE | ID: mdl-31334577

ABSTRACT

Changes in neuronal network activity and increased interindividual variability in memory are among the most consistent features of growing older. Here, we examined the relationship between these hallmarks of aging. Young and aged rats were trained on a water maze task where aged individuals reliably display an increased range of spatial memory capacities relative to young. Two weeks later, neuronal activity was induced pharmacologically with a low dose of pilocarpine and control animals received vehicle. Activity levels were proxied by quantifying the immediate early gene products Arc and c-Fos. While no relationship was observed between basal, resting activity, and individual differences in spatial memory in any brain region, pilocarpine-induced marker expression was tightly coupled with memory in all areas of the prefrontal cortex (PFC) and hippocampus examined. The nature of this association, however, differed across regions and in relation to age-related cognitive outcome. Specifically, in the medial PFC, induced activity was greatest in aged rats with cognitive impairment and correlated with water maze performance across all subjects. In the hippocampus, the range of induced marker expression was comparable between groups and similarly coupled with memory in both impaired and unimpaired aged rats. Together the findings highlight that the dynamic range of neural network activity across multiple brain regions is a critical component of neurocognitive aging.


Subject(s)
Cognitive Aging/physiology , Hippocampus/metabolism , Maze Learning/physiology , Nerve Net/metabolism , Prefrontal Cortex/metabolism , Animals , Cognitive Aging/psychology , Cytoskeletal Proteins/biosynthesis , Hippocampus/drug effects , Male , Maze Learning/drug effects , Muscarinic Agonists/pharmacology , Nerve Net/drug effects , Nerve Tissue Proteins/biosynthesis , Pilocarpine/pharmacology , Prefrontal Cortex/drug effects , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Long-Evans
5.
Behav Brain Res ; 301: 1-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26698400

ABSTRACT

Long-term use of anti-diabetic agents has become commonplace as rates of obesity, metabolic syndrome and diabetes continue to escalate. Metformin, a commonly used anti-diabetic drug, has been shown to have many beneficial effects outside of its therapeutic regulation of glucose metabolism and insulin sensitivity. Studies on metformin's effects on the central nervous system are limited and predominantly consist of in vitro studies and a few in vivo studies with short-term treatment in relatively young animals; some provide support for metformin as a neuroprotective agent while others show evidence that metformin may be deleterious to neuronal survival. In this study, we examined the effect of long-term metformin treatment on brain neurotrophins and cognition in aged male C57Bl/6 mice. Mice were fed control (C), high-fat (HF) or a high-fat diet supplemented with metformin (HFM) for 6 months. Metformin decreased body fat composition and attenuated declines in motor function induced by a HF diet. Performance in the Morris water maze test of hippocampal based memory function, showed that metformin prevented impairment of spatial reference memory associated with the HF diet. Quantitative RT-PCR on brain homogenates revealed decreased transcription of BDNF, NGF and NTF3; however protein levels were not altered. Metformin treatment also decreased expression of the antioxidant pathway regulator, Nrf2. The decrease in transcription of neurotrophic factors and Nrf2 with chronic metformin intake, cautions of the possibility that extended metformin use may alter brain biochemistry in a manner that creates a vulnerable brain environment and warrants further investigation.


Subject(s)
Aging/drug effects , Brain/drug effects , Cognition/drug effects , Hypoglycemic Agents/adverse effects , Metformin/adverse effects , NF-E2-Related Factor 2/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/drug effects , Aging/physiology , Aging/psychology , Animals , Blood Glucose/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cognition/physiology , Diet, High-Fat/adverse effects , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice, Inbred C57BL , Motor Activity/drug effects , Nerve Growth Factor/metabolism , Neurotrophin 3/metabolism , RNA, Messenger/metabolism , Random Allocation
6.
J Neurosci ; 35(33): 11729-42, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26290249

ABSTRACT

The therapeutic potential of histone deacetylase inhibitor (HDACi) treatment has attracted considerable attention in the emerging area of cognitive neuroepigenetics. The possibility that ongoing cognitive experience importantly regulates the cell biological effects of HDACi administration, however, has not been systematically examined. In an initial experiment addressing this issue, we tested whether water maze training influences the gene expression response to acute systemic HDACi administration in the young adult rat hippocampus. Training powerfully modulated the response to HDACi treatment, increasing the total number of genes regulated to nearly 3000, including many not typically linked to neural plasticity, compared with <300 following HDACi administration alone. Although water maze training itself also regulated nearly 1800 genes, the specific mRNAs, gene networks, and biological pathways involved were largely distinct when the same experience was provided together with HDACi administration. Next, we tested whether the synaptic protein response to HDACi treatment is similarly dependent on recent cognitive experience, and whether this plasticity is altered in aged rats with memory impairment. Whereas synaptic protein labeling in the young hippocampus was selectively increased when HDACi administration was provided in conjunction with water maze training, combined treatment had no effect on synaptic proteins in the aged hippocampus. Our findings indicate that ongoing experience potently regulates the molecular consequences of HDACi treatment and that the interaction of recent cognitive experience with histone acetylation dynamics is disrupted in the aged hippocampus. SIGNIFICANCE STATEMENT: The possibility that interventions targeting epigenetic regulation could be effective in treating a range of neurodegenerative disorders has attracted considerable interest. Here we demonstrate in the rat hippocampus that ongoing experience powerfully modifies the molecular response to one such intervention, histone deacetylase inhibitor (HDACi) administration. A single learning episode dramatically shifts the gene expression profile induced by acute HDACi treatment, yielding a qualitatively distinct hippocampal transcriptome compared with the influence of behavioral training alone. The downstream synaptic protein response to HDACi administration is similarly experience-dependent, and we report that this plasticity is disrupted in the aged hippocampus. The findings highlight that accommodating the modulatory influence of ongoing experience represents a challenge for therapeutic development in the area of cognitive neuroepigenetics.


Subject(s)
Aging/physiology , Hippocampus/physiology , Histone Deacetylase Inhibitors/pharmacology , Memory, Long-Term/physiology , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Animals , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Male , Memory, Long-Term/drug effects , Neuronal Plasticity/drug effects , Rats , Rats, Inbred F344
7.
Pharmacol Biochem Behav ; 100(1): 205-11, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21875615

ABSTRACT

Some patients experience enduring cognitive impairment after cancer treatment, a condition termed "chemofog". Animal models allow assessment of chemotherapy effects on learning and memory per se, independent of changes due to cancer itself or associated health consequences such as depression. The present study examined the long-term learning and memory effects of a chemotherapy cocktail used widely in the treatment of breast cancer, consisting of 5-fluorouracil (5FU) and cyclophosphamide (CYP). Eighty 5-month old male F344 rats received contextual and cued fear conditioning before treatment with saline, or a low or high dose drug cocktail (50mg/kg CYP and 75 mg/kg 5FU, or 75 mg/kg CYP and 120 mg/kg 5FU, i.p., respectively) every 30 days for 2 months. After a 2-month, no-drug recovery, both long-term retention and new task acquisition in the water maze and 14-unit T-maze were assessed. Neither dose of the CYP/5FU cocktail impaired retrograde fear memory despite marked toxicity documented by enduring weight loss and 50% mortality at the higher dose. Acquisition in the water maze and Stone maze was also normal relative to controls in rats treated with CYP/5FU. The results contribute to a growing literature suggesting that learning and memory mediated by the hippocampus can be relatively resistant to chemotherapy. Future investigation may need to focus on assessments of processing speed, executive function and attention, and the possible interactive contribution of cancer itself and aging to the post-treatment development of cognitive impairment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Memory/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cognition Disorders/chemically induced , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Learning/drug effects , Learning/physiology , Male , Memory/physiology , Rats , Rats, Inbred F344 , Treatment Outcome
8.
J Biol Chem ; 284(14): 9540-8, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19189968

ABSTRACT

We recently demonstrated mitochondrial localization of estrogen receptor beta (ERbeta). We herein confirm the mitochondrial localization of ERbeta by the loss of mitochondrial ERbeta immunoreactivity in ERbeta knockdown cells. A phenotype change characterized as an increase in resistance to oxidative stressors is associated with ERbeta knockdown. ERbeta knockdown results in a lower resting mitochondrial membrane potential (Deltapsim) and increase in resistance to hydrogen peroxide-induced Deltapsim depolarization in both immortal hippocampal cells and primary hippocampal neurons. ERbeta knockdown cells maintained ATP concentrations despite insults that compromise ATP production and produce less mitochondrial superoxide under oxidative stress. Furthermore, similar mitochondrial phenotype changes were identified in primary hippocampal neurons derived from ERbeta knock-out mice. These data demonstrate that ERbeta is expressed in mitochondria and function as a mitochondrial vulnerability factor involved in Deltapsim maintenance, potentially through a mitochondrial transcription dependent mechanism.


Subject(s)
Estrogen Receptor beta/metabolism , Mitochondria/metabolism , Animals , Cells, Cultured , Estrogen Receptor beta/deficiency , Estrogen Receptor beta/genetics , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , RNA Interference , Rats , Tissue Culture Techniques
9.
Biochim Biophys Acta ; 1772(4): 473-83, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17113760

ABSTRACT

Although neurofibrillary tangle (NFT) formation is a central event in both familial and sporadic Alzheimer's disease (AD), neither cellular origin nor functional consequence of the NFTs are fully understood. This largely is due to the lack of available in vivo models for neurofibrillary degeneration (NFD). NFTs have only been identified in transgenic mice, bearing a transgene for a rare hereditary neurodegenerative disease, frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP17). Epidemiological evidence suggests a much higher occurrence of dementia in stroke patients. This may represent the underlying cause of the pathogenesis of sporadic AD, which accounts for the majority of AD cases. We examined pathological markers of AD in a rodent stroke model. Here we show that after transient cerebral ischemia, hyperphosphorylated tau accumulates in neurons of the cerebral cortex in the ischemic area, forms filaments similar to those present in human neurodegenerative tauopathies and colocalizes with markers of apoptosis. As a potential underlying mechanism, we were able to determine that transient ischemia induced tau hyperphosphorylation and NFT-like conformations are associated with aberrant activation of cyclin dependent kinase 5 (Cdk5) and can be rescued by delivery of a potent, but non-specific cyclin dependent kinase inhibitor, roscovitine to the brain. Our study further indicates that accumulation of p35 and its calpain-mediated cleavage product, p25 may account for the deregulation of Cdk5 induced by transient ischemia. We conclude that Cdk5 may be the principal protein kinase responsible for tau hyperphosphorylation and may be a hallmark of the tauopathies in this stroke model.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Ischemic Attack, Transient/pathology , Animals , Cyclin-Dependent Kinases/antagonists & inhibitors , Disease Models, Animal , Female , Functional Laterality , Neurofibrillary Tangles/pathology , Phosphorylation , Protein Kinase Inhibitors , Purines , Rats , Rats, Sprague-Dawley , Roscovitine , Tauopathies/pathology
10.
Curr Drug Targets CNS Neurol Disord ; 4(2): 169-77, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15857302

ABSTRACT

Estrogens are now recognized as potent neuroprotectants in a variety of in vitro and in vivo model for cerebral ischemia. These protective effects of estrogens are seen in neurons, astrocytes, microglia and vascular endothelial cells and result in a profound protection of the brain during stroke. Herein, we provide a thesis that indicates that the protective effects of estrogens during stroke may be a combined effect on multiple targets of the neurovascular unit (NVU) through a fundamental protective effect of estrogens on the subcellular organelle that defines the fate of cells during insults, the mitochondria. By protecting mitochondria during insult, estrogens are able to reduce or eliminate the signal for cellular necrosis or apoptosis and thereby protect the NVU from ischemia/reperfusion. In this context, estrogens may be unique in their ability to target the cellular site of initiation of damage during stroke and could be a central compound in a multi-drug approach to the prevention and treatment of brain damage from stroke.


Subject(s)
Brain Ischemia/metabolism , Estrogens/metabolism , Mitochondria/metabolism , Neuroprotective Agents/metabolism , Stroke/metabolism , Animals , Brain/blood supply , Brain/cytology , Brain/metabolism , Brain Ischemia/complications , Brain Ischemia/prevention & control , Cerebral Arteries/cytology , Endothelium, Vascular/metabolism , Estrogens/therapeutic use , Humans , Microglia/metabolism , Muscle, Smooth, Vascular/metabolism , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Stroke/complications , Stroke/drug therapy
11.
Neuroreport ; 15(9): 1515-8, 2004 Jun 28.
Article in English | MEDLINE | ID: mdl-15194886

ABSTRACT

Estrogens exert neuroprotective activity in both in vivo and in vitro model systems. Herein, we report that both 17beta-estradiol and low concentrations of nitric oxide (NO) attenuate hydrogen peroxide (H2O2) induced toxicity in SK-N-SH cells, which express the neuronal nitric oxide synthase (nNOS). 17beta-estradiol rapidly induced an increase in NO levels. A nNOS inhibitor was able to block the neuroprotection of 17beta-estradiol. Cyclic guanylyl mono-phosphate (cGMP) also protected against H2O2 induced toxicity, while NO's protection was attenuated by ODQ, a soluble guanylyl cyclase (sGC) inhibitor. In SK-N-SH cells, the major estrogen receptor isoforms is estrogen receptor beta. Our current study suggests that increased activity of nNOS may be involved in the neuroprotection conferred by 17beta-estradiol.


Subject(s)
Estradiol/pharmacology , Neurons/drug effects , Neurons/enzymology , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase/metabolism , Cell Line, Tumor , Cyclic GMP/metabolism , Humans , Hydrogen Peroxide/toxicity , Neuroblastoma , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type I , Nitroso Compounds/pharmacology , Oxidants/toxicity
12.
Proc Natl Acad Sci U S A ; 101(12): 4130-5, 2004 Mar 23.
Article in English | MEDLINE | ID: mdl-15024130

ABSTRACT

Estrogen receptors (ERs) are believed to be ligand-activated transcription factors belonging to the nuclear receptor superfamily, which on ligand binding translocate into the nucleus and activate gene transcription. To date, two ERs have been identified: ERalpha and ERbeta. ERalpha plays major role in the estrogen-mediated genomic actions in both reproductive and nonreproductive tissue, whereas the function of ERbeta is still unclear. In this study, we used immunocytochemistry, immunoblotting, and proteomics to demonstrate that ERbeta localizes to the mitochondria. In immunocytochemistry studies, ERbeta was detected with two ERbeta antibodies and found to colocalize almost exclusively with a mitochondrial marker in rat primary neuron, primary cardiomyocyte, and a murine hippocampal cell line. The colocalization of ERbeta and mitochondrial markers was identified by both fluorescence and confocal microscopy. No translocation of ERbeta into the nucleus on 17beta-estradiol treatment was seen by using immunocytochemistry. Immunoblotting of purified human heart mitochondria showed an intense signal of ERbeta, whereas no signals for nuclear and other organelle markers were found. Finally, purified human heart mitochondrial proteins were separated by SDS/PAGE. The 50,000-65,000 M(r) band was digested with trypsin and subjected to matrix-assisted laser desorption/ionization mass spectrometric analysis, which revealed seven tryptic fragments that matched with those of ERbeta. In summary, this study demonstrated that ERbeta is localized to mitochondria, suggesting a role for mitochondrial ERbeta in estrogen effects on this important organelle.


Subject(s)
Mitochondria/metabolism , Receptors, Estrogen/metabolism , Animals , Estrogen Receptor beta , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunohistochemistry , Mice , Microscopy, Confocal , Myocytes, Cardiac/metabolism , Neurons/metabolism , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Proc Natl Acad Sci U S A ; 100(20): 11741-6, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-14504383

ABSTRACT

Substantial evidence now exists that intrinsic free-radical scavenging contributes to the receptor-independent neuroprotective effects of estrogens. This activity is inherently associated with the presence of a phenolic A-ring in the steroid. We report a previously unrecognized antioxidant cycle that maintains the "chemical shield" raised by estrogens against the most harmful reactive oxygen species, the hydroxyl radical (*OH) produced by the Fenton reaction. In this cycle, the capture of *OH was shown to produce a nonphenolic quinol with no affinity to the estrogen receptors. This quinol is then rapidly converted back to the parent estrogen via an enzyme-catalyzed reduction by using NAD(P)H as a coenzyme (reductant) and, unlike redox cycling of catechol estrogens, without the production of reactive oxygen species. Due to this process, protection of neuronal cells against oxidative stress is also possible by quinols that essentially act as prodrugs for the active hormone. We have shown that the quinol obtained from a 17beta-estradiol derivative was, indeed, able to attenuate glutamate-induced oxidative stress in cultured hippocampus-derived HT-22 cells. Estrone quinol was also equipotent with its parent estrogen in reducing lesion volume in ovariectomized rats after transient middle carotid artery occlusion followed by a 24-h reperfusion. These findings may establish the foundation for a rational design of neuroprotective antioxidants focusing on steroidal quinols as unique molecular leads.


Subject(s)
Antioxidants/pharmacology , Estrogens/pharmacology , Hydroquinones/pharmacology , Neuroprotective Agents/pharmacology , Animals , Brain Ischemia/prevention & control , Chromatography, Liquid , Female , Free Radical Scavengers/pharmacology , Hydroxyl Radical , Mass Spectrometry , Oxidative Stress , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...