Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Cell Mol Neurobiol ; 43(5): 2203-2217, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36227397

RESUMO

Shiga toxin 2 (Stx2) from enterohemorrhagic Escherichia coli (EHEC) produces hemorrhagic colitis, hemolytic uremic syndrome (HUS), and acute encephalopathy. The mortality rate in HUS increases significantly when the central nervous system (CNS) is involved. Besides, EHEC also releases lipopolysaccharide (LPS). Many reports have described cognitive dysfunctions in HUS patients, the hippocampus being one of the brain areas targeted by EHEC infection. In this context, a translational murine model of encephalopathy was employed to establish the deleterious effects of Stx2 and the contribution of LPS in the hippocampus. The purpose of this work is to elucidate the signaling pathways that may activate the inflammatory processes triggered by Stx2, which produces cognitive alterations at the level of the hippocampus. Results demonstrate that Stx2 produced depression-like behavior, pro-inflammatory cytokine release, and NF-kB activation independent of the ERK1/2 signaling pathway, while co-administration of Stx2 and LPS reduced memory index. On the other hand, LPS activated NF-kB dependent on ERK1/2 signaling pathway. Cotreatment of Stx2 with LPS aggravated the pathologic state, while dexamethasone treatment succeeded in preventing behavioral alterations. Our present work suggests that the use of drugs such as corticosteroids or NF-kB signaling inhibitors may serve as neuroprotectors from EHEC infection.


Assuntos
Encefalopatias , Disfunção Cognitiva , Escherichia coli Êntero-Hemorrágica , Infecções por Escherichia coli , Síndrome Hemolítico-Urêmica , Camundongos , Humanos , Animais , Toxina Shiga II/toxicidade , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases , NF-kappa B , Encéfalo/patologia , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/patologia , Hipocampo/patologia , Cognição
2.
J Biomed Sci ; 26(1): 16, 2019 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-30732602

RESUMO

BACKGROUND: Shiga toxin 2 from enterohemorrhagic Escherichia coli is the etiologic agent of bloody diarrhea, hemolytic uremic syndrome and derived encephalopathies that may result to death in patients. Being a Gram negative bacterium, lipopolysaccharide is also released. Particularly, the hippocampus has been found affected in patients intoxicated with Shiga toxin 2. In the current work, the deleterious effects of Shiga toxin 2 and lipopolysaccharide are investigated in detail in hippocampal cells for the first time in a translational murine model, providing conclusive evidences on how these toxins may damage in the observed clinic cases. METHODS: Male NIH mice (25 g) were injected intravenously with saline solution, lipopolysaccharide, Shiga toxin 2 or a combination of Shiga toxin 2 with lipopolysaccharide. Brain water content assay was made to determine brain edema. Another set of animals were intracardially perfused with a fixative solution and their brains were subjected to immunofluorescence with lectins to determine the microvasculature profile, and anti-GFAP, anti-NeuN, anti-MBP and anti-Iba1 to study reactive astrocytes, neuronal damage, myelin dysarrangements and microglial state respectively. Finally, the Thiobarbituric Acid Reactive Substances Assay was made to determine lipid peroxidation. In all assays, statistical significance was performed using the One-way analysis of variance followed by Bonferroni post hoc test. RESULTS: Systemic sublethal administration of Shiga toxin 2 increased the expressions of astrocytic GFAP and microglial Iba1, and decreased the expressions of endothelial glycocalyx, NeuN neurons from CA1 pyramidal layer and oligodendrocytic MBP myelin sheath from the fimbria of the hippocampus. In addition, increased interstitial fluids and Thiobarbituric Acid Reactive Substances-derived lipid peroxidation were also found. The observed outcomes were enhanced when sublethal administration of Shiga toxin 2 was co-administered together with lipopolysaccharide. CONCLUSION: Systemic sublethal administration of Shiga toxin 2 produced a deterioration of the cells that integrate the vascular unit displaying astrocytic and microglial reactive profiles, while edema and lipid peroxidation were also observed. The contribution of lipopolysaccharide to pathogenicity caused by Shiga toxin 2 resulted to enhance the observed hippocampal damage.


Assuntos
Edema/fisiopatologia , Escherichia coli Êntero-Hemorrágica/fisiologia , Hipocampo/fisiopatologia , Peroxidação de Lipídeos , Lipopolissacarídeos/efeitos adversos , Toxina Shiga II/efeitos adversos , Animais , Edema/microbiologia , Hipocampo/efeitos dos fármacos , Hipocampo/microbiologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Neuroglia/efeitos dos fármacos , Neuroglia/microbiologia , Neuroglia/fisiologia
3.
Int J Med Microbiol ; 308(8): 1036-1042, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30314914

RESUMO

Infections by Enterohemorrhagic Escherichia coli may cause in addition to hemolytic uremic syndrome neurological disorders which may lead to fatal outcomes in patients. The brain striatum is usually affected during this outcome. The aim of this study was to determine in this area the role of the microglia in pro-inflammatory events that may occur during Shiga toxin 2 intoxication and consequently to this, whether oligodendrocytes were being affected. In the present paper we demonstrated that anti-inflammatory treatments reduced deleterious effects in brain striatal cells exposed to Shiga toxin 2 and LPS. While dexamethasone treatment decreased microglial activation and recovered myelin integrity in the mice striatum, etanercept treatment decreased neuronal uptake of Stx2 in rat striatal neurons, improving the affected area from toxin-derived injury. In conclusion, microglial activation is related to pro-inflammatory events that may deteriorate the brain function during intoxication with Stx2 and LPS. Consequently, the role of anti-inflammatory agents in the treatment of EHEC-derived encephalopathy should be studied in clinical trials.


Assuntos
Anti-Inflamatórios/administração & dosagem , Encefalopatias/tratamento farmacológico , Escherichia coli Êntero-Hemorrágica/patogenicidade , Infecções por Escherichia coli/tratamento farmacológico , Microglia/efeitos dos fármacos , Toxina Shiga II/toxicidade , Animais , Encefalopatias/microbiologia , Dexametasona/administração & dosagem , Dexametasona/farmacologia , Infecções por Escherichia coli/microbiologia , Etanercepte/administração & dosagem , Etanercepte/farmacologia , Humanos , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Microglia/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia , Ratos , Ratos Sprague-Dawley
4.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1173-R1185, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27681328

RESUMO

Shiga toxin 2 (Stx2)-producing enterohemorrhagic induced brain damage. Since a cerebroprotective action was reported for angiotensin (Ang)-(1-7), our aim was to investigate whether Ang-(1-7) protects from brain damage induced by Stx2-producing enterohemorrhagic Escherichia coli The anterior hypothalamic area of adult male Wistar rats was injected with saline solution or Stx2 or Stx2 plus Ang-(1-7) or Stx2 plus Ang-(1-7) plus A779. Rats received a single injection of Stx2 at the beginning of the experiment, and Ang-(1-7), A779, or saline was administered daily in a single injection for 8 days. Cellular ultrastructural changes were analyzed by transmission electron microscopy. Stx2 induced neurodegeneration, axonal demyelination, alterations in synapse, and oligodendrocyte and astrocyte damage, accompanied by edema. Ang-(1-7) prevented neuronal damage triggered by the toxin in 55.6 ± 9.5% of the neurons and the Stx2-induced synapse dysfunction was reversed. In addition, Ang-(1-7) blocked Stx2-induced demyelination in 92 ± 4% of the axons. Oligodendrocyte damage caused by Stx2 was prevented by Ang-(1-7) but astrocytes were only partially protected by the peptide (38 ± 5% of astrocytes were preserved). Ang-(1-7) treatment resulted in 50% reduction in the number of activated microglial cells induced by Stx2, suggesting an anti-inflammatory action. All these beneficial effects elicited by Ang-(1-7) were blocked by the Mas receptor antagonist and thus it was concluded that Ang-(1-7) protects mainly neurons and oligodendrocytes, and partially astrocytes, in the central nervous system through Mas receptor stimulation.


Assuntos
Angiotensina I/administração & dosagem , Infecções por Escherichia coli/prevenção & controle , Hipotálamo/patologia , Encefalite Infecciosa/induzido quimicamente , Encefalite Infecciosa/prevenção & controle , Fragmentos de Peptídeos/administração & dosagem , Toxina Shiga II/toxicidade , Animais , Infecções por Escherichia coli/induzido quimicamente , Infecções por Escherichia coli/patologia , Hipotálamo/efeitos dos fármacos , Encefalite Infecciosa/patologia , Masculino , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Wistar , Escherichia coli Shiga Toxigênica/metabolismo , Resultado do Tratamento
5.
Infect Immun ; 82(9): 3948-57, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25001607

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) is a food-borne pathogen that produces Shiga toxin (Stx) and causes hemorrhagic colitis. Under some circumstances, Stx produced within the intestinal tract enters the bloodstream, leading to systemic complications that may cause the potentially fatal hemolytic-uremic syndrome. Although retinoids like vitamin A (VA) and retinoic acid (RA) are beneficial to gut integrity and the immune system, the effect of VA supplementation on gastrointestinal infections of different etiologies has been controversial. Thus, the aim of this work was to study the influence of different VA status on the outcome of an EHEC intestinal infection in mice. We report that VA deficiency worsened the intestinal damage during EHEC infection but simultaneously improved survival. Since death is associated mainly with Stx toxicity, Stx was intravenously inoculated to analyze whether retinoid levels affect Stx susceptibility. Interestingly, while VA-deficient (VA-D) mice were resistant to a lethal dose of Stx2, RA-supplemented mice were more susceptible to it. Given that peripheral blood polymorphonuclear cells (PMNs) are known to potentiate Stx2 toxicity, we studied the influence of retinoid levels on the absolute number and function of PMNs. We found that VA-D mice had decreased PMN numbers and a diminished capacity to produce reactive oxygen species, while RA supplementation had the opposite effect. These results are in line with the well-known function of retinoids in maintaining the homeostasis of the gut but support the idea that they have a proinflammatory effect by acting, in part, on the PMN population.


Assuntos
Escherichia coli Êntero-Hemorrágica/metabolismo , Infecções por Escherichia coli/metabolismo , Retinoides/metabolismo , Toxina Shiga II/metabolismo , Animais , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Espécies Reativas de Oxigênio/metabolismo , Vitamina A/metabolismo
6.
Clin Sci (Lond) ; 127(5): 295-306, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24827941

RESUMO

The RAS (renin-angiotensin system) is composed of two arms: the pressor arm containing AngII (angiotensin II)/ACE (angiotensin-converting enzyme)/AT1Rs (AngII type 1 receptors), and the depressor arm represented by Ang-(1-7) [angiotensin-(1-7)]/ACE2/Mas receptors. All of the components of the RAS are present in the brain. Within the brain, Ang-(1-7) contributes to the regulation of BP (blood pressure) by acting at regions that control cardiovascular function such that, when Ang-(1-7) is injected into the nucleus of the solitary tract, caudal ventrolateral medulla, paraventricular nucleus or anterior hypothalamic area, a reduction in BP occurs; however, when injected into the rostral ventrolateral medulla, Ang-(1-7) stimulates an increase in BP. In contrast with AngII, Ang-(1-7) improves baroreflex sensitivity and has an inhibitory neuromodulatory role in hypothalamic noradrenergic neurotransmission. Ang-(1-7) not only exerts effects related to BP regulation, but also acts as a cerebroprotective component of the RAS by reducing cerebral infarct size and neuronal apoptosis. In the present review, we provide an overview of effects elicited by Ang-(1-7) in the brain, which suggest a potential role for Ang-(1-7) in controlling the central development of hypertension.


Assuntos
Encéfalo/fisiologia , Sistema Renina-Angiotensina/fisiologia , Angiotensina I/metabolismo , Angiotensina II/fisiologia , Enzima de Conversão de Angiotensina 2 , Animais , Barorreflexo/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/metabolismo , Neurotransmissores/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptidil Dipeptidase A/fisiologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/fisiologia , Ratos , Receptor Tipo 1 de Angiotensina/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia
7.
Clin Sci (Lond) ; 125(2): 57-65, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23530669

RESUMO

Ang-(1-7) [angiotensin-(1-7)] constitutes an important functional end-product of the RAS (renin-angiotensin system) endogenously formed from AngI (angiotensin I) or AngII (angiotensin II) through the catalytic activity of ACE2 (angiotensin-converting enzyme 2), prolyl carboxypeptidase, neutral endopeptidase or other endopeptidases. Ang-(1-7) lacks the pressor, dipsogenic or stimulatory effect on aldosterone release characteristic of AngII. In contrast, it produces vasodilation, natriuresis and diuresis, and inhibits angiogenesis and cell growth. At the central level, Ang-(1-7) acts at sites involved in the control of cardiovascular function, thus contributing to blood pressure regulation. This action may result from its inhibitory neuromodulatory action on NE [noradrenaline (norepinephrine)] levels at the synaptic cleft, i.e. Ang-(1-7) reduces NE release and synthesis, whereas it causes an increase in NE transporter expression, contributing in this way to central NE neuromodulation. Thus, by selective neurotransmitter release, Ang-(1-7) may contribute to the overall central cardiovascular effects. In the present review, we summarize the central effects of Ang-(1-7) and the mechanism by which the peptide modulates NE levels in the synaptic cleft. We also provide new evidences of its cerebroprotective role.


Assuntos
Angiotensina I/metabolismo , Sistema Nervoso Central/metabolismo , Neurotransmissores/metabolismo , Fragmentos de Peptídeos/metabolismo , Sinapses/metabolismo , Animais , Humanos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
8.
Microb Pathog ; 53(2): 87-94, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22610042

RESUMO

Shiga toxin-producing Escherichia coli produces watery and hemorrhagic diarrhea, and hemolytic uremic syndrome (HUS) characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. Central nervous system (CNS) complications are observed in around 30% of infant population with HUS. Common signs of severe CNS involvement leading to death include seizures, alteration of consciousness, hemiparesis, visual disturbances, and brain stem symptoms. The purpose of the present work was to study the effects of Shiga toxin 2 (Stx2) in the brain of rats intraperitoneally (i.p.) injected with a supernatant from recombinant E. coli expressing Stx2 (sStx2). Neurological alterations such as postural and motor abnormalities including lethargy, abnormal walking, and paralysis of hind legs, were observed in this experimental model of HUS in rats. Neuronal damage, as well as significant decrease in aquaporin 1 (AQP1) and aquaporin 4 (AQP4) expression levels were observed in the brain of rats, 2 days after sStx2 injection, compared to controls. Downregulation of aquaporin protein levels, and neuronal alterations, observed in brain of rats injected with sStx2, may be involved in edema formation and in neurological manifestations characteristic of HUS.


Assuntos
Aquaporina 1/genética , Aquaporina 4/genética , Encéfalo/metabolismo , Infecções por Escherichia coli/genética , Escherichia coli/metabolismo , Síndrome Hemolítico-Urêmica/genética , Neurônios/metabolismo , Toxina Shiga II/metabolismo , Animais , Aquaporina 1/metabolismo , Aquaporina 4/metabolismo , Encéfalo/efeitos dos fármacos , Escherichia coli/genética , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Síndrome Hemolítico-Urêmica/metabolismo , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Masculino , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Toxina Shiga II/administração & dosagem , Toxina Shiga II/genética , Toxina Shiga II/toxicidade
9.
Front Mol Biosci ; 9: 813637, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35372499

RESUMO

At first glance, the biological function of globoside (Gb) clusters appears to be that of glycosphingolipid (GSL) receptors for bacterial toxins that mediate host-pathogen interaction. Indeed, certain bacterial toxin families have been evolutionarily arranged so that they can enter eukaryotic cells through GSL receptors. A closer look reveals this molecular arrangement allocated on a variety of eukaryotic cell membranes, with its role revolving around physiological regulation and pathological processes. What makes Gb such a ubiquitous functional arrangement? Perhaps its peculiarity is underpinned by the molecular structure itself, the nature of Gb-bound ligands, or the intracellular trafficking unleashed by those ligands. Moreover, Gb biological conspicuousness may not lie on intrinsic properties or on its enzymatic synthesis/degradation pathways. The present review traverses these biological aspects, focusing mainly on globotriaosylceramide (Gb3), a GSL molecule present in cell membranes of distinct cell types, and proposes a wrap-up discussion with a phylogenetic view and the physiological and pathological functional alternatives.

10.
Toxicon ; 216: 115-124, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35835234

RESUMO

Encephalopathy associated with hemolytic uremic syndrome is produced by enterohemorrhagic E. coli (EHEC) infection, which releases the virulence factors Shiga toxin (Stx) and lipopolysaccharide (LPS). Neurological compromise is a poor prognosis and mortality factor of the disease, and the thalamus is one of the brain areas most frequently affected. We have previously demonstrated the effectiveness of anti-inflammatory drugs to ameliorate the deleterious effects of these toxins. However, the thalamic production of cytokines involved in pro-inflammatory processes has not yet been acknowledged. The aim of this work attempts to determine whether systemic sublethal Stx2a or co-administration of Stx2a with LPS are able to rise a proinflammatory profile accompanying alterations of the neurovascular unit in anterior and lateral ventral nuclei of the thalamus (VA-VL) and motor behavior in mice. After 4 days of treatment, Stx2a affected the lectin-bound microvasculature distribution while increasing the expression of GFAP in reactive astrocytes and producing aberrant NeuN distribution in degenerative neurons. In addition, increased swimming latency was observed in a motor behavioral test. All these alterations were heightened when Stx2a was co-administered with LPS. The expression of pro-inflammatory cytokines TNFα, INF-γ and IL-2 was detected in VA-VL. All these effects were concomitant with increased expression of the Stx receptor globotriaosylceramide (Gb3), which hints at receptor involvement in the neuroinflammatory process as a key finding of this study. In conclusion, Stx2a to Gb3 may be determinant in triggering a neuroinflammatory event, which may resemble clinical outcomes and should thus be considered in the development of preventive strategies.


Assuntos
Infecções por Escherichia coli , Toxina Shiga II , Animais , Citocinas/metabolismo , Escherichia coli/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Toxina Shiga/metabolismo , Toxina Shiga II/toxicidade , Tálamo/metabolismo , Triexosilceramidas
11.
Curr Neuropharmacol ; 19(1): 24-44, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32077828

RESUMO

Infection with Shiga toxin-producing Escherichia coli (STEC) may cause hemorrhagic colitis, hemolytic uremic syndrome (HUS) and encephalopathy. The mortality rate derived from HUS adds up to 5% of the cases, and up to 40% when the central nervous system (CNS) is involved. In addition to the well-known deleterious effect of Stx, the gram-negative STEC releases lipopolysaccharides (LPS) and may induce a variety of inflammatory responses when released in the gut. Common clinical signs of severe CNS injury include sensorimotor, cognitive, emotional and/or autonomic alterations. In the last few years, a number of drugs have been experimentally employed to establish the pathogenesis of, prevent or treat CNS injury by STEC. The strategies in these approaches focus on: 1) inhibition of Stx production and release by STEC, 2) inhibition of Stx bloodstream transport, 3) inhibition of Stx entry into the CNS parenchyma, 4) blockade of deleterious Stx action in neural cells, and 5) inhibition of immune system activation and CNS inflammation. Fast diagnosis of STEC infection, as well as the establishment of early CNS biomarkers of damage, may be determinants of adequate neuropharmacological treatment in time.


Assuntos
Escherichia coli Êntero-Hemorrágica , Infecções por Escherichia coli , Escherichia coli Shiga Toxigênica , Sistema Nervoso Central , Infecções por Escherichia coli/tratamento farmacológico , Humanos , Toxina Shiga
12.
Artigo em Inglês | MEDLINE | ID: mdl-31970091

RESUMO

Shiga toxin (Stx) produced by enterohemorrhagic E. coli produces hemolytic uremic syndrome and encephalopathies in patients, which can lead to either reversible or permanent neurological abnormalities, or even fatal cases depending on the degree of intoxication. It has been observed that the inflammatory component plays a decisive role in the severity of the disease. Therefore, the objective of this work was to evaluate the behavior of microglial cell primary cultures upon Stx2 exposure and heat shock or lipopolysaccharide challenges, as cues which modulate cellular environments, mimicking fever and inflammation states, respectively. In these contexts, activated microglial cells incorporated Stx2, increased their metabolism, phagocytic capacity, and pro-inflammatory profile. Stx2 uptake was associated to receptor globotriaosylceramide (Gb3)-pathway. Gb3 had three clearly distinguishable distribution patterns which varied according to different contexts. In addition, toxin uptake exhibited both a Gb3-dependent and a Gb3-independent binding depending on those contexts. Altogether, these results suggest a fundamental role for microglial cells in pro-inflammatory processes in encephalopathies due to Stx2 intoxication and highlight the impact of environmental cues.


Assuntos
Escherichia coli Êntero-Hemorrágica/metabolismo , Microglia/metabolismo , Toxina Shiga II/metabolismo , Animais , Encefalopatias , Citocinas/metabolismo , Modelos Animais de Doenças , Infecções por Escherichia coli , Resposta ao Choque Térmico , Síndrome Hemolítico-Urêmica , Inflamação , Lipopolissacarídeos , Macrófagos/metabolismo , Ratos , Ratos Wistar , Triexosilceramidas/metabolismo
13.
Brain Res ; 1230: 320-33, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18675791

RESUMO

Shiga toxin (Stx) from enterohemorrhagic Escherichia coli (STEC) is the main cause of hemorrhagic colitis which may derive into Hemolytic Uremic Syndrome (HUS) and acute encephalopathy, one of the major risk factors for infant death caused by the toxin. We have previously demonstrated that intracerebroventricular administration of Stx2 causes neuronal death and glial cell damage in rat brains. In the present work, we observed that the intracerebroventricular administration of Stx2 increased the expression of glial fibrillary acidic protein (GFAP) leading to astrogliosis. Confocal microscopy showed reactive astrocytes in contact with Stx2-containing neurons. Immunocolocalization of increased GFAP and Stx2 in astrocytes was also observed. This insult in the brain was correlated with changes in the expression and activity of neuronal nitric oxide synthase (nNOS) by using the NADPH-diaphorase histochemical technique (NADPH-d HT). A significant decrease in NOS/NADPH-d-positive neurons and NOS/NADPH-d activity was observed in cerebral cortex and striatum, whereas an opposite effect was found in the hypothalamic paraventricular nucleus. We concluded that the i.c.v. administration of Stx2 promotes a typical pattern of brain injury showing reactive astrocytes and an alteration in the number and activity of nNOS/NADPH-d. According to the functional state of nNOS/NADPH-d and to brain cell morphology data, it could be inferred that the i.c.v. administration of Stx2 leads to either a neurodegenerative or a neuroprotective mechanism in the affected brain areas. The present animal model resembles the encephalopathy developed in Hemolytic Uremic Syndrome (HUS) patients by STEC intoxication.


Assuntos
Química Encefálica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/biossíntese , Óxido Nítrico Sintase Tipo I/biossíntese , Toxina Shiga II/toxicidade , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Relação Dose-Resposta a Droga , Proteína Glial Fibrilar Ácida/genética , Imuno-Histoquímica , Injeções Intraventriculares , Masculino , Microscopia Confocal , Microscopia Eletrônica de Transmissão , NADPH Desidrogenase/metabolismo , Neostriado/fisiologia , Óxido Nítrico Sintase Tipo I/genética , Células Piramidais/efeitos dos fármacos , Células Piramidais/enzimologia , Células Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley , Toxina Shiga II/administração & dosagem , Toxina Shiga II/isolamento & purificação
14.
Microorganisms ; 6(4)2018 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-30274180

RESUMO

Pathogenic Escherichia coli are known to be a common cause of diarrheal disease and a frequently occurring bacterial infection in children and adults in Latin America. Despite the effort to combat diarrheal infections, the south of the American continent remains a hot spot for infections and sequelae associated with the acquisition of one category of pathogenic E. coli, the Shiga toxin-producing E. coli (STEC). This review will focus on an overview of the prevalence of different STEC serotypes in human, animals and food products, focusing on recent reports from Latin America outlining the recent research progress achieved in this region to combat disease and endemicity in affected countries and to improve understanding on emerging serotypes and their virulence factors. Furthermore, this review will highlight the progress done in vaccine development and treatment and will also discuss the effort of the Latin American investigators to respond to the thread of STEC infections by establishing a multidisciplinary network of experts that are addressing STEC-associated animal, human and environmental health issues, while trying to reduce human disease. Regardless of the significant scientific contributions to understand and combat STEC infections worldwide, many significant challenges still exist and this review has focus in the Latin American efforts as an example of what can be accomplished when multiple groups have a common goal.

15.
Front Microbiol ; 9: 3104, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619183

RESUMO

Hemolytic uremic syndrome (HUS), principally caused by shiga toxins (Stxs), is associated with Shiga toxin-producing Escherichia coli (STEC) infections. We previously reported Stx2 expression by host cells in vitro and in vivo. As the genes encoding the two Stx subunits are located in bacteriophage genomes, the aim of the current study was to evaluate the role of bacteriophage induction in HUS development in absence of an E. coli O157:H7 genomic background. Mice were inoculated with a non-pathogenic E. coli strain carrying the lysogenic bacteriophage 933W (C600Φ933W), and bacteriophage excision was induced by an antibiotic. The mice died 72 h after inoculation, having developed pathogenic damage typical of STEC infection. As well as renal and intestinal damage, markers of central nervous system (CNS) injury were observed, including aberrant immunolocalization of neuronal nuclei (NeuN) and increased expression of glial fibrillary acidic protein (GFAP). These results show that bacteriophage 933W without an E. coli O157:H7 background is capable of inducing the pathogenic damage associated with STEC infection. In addition, a novel mouse model was developed to evaluate therapeutic approaches focused on the bacteriophage as a new target.

16.
J Clin Invest ; 114(3): 357-69, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15286802

RESUMO

Regulation of energy balance by leptin involves regulation of several neuropeptides, including thyrotropin-releasing hormone (TRH). Synthesized from a larger inactive precursor, its maturation requires proteolytic cleavage by prohormone convertases 1 and 2 (PC1 and PC2). Since this maturation in response to leptin requires prohormone processing, we hypothesized that leptin might regulate hypothalamic PC1 and PC2 expression, ultimately leading to coordinated processing of prohormones into mature peptides. Using hypothalamic neurons, we found that leptin stimulated PC1 and PC2 mRNA and protein expression and also increased PC1 and PC2 promoter activities in transfected 293T cells. Starvation of rats, leading to low serum leptin levels, decreased PC1 and PC2 gene and protein expression in the paraventricular nucleus (PVN) of the hypothalamus. Exogenous administration of leptin to fasted animals restored PC1 levels in the median eminence (ME) and the PVN to approximately the level found in fed control animals. Consistent with this regulation of PCs in the PVN, concentrations of TRH in the PVN and ME were substantially reduced in the fasted animals relative to the fed animals, and leptin reversed this decrease. Further analysis showed that proteolytic cleavage of pro-thyrotropin-releasing hormone (proTRH) at known PC cleavage sites was reduced by fasting and increased in animals given leptin. Combined, these findings suggest that leptin-dependent stimulation of hypothalamic TRH expression involves both activation of trh transcription and stimulation of PC1 and PC2 expression, which lead to enhanced processing of proTRH into mature TRH.


Assuntos
Regulação Enzimológica da Expressão Gênica , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 2/genética , Processamento de Proteína Pós-Traducional , Hormônio Liberador de Tireotropina/metabolismo , Animais , Células Cultivadas , Ingestão de Energia , Feminino , Hipotálamo/citologia , Hipotálamo/embriologia , Imuno-Histoquímica , Injeções Intraperitoneais , Leptina/administração & dosagem , Leptina/farmacologia , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Gravidez , Pró-Proteína Convertase 1/biossíntese , Pró-Proteína Convertase 1/efeitos dos fármacos , Pró-Proteína Convertase 2/biossíntese , Pró-Proteína Convertase 2/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Hormônio Liberador de Tireotropina/genética , Tiroxina/sangue , Tri-Iodotironina/sangue
17.
J Mol Neurosci ; 31(1): 69-82, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17416971

RESUMO

ProThyrotropin-releasing hormone (proTRH) is a prohormone widely distributed in many areas of the brain. After biosynthesis, proTRH is subjected to post-translational processing to generate TRH and seven non-TRH peptides. Among these non-TRH sequences, we found previously that preproTRH178-199 could regulate the secretion of prolactin in suckled rats by their pups. Dopamine (DA), the main regulator of prolactin secretion, is produced in dopaminergic tyrosine hydroxylase (TH)-positive neurons in the hypothalamic arcuate nucleus (ARC). In this study we investigated whether prolactin release during the estrous sexual cycle is regulated by preproTRH178-199 through its effect on DA neurons of the ARC. We observed that biotinylated preproTRH178-199 bound to neurons in the ARC; this was higher during proestrus than during diestrus. Binding of preproTRH178-199 to DA neurons was seen only during proestrus in the ARC. Using primary neuronal hypothalamic cultures we found that preproTRH178-199 peptide decreased TH levels in a dose-responsive manner, whereas intra-ARC administration of preproTRH178-199 induced a 20-fold increase in plasma prolactin levels. Together, these results suggest a potential role for preproTRH178-199 in regulating dopaminergic neurons involved in the inhibition of pituitary prolactin release.


Assuntos
Produtos do Gene env/metabolismo , Hipotálamo/citologia , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Precursores de Proteínas/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Células Cultivadas , Dopamina/metabolismo , Ciclo Estral/fisiologia , Feminino , Hipotálamo/metabolismo , Neurônios/química , Neurônios/citologia , Gravidez , Prolactina/sangue , Prolactina/metabolismo , Ratos , Ratos Sprague-Dawley
18.
Brain Res ; 1161: 106-15, 2007 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-17610852

RESUMO

Shiga toxin (Stx) from enterohemorrhagic Escherichia coli (STEC) is the main cause of hemorrhagic colitis which may derive to hemolytic-uremic syndrome (HUS). HUS is characterized by acute renal failure, thrombocytopenia and microangiopathic hemolytic anemia. Mortality in the acute stage has been lower than 5% of total affected argentine children with endemic HUS. Common signs of severe CNS involvement leading to death included seizures, alteration of consciousness, hemiparesis, visual disturbances, and brainstem symptoms. The main purpose of the present work was to study the direct involvement of Stx2 in brain cells by intracerebroventricular (i.c.v.) administration of Stx2. Immunodetection of Stx2 was confirmed by immunoelectron cytochemistry in different subsets and compartments of affected caudate putamen cells of corpus striatum. Transmission electron microscopy (TEM) studies revealed apoptotic neurons, glial ultrastructural alterations and demyelinated fibers. The i.c.v. microinfusion was applied for the first time in rats to demonstrate the direct action of Stx2 in neurons and glial cells. The toxin may affect brain neuroglial cells without the involvement of proinflammatory or systemic neurotoxic elements.


Assuntos
Corpo Estriado/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/ultraestrutura , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Toxina Shiga II/administração & dosagem , Análise de Variância , Animais , Relação Dose-Resposta a Droga , Proteína Glial Fibrilar Ácida/metabolismo , Injeções Intraventriculares/métodos , Masculino , Microscopia Imunoeletrônica/métodos , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Toxina Shiga II/metabolismo
20.
Neuroscience ; 344: 25-38, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28042026

RESUMO

Shiga toxin 2 (Stx2) from enterohemorrhagic Escherichia coli (EHEC) causes bloody diarrhea and Hemolytic Uremic Syndrome (HUS) that may derive to fatal neurological outcomes. Neurological abnormalities in the striatum are frequently observed in affected patients and in studies with animal models while motor disorders are usually associated with pyramidal and extra pyramidal systems. A translational murine model of encephalopathy was employed to demonstrate that systemic administration of a sublethal dose of Stx2 damaged the striatal microvasculature and astrocytes, increase the blood brain barrier permeability and caused neuronal degeneration. All these events were aggravated by lipopolysaccharide (LPS). The injury observed in the striatum coincided with locomotor behavioral alterations. The anti-inflammatory Dexamethasone resulted to prevent the observed neurologic and clinical signs, proving to be an effective drug. Therefore, the present work demonstrates that: (i) systemic sub-lethal Stx2 damages the striatal neurovascular unit as it succeeds to pass through the blood brain barrier. (ii) This damage is aggravated by the contribution of LPS which is also produced and secreted by EHEC, and (iii) the observed neurological alterations may be prevented by an anti-inflammatory treatment.


Assuntos
Anti-Inflamatórios/farmacologia , Transtornos Cerebrovasculares/tratamento farmacológico , Dexametasona/farmacologia , Lipopolissacarídeos/toxicidade , Transtornos dos Movimentos/tratamento farmacológico , Toxina Shiga II/toxicidade , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Astrócitos/patologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Transtornos Cerebrovasculares/etiologia , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/patologia , Corpo Estriado/irrigação sanguínea , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/imunologia , Corpo Estriado/patologia , Modelos Animais de Doenças , Escherichia coli , Feminino , Camundongos , Microvasos/efeitos dos fármacos , Microvasos/imunologia , Microvasos/patologia , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/imunologia , Transtornos dos Movimentos/patologia , Fármacos Neuroprotetores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA