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










Publication year range
1.
Mol Neurobiol ; 61(1): 239-251, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37603152

ABSTRACT

Bacterial meningitis is considered a life-threatening condition with high mortality rates. In response to the infection, signaling cascades, producing pro-inflammatory mediators trigger an exacerbated host immune response. Another inflammatory pathway occurs through the activation of inflammasomes. Studies highlight the role of the NLR family pyrin domain containing 3 (NLRP3) in central nervous system disorders commonly involved in neuroinflammation. We aimed to investigate the role of NLRP3 and its inhibitor MCC950 on neurochemical, immunological, and behavioral parameters in the early and late stages of experimental pneumococcal meningitis. For this, adult male Wistar rats received an intracisternal injection of Streptococcus pneumoniae or artificial cerebrospinal fluid as a placebo. The animals were divided into control/saline, control/MCC950, meningitis/saline, and meningitis/MCC950. Immediately after the meningitis induction, the animals received 140 ng/kg MCC950 via intracisternal injection. For the acute protocol, 24 h after induction, brain structures were collected to evaluate cytokines, NLRP3, and microglia. In the long-term group, the animals were submitted to open field and recognition of new objects tests at ten days after the meningitis induction. After the behavioral tests, the same markers were evaluated. The animals in the meningitis group at 24 h showed increased levels of cytokines, NLRP3, and IBA-1 expression, and the use of the MCC950 significantly reduced those levels. Although free from infection, ten days after meningitis induction, the animals in the meningitis group had elevated cytokine levels and demonstrated behavioral deficits; however, the single dose of NLRP3 inhibitor rescued the behavior deficits and decreased the brain inflammatory profile.


Subject(s)
Meningitis, Pneumococcal , Animals , Male , Rats , Cytokines/metabolism , Inflammasomes/metabolism , Memory Disorders , Meningitis, Pneumococcal/complications , Meningitis, Pneumococcal/drug therapy , Models, Theoretical , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats, Wistar , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
2.
Pharmacol Biochem Behav ; 220: 173468, 2022 10.
Article in English | MEDLINE | ID: mdl-36174752

ABSTRACT

Major depressive disorder (MDD) is the most prevalent mood disorder globally. Most antidepressants available for the treatment of MDD increase the concentration of monoamines in the synaptic cleft. However, such drugs have a high latency time to obtain benefits. Thus, new antidepressants with fast action and robust efficacy are very important. This study evaluated the effects of escitalopram, ketamine, and probiotic Bifidobacterium infantis in rats submitted to the maternal deprivation (MD). MD rats received saline, escitalopram, ketamine, or probiotic for 10, 30, or 50 days, depending on the postnatal day (PND):21, 41, and 61. Following behavior, this study examined the integrity of the blood-brain barrier (BBB) and oxidative stress markers. MD induced depressive-like behavior in females with PND21 and males with PND61. All treatments reversed depressive-like behavior in females and escitalopram and ketamine in males. MD induced an increase in the permeability of the BBB, an imbalance between oxidative stress and antioxidant defenses. Treatments regulated the oxidative damage and the integrity of the BBB induced by MD. The treatment with escitalopram, ketamine, or probiotics may prevent behavioral and neurochemical changes associated with MDD, depending on the developmental period and gender.


Subject(s)
Antidepressive Agents , Depressive Disorder, Major , Sex Characteristics , Stress, Psychological , Animals , Female , Male , Rats , Antidepressive Agents/therapeutic use , Antioxidants/metabolism , Depressive Disorder, Major/drug therapy , Ketamine , Rats, Wistar , Stress, Psychological/drug therapy , Escitalopram
3.
mBio ; 13(5): e0188622, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36036510

ABSTRACT

Pneumococcal meningitis, inflammation of the meninges due to an infection of the Central Nervous System caused by Streptococcus pneumoniae (the pneumococcus), is the most common form of community-acquired bacterial meningitis globally. Aquaporin 4 (AQP4) water channels on astrocytic end feet regulate the solute transport of the glymphatic system, facilitating the exchange of compounds between the brain parenchyma and the cerebrospinal fluid (CSF), which is important for the clearance of waste away from the brain. Wistar rats, subjected to either pneumococcal meningitis or artificial CSF (sham control), received Evans blue-albumin (EBA) intracisternally. Overall, the meningitis group presented a significant impairment of the glymphatic system by retaining the EBA in the CSF compartments compared to the uninfected sham group. Our results clearly showed that during pneumococcal meningitis, the glymphatic system does not function because of a detachment of the astrocytic end feet from the blood-brain barrier (BBB) vascular endothelium, which leads to misplacement of AQP4 with the consequent loss of the AQP4 water channel's functionality. IMPORTANCE The lack of solute drainage due to a dysfunctional glymphatic system leads to an increase of the neurotoxic bacterial material in the CSF compartments of the brain, ultimately leading to brain-wide neuroinflammation and neuronal damage with consequent impairment of neurological functions. The loss of function of the glymphatic system can therefore be a leading cause of the neurological sequelae developing post-bacterial meningitis.


Subject(s)
Glymphatic System , Meningitis, Pneumococcal , Animals , Rats , Albumins/metabolism , Aquaporin 4/genetics , Aquaporin 4/metabolism , Astrocytes/metabolism , Brain/metabolism , Glymphatic System/metabolism , Meningitis, Pneumococcal/metabolism , Rats, Wistar
4.
Braz J Psychiatry ; 43(6): 650-664, 2021.
Article in English | MEDLINE | ID: mdl-33605367

ABSTRACT

Severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) causes the coronavirus disease 2019 (COVID-19), which has been declared a public health emergency of international interest, with confirmed cases in most countries. COVID-19 presents manifestations that can range from asymptomatic or mild infections up to severe manifestations that lead to hospitalization and death. A growing amount of evidence indicates that the virus may cause neuroinvasion. Postmortem brain study findings have included edema, hemorrhage, hydrocephalus, atrophy, encephalitis, infarcts, swollen axons, myelin loss, gliosis, neuronal satellitosis, hypoxic-ischemic damage, arteriolosclerosis, leptomeningeal inflammation, neuronal loss, and axon degeneration. In addition, the COVID-19 pandemic is causing dangerous effects on the mental health of the world population, some of which can be attributed to its social impact (social distancing, financial issues, and quarantine). There is also a concern that environmental stressors, enhanced by psychological factors, are contributing to the emergence of psychiatric outcomes during the pandemic. Although clinical studies and diagnosing SARS-CoV-2-related neurological disease can be challenging, they are necessary to help define the manifestations and burden of COVID-19 in neurological and psychiatric symptoms during and after the pandemic. This review aims to present the neurobiology of coronavirus and postmortem neuropathological hallmarks.


Subject(s)
COVID-19 , Brain , Humans , Pandemics , Quarantine , SARS-CoV-2
5.
Tissue Barriers ; 9(1): 1840912, 2021 01 02.
Article in English | MEDLINE | ID: mdl-33319634

ABSTRACT

Sepsis is a life-threatening organ dysfunction triggered by a dysregulated host immune response attempting to eliminate the infection. After hospital discharge, half of the sepsis survivors recover, one-third of the patients die the following year, and one-sixth have a long-term cognitive impairment, including memory dysfunction, anxiety, depression, and post-traumatic stress disorder. The infection triggers the host immune response, and both can cause vascular endothelial damage, interrupting tight junctions proteins; consequently, the blood-brain barrier (BBB) breaks down, allowing and facilitating the entry of peripheral immune cells into the brain, which triggers or exacerbates the activation of glial cells and neuroinflammation. The focus of this review is to identify biochemical abnormalities induced by sepsis, which is associated with BBB dysfunction; provide evidence of biomarkers involved in the tight junction disruption and BBB damage, and draw attention to the role of the BBB as a bridge between systemic infection and brain inflammation.


Subject(s)
Blood-Brain Barrier/physiopathology , Sepsis/physiopathology , Humans
6.
Neurotherapeutics ; 18(1): 640-653, 2021 01.
Article in English | MEDLINE | ID: mdl-32886341

ABSTRACT

Pneumococcal meningitis is a life-threatening infection of the central nervous system (CNS), and half of the survivors of meningitis suffer from neurological sequelae. We hypothesized that pneumococcal meningitis causes CNS inflammation via the disruption of the blood-brain barrier (BBB) and by increasing the receptor for advanced glycation end product (RAGE) expression in the brain, which causes glial cell activation, leading to cognitive impairment. To test our hypothesis, 60-day-old Wistar rats were subjected to meningitis by receiving an intracisternal injection of Streptococcus pneumoniae or artificial cerebrospinal fluid as a control group and were treated with a RAGE-specific inhibitor (FPS-ZM1) in saline. The rats also received ceftriaxone 100 mg/kg intraperitoneally, bid, and fluid replacements. Experimental pneumococcal meningitis triggered BBB disruption after meningitis induction, and FPS-ZM1 treatment significantly suppressed BBB disruption. Ten days after meningitis induction, surviving animals were free from infection, but they presented increased levels of TNF-α and IL-1ß in the prefrontal cortex (PFC); high expression levels of RAGE, amyloid-ß (Aß1-42), and microglial cell activation in the PFC and hippocampus; and memory impairment, as evaluated by the open-field, novel object recognition task and Morris water maze behavioral tasks. Targeted RAGE inhibition was able to reduce cytokine levels, decrease the expression of RAGE and Aß1-42, inhibit microglial cell activation, and improve cognitive deficits in meningitis survivor rats. The sequence of events generated by pneumococcal meningitis can persist long after recovery, triggering neurocognitive decline; however, RAGE blocker attenuated the development of brain inflammation and cognitive impairment in experimental meningitis.


Subject(s)
Cognitive Dysfunction/etiology , Meningitis, Pneumococcal/complications , Receptor for Advanced Glycation End Products/metabolism , Animals , Benzamides/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blotting, Western , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Disease Models, Animal , Interleukin-1beta/metabolism , Male , Meningitis, Pneumococcal/drug therapy , Morris Water Maze Test/drug effects , Neuroprotective Agents/pharmacology , Open Field Test/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
7.
Pharmacol Res ; 164: 105314, 2021 02.
Article in English | MEDLINE | ID: mdl-33246175

ABSTRACT

The gut microbiota is a complex ecosystem that comprises of more than 100 trillion symbiotic microbial cells. The microbiota, the gut, and the brain form an association, 'the microbiota-gut-brain axis,' and synchronize the gut with the central nervous system and modify the behavior and brain immune homeostasis. The bidirectional communication between gut and brain occurs via the immune system, the vagus nerve, the enteric nervous system, and microbial metabolites, including short-chain fatty acids (SCFAs), proteins, and tryptophan metabolites. Recent studies have implicated the gut microbiota in many neurodegenerative diseases, including Alzheimer's disease (AD). In this review, we present an overview of gut microbiota, including Firmicutes, Bacteroidetes, SCFA, tryptophan, bacterial composition, besides age-related changes in gut microbiota composition, the microbiota-gut-brain axis pathways, the role of gut metabolites in amyloid-beta clearance, and gut microbiota modulation from experimental and clinical AD models. Understanding the role of the microbiota may provide new targets for treatment to delay the onset, progression, or reverse AD, and may help in reducing the prevalence of AD.


Subject(s)
Alzheimer Disease/microbiology , Brain , Gastrointestinal Microbiome , Animals , Humans
8.
J Neuroinflammation ; 17(1): 5, 2020 Jan 04.
Article in English | MEDLINE | ID: mdl-31901235

ABSTRACT

BACKGROUND: Bacterial meningitis is a devastating central nervous system (CNS) infection with acute and long-term neurological consequences, including cognitive impairment. The aim of this study was to understand the association between activated microglia-induced neuroinflammation and post-meningitis cognitive impairment. METHOD: Meningitis was induced in male Wistar rats by injecting Streptococcus pneumoniae into the brain through the cisterna magna, and rats were then treated with ceftriaxone. Twenty-four hours and 10 days after meningitis induction, rats were imaged with positron emission tomography (PET) using [11C]PBR28, a specific translocator protein (TSPO) radiotracer, to determine in vivo microglial activation. Following imaging, the expression of TSPO, cardiolipin, and cytochrome c, inflammatory mediators, oxidative stress markers, and glial activation markers were evaluated in the prefrontal cortex and hippocampus. Ten days after meningitis induction, animals were subjected to behavioral tests, such as the open-field, step-down inhibitory avoidance, and novel object recognition tests. RESULTS: Both 24-h (acute) and 10-day (long-term) groups of rats demonstrated increased [11C]PBR28 uptake and microglial activation in the whole brain compared to levels in the control group. Although free from infection, 10-day group rats exhibited increased expression levels of cytokines and markers of oxidative stress, microglial activation (IBA-1), and astrocyte activation (GFAP) similar to those seen in the 24-h group. Acute meningitis induction also elevated TSPO, cytochrome c, and caspase-3 levels with no change in caspase-9 levels. Furthermore, upregulated levels of TSPO, cytochrome c, and caspase-3 and caspase-9 were observed in the rat hippocampus 10 days after meningitis induction with a simultaneous reduction in cardiolipin levels. Animals showed a cognitive decline in all tasks compared with the control group, and this impairment may be at least partially mediated by activating a glia-mediated immune response and upregulating TSPO. CONCLUSIONS: TSPO-PET could potentially be used as an imaging biomarker for microglial activation and long-term cognitive impairment post-meningitis. Additionally, this study opens a new avenue for the potential use of TSPO ligands after infection-induced neurological sequelae.


Subject(s)
Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/metabolism , Inflammation Mediators/metabolism , Meningitis/diagnostic imaging , Meningitis/metabolism , Positron-Emission Tomography/methods , Animals , Avoidance Learning/physiology , Cognitive Dysfunction/microbiology , Male , Meningitis/microbiology , Rats , Rats, Wistar , Streptococcus pneumoniae
9.
Mediators Inflamm ; 2017: 6490652, 2017.
Article in English | MEDLINE | ID: mdl-29200666

ABSTRACT

The aim of this study was to investigate the effects of lithium on brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and glial cell line-derived neurotrophic factor (GDNF) expression in the hippocampus and on memory in experimental pneumococcal meningitis. The mood-stabilizer lithium is known as a neuroprotective agent with many effects on the brain. In this study, animals received either artificial cerebrospinal fluid or Streptococcus pneumoniae suspension at a concentration of 5 × 109 CFU/mL. Eighteen hours after induction, all animals received ceftriaxone. The animals received saline or lithium (47.5 mg/kg) or tamoxifen (1 mg/kg) as adjuvant treatment, and they were separated into six groups: control/saline, control/lithium, control/tamoxifen, meningitis/saline, meningitis/lithium, and meningitis/tamoxifen. Ten days after meningitis induction, animals were subjected to open-field habituation and the step-down inhibitory avoidance tasks. Immediately after these tasks, the animals were killed and their hippocampus was removed to evaluate the expression of BDNF, NGF, and GDNF. In the meningitis group, treatment with lithium and tamoxifen resulted in improvement in memory. Meningitis group showed decreased expression of BDNF and GDNF in the hippocampus while lithium reestablished the neurotrophin expression. Lithium was able to prevent memory impairment and reestablishes hippocampal neurotrophin expression in experimental pneumococcal meningitis.


Subject(s)
Hippocampus/metabolism , Lithium/therapeutic use , Memory Disorders/metabolism , Memory Disorders/prevention & control , Meningitis, Pneumococcal/metabolism , Neuroprotective Agents/therapeutic use , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hippocampus/drug effects , Male , Rats , Rats, Wistar
10.
Neurosci Biobehav Rev ; 83: 183-199, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29054730

ABSTRACT

Translocator protein (TSPO) is an 18kDa translocator membrane protein expressed in the outer mitochondrial membrane of steroid-synthesizing cells in the central and peripheral nervous systems. TSPO is involved in cellular functions, including the regulation of cell proliferation, transport of cholesterol to the inner mitochondrial membranes of glial cells, regulation of mitochondrial quality control, and haem synthesis. In the brain, TSPO has been extensively used as a biomarker of injury and inflammation. Indeed, TSPO was up-regulated in several inflammatory and neurodegenerative diseases. In contrast, the expression of TSPO was decreased in peripheral blood from psychiatric patients. Since TSPO is involved in several mechanisms related to mitochondrial function and inflammatory alterations, therapeutic approaches focusing on the regulation of TSPO may provide a new avenue for the treatment of neuropsychiatric disorders. Based on the involvement of mitochondrial alterations in the neurobiology of neuropsychiatric disorders, this review will focus on the functions and physiological roles of TSPO and the potential of TSPO ligands as therapeutic strategies for the treatment of neuropsychiatric disorders.


Subject(s)
Mental Disorders/metabolism , Receptors, GABA/metabolism , Animals , Humans , Ligands , Mental Disorders/therapy , Up-Regulation
11.
Neurosci Lett ; 653: 288-295, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28583583

ABSTRACT

Despite advances in antimicrobial therapy and advanced critical care neonatal bacterial meningitis has a mortality rate of over 10% and induces neurological sequelae in 20-50% of cases. Escherichia coli K1 (E. coli K1) is the most common gram-negative organism causing neonatal meningitis and is the second most common cause behind group B streptococcus. We previously reported that an E. coli K1 experimental meningitis infection in neonatal rats resulted in habituation and aversive memory impairment and a significant increase in cytokine levels in adulthood. In this present study, we investigated the oxidative stress profile including malondialdehyde (MDA) levels, carbonyl protein formation, myeloperoxidase activity (MPO) activity, superoxide dismutase (SOD) activity and catalase (CAT) activity 6, 12, 24, 48, 72 and 96h after E. coli K1 experimental meningitis infection. In addition, sulfhydryl groups, nitrite and nitrate levels and activity of the mitochondrial respiratory chain enzymes were also measured in the frontal cortex and hippocampus of neonatal rats. The results from this study demonstrated a significant increase in MDA, protein carbonyls and MPO activity and a simultaneous decrease in SOD activity in the hippocampus of the neonatal meningitis survivors but the same was not observed in frontal cortex. In addition, we also observed a significant increase in complex IV activity in the hippocampus and frontal cortex of meningitis survivor rats. Thus, the results from this study reaffirmed the possible role of oxidative stress, nitric oxide and its related compounds in the complex pathophysiology of E. coli K1-induced bacterial meningitis.


Subject(s)
Escherichia coli/pathogenicity , Frontal Lobe/metabolism , Hippocampus/metabolism , Meningitis, Escherichia coli/metabolism , Oxidative Stress/physiology , Animals , Animals, Newborn , Disease Models, Animal , Frontal Lobe/enzymology , Hippocampus/enzymology , Male , Malondialdehyde/metabolism , Meningitis, Escherichia coli/enzymology , Peroxidase/metabolism , Rats , Rats, Wistar
12.
Aging Dis ; 6(5): 342-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26425389

ABSTRACT

The central nervous system (CNS) is protected by a complex blood-brain barrier system; however, a broad diversity of virus, bacteria, fungi, and protozoa can gain access and cause illness. As pathogens replicate, they release molecules that can be recognized by innate immune cells. These molecules are pathogen-associated molecular patterns (PAMP) and they are identified by pattern-recognition receptors (PRR) expressed on antigen-presenting cells. Examples of PRR include toll-like receptors (TLR), receptors for advanced glycation endproducts (RAGE), nucleotide binding oligomerisation domain (NOD)-like receptors (NLR), c-type lectin receptors (CLR), RIG-I-like receptors (RLR), and intra-cytosolic DNA sensors. The reciprocal action between PAMP and PRR triggers the release of inflammatory mediators that regulate the elimination of invasive pathogens. Damage-associated molecular patterns (DAMP) are endogenous constituents released from damaged cells that also have the ability to activate the innate immune response. An increase of RAGE expression levels on neurons, astrocytes, microglia, and endothelial cells could be responsible for the accumulation of αß-amyloid in dementia and related to the chronic inflammatory state that is found in neurodegenerative disorders.

13.
J Neuroimmunol ; 278: 262-70, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25468775

ABSTRACT

Pneumococcal meningitis is a severe infectious disease of the central nervous system (CNS) and a significant cause of morbidity and mortality worldwide. The inflammatory reaction to the disease contributes to neuronal injury and involves the meninges, the subarachnoid space and the brain parenchymal vessels. Bacterial pathogens may reach the blood-brain barrier and be recognized by antigen-presenting cells through the binding of Toll-like receptors, triggering an inflammatory cascade. This in turn produces cytokines and chemokines, increases adhesion molecule expression and attracts leukocytes from the blood. This cascade leads to lipid peroxidation, mitochondrial damage and blood-brain barrier permeability. In spite of effective antibacterial treatments, approximately one third of survivors suffer from long-term sequelae, such as hearing loss, cerebral palsy, seizures, hydrocephaly or cognitive impairment. This review summarizes the information on targets of adjuvant treatments of acute pneumococcal meningitis.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Brain/metabolism , Meningitis, Pneumococcal/metabolism , Meningitis, Pneumococcal/therapy , Streptococcus pneumoniae/pathogenicity , Anti-Bacterial Agents/pharmacology , Brain/drug effects , Brain/microbiology , Humans , Meningitis, Pneumococcal/complications , Meningitis, Pneumococcal/diagnosis
14.
Brain Res Bull ; 92: 56-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22683802

ABSTRACT

Streptococcus agalactiae (GBS) is a major cause of severe morbidity and mortality in neonates and young infants, causing sepsis, pneumonia and meningitis. The survivors from this meningitis can suffer serious long-term neurological consequences, such as, seizures, hearing loss, learning and memory impairments. Neurotrophins, such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) control the neuronal cell death during the brain development and play an important role in neuronal differentiation, survival and growth of neurons. Neonate Wistar rats, received either 10µL of sterile saline as a placebo or an equivalent volume of GBS suspension at a concentration of 1×10(6)cfu/mL. Sixty days after induction of meningitis, the animals underwent behavioral tests, after were killed and the hippocampus and cortex were retired for analyze of the BDNF and NGF levels. In the open-field demonstrated no difference in motor, exploratory activity and habituation memory between the groups. The step-down inhibitory avoidance, when we evaluated the long-term memory at 24h after training session, we found that the meningitis group had a decrease in aversive memory when compared with the long-term memory test of the sham group. BDNF levels decreased in hippocampus and cortex; however the NGF levels decreased only in hippocampus. These findings suggest that the meningitis model could be a good research tool for the study of the biological mechanisms involved in the behavioral alterations secondary to GBS meningitis.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Memory Disorders/etiology , Meningitis/complications , Meningitis/metabolism , Meningitis/mortality , Nerve Growth Factor/metabolism , Animals , Animals, Newborn , Avoidance Learning , Brain/metabolism , Brain/microbiology , Brain/pathology , Disease Models, Animal , Exploratory Behavior , Gene Expression Regulation , Inhibition, Psychological , Male , Memory Disorders/microbiology , Meningitis/etiology , Rats , Rats, Wistar , Reaction Time , Statistics, Nonparametric , Streptococcal Infections/complications
15.
Arq Neuropsiquiatr ; 70(5): 366-72, 2012 May.
Article in English | MEDLINE | ID: mdl-22618789

ABSTRACT

Pneumococcal meningitis is a life-threatening disease characterized by an acute purulent infection affecting piamater, arachnoid and the subarachnoid space. The intense inflammatory host's response is potentially fatal and contributes to the neurological sequelae. Streptococcus pneumoniae colonizes the nasopharynx, followed by bacteremia, microbial invasion and blood-brain barrier traversal. S. pneumoniae is recognized by antigen-presenting cells through the binding of Toll-like receptors inducing the activation of factor nuclear kappa B or mitogen-activated protein kinase pathways and subsequent up-regulation of lymphocyte populations and expression of numerous proteins involved in inflammation and immune response. Many brain cells can produce cytokines, chemokines and others pro-inflammatory molecules in response to bacteria stimuli, as consequence, polymorphonuclear are attracted, activated and released in large amounts of superoxide anion and nitric oxide, leading to the peroxynitrite formation, generating oxidative stress. This cascade leads to lipid peroxidation, mitochondrial damage, blood-brain barrier breakdown contributing to cell injury during pneumococcal meningitis.


Subject(s)
Meningitis, Pneumococcal , Acute Disease , Adrenal Cortex Hormones/therapeutic use , Cytokines/metabolism , Humans , Matrix Metalloproteinases/metabolism , Meningitis, Pneumococcal/drug therapy , Meningitis, Pneumococcal/immunology , Meningitis, Pneumococcal/microbiology , Meningitis, Pneumococcal/physiopathology , Nose/microbiology , Oxidative Stress/physiology , Streptococcus pneumoniae
16.
Arq. neuropsiquiatr ; 70(5): 366-372, May 2012. ilus, tab
Article in English | LILACS | ID: lil-622576

ABSTRACT

Pneumococcal meningitis is a life-threatening disease characterized by an acute purulent infection affecting piamater, arachnoid and the subarachnoid space. The intense inflammatory host's response is potentially fatal and contributes to the neurological sequelae. Streptococcus pneumoniae colonizes the nasopharynx, followed by bacteremia, microbial invasion and blood-brain barrier traversal. S. pneumoniae is recognized by antigen-presenting cells through the binding of Toll-like receptors inducing the activation of factor nuclear kappa B or mitogen-activated protein kinase pathways and subsequent up-regulation of lymphocyte populations and expression of numerous proteins involved in inflammation and immune response. Many brain cells can produce cytokines, chemokines and others pro-inflammatory molecules in response to bacteria stimuli, as consequence, polymorphonuclear are attracted, activated and released in large amounts of superoxide anion and nitric oxide, leading to the peroxynitrite formation, generating oxidative stress. This cascade leads to lipid peroxidation, mitochondrial damage, blood-brain barrier breakdown contributing to cell injury during pneumococcal meningitis.


A meningite pneumocócica é doença potencialmente fatal caracterizada por infecção aguda purulenta que afeta a pia-máter, a aracnoide e o espaço subaracnoide. A resposta inflamatória do hospedeiro é potencialmente fatal e contribui para as sequelas neurológicas. O processo inicia-se com a colonização da nasofaringe pelo Streptococcus pneumoniae, seguida de invasão, bacteremia e passagem através da barreira hematoencefálica. O S. pneumoniae é reconhecido por células apresentadoras de antígenos através da ligação aos receptores Toll-like. Isto induz a ativação do fator nuclear kappa B ou proteína quinase ativada por mitógenos. Muitas células cerebrais também podem produzir citocinas, quimiocinas e outras moléculas pró-inflamatórias em resposta aos estímulos bacterianos. Como consequência, são atraídos polimorfonucleares, ocorrendo a liberação de grandes quantidades de ânion superóxido e óxido nítrico, o que leva à formação de peroxinitrito e ocasiona o estresse oxidativo. Esta cascata pró-inflamatória leva à peroxidação lipídica, a danos mitocondriais e à ruptura da barreira hematoencefálica, contribuindo para o dano celular em meningite pneumocócica.


Subject(s)
Humans , Meningitis, Pneumococcal , Acute Disease , Adrenal Cortex Hormones/therapeutic use , Cytokines/metabolism , Matrix Metalloproteinases/metabolism , Meningitis, Pneumococcal/drug therapy , Meningitis, Pneumococcal/immunology , Meningitis, Pneumococcal/microbiology , Meningitis, Pneumococcal/physiopathology , Nose/microbiology , Oxidative Stress/physiology , Streptococcus pneumoniae
17.
Microbiol Immunol ; 56(3): 191-4, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22188584

ABSTRACT

Pneumococcal meningitis is a life-threatening disease characterized by acute purulent infection of the meninges causing neuronal injury, cortical necrosis and hippocampal apoptosis. Cholinergic neurons and their projections are extensively distributed throughout the central nervous system. The aim of this study was to assess acetylcholinesterase activity in the rat brain after pneumococcal meningitis. In the hippocampus, frontal cortex and cerebrospinal fluid, acetylcholinesterase activity was found to be increased at 6, 12, 24, 48 and 96 hr without antibiotic treatment, and at 48 and 96 hr with antibiotic treatment. Our data suggest that acetylcholinesterase activity could be related to neuronal damage induced by pneumococcal meningitis.


Subject(s)
Acetylcholinesterase/biosynthesis , Brain/enzymology , Brain/microbiology , Meningitis, Pneumococcal/pathology , Animals , Brain/pathology , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebrospinal Fluid/enzymology , Disease Models, Animal , GPI-Linked Proteins/biosynthesis , Hippocampus/enzymology , Hippocampus/pathology , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...