Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 24(23)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38069296

ABSTRACT

Despite researchers' and clinicians' exponential understanding of chronic diseases' complexity, ranging from cancer, diabetes, and neurodegenerative disorders, we still have a lot of unanswered questions on pathobiology mechanisms, wherein inflammation is central [...].


Subject(s)
Cognition , Diabetes Mellitus , Humans , Inflammation
2.
BMC Biol ; 21(1): 256, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37953247

ABSTRACT

BACKGROUND: Traditionally, in biomedical animal research, laboratory rodents are individually examined in test apparatuses outside of their home cages at selected time points. However, the outcome of such tests can be influenced by various factors and valuable information may be missed when the animals are only monitored for short periods. These issues can be overcome by longitudinally monitoring mice and rats in their home cages. To shed light on the development of home cage monitoring (HCM) and the current state-of-the-art, a systematic review was carried out on 521 publications retrieved through PubMed and Web of Science. RESULTS: Both the absolute (~ × 26) and relative (~ × 7) number of HCM-related publications increased from 1974 to 2020. There was a clear bias towards males and individually housed animals, but during the past decade (2011-2020), an increasing number of studies used both sexes and group housing. In most studies, animals were kept for short (up to 4 weeks) time periods in the HCM systems; intermediate time periods (4-12 weeks) increased in frequency in the years between 2011 and 2020. Before the 2000s, HCM techniques were predominantly applied for less than 12 h, while 24-h measurements have been more frequent since the 2000s. The systematic review demonstrated that manual monitoring is decreasing in relation to automatic techniques but still relevant. Until (and including) the 1990s, most techniques were applied manually but have been progressively replaced by automation since the 2000s. Independent of the year of publication, the main behavioral parameters measured were locomotor activity, feeding, and social behaviors; the main physiological parameters were heart rate and electrocardiography. External appearance-related parameters were rarely examined in the home cages. Due to technological progress and application of artificial intelligence, more refined and detailed behavioral parameters have been investigated in the home cage more recently. CONCLUSIONS: Over the period covered in this study, techniques for HCM of mice and rats have improved considerably. This development is ongoing and further progress as well as validation of HCM systems will extend the applications to allow for continuous, longitudinal, non-invasive monitoring of an increasing range of parameters in group-housed small rodents in their home cages.


Subject(s)
Artificial Intelligence , Behavior, Animal , Male , Female , Mice , Animals , Rats , Behavior, Animal/physiology , Social Behavior , Heart Rate/physiology , Animals, Domestic
3.
Int J Mol Sci ; 24(10)2023 May 17.
Article in English | MEDLINE | ID: mdl-37240254

ABSTRACT

Traumatic brain injury (TBI) remains one of the leading causes of death and disability in young adults worldwide. Despite growing evidence and advances in our knowledge regarding the multifaceted pathophysiology of TBI, the underlying mechanisms, though, are still to be fully elucidated. Whereas initial brain insult involves acute and irreversible primary damage to the brain, the processes of subsequent secondary brain injury progress gradually over months to years, providing a window of opportunity for therapeutic interventions. To date, extensive research has been focused on the identification of druggable targets involved in these processes. Despite several decades of successful pre-clinical studies and very promising results, when transferred to clinics, these drugs showed, at best, modest beneficial effects, but more often, an absence of effects or even very harsh side effects in TBI patients. This reality has highlighted the need for novel approaches that will be able to respond to the complexity of the TBI and tackle TBI pathological processes on multiple levels. Recent evidence strongly indicates that nutritional interventions may provide a unique opportunity to enhance the repair processes after TBI. Dietary (poly)phenols, a big class of compounds abundantly found in fruits and vegetables, have emerged in the past few years as promising agents to be used in TBI settings due to their proven pleiotropic effects. Here, we give an overview of the pathophysiology of TBI and the underlying molecular mechanisms, followed by a state-of-the-art summary of the studies that have evaluated the efficacy of (poly)phenols administration to decrease TBI-associated damage in various animal TBI models and in a limited number of clinical trials. The current limitations on our knowledge concerning (poly)phenol effects in TBI in the pre-clinical studies are also discussed.


Subject(s)
Brain Injuries, Traumatic , Brain Neoplasms , Animals , Phenols/therapeutic use , Brain/pathology , Models, Animal , Brain Neoplasms/pathology
4.
Life Sci ; 297: 120470, 2022 May 15.
Article in English | MEDLINE | ID: mdl-35283177

ABSTRACT

The brain is the softest organ in the body, and any change in the mechanical properties of the tissue induces the activation of glial cells, astrocytes and microglia. Amyloid plaques, one of the main pathological features of Alzheimer's disease (AD), are substantially harder than the surrounding brain tissue and can activate astrocytes and microglia resulting in the glial engulfment of plaques. Durotaxis, a migratory preference towards stiffer tissue, is prompting microglia to form a mechanical barrier around plaques reducing amyloid ß (Aß) induced neurotoxicity. Mechanoreceptors are highly expressed in the brain, particularly in microglia. The large increase in the expression of the mechanoreceptor Piezo1 was observed in the brains from AD animal models and AD patients in plaque encompassing glia. Importantly, Piezo1 function is regulated via force-from-lipids through the lipid composition of the membrane and membranous incorporation of polyunsaturated fatty acids (PUFAs) can affect the function of Piezo1 altering mechanosensitive properties of the cell. On the other hand, PUFAs dietary supplementation can alter microglial polarization, the envelopment of amyloid plaques, and immune response and Piezo1 activity was implicated in the similar modulations of microglia behavior. Finally, PUFAs treatment is currently in use in medical trials as the therapy for sickle cell anemia, a disease linked with the mutations in Piezo1. Further studies are needed to elucidate the connection between PUFAs, Piezo1 expression, and microglia behavior in the AD brain. These findings could open new possibilities in harnessing microglia in AD and in developing novel therapeutic strategies.


Subject(s)
Alzheimer Disease , Fatty Acids , Ion Channels , Microglia , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Fatty Acids/metabolism , Humans , Ion Channels/genetics , Ion Channels/metabolism , Mice , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/pathology
5.
Nutr Neurosci ; 25(3): 537-549, 2022 Mar.
Article in English | MEDLINE | ID: mdl-32476608

ABSTRACT

OBJECTIVE: Traumatic brain injury (TBI) is one of the most common causes of neurological damage in young and middle aged people. Food restriction (FR) has been shown to act neuroprotectively in animal models of stroke and TBI. Indeed, our previous studies showed that FR attenuates inflammation, through suppression of microglial activation and TNF-α production, suppresses caspase-3-induced neuronal cell death and enhances neuroplasticity in the rat model of TBI. Glucocorticoids (GCs) play a central role in mediating both molecular and behavioral responses to food restriction. However, the exact mechanisms of FR neuroprotection in TBI are still unclear. The goal of the present study was to examine whether FR exerts its beneficial effects by altering the glucocorticoid receptor (GR) signaling alone and/or together with other protective factors. METHODS: To this end, we examined the effects of FR (50% of regular daily food intake for 3 months prior to TBI) on the protein levels of total GR, GR phosphoisoform Ser232 (p-GR) and its transcriptional activity, as well as 11ß-HSD1, NFκB (p65) and HSP70 as factors related to the GR signaling. RESULTS: Our results demonstrate that FR applied prior to TBI significantly changes p-GR levels, and it's transcriptional activity during the recovery period after TBI. Moreover, as a pretreatment, FR modulates other protective factors in response to TBI, such as 11ß-HSD1, NF-κB (p65) and HSP70 that act in parallel with GR in it's anti-inflammatory and neuroprotective effects in the rat model of brain injury. CONCLUSION: Our results suggest that prophylactic FR represents a potent non-invasive approach capable of changing GR signalling, together with other factors related to the GR signaling in the model of TBI.


Subject(s)
Brain Injuries, Traumatic , Neuroprotective Agents , Animals , Disease Models, Animal , Glucocorticoids , Humans , Middle Aged , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley
6.
Nutrients ; 13(9)2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34578818

ABSTRACT

The rise of neurodegenerative diseases in an aging population is an increasing problem of health, social and economic consequences. Epidemiological and intervention studies have demonstrated that diets rich in (poly)phenols can have potent health benefits on cognitive decline and neurodegenerative diseases. Meanwhile, the role of gut microbiota is ever more evident in modulating the catabolism of (poly)phenols to dozens of low molecular weight (poly)phenol metabolites that have been identified in plasma and urine. These metabolites can reach circulation in higher concentrations than parent (poly)phenols and persist for longer periods of time. However, studies addressing their potential brain effects are still lacking. In this review, we will discuss different model organisms that have been used to study how low molecular weight (poly)phenol metabolites affect neuronal related mechanisms gathering critical insight on their potential to tackle the major hallmarks of neurodegeneration.


Subject(s)
Diet/methods , Gastrointestinal Microbiome , Neurodegenerative Diseases/blood , Polyphenols/blood , Aged , Humans
7.
Antioxid Redox Signal ; 34(5): 421-438, 2021 02 10.
Article in English | MEDLINE | ID: mdl-32242468

ABSTRACT

Significance: It is well established that lifestyle and dietary habits have a tremendous impact on life span, the rate of aging, and the onset/progression of age-related diseases. Specifically, dietary restriction (DR) and other healthy dietary patterns are usually accompanied by physical activity and differ from Western diet that is rich in fat and sugars. Moreover, as the generation of reactive oxidative species is the major causative factor of aging, while DR could modify the level of oxidative stress, it has been proposed that DR increases both survival and longevity. Recent Advances: Despite the documented links between DR, aging, and oxidative stress, many issues remain to be addressed. For instance, the free radical theory of aging is under "re-evaluation," while DR as a golden standard for prolonging life span and ameliorating the effects of aging is also under debate. Critical Issues: This review article pays special attention to highlight the link between DR and oxidative stress in both aging and age-related diseases. We discuss in particular DR's capability to counteract the consequences of oxidative stress and the molecular mechanisms involved in these processes. Future Directions: Although DR is undoubtedly beneficial, several considerations must be taken into account when designing the best dietary intervention. Use of intermittent fasting, daily food reduction, or DR mimetics? Future research should unravel the pros and cons of all these processes. Antioxid. Redox Signal. 34, 421-438.


Subject(s)
Caloric Restriction , Oxidative Stress , Aging , Animals , Diet , Disease Susceptibility , Energy Metabolism , Humans , Longevity , Mitochondria/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
8.
Article in English | MEDLINE | ID: mdl-33238164

ABSTRACT

Reduction in direct social contact with peers during early adolescence is thought to be a risk factor for an increase in depressive symptoms, but there is still no clear evidence to suggest early behavioral manifestations and their association with the later outcome of social distancing during this period. To address this question, we used social isolation paradigm in peripubertal rats as the rodent model of adolescence. The litter was an experimental unit. On postnatal day 29, each litter gave group-housed and single-housed males, which were reared and tested one week and two weeks thereafter. Psychomotor/emotional response to novelty in exploration-based tasks, behavioral and neuronal responses to the drug reward (D-amphetamine), motivation/hedonic behavior, physiological and response to physiological stress were examined. Social isolation in peripubertal rats manifested through: hyper-reactivity/agitation and the state anxiety/risk-taking at an early stage; reduced behavioral response to D-amphetamine and altered neural processing of this stimulus, at a later stage; consummatory hypohedonia that deepened over time without changing the motivation to eat; unchanged body weight gain and resting blood corticosterone, cortisol and glucose levels over time; altered blood biochemistry (silenced corticosterone and increased glucose) due to overnight fasting only at an early stage. Our results highlight that the outcome of reduced direct social contact with peers during peripuberty is dynamic, with the cluster of atypical early symptoms that evolve into the syndrome that is delicate for assessment through routinely measurable behavior and biomarkers of stress, but with progressive consummatory hypohedonia and unaffected motivation to eat as stable marks.


Subject(s)
Dextroamphetamine/pharmacology , Exploratory Behavior/drug effects , Sexual Maturation/genetics , Social Isolation , Stress, Psychological/drug therapy , Animals , Corticosterone/blood , Emotions/drug effects , Male , Motor Activity/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Rats , Rats, Wistar , Stress, Psychological/psychology
9.
Free Radic Biol Med ; 162: 88-103, 2021 01.
Article in English | MEDLINE | ID: mdl-33279620

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia worldwide, characterized by a progressive decline in a variety of cognitive and non-cognitive functions. The amyloid beta protein cascade hypothesis places the formation of amyloid beta protein aggregates on the first position in the complex pathological cascade leading to neurodegeneration, and therefore AD might be considered to be a protein-misfolding disease. The Ubiquitin Proteasome System (UPS), being the primary protein degradation mechanism with a fundamental role in the maintenance of proteostasis, has been identified as a putative therapeutic target to delay and/or to decelerate the progression of neurodegenerative disorders that are characterized by accumulated/aggregated proteins. The purpose of this study was to test if the activation of proteasome in vivo can alleviate AD pathology. Specifically by using two compounds with complementary modes of proteasome activation and documented antioxidant and redox regulating properties in the 5xFAD transgenic mice model of AD, we ameliorated a number of AD related deficits. Shortly after proteasome activation we detected significantly reduced amyloid-beta load correlated with improved motor functions, reduced anxiety and frailty level. Essentially, to our knowledge this is the first report to demonstrate a dual activation of the proteasome and its downstream effects. In conclusion, these findings open up new directions for future therapeutic potential of proteasome-mediated proteolysis enhancement.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Phenotype , Proteasome Endopeptidase Complex
10.
Mech Ageing Dev ; 185: 111195, 2020 01.
Article in English | MEDLINE | ID: mdl-31837369

ABSTRACT

Alzheimer's disease patients (AD), as well as AD transgenic mice, are characterized by increased frailty. Furthermore, the assessment of frailty status represents a feasible approach for detecting individuals prone to develop more severe form of AD and for measuring the outcome of existing and putative AD therapeutics. The 5xFAD mouse is one of the widely used transgenic animal models of AD, but frailty in this model is scantly investigated. We used two validated mouse frailty assessment tools: phenotypic frailty score (FS) and clinical frailty index (FI) to investigate age- and sex- related differences in frailty status in 5xFAD mice. These tools measure different age-related deficits and do not necessarily identify the same subpopulations as frail. We detected a significant increase in frailty with age in both sexes, although females were surprisingly less frail than males. Depending on the tools used, a notable difference in frailty status was detected, with frailty index and frailty score identifying different mice as frail. These results warrant great caution when choosing the frailty tool and point to the need for further adaptation of frailty measurements in mouse models of AD.


Subject(s)
Alzheimer Disease/physiopathology , Disease Models, Animal , Frailty/diagnosis , Mice , Age Factors , Animals , Biological Variation, Population , Health Status Indicators , Humans , Mice, Transgenic , Research Design , Sex Factors
11.
PLoS One ; 14(5): e0216726, 2019.
Article in English | MEDLINE | ID: mdl-31095617

ABSTRACT

Dystrophic neurites and activated microglia are one of the main neuropathological characteristics of Alzheimer's disease (AD). Although the use of supplements with omega-3 fatty acids has been associated with reduced risk and lessened AD pathology, it still remains elusive whether such a treatment could affect dystrophic neurites (DNs) formation and microglia/macrophage behavior in the early phase of disease. We analyzed the effects of short-term (3 weeks) fish oil supplementation on DNs formation, tau hyperphosphorylation, Amyloid-beta peptide 1-42 (Aß42) levels and microglial/macrophage response to AD pathology in the parietal cortex of 4-month-old 5xFAD mice, a mouse model of AD. The present study shows for the first time that short-term FO supplementation applied in presymptomatic stage of AD, alters the behaviour of microglia/macrophages prompting them to establish a physical barrier around amyloid plaques. This barrier significantly suppresses DNs formation through the reduction of both Aß content and tau hyperphosphorylation. Moreover, the short-term FO treatment neither suppresses inflammation nor enhances phagocytic properties of microglia/macrophages in the response to Aß pathology, the effects most commonly attributed to the fish oil supplementation. Our findings suggest that fish oil consumption may play an important role in modulating microglial/macrophage response and ameliorating the AD pathology in presymptomatic stage of Alzheimer's disease.


Subject(s)
Alzheimer Disease/pathology , Asymptomatic Diseases , Fish Oils/pharmacology , Macrophages/drug effects , Microglia/drug effects , Neurites/pathology , Parietal Lobe/pathology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Atrophy/prevention & control , Cell Count , Cytokines/metabolism , Dietary Supplements , Disease Models, Animal , Macrophages/immunology , Mice , Microglia/pathology , Neurites/drug effects , Parietal Lobe/drug effects , Peptide Fragments/metabolism , Phagocytosis/drug effects , Phosphoproteins/metabolism , Time Factors , tau Proteins/metabolism
12.
Neurotox Res ; 32(2): 247-263, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28435999

ABSTRACT

Propofol is a general anesthetic commonly used in pediatric clinical practices. Experimental findings demonstrate that anesthetics induce widespread apoptosis and cognitive decline in a developing brain. Although anesthesia-mediated neurotoxicity is the most prominent during intense period of synaptogenesis, the effects of an early anesthesia exposure on the synapses are not well understood. The aim of this study was to examine the effects of neonatal propofol anesthesia on the expression of key proteins that participate in synaptogenesis and synaptic plasticity and to evaluate long-term neurobehavioral abnormalities in the mature adult brain. Propofol-injected 7-day-old rats were maintained under 2-, 4-, and 6-h-long anesthesia and sacrificed 0, 4, 16, and 24 h after the termination of each exposure. We showed that propofol anesthesia strongly influenced spatiotemporal expression and/or proteolytic processing of crucial presynaptic (GAP-43, synaptophysin, α-synuclein), trans-synaptic (N-cadherin), and postsynaptic (drebrin, MAP-2) proteins in the cortex and thalamus. An overall decrease of synaptophysin, α-synuclein, N-cadherin, and drebrin indicated impaired function and structure of the synaptic contacts immediately after anesthesia cessation. GAP-43 and MAP-2 adult and juvenile isoforms are upregulated following anesthesia, suggesting compensatory mechanism in the maintaining of the structural integrity and stabilization of developing axons and dendritic arbors. Neonatal propofol exposure significantly altered spontaneous motor activity (increased stereotypic/repetitive movements) and changed emotional behavior (reduced anxiety-like response) in the adulthood, 6 months later. These findings suggest that propofol anesthesia is synaptotoxic in the developing brain, disturbing synaptic dynamics and producing neuroplastic changes permanently incorporated into existing networks with long-lasting functional consequences.


Subject(s)
Anxiety/chemically induced , Hypnotics and Sedatives/toxicity , Propofol/toxicity , Stereotyped Behavior/drug effects , Synapses/metabolism , alpha-Synuclein/metabolism , Adaptation, Ocular/drug effects , Animals , Animals, Newborn , Anxiety/pathology , Brain/drug effects , Brain/growth & development , Brain/metabolism , Cadherins/metabolism , Exploratory Behavior/drug effects , Gene Expression Regulation, Developmental/drug effects , Male , Microtubule-Associated Proteins/metabolism , Motor Activity/drug effects , Neuropeptides/metabolism , Rats , Rats, Wistar , Synapses/drug effects , Synaptophysin/metabolism , Time Factors
13.
Neurotox Res ; 30(3): 434-52, 2016 10.
Article in English | MEDLINE | ID: mdl-27189477

ABSTRACT

A number of experimental studies have reported that exposure to common, clinically used anesthetics induce extensive neuroapoptosis and cognitive impairment when applied to young rodents, up to 2 weeks old, in phase of rapid synaptogenesis. Propofol is the most used general anesthetic in clinical practice whose mechanisms of neurotoxicity on the developing brain remains to be examined in depth. This study investigated effects of different exposures to propofol anesthesia on Fas receptor and Fas ligand expressions, which mediate proapoptotic and proinflammation signaling in the brain. Propofol (20 mg/kg) was administered to 7-day-old rats in multiple doses sufficient to maintain 2-, 4- and 6-h duration of anesthesia. Animals were sacrificed at 0, 4, 16 and 24 h after termination of anesthesia. It was found that propofol anesthesia induced Fas/FasL and downstream caspase-8 expression more prominently in the thalamus than in the cortex. Opposite, Bcl-2 and caspase-9, markers of intrinsic pathway activation, were shown to be more influenced by propofol treatment in the cortex. Further, we have established upregulation of caspase-1 and IL-1ß cytokine transcription as well as subsequent activation of microglia that is potentially associated with brain inflammation. Behavioral analyses revealed that P35 and P60 animals, neonatally exposed to propofol, had significantly higher motor activity during three consecutive days of testing in the open field, though formation of the intersession habituation was not prevented. This data, together with our previous results, contributes to elucidation of complex mechanisms of propofol toxicity in developing brain.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Brain/growth & development , Fas Ligand Protein/metabolism , Propofol/toxicity , fas Receptor/metabolism , Anesthetics, Intravenous/toxicity , Animals , Animals, Newborn , Apoptosis/physiology , Brain/immunology , Brain/pathology , Caspase 1/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Interleukin-1beta/metabolism , Male , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Motor Activity/drug effects , Motor Activity/physiology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Rats, Wistar , Time Factors
14.
Neurochem Int ; 96: 69-76, 2016 06.
Article in English | MEDLINE | ID: mdl-26939764

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death and disability in humans. Subsequent pathological events occurring in the brain after TBI, referred to as secondary injury, continue to damage surrounding tissue resulting in substantial neuronal loss. Using an animal model of TBI we examined the effect of dietary restriction (DR) on the neuroapoptosis and Bcl-2 family genes as the main regulators of the intrinsic apoptotic pathway. Bcl-2, Bcl-xl and Bax mRNA and protein expression in the ipsilateral cortex of adult Wistar rats exposed to DR before TBI were studied from 2 to 28 days post injury. Our results showed that DR suppressed neuroapoptosis and promoted significant upregulation of antiapoptotic Bcl-2 and Bcl-xl mRNAs in the ipsilateral cortex following injury. Expression of the proapoptotic Bax gene increased in ad libitum (AL) fed rats but remained unchanged in rats exposed to DR. Although the expression of Bcl-2, Bcl-xl and Bax proteins was changed in a similar manner in both experimental groups, DR promoted a continuous increase in the Bcl-2:Bax protein ratio throughout the recovery period. Together with our previous finding that DR mediates inhibition of the extrinsic apoptotic pathway the present work reveals that modulation of the intrinsic pathway contributes to the beneficial effect of DR in brain injury. These findings provide new insight into the effects of DR on pro-survival signaling after injury, lending further support to its neuroprotective effect.


Subject(s)
Brain Injuries/metabolism , Caloric Restriction/trends , Cerebral Cortex/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , bcl-2-Associated X Protein/biosynthesis , bcl-X Protein/biosynthesis , Animals , Apoptosis/physiology , Brain Injuries/pathology , Brain Injuries/prevention & control , Cerebral Cortex/pathology , Male , Random Allocation , Rats , Rats, Wistar
15.
Int J Dev Neurosci ; 44: 22-32, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25980792

ABSTRACT

Propofol anesthesia can trigger pro- and anti-apoptotic signaling pathways in the rat brain. In our previous work, we demonstrated that propofol causes widespread apoptotic neurodegeneration in 7-postnatal-day-old (PND7) but not in PND14 rat neurons. The mechanism responsible for these opposing outcomes is unknown, apparently linked to the specific stage of brain development. The present study aims to elucidate the anti-apoptotic process that is activated in the cortex and thalamus of PND14 Wistar rats during the first 48 h after the onset of propofol anesthesia. We showed that the expression of tumor necrosis factor-α (TNF-α) and several components of its pathway, TNFR1 and caspase-8, was significantly increased in the cortex and thalamus. Nuclear factor kappa B (NF-κB) p65 was downregulated in the cortex and upregulated in the thalamus. The expression of c-Fos was upregulated only in the cortex, showing opposed profile compared to NF-κB p65. Double immunofluorescence staining revealed the colocalization of NF-κB p65 with neuronal marker (NeuN), but with predominantly cytoplasmic localization. Finally, X-linked inhibitor of apoptosis protein (XIAP) was upregulated in both examined structures. Immunohistochemical staining with Iba-1 revealed that the treatment did not induce changes in microglial morphology. Our results (i) reveal that the simultaneous activation of pro- and anti-apoptotic signaling occurs after propofol anesthesia, and (ii) pinpoint the potential neuroprotective role of XIAP in anesthesia-induced neurotoxicity.


Subject(s)
Brain/drug effects , Hypnotics and Sedatives/pharmacology , Propofol/pharmacology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Analysis of Variance , Animals , Animals, Newborn , Caspase 8/metabolism , Gene Expression Regulation, Developmental/drug effects , Inhibitor of Apoptosis Proteins/metabolism , Male , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar , Time Factors , Tumor Necrosis Factor-alpha/genetics
16.
Lipids ; 48(11): 1069-77, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24057446

ABSTRACT

Disturbance of cholesterol homeostasis in the brain is coupled to age-related brain dysfunction. In the present work, we studied the relationship between aging and cholesterol metabolism in two brain regions, the cortex and hippocampus, as well as in the sera and liver of 6-, 12-, 18- and 24-month-old male Wistar rats. Using gas chromatography-mass spectrometry, we undertook a comparative analysis of the concentrations of cholesterol, its precursors and metabolites, as well as dietary-derived phytosterols. During aging, the concentrations of the three cholesterol precursors examined (lanosterol, lathosterol and desmosterol) were unchanged in the cortex, except for desmosterol which decreased (44 %) in 18-month-old rats. In the hippocampus, aging was associated with a significant reduction in lanosterol and lathosterol concentrations at 24 months (28 and 25 %, respectively), as well as by a significant decrease of desmosterol concentration at 18 and 24 months (36 and 51 %, respectively). In contrast, in the liver we detected age-induced increases in lanosterol and lathosterol concentrations, and no change in desmosterol concentration. The amounts of these sterols were lower than in the brain regions. In the cortex and hippocampus, desmosterol was the predominant cholesterol precursor. In the liver, lathosterol was the most abundant precursor. This ratio remained stable during aging. The most striking effect of aging observed in our study was a significant decrease in desmosterol concentration in the hippocampus which could reflect age-related reduced synaptic plasticity, thus representing one of the detrimental effects of advanced age.


Subject(s)
Aging , Cerebral Cortex/metabolism , Cholesterol/blood , Hippocampus/metabolism , Liver/metabolism , Animals , Lanosterol/blood , Male , Organ Specificity , Rats , Rats, Wistar
17.
Age (Dordr) ; 35(6): 2057-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23255148

ABSTRACT

Neurotrophins are established molecular mediators of neuronal plasticity in the adult brain. We analyzed the impact of aging on brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) protein isoforms, their receptors, and on the expression patterns of multiple 5' exon-specific BDNF transcripts in the rat cortex and hippocampus throughout the life span of the rat (6, 12, 18, and 24 months of age). ProNGF was increased during aging in both structures. Mature NGF gradually decreased in the cortex, and, in 24-month-old animals, it was 30% lower than that in adult 6-month-old rats. The BDNF expression did not change during aging, while proBDNF accumulated in the hippocampus of aged rats. Hippocampal total BDNF mRNA was lower in 12-month-old animals, mostly as a result of a decrease of BDNF transcripts 1 and 2. In contrast to the region-specific regulation of specific exon-containing BDNF mRNAs in adult animals, the same BDNF RNA isoforms (containing exons III, IV, or VI) were present in both brain structures of aged animals. Deficits in neurotrophin signaling were supported by the observed decrease in Trk receptor expression which was accompanied by lower levels of the two main downstream effector kinases, pAkt and protein kinase C. The proteolytic processing of p75NTR observed in 12-month-old rats points to an additional regulatory mechanism in early aging. The changes described herein could contribute to reduced brain plasticity underlying the age-dependent decline in cognitive function.


Subject(s)
Aging/genetics , Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/growth & development , Gene Expression Regulation, Developmental , Hippocampus/growth & development , Nerve Growth Factors/genetics , Protein Precursors/genetics , Aging/metabolism , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/biosynthesis , Cerebral Cortex/metabolism , Hippocampus/metabolism , Immunohistochemistry , Nerve Growth Factors/biosynthesis , Protein Precursors/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Transcription, Genetic
18.
PLoS One ; 7(5): e37215, 2012.
Article in English | MEDLINE | ID: mdl-22615943

ABSTRACT

Traumatic brain injury (TBI) is a widespread cause of death and a major source of adult disability. Subsequent pathological events occurring in the brain after TBI, referred to as secondary injury, continue to damage surrounding tissue resulting in substantial neuronal loss. One of the hallmarks of the secondary injury process is microglial activation resulting in increased cytokine production. Notwithstanding that recent studies demonstrated that caloric restriction (CR) lasting several months prior to an acute TBI exhibits neuroprotective properties, understanding how exactly CR influences secondary injury is still unclear. The goal of the present study was to examine whether CR (50% of daily food intake for 3 months) alleviates the effects of secondary injury on neuronal loss following cortical stab injury (CSI). To this end, we examined the effects of CR on the microglial activation, tumor necrosis factor-α (TNF-α) and caspase-3 expression in the ipsilateral (injured) cortex of the adult rats during the recovery period (from 2 to 28 days) after injury. Our results demonstrate that CR prior to CSI suppresses microglial activation, induction of TNF-α and caspase-3, as well as neurodegeneration following injury. These results indicate that CR strongly attenuates the effects of secondary injury, thus suggesting that CR may increase the successful outcome following TBI.


Subject(s)
Brain Injuries/pathology , Caloric Restriction , Microglia/pathology , Animals , Apoptosis/physiology , Brain Injuries/complications , Brain Injuries/metabolism , Caspase 3/biosynthesis , Male , Rats , Tumor Necrosis Factor-alpha/biosynthesis
19.
Dev Neurosci ; 32(4): 288-301, 2010.
Article in English | MEDLINE | ID: mdl-20714114

ABSTRACT

Exposure of newborn rats to a variety of anesthetics has been shown to induce apoptotic neurodegeneration in the developing brain. We investigated the effect of the general anesthetic propofol on the brain of 7-day-old (P7) Wistar rats during the peak of synaptic growth. Caspase and calpain protease families most likely participate in neuronal cell death. Our objective was to examine regional and temporal patterns of caspase-3 and calpain activity following repeated propofol administration (20 mg/kg). P7 rats were exposed for 2, 4 or 6 h to propofol and killed 0, 4, 16 and 24 h after exposure. Relative caspase-3 and calpain activities were estimated by Western blot analysis of the proteolytic cleavage products of α-II-spectrin, protein kinase C and poly(ADP-ribose) polymerase 1. Caspase-3 activity and expression displayed a biphasic pattern of activation. Calpain activity changed in a region- and time-specific manner that was distinct from that observed for caspase-3. The time profile of calpain activity exhibited substrate specificity. Fluoro-Jade B staining revealed an immediate neurodegenerative response that was in direct relationship to the duration of anesthesia in the cortex and inversely related to the duration of anesthesia in the thalamus. At later post-treatment intervals, dead neurons were detected only in the thalamus 24 h following the 6-hour propofol exposure. Strong caspase-3 expression that was detected at 24 h was not followed by cell death after 2- and 4-hour exposures to propofol. These results revealed complex patterns of caspase-3 and calpain activities following prolonged propofol anesthesia and suggest that both are a manifestation of propofol neurotoxicity at a critical developmental stage.


Subject(s)
Anesthetics, Intravenous/toxicity , Brain/drug effects , Calpain/metabolism , Caspase 3/metabolism , Propofol/toxicity , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Immunohistochemistry , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Rats , Rats, Wistar
20.
Brain Res ; 1321: 96-104, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20114033

ABSTRACT

The changes that occur during adolescence have a profound impact on the brain and behavior later in life. In this work we examined changes in motor activity during habituation to a novel environment and after treatment with MK-801 (0.025, 0.05, 0.1mg/kg) in peripubertal, pubertal and adult Wistar rats. The involvement of the motor cortex and striatum in motor activity was assessed by analyzing changes in c-Fos protein levels that served as an indicator of neuronal activity. During the habituation period, locomotor activity in peripubertal rats was higher during the first 10 min than in other groups. The same amount of stereotypy-like movements was detected in all three groups. MK-801 induced dose- and age-dependent changes in motor activity. Peripubertal rats were the most sensitive to treatment with MK-801. We also report a surprising finding that systemic application of MK-801 induced a similar age-related profile of changes in motor activity and c-Fos protein expression in the motor cortex but no c-Fos induction in the striatum. Our results demonstrate that, depending on the phase of adolescence the same dose of MK-801 affected behavioral functions in a different manner and that activity of the motor cortex rather than striatal activity was linked to drug-motor activity interactions.


Subject(s)
Brain/drug effects , Brain/metabolism , Dizocilpine Maleate/pharmacology , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Behavior, Animal/drug effects , Blotting, Western , Brain/growth & development , Habituation, Psychophysiologic/physiology , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL