Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Neurol ; 28(9): 3100-3112, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34157194

RESUMO

BACKGROUND: Current drugs for epilepsy affect seizures, but no antiepileptogenic or disease-modifying drugs are available that prevent or slow down epileptogenesis, which is characterized by neuronal cell loss, inflammation and aberrant network formation. Ghrelin and ghrelin receptor (ghrelin-R) agonists were previously found to exert anticonvulsant, neuroprotective and anti-inflammatory effects in seizure models and immediately after status epilepticus (SE). Therefore, the aim of this study was to assess whether the ghrelin-R agonist macimorelin is antiepileptogenic in the pharmacoresistant intrahippocampal kainic acid (IHKA) mouse model. METHODS: SE was induced in C57BL/6 mice by unilateral IHKA injection. Starting 24 h after SE, mice were treated intraperitoneally with macimorelin (5 mg/kg) or saline twice daily for 2 weeks, followed by a 2-week wash-out. Mice were continuously electroencephalogram-monitored, and at the end of the experiment neuroprotection and gliosis were assessed. RESULTS: Macimorelin significantly decreased the number and duration of seizures during the treatment period, but had no antiepileptogenic or disease-modifying effect in this dose regimen. While macimorelin did not significantly affect food intake or body weight over a 2-week treatment period, its acute orexigenic effect was preserved in epileptic mice but not in sham mice. CONCLUSIONS: While the full ghrelin-R agonist macimorelin was not significantly antiepileptogenic nor disease-modifying, this is the first study to demonstrate its anticonvulsant effects in the IHKA model of drug-refractory temporal lobe epilepsy. These findings highlight the potential use of macimorelin as a novel treatment option for seizure suppression in pharmacoresistant epilepsy.


Assuntos
Epilepsia do Lobo Temporal , Estado Epiléptico , Animais , Modelos Animais de Doenças , Eletroencefalografia , Hipocampo , Humanos , Indóis , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Grelina , Convulsões/tratamento farmacológico , Estado Epiléptico/tratamento farmacológico , Triptofano/análogos & derivados
2.
Int J Mol Sci ; 20(10)2019 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-31137460

RESUMO

The ghrelin system has received substantial recognition as a potential target for novel anti-seizure drugs. Ghrelin receptor (ghrelin-R) signaling is complex, involving Gαq/11, Gαi/o, Gα12/13, and ß-arrestin pathways. In this study, we aimed to deepen our understanding regarding signaling pathways downstream the ghrelin-R responsible for mediating anticonvulsive effects in a kindling model. Mice were administered the proconvulsive dopamine 1 receptor-agonist, SKF81297, to gradually induce a kindled state. Prior to every SKF81297 injection, mice were treated with a ghrelin-R full agonist (JMV-1843), a Gαq and Gα12 biased ligand unable to recruit ß-arrestin (YIL781), a ghrelin-R antagonist (JMV-2959), or saline. Mice treated with JMV-1843 had fewer and less severe seizures compared to saline-treated controls, while mice treated with YIL781 experienced longer and more severe seizures. JMV-2959 treatment did not lead to differences in seizure severity and number. Altogether, these results indicate that the Gαq or Gα12 signaling pathways are not responsible for mediating JMV-1843's anticonvulsive effects and suggest a possible involvement of ß-arrestin signaling in the anticonvulsive effects mediated by ghrelin-R modulation.


Assuntos
Encéfalo/metabolismo , Excitação Neurológica , Receptores de Grelina/agonistas , Animais , Benzazepinas/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Agonistas de Dopamina/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Indóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piperidinas/farmacologia , Quinazolinonas/farmacologia , Receptores de Grelina/antagonistas & inibidores , Triazóis/farmacologia , Triptofano/análogos & derivados , Triptofano/farmacologia , beta-Arrestinas/farmacologia
3.
J Neuroinflammation ; 9: 114, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22647642

RESUMO

BACKGROUND: For successful translation to clinical stroke studies, the Stroke Therapy Academic Industry Round Table criteria have been proposed. Two important criteria are testing of therapeutic interventions in conscious animals and the presence of a co-morbidity factor. We chose to work with hypertensive rats since hypertension is an important modifiable risk factor for stroke and influences the clinical outcome. We aimed to compare the susceptibility to ischemia in hypertensive rats with those in normotensive controls in a rat model for induction of ischemic stroke in conscious animals. METHODS: The vasoconstrictor endothelin-1 was stereotactically applied in the vicinity of the middle cerebral artery of control Wistar Kyoto rats (WKYRs) and spontaneously hypertensive rats (SHRs) to induce a transient decrease in striatal blood flow, which was measured by the laser Doppler technique. Infarct size was assessed histologically by cresyl violet staining. Sensory-motor functions were measured at several time points using the neurological deficit score. Activation of microglia and astrocytes in the striatum and cortex was investigated by immunohistochemistry using antibodies against CD68/Iba-1 and glial fibrillary acidic protein. RESULTS AND CONCLUSIONS: The SHRs showed significantly larger infarct volumes and more pronounced sensory-motor deficits, compared to the WKYRs at 24 h after the insult. However, both differences disappeared between 24 and 72 h. In SHRs, microglia were less susceptible to activation by lipopolysaccharide and there was a reduced microglial activation after induction of ischemic stroke. These quantitative and qualitative differences may be relevant for studying the efficacy of new treatments for stroke in accordance to the Stroke Therapy Academic Industry Round Table criteria.


Assuntos
Isquemia Encefálica/metabolismo , Regulação para Baixo/fisiologia , Hipertensão/metabolismo , Lipopolissacarídeos/toxicidade , Microglia/metabolismo , Microglia/patologia , Acidente Vascular Cerebral/metabolismo , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Regulação para Baixo/efeitos dos fármacos , Endotelina-1/toxicidade , Predisposição Genética para Doença/etiologia , Hipertensão/complicações , Hipertensão/patologia , Lipopolissacarídeos/administração & dosagem , Masculino , Microglia/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
4.
BMC Neurosci ; 13: 105, 2012 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-22920191

RESUMO

BACKGROUND: Stroke remains one of the most common diseases with a serious impact on quality of life but few effective treatments exist. Mild hypothermia (33°C) is a promising neuroprotective therapy in stroke management. This study investigated whether a delayed short mild hypothermic treatment is still beneficial as neuroprotective strategy in the endothelin-1 (Et-1) rat model for a transient focal cerebral ischemia. Two hours of mild hypothermia (33°C) was induced 20, 60 or 120 minutes after Et-1 infusion. During the experiment the cerebral blood flow (CBF) was measured via Laser Doppler Flowmetry in the striatum, which represents the core of the infarct. Functional outcome and infarct volume were assessed 24 hours after the insult. In this sub-acute phase following stroke induction, the effects of the hypothermic treatment on apoptosis, phagocytosis and astrogliosis were assessed as well. Apoptosis was determined using caspase-3 immunohistochemistry, phagocytic cells were visualized by CD-68 expression and astrogliosis was studied by glial fibrillary acidic protein (GFAP) staining. RESULTS: Cooling could be postponed up to 1 hour after the onset of the insult without losing its positive effects on neurological deficit and infarct volume. These results correlated with the caspase-3 staining. In contrast, the increased CD-68 expression post-stroke was reduced in the core of the insult with all treatment protocols. Hypothermia also reduced the increased levels of GFAP staining, even when it was delayed up to 2 hours after the insult. The study confirmed that the induction of the hypothermia treatment in the Et-1 model does not affect the CBF. CONCLUSIONS: These data indicate that in the Et-1 rat model, a short mild hypothermic treatment delayed for 1 hour is still neuroprotective and correlates with apoptosis. At the same time, hypothermia also establishes a lasting inhibitory effect on the activation of astrogliosis.


Assuntos
Apoptose/efeitos dos fármacos , Isquemia Encefálica , Endotelina-1/efeitos adversos , Gliose/etiologia , Hipotermia Induzida/métodos , Estatística como Assunto , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Apoptose/fisiologia , Infarto Encefálico/etiologia , Isquemia Encefálica/induzido quimicamente , Isquemia Encefálica/complicações , Isquemia Encefálica/terapia , Caspase 3/metabolismo , Contagem de Células , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/terapia , Fluxometria por Laser-Doppler , Masculino , Transtornos dos Movimentos/etiologia , Exame Neurológico , Distribuição Aleatória , Ratos , Ratos Wistar , Fatores de Tempo
5.
Cell Mol Life Sci ; 68(23): 3823-41, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21744067

RESUMO

It has been known for several decades that cyclic AMP (cAMP), a prototypical second messenger, transducing the action of a variety of G-protein-coupled receptor ligands, has potent immunosuppressive and anti-inflammatory actions. These actions have been attributed in part to the ability of cAMP-induced signals to interfere with the function of the proinflammatory transcription factor Nuclear Factor-kappaB (NF-κB). NF-κB plays a crucial role in switching on the gene expression of a plethora of inflammatory and immune mediators, and as such is one of the master regulators of the immune response and a key target for anti-inflammatory drug design. A number of fundamental molecular mechanisms, contributing to the overall inhibitory actions of cAMP on NF-κB function, are well established. Paradoxically, recent reports indicate that cAMP, via its main effector, the protein kinase A (PKA), also promotes NF-κB activity. Indeed, cAMP actions appear to be highly cell type- and context-dependent. Importantly, several novel players in the cAMP/NF-κB connection, which selectively direct cAMP action, have been recently identified. These findings not only open up exciting new research avenues but also reveal novel opportunities for the design of more selective, NF-κB-targeting, anti-inflammatory drugs.


Assuntos
AMP Cíclico/metabolismo , NF-kappa B/metabolismo , Animais , AMP Cíclico/imunologia , AMP Cíclico/farmacologia , Humanos , NF-kappa B/química , NF-kappa B/imunologia , Transdução de Sinais/efeitos dos fármacos
6.
Cells ; 11(3)2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35159340

RESUMO

Here, we investigate changes in inflammation-related gene-expression in peripheral mononuclear blood cells (PBMC) by strength training. A total of 14 women aged ≥65 years were randomized into 3 months of either 3×/week intensive strength training (IST: 3×10 rep at 80% 1RM), strength endurance training (SET: 2×30 reps at 40% 1RM) or control (CON: 3×30 sec stretching). Differentially expressed genes (fold change ≤0.67 or ≥1.5) were identified by targeted RNA-sequencing of 407 inflammation-related genes. A total of 98 genes (n = 61 pro-inflammatory) were significantly affected. IST and SET altered 14 genes in a similar direction and 19 genes in the opposite direction. Compared to CON, IST changed the expression of 6 genes in the same direction, and 17 genes in the SET. Likewise, 18 and 13 genes were oppositely expressed for, respectively, IST and SET compared to CON. Changes in gene expression affected 33 canonical pathways related to chronic inflammation. None of the altered pathways overlapped between IST and SET. Liver X Receptor/Retinoid X Receptor Activation (LXR/RXR) and Triggering Receptor Expressed On Myeloid Cells 1 (TREM1) pathways were enriched oppositely in both training groups. We conclude that three months IST and SET can induce changes in CLIP-related gene expression in PBMC, but by affecting different genes and related pathways.


Assuntos
Treinamento Resistido , Idoso , Feminino , Expressão Gênica , Humanos , Inflamação/genética , Leucócitos Mononucleares , Receptor Gatilho 1 Expresso em Células Mieloides
7.
J Neuroinflammation ; 8: 60, 2011 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21627837

RESUMO

BACKGROUND: Stroke is an important cause of morbidity and mortality and few therapies exist thus far. Mild hypothermia (33°C) is a promising neuroprotective strategy to improve outcome after ischemic stroke. However, its complete mechanism of action has not yet been fully elaborated. This study is the first to investigate whether this neuroprotection occurs through modulation of the neuroinflammatory response after stroke in a time-dependent manner. METHODS: The Endothelin-1 (Et-1) model was used to elicit a transient focal cerebral ischemia in male Wistar rats. In this model, the core and penumbra of the insult are represented by the striatum and the cortex respectively. We assessed the effects of 2 hours of hypothermia, started 20 minutes after Et-1 injection on neurological outcome and infarct volume. Furthermore, pro- and anti-inflammatory cytokine expression was determined using ELISA. Microgliosis and astrogliosis were investigated using CD-68 and GFAP staining respectively. All parameters were determined 8, 24, 72 hours and 1 week after the administration of Et-1. RESULTS: Et-1 infusion caused neurological deficit and a reproducible infarct size which increased up to 3 days after the insult. Both parameters were significantly reduced by hypothermia. The strongest reduction in infarct volume with hypothermia, at 3 days, corresponded with increased microglial activation. Reducing the brain temperature affected the stroke induced increase in interleukin-1ß and tumor necrosis factor α in the striatum, 8 hours after its induction, but not at later time points. Transforming growth factor ß increased as a function of time after the Et-1-induced insult and was not influenced by cooling. Hypothermia reduced astrogliosis at 1 and 3 days after stroke onset. CONCLUSIONS: The beneficial effects of hypothermia after stroke on infarct volume and functional outcome coincide with a time-dependent modulation of the cytokine expression and gliosis.


Assuntos
Citocinas/metabolismo , Endotelina-1/farmacologia , Gliose , Hipotermia/metabolismo , Ataque Isquêmico Transitório/induzido quimicamente , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Gliose/induzido quimicamente , Gliose/metabolismo , Gliose/patologia , Interleucina-1beta/metabolismo , Ataque Isquêmico Transitório/fisiopatologia , Masculino , Ratos , Ratos Wistar , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Biomedicines ; 10(1)2021 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-35052733

RESUMO

Epilepsy is a neurological disease affecting more than 50 million individuals worldwide. Notwithstanding the availability of a broad array of antiseizure drugs (ASDs), 30% of patients suffer from pharmacoresistant epilepsy. This highlights the urgent need for novel therapeutic options, preferably with an emphasis on new targets, since "me too" drugs have been shown to be of no avail. One of the appealing novel targets for ASDs is the ghrelin receptor (ghrelin-R). In epilepsy patients, alterations in the plasma levels of its endogenous ligand, ghrelin, have been described, and various ghrelin-R ligands are anticonvulsant in preclinical seizure and epilepsy models. Up until now, the exact mechanism-of-action of ghrelin-R-mediated anticonvulsant effects has remained poorly understood and is further complicated by multiple downstream signaling pathways and the heteromerization properties of the receptor. This review compiles current knowledge, and discusses the potential mechanisms-of-action of the anticonvulsant effects mediated by the ghrelin-R.

9.
J Neuroinflammation ; 7: 74, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21040547

RESUMO

Neuroinflammation is a key element in the ischemic cascade after cerebral ischemia that results in cell damage and death in the subacute phase. However, anti-inflammatory drugs do not improve outcome in clinical settings suggesting that the neuroinflammatory response after an ischemic stroke is not entirely detrimental. This review describes the different key players in neuroinflammation and their possible detrimental and protective effects in stroke. Because of its inhibitory influence on several pathways of the ischemic cascade, hypothermia has been introduced as a promising neuroprotective strategy. This review also discusses the influence of hypothermia on the neuroinflammatory response. We conclude that hypothermia exerts both stimulating and inhibiting effects on different aspects of neuroinflammation and hypothesize that these effects are key to neuroprotection.


Assuntos
Isquemia Encefálica/patologia , Hipotermia Induzida , Inflamação/patologia , Inflamação/terapia , Acidente Vascular Cerebral/patologia , Animais , Anti-Inflamatórios/uso terapêutico , Isquemia Encefálica/complicações , Isquemia Encefálica/imunologia , Isquemia Encefálica/fisiopatologia , Quimiocinas/imunologia , Citocinas/imunologia , Proteínas HMGB/imunologia , Humanos , Inflamação/etiologia , Inflamação/imunologia , Integrinas/imunologia , Metaloproteinases da Matriz/imunologia , Fármacos Neuroprotetores/uso terapêutico , Espécies Reativas de Oxigênio/imunologia , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/fisiopatologia
10.
Neuroscience ; 426: 101-114, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31846748

RESUMO

We and others have shown that insulin-like growth factor-1 (IGF-1) is neuroprotective when administered systemically shortly following stroke. In the current study, we addressed the hypothesis that microglia mediate neuroprotection by IGF-1 following ischemic stroke. Furthermore, we investigated whether IGF-1 modulates pro- and anti-inflammatory mediators in ischemic brain with a special reference to microglia. Ischemic stroke was induced in normal conscious Wistar rats by infusing the vasoconstrictor, endothelin-1 (Et-1), next to middle cerebral artery (MCA). IGF-1 (300 µg) was injected subcutaneously (SC) at 30 and 120 min following stroke. Microglial inhibitor, minocycline, was injected intraperitoneally (IP) at 1 h before stroke (25 mg/kg) and 11 h after stroke (45 mg/kg). Post-stroke IGF-1 treatment reduced the infarct size and increased the sensorimotor function which coincided with an increase in the number of ameboid microglia in the ischemic cortex. Minocycline treatment abrogated the increase in ameboid microglia by IGF-1, while the effect of IGF-1 in the reduction of infarct size was only partially affected. IGF-1 suppressed mRNA expression of inducible nitric oxide synthase (iNOS) and interleukin (IL)-1ß in the ischemic hemisphere, while in purified microglia, only iNOS expression levels were reduced. Our findings show that microglia are a target for IGF-1 and that neuroprotection by IGF-1 coincides with down-regulation of inflammatory mediators which could be instrumental to the beneficial effects.


Assuntos
Anti-Inflamatórios/farmacologia , Isquemia Encefálica/tratamento farmacológico , Inflamação/tratamento farmacológico , Fator de Crescimento Insulin-Like I/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Inflamação/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos Wistar
11.
Stroke ; 40(4): e83-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19197073

RESUMO

BACKGROUND AND PURPOSE: Insulin-like growth factor I (IGF-I) exerts neuroprotective effects in both white and gray matter under different detrimental conditions. The purpose of this review is to collect the evidence whether IGF-I is a candidate neuroprotective drug in patients with acute ischemic stroke. RESULTS: IGF-I was found to be neuroprotective in animal models of focal brain ischemia when given >or=2 hours after the insult. Different routes of administration (eg, cerebroventricular, intravenous, and intranasal) were found to be effective. In addition to inhibition of apoptosis and reduction of the infarct volume, IGF-I also improved neurological outcome. Furthermore, there are strong indications that IGF-I can also stimulate the regeneration of neural tissue. CONCLUSIONS: Additional studies are required to reveal the neuroprotective mechanisms of IGF-I in detail and to elucidate the role of IGF-binding proteins. Preclinical studies in relevant animal models for studying stroke (ie, hypertensive, diabetic, or aged animals) should be done testing different doses and routes of IGF-I administration and different combinations of IGF-I and IGF-binding proteins.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fator de Crescimento Insulin-Like I/farmacologia , Fármacos Neuroprotetores/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Doença Aguda , Animais , Modelos Animais de Doenças , Humanos
12.
Cytokine Growth Factor Rev ; 17(4): 305-23, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16621671

RESUMO

IGF-I is important for fetal and post-natal development, but it also controls tissue homeostasis throughout life via regulation of cell proliferation and apoptosis. This review summarizes our current understanding of how IGF-I receptor signaling interferes with the apoptotic machinery of the cell. IGF-I acts at different control points of apoptosis, including the Bcl-2 family proteins, inhibitors of caspases and signaling of death-inducing receptors. The main focal point of IGF-I is the regulation of Bcl-2 family proteins. Several signaling pathways converge to both the phosphorylation and transcriptional regulation of these proteins. This phenomenon may explain the efficacy of IGF-I as an inhibitor of apoptosis in many different cell types and in the presence of different apoptogenic stimuli.


Assuntos
Apoptose , Fator de Crescimento Insulin-Like I/fisiologia , Animais , Humanos , Fator de Crescimento Insulin-Like I/uso terapêutico , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais
13.
Biofactors ; 35(1): 76-81, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19319849

RESUMO

The insulin-like growth factor IGF-I is an important fetal and postnatal growth factor, which is also involved in tissue homeostasis via regulation of proliferation, differentiation, and cell survival. To understand the role of IGF-I in the pathophysiology of a variety of disorders, including growth disorders, cancer, and neurodegenerative diseases, a detailed knowledge of IGF-I signal transduction is required. This knowledge may also contribute to the development of new therapies directed at the IGF-I receptor or other signaling molecules. In this review, we will address IGF-I receptor signaling through the JAK/STAT pathway in IGF-I signaling and the role of cytokine-induced inhibitors of signaling (CIS) and suppressors of cytokine signaling (SOCS). It appears that, in addition to the canonical IGF-I signaling pathways through extracellular-regulated kinase (ERK) and phosphatidylinositol-3 kinase (PI3K)-Akt, IGF-I also signals through the JAK/STAT pathway. Activation of this pathway may lead to induction of SOCS molecules, well-known feedback inhibitors of the JAK/STAT pathway, which also suppress of IGF-I-induced JAK/STAT signaling. Furthermore, other IGF-I-induced signaling pathways may also be modulated by SOCS. It is conceivable that the effect of these classical inhibitors of cytokine signaling directly affect IGF-I receptor signaling, because they are able to associate to the intracellular part of the IGF-I receptor. These observations indicate that CIS and SOCS molecules are key to cross-talk between IGF-I receptor signaling and signaling through receptors belonging to the hematopoietic/cytokine receptor superfamily. Theoretically, dysregulation of CIS or SOCS may affect IGF-I-mediated effects on body growth, cell differentiation, proliferation, and cell survival.


Assuntos
Janus Quinases/metabolismo , Receptor IGF Tipo 1/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Animais , Citocinas/metabolismo , Humanos , Modelos Biológicos , Proteínas Supressoras da Sinalização de Citocina/metabolismo
14.
Front Aging Neurosci ; 11: 349, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31920629

RESUMO

Post-stroke systemic injections of insulin-like growth factor-1 (IGF-1) exert neuroprotective effects in rats. In the current study, we aimed to test the efficacy of IGF-1 neuroprotection in aged rats (24-25 months old) and to compare the results with adult rats (6-7 months old). Furthermore, we addressed putative differences in microglial responses to IGF-1 in adult and aged rats. Rats were subjected to ischemic stroke while they were conscious by infusing endothelin-1 (Et-1) through a guide cannula that was implemented in the vicinity of the middle cerebral artery (MCA). Rats were given subcutaneous injections of IGF-1 (1 mg/kg) at 30 min and 120 min after the insult. Post-stroke IGF-1 treatment reduced the infarct size by 34% and 38% in aged and adult rats, respectively. The IGF-1 treated adult rats also showed significant improvement in sensorimotor function following stroke, while this function was not significantly affected in aged rats. Furthermore, aged rats displayed exaggerated activation of microglia in the ischemic hemisphere. Significant reduction of microglial activation by IGF-1 was only detected at specific regions in the ipsilateral hemisphere of adult rats. We show that IGF-1 reduced infarct size in aged rats with an ischemic stroke. It remains to be established, however, whether the age-related changes in microglial function affect the improvement in behavioral outcomes.

15.
Front Pharmacol ; 10: 1491, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31969819

RESUMO

Background: In the human central nervous system (CN), resting astrocytes do not visually show endothelin-1 (ET-1)-like immunoreactivity. In patients with multiple sclerosis (MS), an inflammatory disorder of the CNS, high levels of ET-1 are found in reactive astrocytes in demyelinated plaques. ET-1 may contribute to the pathology of MS by interrupting the blood-brain-barrier, enhancing inflammatory responses, excitotoxicity and reducing cerebral blood flow. Methods: We used the human astrocytoma cell line 1321N1 to investigate the role of inflammatory cytokines involved in MS lesions (IL-1ß, TNF-α, IFN-γ, LPS, IL-10, TGF-ß) on astrocytic ET-1 upregulation. Prucalopride, rolipram, fenofibrate, fluoxetine, simvastatin, daglutril, and resveratrol were investigated as potential candidate drugs to suppress cytokine-induced astrocytic ET-1 production. Effects on ET-1 production were measured using both ELISA and RT-qPCR. Results and Conclusions: ET-1 secretion by astrocytoma cells was only stimulated by the pro-inflammatory cytokines IL-1ß and TNF-α. Fluoxetine, simvastatin, and resveratrol significantly inhibited this IL-1ß- and TNF-α-induced ET-1 production. Simvastatin and resveratrol significantly reduced ET-1 mRNA levels, indicating an effect at the level of transcription. Fluoxetine significantly reduced endothelin converting enzyme-1 mRNA levels, suggesting and effect at the level of protein-processing. The required concentrations of simvastatin (>0.1 µM) and resveratrol (>10 µM) cannot be achieved in humans using pharmacologically accepted doses. Fluoxetine exerted a significant inhibitory effect on ET-1 secretion at a concentration of 5 µM, which is pharmacologically achievable in human brain, but the effect was modest (<50% suppression) and probably not sufficient to obtain a clinically relevant ET-1 effect. Our in vitro model can be a useful screening tool in the development of new drugs to suppress astrocytic ET-1 production. The effect of simvastatin was for the most part mediated via the mevalonate pathway, suggesting that this might be an interesting target for further drug development.

16.
Growth Horm IGF Res ; 17(5): 383-91, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17513150

RESUMO

OBJECTIVE: Since serum IGF-I levels are related to risks of prostate cancer and cytokines like interleukin (IL)-6 and IL-8 have been implicated in prostate cancer progression, we investigated the effects of IGF-I on IL-6 and IL-8 expression in the prostate cancer cell. DESIGN: In order to address the regulation by IGF-I of cytokine expression in prostate cancer cells we used cell cultures of established androgen dependent (LNCaP) and androgen-independent cell lines (DU-145 and PC-3). RESULTS: We found that IGF-I stimulates IL-8 mRNA expression and secretion in DU-145 cells, whereas the secretion of IL-6 was hardly affected. IGF-I enhances IL-8 expression in synergy with IL-1beta, but not with tumour necrosis factor (TNF)alpha. Similarly, on IL-8 promoter activity, IGF-I exerted synergistic effects with IL-1beta, but not with TNFalpha. Although IGF-I stimulated the phosphorylation of both Akt (protein kinase B) and extracellular-regulated kinase (ERK), the effect of IGF-I at IL-8 expression was inhibited only by U0126, a pharmacological inhibitor of MAPK/ERK kinase (MEK) and not by inhibition of the upstream activator of Akt, phosphatidylinositol-3 kinase (PI3K). CONCLUSIONS: Our results indicate that IGF-I stimulates IL-8 expression through the MEK-ERK pathway in DU-145 cells, at least in part, by augmentation of transcriptional activity. This finding is in accordance with our observations that IGF-I did not influence cytokine secretion and phosphorylation of ERK in LNCaP or PC-3 cells. It remains to be established whether IL-8 mediates certain effects of IGF-I on prostate cancer cells and whether differential responsiveness of prostate cancer cells to IGF-I relates to certain stages of prostate cancer.


Assuntos
Citocinas/farmacologia , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-8/genética , Neoplasias da Próstata/genética , Androstadienos/farmacologia , Butadienos/farmacologia , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/fisiopatologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Masculino , Nitrilas/farmacologia , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Proteínas Recombinantes/farmacologia , Transfecção , Wortmanina
17.
Neuroscience ; 348: 335-345, 2017 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-28257893

RESUMO

Although ischemic stroke is a major cause of death worldwide and the predominant cause of acquired disability, the only effective drug therapy that has been developed thus far is reperfusion by tissue plasminogen activator. Since most patients do not qualify for this treatment, new methods have to be developed. It is well known that estradiol (E2) exerts neuroprotective effects in different models of cerebral ischemia, but post-stroke treatment after an acute stroke has hardly been investigated. As many patients with an acute ischemic stroke have arterial hypertension, it is also of interest to evaluate the influence of this co-morbidity on the treatment efficacy of E2. The effects of E2 administered 30min after a transient middle cerebral artery occlusion (tMCAO) induced by an intracerebral injection of endothelin-1 were assessed in male normotensive Wistar Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs). Treatment with E2 reduced infarct size in both WKY and SHRs and decreased the number of degenerating neurons, indicating that acute treatment with E2 is indeed neuroprotective. To address the role of glia in neuroprotection, the effects of E2 on the activation of microglia and astrocytes was determined. It appeared that E2 had no effect on microglial activation, but reduced the activation of astrocytes in SHRs but not in the normotensive controls. We conclude that post-stroke E2 treatment in both normotensive and hypertensive rats is neuroprotective. Although the presence of hypertension changed the astrocytic response to E2, it did not affect treatment efficacy.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Estradiol/uso terapêutico , Hipertensão/complicações , Fármacos Neuroprotetores/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Astrócitos/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Isquemia Encefálica/complicações , Estradiol/farmacologia , Masculino , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Acidente Vascular Cerebral/complicações
18.
Int J Biochem Cell Biol ; 38(11): 1957-64, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16846747

RESUMO

We previously established that stimulation by IGF-I of interleukin (IL)-8 expression in leukocytes required activation of extracellular-regulated kinase (ERK) and basal activity of c-Jun N-terminal kinase (JNK). In this study, we tested the hypothesis that IGF-I stimulates IL-8 expression at the transcriptional level through induction of Fos/Jun activator protein (AP)-1 complex formation. Inhibition studies using the transcriptional inhibitor actinomycin D and IL-8 promoter activation studies indicate that IGF-I act at the transcriptional level. Using gel shift assays we demonstrate that IGF-I induces the formation of active c-Jun/c-Fos AP-1 complexes. Promoter activation studies using mutated IL-8 promoter constructs show that the AP-1 response element is required for promoter activation by IGF-I whereas CAAT-enhancer binding protein (C/EBP) and nuclear factor of kappa B (NFkappaB) sites were not essential. These results indicate that IGF-I can augment IL-8 expression through activation of AP-1 independent of other inducible transcription factors which have shown to be involved in IL-8 regulation by immune stimuli. This finding is in agreement with our previous observation that IGF-I is able to enhance basal IL-8 production in peripheral blood mononuclear cells (PBMC) in the absence of other stimuli.


Assuntos
Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-8/genética , Regiões Promotoras Genéticas/genética , Fator de Transcrição AP-1/genética , Linhagem Celular , Dactinomicina/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica/efeitos dos fármacos , Células HL-60 , Humanos , Interleucina-8/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/efeitos dos fármacos
19.
J Neuroimmunol ; 172(1-2): 206-10, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16325274

RESUMO

The pituitary hormone prolactin (PRL) is also produced extrapituitarily by cells of the immune system. In leukocytes PRL expression is directed by an alternative promoter, located 5800 bp upstream of the pituitary promoter. We have shown here that this alternative promoter is activated in myeloid leukemic cells by the proinflammatory cytokine tumor necrosis factor (TNF)-alpha. The effect of TNF-alpha on promoter activation was blocked by the protein kinase C (PKC)-inhibitor GFX109203. In addition, we have shown that the TNF-alpha-responsive region is located between -1842 and -1662 of the extrapituitary PRL promoter. Our findings might have clinical relevance in view of recent data indicating leukocyte-derived PRL is involved in auto-immune and hematological disorders.


Assuntos
Regulação da Expressão Gênica/fisiologia , Leucemia Mieloide/patologia , Prolactina/genética , Regiões Promotoras Genéticas/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Análise de Variância , Linhagem Celular , Interações Medicamentosas , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Mutagênese/fisiologia , Prolactina/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Transfecção/métodos
20.
Cell Signal ; 17(7): 901-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15763432

RESUMO

Besides its pivotal role in reproduction, the polypeptide hormone prolactin (PRL) has been attributed an immunomodulatory function. Extrapituitary PRL expression is regulated differently from that in the pituitary, due to the use of an alternative promoter. In leukocytes, cAMP is an important regulator of PRL expression. We report that in the human eosinophilic cell line Eol-1, cAMP-induced PRL expression is partially abrogated by two protein kinase A (PKA) inhibitors (H89, PKI) and by the p38 inhibitor SB203580. Phosphorylation of p38 was PKA-independent and could be stimulated by a methylated cAMP analogue, which specifically activates the exchange factor directly activated by cAMP (EPAC). Furthermore, cAMP induced a PKA-dependent phosphorylation of cAMP-responsive element binding protein (CREB). We postulate that cAMP induces PRL expression via two different signalling pathways: a PKA-dependent pathway leading to the phosphorylation of CREB, and a PKA-independent pathway leading to the phosphorylation of p38.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , AMP Cíclico/fisiologia , Eosinófilos/metabolismo , Prolactina/biossíntese , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Proteínas do Leite/metabolismo , Fosforilação , Fator de Transcrição STAT5 , Transdução de Sinais , Transativadores/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA