Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-37889220

ABSTRACT

Throughout the years, anatomic studies have demonstrated numerous variations in the course of the cephalic vein (CV). There are, however, very rare cases of uncommon formation, course or termination of the vein to which our attention should be drawn. During a routine dissections conducted in the Department of Anatomy and Neurobiology, in two formalin-fixed cadavers, the very rare anatomical variants were found. In 80 year-old Caucasian female the right cephalic vein, after crossing the clavipectoral triangle, ascended anterior and superior to the clavicle and drained into the lateral branch of the right external jugular vein, which in turn opened to the right subclavian vein. In the second case, the dissection of 83 year-old Caucasian male cadaver revealed that after passing through the deltopectoral groove, the left cephalic vein run between clavicle and subclavius muscle to terminate in the left subclavian vein. Understanding of the topography, morphology and anatomical variations of the cephalic vein is important not only for the anatomists but for the clinicians and nurses as well. Such knowledge can prevent multiple complications during many invasive procedures including implantation of Cardiac Implantable Electronic Devices, central venous access, arteriovenous fistula creation or even iatrogenic injuries during clavicle or glenohumeral joint surgery.

2.
Cell Mol Neurobiol ; 43(8): 4245-4259, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37801200

ABSTRACT

Hypercholesterolemia affects the neurovascular unit, including the cerebral blood vessel endothelium. Operation of this system, especially in the context of energy metabolism, is controlled by extracellular concentration of purines, regulated by ecto-enzymes, such as e-NTPDase-1/CD39, ecto-5'-NT/CD73, and eADA. We hypothesize that hypercholesterolemia, via modulation of the activity of nucleotide metabolism-regulating ecto-enzymes, deteriorates glycolytic efficiency and energy metabolism of endothelial cells, which may potentially contribute to development of neurodegenerative processes. We aimed to determine the effect of hypercholesterolemia on the concentration of purine nucleotides, glycolytic activity, and activity of ecto-enzymes in the murine brain microvascular endothelial cells (mBMECs). We used 3-month-old male LDLR-/-/Apo E-/- double knockout mice to model hypercholesterolemia and atherosclerosis. The age-matched wild-type C57/BL6 mice were a control group. The intracellular concentration of ATP and NAD and extracellular activity of the ecto-enzymes were measured by HPLC. The glycolytic function of mBMECs was assessed by means of the extracellular acidification rate (ECAR) using the glycolysis stress test. The results showed an increased activity of ecto-5'-NT and eADA in mBMECs of the hypercholesterolemic mice, but no differences in intracellular concentration of ATP, NAD, and ECAR between the hypercholesterolemic and control groups. The changed activity of ecto-5'-NT and eADA leads to increased purine nucleotides turnover and a shift in their concentration balance towards adenosine and inosine in the extracellular space. However, no changes in the energetic metabolism of the mBMECs are reported. Our results confirm the influence of hypercholesterolemia on regulation of purine nucleotides metabolism, which may impair the function of the cerebral vascular endothelium. The effect of hypercholesterolemia on the murine brain microvascular endothelial cells (mBMECs). An increased activity of ecto-5'-NT and eADA in mBMECs of the LDLR-/-/Apo E-/- mice leads to a shift in the concentration balance towards adenosine and inosine in the extracellular space with no differences in intracellular concentration of ATP. Figure was created with Biorender.com.


Subject(s)
Hypercholesterolemia , Male , Mice , Animals , Endothelial Cells/metabolism , NAD/metabolism , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Brain/metabolism , Mice, Knockout , Endothelium/metabolism , Inosine , Apolipoproteins E , 5'-Nucleotidase/metabolism
3.
Mol Cell Neurosci ; 125: 103860, 2023 06.
Article in English | MEDLINE | ID: mdl-37182573

ABSTRACT

One of the effects of hypercholesterolemia (Hch) exerted on the central nervous system (CNS) is damage to the blood-brain barrier (BBB). Increased permeability of BBB results from structural changes in the vascular wall, loss of the tight junctions and barrier function, as well as alterations in the concentration of proteins located in the layers of the vascular wall. These changes occur in the course of metabolic and neurodegenerative diseases. The important role in the course of these processes is attributed to agrin, matrix metalloproteinase-9, and aquaporin-4. In this study, we aimed to determine: 1) the extent of Hch-induced damage to the BBB during maturation, and 2) the distribution of the above-mentioned markers in the vascular wall. Immunohistochemical staining and confocal microscopy were used for vascular wall protein assessment. The size of BBB damage was studied based on perivascular leakage of fluorescently labeled dextran. Three- and twelve-month-old male LDLR-/-/Apo E-/- double knockout mice (EX) developing Hch were used in the study. Age-matched male wild-type (WT) C57BL/6 mice were used as a control group. Differences in the concentration of studied markers coexisted with BBB disintegration, especially in younger mice. A relationship between the maturation of the vascular system and reduction of the BBB damage was also observed. We conclude that the extent of BBB permeability depends on animal age, duration of Hch, and brain region. These may explain different susceptibility of various brain areas to Hch, and different presentation of this pathology depending on age and its duration.


Subject(s)
Blood-Brain Barrier , Brain , Animals , Male , Mice , Apolipoproteins E/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/metabolism
4.
Mol Neurobiol ; 60(9): 4935-4951, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37204689

ABSTRACT

Statins are lipid-lowering drugs that act by inhibiting 3-hydroxy-3-methylglutaryl coenzyme A reductase, a rate-limiting enzyme in cholesterol biosynthesis. Animal studies have shown neuroprotective effects of statins in cerebral stroke. However, the underlying mechanisms are not fully understood. The nuclear factor-kappa B (NF-κB) transcription factor is involved in the regulation of apoptosis in stroke. Different dimers of NF-κB regulate the gene expression of proteins involved in both neurodegeneration and neuroprotection. We aimed to determine whether simvastatin improves stroke outcome via inhibition of the RelA/p65-containing subunit and downregulation of stroke-induced pro-apoptotic genes or via activation of NF-κB dimers containing the c-Rel subunit and upregulation of anti-apoptotic genes during the acute stroke phase. Eighteen-month-old Wistar rats, subjected to permanent MCAO or sham surgery, were administered simvastatin (20 mg/kg b.w.) or saline for 5 days before the procedure. Stroke outcome was determined by measuring cerebral infarct and assessing motor functions. The expression of NF-κB subunits in various cell populations was investigated using immunofluorescence/confocal microscopy. RelA and c-Rel were detected by WB. The NF-κB-DNA binding activity was investigated using EMSA, and expression of Noxa, Puma, Bcl-2, and Bcl-x genes was analyzed by qRT-PCR. Results showed a 50% infarct size reduction and significant motor function improvement in the simvastatin-treated animals which correlated with a decrease in RelA and a transient increase in the c-Rel level in the nucleus, normalization of the NF-κB-DNA binding activity, and downregulation of the NF-κB-regulated genes. Our results provide new insights into the statin-mediated neuroprotective action against stroke based on NF-κB pathway inhibition.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Ischemic Stroke , Stroke , Rats , Animals , NF-kappa B/metabolism , Simvastatin/pharmacology , Simvastatin/therapeutic use , Neuroprotection , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Rats, Wistar , Transcription Factor RelA/metabolism , Stroke/complications , Stroke/drug therapy , DNA
5.
J Chem Neuroanat ; 127: 102192, 2023 01.
Article in English | MEDLINE | ID: mdl-36403746

ABSTRACT

Brain injury triggers a complex response involving morphological changes, cellular proliferation, and differentiation of newly formed neuroglial subpopulations. These processes have been extensively studied in animal stroke models with permanent large vessel occlusion. However, less is known about neuroglial response after transient cerebral ischemia. Herein, we aimed to determine an astrocytic and NG2 glial proliferative response, potential changes in expression of developmental neuroglial markers: vimentin, nestin, oligodendrocyte transcription marker (Olig2), and a role of neuroglial subpopulations as a source of cells replenishing structural deficiencies in the ischemic brain. Results showed an induction of a proliferative neuroglial response in the peri-infarct area reflected in an increased percentage of GFAP/Ki67 + and NG2/Ki67 + cells within 4 weeks after transient MCAO. The peak of GFAP+ astrocytes proliferation of 30.3 ± 10.3% was observed in the first week, and a peak of NG2 + cells proliferation of 23.1 ± 11.8% in the second week after stroke. The presence of GFAP/Vimentin+ and GFAP/Nestin+ cells, as well as GFAP/Olig2 + and NG2/Olig2 + cells indicated an induction of developmental phenotypes with a differentiation potential. Finally, observed between day 1 and week 3 transient GFAP/NG2 + colocalization suggests the heterogeneous source of the reactive neuroglia after transient MCAO. Altogether, one-hour MCAO is a sufficient pathological stimulus to trigger a strong proliferative response of GFAP+ and NG2 + neuroglial cells and induce their early developmental phenotype. Our results suggest that transient ischemia may initiate a change in the direction of differentiation within the neuroglia cell population.


Subject(s)
Ischemic Attack, Transient , Stroke , Animals , Ischemic Attack, Transient/pathology , Nestin/metabolism , Vimentin/metabolism , Ki-67 Antigen/metabolism , Glial Fibrillary Acidic Protein/metabolism , Neuroglia/metabolism , Astrocytes/metabolism , Cell Differentiation/physiology , Stroke/metabolism , Cell Proliferation
6.
Int J Mol Sci ; 23(5)2022 Feb 27.
Article in English | MEDLINE | ID: mdl-35269761

ABSTRACT

Stress and negative emotions evoked by social relationships and working conditions, frequently accompanied by the consumption of addictive substances, and metabolic and/or genetic predispositions, negatively affect brain function. One of the affected structures is nucleus accumbens (NAc). Although its function is commonly known to be associated with brain reward responses and addiction, a growing body of evidence also suggests its role in some mental disorders, such as depression and schizophrenia, as well as neurodegenerative diseases, such as Alzheimer's, Huntington's, and Parkinson's. This may result from disintegration of the extensive connections based on numerous neurotransmitter systems, as well as impairment of some neuroplasticity mechanisms in the NAc. The consequences of NAc lesions are both morphological and functional. They include changes in the NAc's volume, cell number, modifications of the neuronal dendritic tree and dendritic spines, and changes in the number of synapses. Alterations in the synaptic plasticity affect the efficiency of synaptic transmission. Modification of the number and structure of the receptors affects signaling pathways, the content of neuromodulators (e.g., BDNF) and transcription factors (e.g., pCREB, DeltaFosB, NFκB), and gene expression. Interestingly, changes in the NAc often have a different character and intensity compared to the changes observed in the other parts of the basal ganglia, in particular the dorsal striatum. In this review, we highlight the role of the NAc in various pathological processes in the context of its structural and functional damage, impaired connections with the other brain areas cooperating within functional systems, and progression of the pathological processes.


Subject(s)
Behavior, Addictive , Mental Disorders , Neurodegenerative Diseases , Humans , Mental Disorders/etiology , Mental Disorders/metabolism , Neurodegenerative Diseases/metabolism , Nucleus Accumbens/metabolism , Reward
7.
Int J Mol Sci ; 22(18)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34575969

ABSTRACT

A growing body of evidence suggests that nucleus accumbens (NAc) plays a significant role not only in the physiological processes associated with reward and satisfaction but also in many diseases of the central nervous system. Summary of the current state of knowledge on the morphological and functional basis of such a diverse function of this structure may be a good starting point for further basic and clinical research. The NAc is a part of the brain reward system (BRS) characterized by multilevel organization, extensive connections, and several neurotransmitter systems. The unique role of NAc in the BRS is a result of: (1) hierarchical connections with the other brain areas, (2) a well-developed morphological and functional plasticity regulating short- and long-term synaptic potentiation and signalling pathways, (3) cooperation among several neurotransmitter systems, and (4) a supportive role of neuroglia involved in both physiological and pathological processes. Understanding the complex function of NAc is possible by combining the results of morphological studies with molecular, genetic, and behavioral data. In this review, we present the current views on the NAc function in physiological conditions, emphasizing the role of its connections, neuroplasticity processes, and neurotransmitter systems.


Subject(s)
Brain/physiology , Central Nervous System/physiology , Neuronal Plasticity/physiology , Nucleus Accumbens/physiology , Animals , Humans , Personal Satisfaction , Reward
8.
Int J Mol Sci ; 22(13)2021 Jun 27.
Article in English | MEDLINE | ID: mdl-34199004

ABSTRACT

Guanosine (Guo) is a nucleotide metabolite that acts as a potent neuromodulator with neurotrophic and regenerative properties in neurological disorders. Under brain ischemia or trauma, Guo is released to the extracellular milieu and its concentration substantially raises. In vitro studies on brain tissue slices or cell lines subjected to ischemic conditions demonstrated that Guo counteracts destructive events that occur during ischemic conditions, e.g., glutaminergic excitotoxicity, reactive oxygen and nitrogen species production. Moreover, Guo mitigates neuroinflammation and regulates post-translational processing. Guo asserts its neuroprotective effects via interplay with adenosine receptors, potassium channels, and excitatory amino acid transporters. Subsequently, guanosine activates several prosurvival molecular pathways including PI3K/Akt (PI3K) and MEK/ERK. Due to systemic degradation, the half-life of exogenous Guo is relatively low, thus creating difficulty regarding adequate exogenous Guo distribution. Nevertheless, in vivo studies performed on ischemic stroke rodent models provide promising results presenting a sustained decrease in infarct volume, improved neurological outcome, decrease in proinflammatory events, and stimulation of neuroregeneration through the release of neurotrophic factors. In this comprehensive review, we discuss molecular signaling related to Guo protection against brain ischemia. We present recent advances, limitations, and prospects in exogenous guanosine therapy in the context of ischemic stroke.


Subject(s)
Guanosine/pharmacology , Ischemic Stroke/metabolism , Neuroprotective Agents/pharmacology , Animals , Biomarkers , Central Nervous System/drug effects , Central Nervous System/metabolism , Disease Management , Disease Susceptibility , Humans , Ischemic Stroke/drug therapy , Ischemic Stroke/etiology , Ischemic Stroke/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Signal Transduction
9.
Diagnostics (Basel) ; 10(6)2020 May 26.
Article in English | MEDLINE | ID: mdl-32466525

ABSTRACT

BACKGROUND: the goal of the study is to ascertain the influence of discopathy in the lumbosacral (L-S) segment on the gait parameters. The inertial sensors are used to determine the pathologic parameters of gait. METHODS: the study involved four patients (44, 46, 42, and 38 years). First, the goal of the survey was to analyze by a noninvasive medical test magnetic resonance imaging (MRI) of each patient. Next, by using inertial sensors, the flexion-extension of joint angles of the left and right knees were calculated. The statistical analysis was performed. The wavelet transform was applied to analyze periodic information in the acceleration data. RESULTS: in the patients with discopathy, the amount of knee flexion attained during stance phase is significantly lower than that of normal (health side), which could indicate poor eccentric control or a pain avoidance mechanism. The biggest differences are observed in the Initial Swing phase. Bending of the lower limb in the knee joint at this stage reaches maximum values during the entire gait cycle. CONCLUSIONS: It has been difficult to quantify the knee angle during gait by visual inspection. The inertial measurement unit (IMU) system can be useful in determining the level of spine damage and its degree. In patients in the first stages of the intervertebral disc disease who may undergo conservative treatment, it may also partially delay or completely exclude the decision to perform a complicated imaging examination which is MRI, often showing a false positive result in this phase of the disease.

10.
Transl Stroke Res ; 11(4): 553-579, 2020 08.
Article in English | MEDLINE | ID: mdl-31701356

ABSTRACT

Cerebral stroke, which is one of the most frequent causes of mortality and leading cause of disability in developed countries, often leads to devastating and irreversible brain damage. Neurological and neuroradiological diagnosis of stroke, especially in its acute phase, is frequently uncertain or inconclusive. This results in difficulties in identification of patients with poor prognosis or being at high risk for complications. It also makes difficult identification of these stroke patients who could benefit from more aggressive therapies. In contrary to the cardiovascular disease, no single biomarker is available for the ischemic stroke, addressing the abovementioned issues. This justifies the need for identifying of effective diagnostic measures characterized by high specificity and sensitivity. One of the promising avenues in this area is studies on the panels of biomarkers characteristic for processes which occur in different types and phases of ischemic stroke and represent all morphological constituents of the brains' neurovascular unit (NVU). In this review, we present the current state of knowledge concerning already-used or potentially applicable biomarkers of the ischemic stroke. We also discuss the perspectives for identification of biomarkers representative for different types and phases of the ischemic stroke, as well as for different constituents of NVU, which concentration levels correlate with extent of brain damage and patients' neurological status. Finally, a critical analysis of perspectives on further improvement of the ischemic stroke diagnosis is presented.


Subject(s)
Brain Ischemia/diagnosis , Ischemic Stroke/diagnosis , Ischemic Stroke/physiopathology , Neurovascular Coupling , Animals , Biomarkers/metabolism , Brain/blood supply , Brain/metabolism , Brain/physiopathology , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Encephalitis/complications , Encephalitis/metabolism , Humans , Ischemic Stroke/complications , Ischemic Stroke/metabolism , Translational Research, Biomedical
11.
Anaesthesiol Intensive Ther ; 50(2): 95-102, 2018.
Article in English | MEDLINE | ID: mdl-29953572

ABSTRACT

BACKGROUND: In nearly all cases of general anaesthesia with a volatile agent, the anaesthetic concentration has to be increased. Since the anaesthetic affects both the factors determining intracranial homeostasis and the systemic circulation, it is crucial that cerebral perfusion pressure (CPP) is protected. The aim of the present study was to assess the influence of gradually increased concentrations of desflurane on the cerebral and systemic circulations based on CPP, mean arterial pressure (MAP), intracranial pressure (ICP) and their correlations. METHODS: The study was carried out on 25 rabbits of the same gender (male) randomly assigned to two groups: control (n = 10) and group I (n = 15). Over three 15-minute periods, the animals were exposed to increase concentrations of desflurane so as to achieve 1/3, 2/3 and 1 MAC Minimal Alveolar Concentration (3, 6, 9 vol%) of the effective end-tidal concentration of desflurane (Et) at the end of each period, respectively. RESULTS: Intragroup analysis of CPP changes demonstrated decreases in its successive values from minute 18, compared with baseline values. The mean values of ICP did not differ throughout the experiment. From minute 19 on, all successive values of MAP decreased compared with baseline values. A weak correlation (r = -0.2179) was found between ICP and CPP and a strong correlation between MAP and CPP (r = 0.98829). Moreover, there was a strong correlation between Etdesflurane vs. CPP (r = -0.8769) and MAP (r = -0.8224) and a weak correlation versus ICP (r = 0.15755). CONCLUSIONS: A decrease in CPP induced by desflurane was associated with a decrease in MAP but not an increase in ICP. The depressive effect of desflurane on the cerebral and systemic circulations is a consequence of its effector site concentration.


Subject(s)
Anesthetics, Inhalation/pharmacology , Blood Pressure/drug effects , Cerebrovascular Circulation/drug effects , Desflurane/pharmacology , Anesthetics, Inhalation/administration & dosage , Animals , Arterial Pressure/drug effects , Desflurane/administration & dosage , Intracranial Pressure/drug effects , Male , Rabbits
12.
J Chem Neuroanat ; 88: 1-12, 2018 03.
Article in English | MEDLINE | ID: mdl-29100904

ABSTRACT

Coffee and nicotine consumption are frequently combined, indicating possible intensifying effect of caffeine on smoking behavior, although neurobiological background of this phenomenon remains unknown. We aimed at determining the effect of caffeine and nicotine, applied separately or simultaneously, on activation of six structures of the brain reward system: nucleus accumbens (NAc), ventral tegmental area (VTA), amygdala (Amg), hippocampus (Hip), medial prefrontal cortex (mPfr) and dorsal striatum (CdP) in the adult male Wistar rats. Activation of two transcription factors, the phosphorylated form of cyclic AMP-response element binding protein (pCREB) and DeltaFosB (ΔFosB) was assessed by immunohistochemistry after multiple-dose five-days psychostimulants administration followed by 20min and 24h survival, respectively. Nicotine evoked the highest increase of pCREB-immunoreactivity (-ir) in NAc, while caffeine exerted the weakest effect in mPfr and CdP. Nicotine/caffeine co-administration resulted in decrease of pCREB-ir in NAc and increase in Amg, compared with the effect of each psychostimulant used separately. Nicotine was the strongest psychostimulant activating ΔFosB-ir in Amg, whereas caffeine - in Hip. Nicotine/caffeine-exerted effect upon ΔFosB-ir in Amg was weaker, whereas in mPfr stronger, than nicotine-evoked effect in these structures. In summary, pCREB and ΔFosB activation is dependent on the type of stimulus, brain structure and functional context. Activation of both transcription factors is responsible for caffeine's modifying effect upon nicotine-related behaviors and must be taken into account while quitting cigarette smoking.


Subject(s)
Brain/drug effects , Brain/metabolism , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Nicotine/pharmacology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Nicotinic Agonists/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Reward
13.
Cell Mol Neurobiol ; 38(3): 579-593, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28623429

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is one of the most widely distributed and extensively studied neurotrophins in the mammalian brain. Among its prominent functions, one can mention control of neuronal and glial development, neuroprotection, and modulation of both short- and long-lasting synaptic interactions, which are critical for cognition and memory. A wide spectrum of processes are controlled by BDNF, and the sometimes contradictory effects of its action can be explained based on its specific pattern of synthesis, comprising several intermediate biologically active isoforms that bind to different types of receptor, triggering several signaling pathways. The functions of BDNF must be discussed in close relation to the stage of brain development, the different cellular components of nervous tissue, as well as the molecular mechanisms of signal transduction activated under physiological and pathological conditions. In this review, we briefly summarize the current state of knowledge regarding the impact of BDNF on regulation of neurophysiological processes. The importance of BDNF for future studies aimed at disclosing mechanisms of activation of signaling pathways, neuro- and gliogenesis, as well as synaptic plasticity is highlighted.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Neuronal Plasticity/physiology , Signal Transduction/physiology , Synapses/metabolism , Animals , Brain/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Humans , Neuronal Plasticity/drug effects , Receptor, trkB/metabolism , Signal Transduction/drug effects , Synapses/drug effects
14.
Transl Neurosci ; 8: 70-75, 2017.
Article in English | MEDLINE | ID: mdl-28828183

ABSTRACT

Clinicians often rely on selected small molecular compounds from body fluids for the detection, screening or monitoring of numerous life-threatening diseases. Among others, important monoamines - biogenic amines (BAs) - and their metabolites serve as sensitive biomarkers to study the progression or even early detection of on-going brain pathologies or tumors of neuroendocrine origins. Undertaking the task to optimize a reliable method for the simultaneous analysis of the most relevant BAs in biological matrices is of utmost importance for scientists. Hydrophilic interaction liquid chromatography (HILIC) with mass spectrometry (MS) detection provides a specific and sensitive technique for the separation and assessment of several neurotransmitter concentrations in body fluids (blood, urine, tissues). The present study was focused on the optimization of a straightforward, sensitive and reliable method for the simultaneous analysis of the ten most important BAs and their acidic metabolites from homogenates of rat brain tissues by use of HILIC-MS. Here, we present the optimized experimental workflow in terms of sample preparation, buffer compositions, HILIC and MS settings and data analysis. The presented method is reliable, straightforward and sensitive. Our method permits the unbiased, qualitative and quantitative determination of several BAs and their metabolites simultaneously. The optimized method was applied to the analysis of rat brain tissue samples from healthy hemispheres or those with induced transient ischemic attack (TIA). The undertaken pilot study demonstrated that the proposed approach could be applied to reveal the perturbation in neurotransmitters concentration after TIA in rat brains.

15.
Metab Brain Dis ; 32(4): 935-948, 2017 08.
Article in English | MEDLINE | ID: mdl-28432486

ABSTRACT

The brain, demanding constant level of cholesterol, precisely controls its synthesis and homeostasis. The brain cholesterol pool is almost completely separated from the rest of the body by the functional blood-brain barrier (BBB). Only a part of cholesterol pool can be exchanged with the blood circulation in the form of the oxysterol metabolites such, as 27-hydroxycholesterol (27-OHC) and 24S-hydroxycholesterol (24S-OHC). Not only neurons but also blood vessels and neuroglia, constituting neurovascular unit (NVU), are crucial for the brain cholesterol metabolism and undergo precise regulation by numerous modulators, metabolites and signal molecules. In physiological conditions maintaining the optimal cholesterol concentration is important for the energetic metabolism, composition of cell membranes and myelination. However, a growing body of evidence indicates the consequences of the cholesterol homeostasis dysregulation in several pathophysiological processes. There is a causal relationship between hypercholesterolemia and 1) development of type 2 diabetes due to long-term high-fat diet consumption, 2) significance of the oxidative stress consequences for cerebral amyloid angiopathy and neurodegenerative diseases, 3) insulin resistance on progression of the neurodegenerative brain diseases. In this review, we summarize the current state of knowledge concerning the cholesterol influence upon functioning of the NVU under physiological and pathological conditions.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Diseases/metabolism , Cholesterol/metabolism , Nerve Degeneration/metabolism , Animals , Blood-Brain Barrier/pathology , Brain Diseases/pathology , Humans , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology
16.
Naunyn Schmiedebergs Arch Pharmacol ; 388(1): 79-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25344204

ABSTRACT

This study evaluated the therapeutic potential of masitinib, an oral tyrosine kinase inhibitor with activity against c-Kit and platelet-derived growth factor receptors (PDGFR), to reduce ischemic brain area and neurological deficit. Using a well-established filament model of ischemic stroke in rats, the responses to oral treatment with masitinib alone or in combination with recombinant tissue plasminogen activator (rt-PA) were compared to those after rt-PA (10 mg/kg intravenously (i.v.)) monotherapy. In both cases, two doses of masitinib were used--25 or 100 mg/kg, twice per day. Ischemic brain area and the neurological deficit were assessed using the triphenyltetrazolium chloride (TTC) method and behavioral neurological tests, respectively. Masitinib, as a single agent, reduced significantly the infarct size, as compared with the stroke control group. Brain ischemic area decreased from 9.14 to 4.36 % (25 mg/kg) or 2.60 % (100 mg/kg). Moreover, a combined treatment of masitinib with rt-PA produced a stronger effect than the one observed after each of the compound alone. The size of the brain ischemic area (rt-PA 1.67 %) was further reduced to 0.83 or 0.7 % at masitinib doses of 25 and 100 mg/kg, respectively. Masitinib reduced significantly brain ischemia induced by experimental stroke and potentiated the therapeutic effect of rt-PA.


Subject(s)
Brain Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Thiazoles/therapeutic use , Tissue Plasminogen Activator/therapeutic use , Animals , Behavior, Animal/drug effects , Benzamides , Brain/drug effects , Brain/pathology , Brain Ischemia/pathology , Drug Synergism , Drug Therapy, Combination , Hypnotics and Sedatives/therapeutic use , Male , Piperidines , Pyridines , Rats, Wistar , Stroke/pathology
17.
Folia Neuropathol ; 51(3): 203-13, 2013.
Article in English | MEDLINE | ID: mdl-24114637

ABSTRACT

Reactive astrogliosis is regarded as an universal astrocytic response to different kinds of lesions, concerned with glial fibrillary acidic protein (GFAP) up-regulation, cellular hypertrophy and proliferation. The origin of reactive and proliferating cells in the adult brain is still disputable. Persistent progenitors as well as de-differentiating adult cells of various glial lineages are regarded as possible candidates. Pax6 transcription factor is one of the characteristic markers of astroglial de-differentiation, also important for regulation of neural and glial proliferation. Various kinds of pathological stimuli evoke reactive response, differentiated in its morphological, biochemical and immunological character. The aim of this study was to assess the dynamics of astroglial morphological and proliferative response to ischemic injury. One-hour transient focal cerebral ischemia was applied to evoke the reactive astrogliosis in twenty five adult male Wistar rats. The astrocytic morphological and proliferative reactions to ischemia were studied in the period of 6 weeks by means of GFAP and Pax6 immunofluorescent staining. A strong reactive astroglial response was observed in the cerebral cortex and striatum, manifested by GFAP and Pax6 up-regulation and astrocytic hypertrophy. Apparent morphological changes appeared within 24 hrs after ischemia. The GFAP/Pax6 colocalization was numerous and observed 24 hrs after ischemia. A characteristic spatial distribution of GFAP/Pax6 double-labelled astrocytes and Pax6 single-labelled nuclei was revealed, with the latter situated more distantly from the ischemic core. The maximal intensity of astrocytic reaction was present from the first post-ischemic week. Astroglial hypertrophic changes and proliferative reaction were more intense in the striatum than in the cerebral cortex. Our observations reveal intensive astroglial de-differentiation and proliferative response, reflected by dynamic Pax6 up-regulation within GFAP-immunoreactive astrocytes. Transient cerebral ischemia evokes strong reactive astrogliosis, which is apparently differentiated in respect to the post-ischemic period and particular brain structure.


Subject(s)
Astrocytes/metabolism , Eye Proteins/biosynthesis , Homeodomain Proteins/biosynthesis , Ischemic Attack, Transient/metabolism , Paired Box Transcription Factors/biosynthesis , Repressor Proteins/biosynthesis , Animals , Astrocytes/pathology , Disease Models, Animal , Glial Fibrillary Acidic Protein/biosynthesis , Immunohistochemistry , Ischemic Attack, Transient/pathology , Male , Microscopy, Confocal , PAX6 Transcription Factor , Rats , Rats, Wistar
18.
Neurosci Res ; 75(3): 171-83, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23419863

ABSTRACT

Cerebral blood flow adequate for brain activity and metabolic demand is maintained through the processes of autoregulation and neurovascular coupling. Astrocytes undoubtedly make an important contribution to these processes. The critical factors that determine the polarity of astrocytic response include: metabolites (e.g., arachidonic acid and its derivatives, lactate and oxygen concentrations), ions (H(+), Ca(2+) and K(+)), gliotransmitters (glutamate, Glu; gamma-aminobutyric acid, GABA; d-serine; adenosine 5'-triphosphate, ATP and brain derived neurotrophic factor, BDNF), neuronal activity and vascular tone. Although the astrocytic contribution to neurovascular coupling has been intensively studied, a few important questions still remain, such as: (1) the modulatory function of astrocytes in tripartite synapses, including effects related to the strength of synaptic stimulation and the particular signaling pathway (astrocytic or neuronal) that becomes activated, (2) the significance of the vasoconstrictive reaction evoked by arachidonic acid metabolites (e.g., 20-hydroxyeicosatetraenoic acid, 20-HETE) under both physiological and pathological conditions, (3) the relationship between brain activity level and metabolic processes occurring in astrocytes, which is studied using neuroradiological techniques and (4) the astrocytic contribution to the neurovascular response under pathological conditions. Hence, the function of astrocytes in neurovascular coupling remains ambiguous. The function of astrocytes is beneficial and integrative in physiological conditions, but under definitive pathological conditions may become detrimental and involved in the development of diseases like ischemic stroke, arterial hypertension and Alzheimer's disease.


Subject(s)
Astrocytes/physiology , Brain/blood supply , Brain/cytology , Cerebrovascular Circulation/physiology , Animals , Astrocytes/cytology , Brain/metabolism , Humans
20.
Metab Brain Dis ; 24(3): 507-23, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19693659

ABSTRACT

There is large body of evidence suggesting distinct susceptibility to ischemia in various brain regions. However, the reason for this remains unexplained. Comparative studies of programmed cell death (PCD) pathways indicate their differentiated evolutional origin. The caspase-independent pathway is regarded as an older, whereas the caspase-dependent--as more advanced. In our study we address the question of whether there are any characteristic differences in the activation and course of PCD in phylogenetically and morphologically distinguished brain structures after transient focal ischemia. Using Western blot, we studied changes in expression of caspases: 3, 8, 9, and AIF in the frontoparietal neocortex, archicortex (CA1 and CA2 sectors of the hippocampus) and striatum, during reperfusion after 1 h occlusion of the middle cerebral artery. The caspase and AIF expression were differentiated between the studied structures. The activation of only the caspase-dependent pathway was observed in the neocortex. In the archicortex and striatum both caspase-dependent and caspase-independent pathways were activated, although in the latter the extrinsic apoptotic pathway was not activated. In summary, it is conceivable that structures of different evolutionary origin undergo cell-death processes with the participation of phylogenetically distinguished mechanisms. The previously reported unequal susceptibility to ischemia may co-exist with activation of different cell death pathways.


Subject(s)
Brain/pathology , Cell Death/genetics , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/pathology , Animals , Biological Evolution , Blotting, Western , Caspases/metabolism , Cerebrovascular Circulation/physiology , Female , Infarction, Middle Cerebral Artery/pathology , Male , Neurons/pathology , Phylogeny , Rats , Rats, Wistar , Reperfusion Injury/pathology , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...