Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neuroimmune Pharmacol ; 19(1): 17, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38717643

RESUMO

In our previous study, we concluded that sirtuin 5 (SIRT5) was highly expressed in microglia following ischaemic stroke, which induced excessive neuroinflammation and neuronal injury. Therefore, SIRT5-targeting interventions should reduce neuroinflammation and protect against ischaemic brain injury. Here, we showed that treatment with a specific SIRT5 inhibitor, MC3482, alleviated microglia-induced neuroinflammation and improved long-term neurological function in a mouse model of stroke. The mice were administrated with either vehicle or 2 mg/kg MC3482 daily for 7 days via lateral ventricular injection following the onset of middle cerebral artery occlusion. The outcome was assessed by a panel of tests, including a neurological outcome score, declarative memory, sensorimotor tests, anxiety-like behavior and a series of inflammatory factors. We observed a significant reduction of infarct size and inflammatory factors, and the improvement of long-term neurological function in the early stages during ischaemic stroke when the mice were treated with MC3482. Mechanistically, the administration of MC3482 suppressed the desuccinylation of annexin-A1, thereby promoting its membrane recruitment and extracellular secretion, which in turn alleviated neuroinflammation during ischaemic stroke. Based on our findings, MC3482 offers promise as an anti-ischaemic stroke treatment that targets directly the disease's underlying factors.


Assuntos
Anexina A1 , AVC Isquêmico , Microglia , Doenças Neuroinflamatórias , Sirtuínas , Animais , Masculino , Camundongos , Anexina A1/efeitos dos fármacos , Anexina A1/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Sirtuínas/antagonistas & inibidores , Sirtuínas/metabolismo , Regulação para Cima/efeitos dos fármacos
2.
Cells ; 9(11)2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33202930

RESUMO

Stroke is a leading cause of death and disability globally and is associated with a number of co-morbidities including sepsis and sickle cell disease (SCD). Despite thrombo-inflammation underlying these co-morbidities, its pathogenesis remains complicated and drug discovery programs aimed at reducing and resolving the detrimental effects remain a major therapeutic challenge. The objective of this study was to assess whether the anti-inflammatory pro-resolving protein Annexin A1 (AnxA1) was able to reduce inflammation-induced thrombosis and suppress platelet activation and thrombus formation in the cerebral microvasculature. Using two distinct models of pathological thrombo-inflammation (lipopolysaccharide (LPS) and sickle transgenic mice (STM)), thrombosis was induced in the murine brain using photoactivation (light/dye) coupled with intravital microscopy. The heightened inflammation-induced microvascular thrombosis present in these two distinct thrombo-inflammatory models was inhibited significantly by the administration of AnxA1 mimetic peptide AnxA1Ac2-26 (an effect more pronounced in the SCD model vs. the endotoxin model) and mediated by the key resolution receptor, Fpr2/ALX. Furthermore, AnxA1Ac2-26 treatment was able to hamper platelet aggregation by reducing platelet stimulation and aggregation (by moderating αIIbß3 and P-selectin). These findings suggest that targeting the AnxA1/Fpr2/ALX pathway represents an attractive novel treatment strategy for resolving thrombo-inflammation, counteracting e.g., stroke in high-risk patient cohorts.


Assuntos
Anexina A1/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Receptores de Formil Peptídeo/efeitos dos fármacos , Trombose/tratamento farmacológico , Animais , Anexina A1/metabolismo , Humanos , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fragmentos de Peptídeos/farmacologia , Receptores de Formil Peptídeo/metabolismo
3.
Nutrients ; 12(8)2020 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-32823596

RESUMO

Punicalagin (PU), a polyphenol extracted from pomegranate (Punica granatum) husk is proven to have anti-cancer effects on different types of cancer including colorectal cancer (CRC). Its role in modulating endogenous protein as a means of eliciting its anti-cancer effects, however, has not been explored to date. Hence, this study aimed to investigate the role of PU in modulating the interplay between apoptosis and autophagy by regulating Annexin A1 (Anx-A1) expression in HCT 116 colorectal adenocarcinoma cells. In the study, selective cytotoxicity, pro-apoptotic, autophagic and Anx-A1 downregulating properties of PU were shown which indicate therapeutic potential that this polyphenol has against CRC. Autophagy flux analysis via flow cytometry showed significant autophagosomes degradation in treated cells, proving the involvement of autophagy. Proteome profiling of 35 different proteins in the presence and absence of Anx-A1 antagonists in PU-treated cells demonstrated a complex interplay that happens between apoptosis and autophagy that suggests the possible simultaneous induction and inhibition of these two cell death mechanisms by PU. Overall, this study suggests that PU induces autophagy while maintaining basal level of apoptosis as the main mechanisms of cytotoxicity via the modulation of Anx-A1 expression in HCT 116 cells, and thus has a promising translational potential.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Taninos Hidrolisáveis/farmacologia , Extratos Vegetais/farmacologia , Punica granatum/química , Anexina A1/efeitos dos fármacos , Morte Celular Autofágica/efeitos dos fármacos , Células HCT116 , Humanos
4.
J Med Chem ; 61(4): 1609-1621, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29406740

RESUMO

The plant metabolite 3,4,5-tri-O-galloylquinic acid methyl ester (TGAME, compound 6) was synthesized, and its potential effect on calcium oxalate monohydrate (COM) crystal binding to the surface of Madin-Darby canine kidney cells type I (MDCKI) and crystal growth in a Drosophila melanogaster Malpighian tubule (MT) model were investigated. Membrane, cytosolic, and total annexin A1 (AxA1), α-enolase, and heat shock protein 90 (HSP90) amounts were examined by Western blot analysis after subcellular fractionation, then confirmed by immunofluorescence staining of cultured cells. Pretreatment of MDCKI cells with TGAME for up to 6 h significantly diminished COM crystal binding in a concentration-dependent manner. TGAME significantly inhibited AxA1 surface expression by immunofluorescence microscopy, whereas intracellular AxA1 increased. Western blot analysis confirmed AxA1 expression changes in the membrane and cytosolic fractions of compound-treated cells, whereas whole cell AxA1 remained unchanged. TGAME also significantly decreased the size, number, and growth of calcium oxalate (CaOx) crystals induced in a Drosophila melanogaster MT model and possessed a potent antioxidant activity in a DPPH assay.


Assuntos
Anexina A1/efeitos dos fármacos , Oxalato de Cálcio/química , Adesão Celular/efeitos dos fármacos , Ácido Gálico/análogos & derivados , Ácido Quínico/análogos & derivados , Animais , Anexina A1/metabolismo , Antioxidantes , Linhagem Celular , Cristalização , Cães , Drosophila melanogaster , Ácido Gálico/síntese química , Ácido Gálico/química , Ácido Gálico/farmacologia , Células Madin Darby de Rim Canino/efeitos dos fármacos , Células Madin Darby de Rim Canino/metabolismo , Túbulos de Malpighi/química , Ácido Quínico/síntese química , Ácido Quínico/química , Ácido Quínico/farmacologia , Frações Subcelulares/química , Frações Subcelulares/metabolismo
5.
Br J Pharmacol ; 158(7): 1820-34, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19912231

RESUMO

BACKGROUND AND PURPOSE: The current clinical strategy to protect the auditory organ against inflammatory damage by migrating leukocytes is the local delivery of glucocorticoids. However, the mechanism by which glucocorticoids confer this protection remains unknown. Therefore, we investigated the cellular and molecular targets of glucocorticoids in the cochlea that could be involved in preventing leukocyte migration. EXPERIMENTAL APPROACH: We used microscopy as well as immunocytochemical and microfluidic techniques to elucidate the effect of dexamethasone, hydrocortisone and prednisolone on the cellular and intracellular distribution of annexin A1 (ANXA1) - a glucocorticoid target known to inhibit leukocyte migration by receptor-mediated signalling - in the cochlea and isolated cochlear cells of guinea pigs. KEY RESULTS: All the cells lining the scala media - the cochlear compartment containing the auditory organ - express ANXA1 and the ANXA1 receptor FPR2/ALX is present in the scala media, as well as in other cochlear ducts. The majority of ANXA1 in the scala media is stored inside lipid droplets within cochlear Hensen cells. Glucocorticoids activate a myosin IIC-mediated mechanism that drives ANXA1 from the lipid droplets to the apical region of the Hensen cells, where ANXA1 is released to the external milieu by a process involving ABC transporters. CONCLUSIONS AND IMPLICATIONS: These findings suggest that ANXA1 could be a major mediator of the anti-inflammatory effects of glucocorticoids in the cochlea and identify new molecular targets for prevention of sudden sensorineural hearing loss.


Assuntos
Anexina A1/efeitos dos fármacos , Cóclea/efeitos dos fármacos , Glucocorticoides/farmacologia , Animais , Anexina A1/metabolismo , Cóclea/citologia , Cóclea/metabolismo , Dexametasona/farmacologia , Sistemas de Liberação de Medicamentos , Cobaias , Hidrocortisona/farmacologia , Microscopia , Miosina Tipo II/metabolismo , Prednisolona/farmacologia , Receptores de Formil Peptídeo/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Biochem Pharmacol ; 77(12): 1814-26, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19428336

RESUMO

Using biochemical, epifluorescence and electron microscopic techniques in a U937 model system, we investigated the effect of anti-allergic drugs di-sodium cromoglycate and sodium nedocromil on the trafficking and release of the anti-inflammatory protein Annexin-A1 (Anx-A1) when this was triggered by glucocorticoid (GC) treatment. GCs alone produced a rapid (within 5min) concentration-dependent activation of PKCalpha/beta (Protein Kinase C; EC 2.7.11.13) and phosphorylation of Anx-A1 on Ser(27). Both phosphoproteins accumulated at the plasma membrane and Anx-A1 was subsequently externalised thereby inhibiting thromboxane (Tx) B(2) generation. When administered alone, cromoglycate or nedocromil had little effect on this pathway however, in the presence of a fixed sub-maximal concentration of GCs, increasing amounts of the cromoglycate-like drugs caused a striking concentration-dependent enhancement of Anx-A1 and PKCalpha/beta phosphorylation, membrane recruitment and Anx-A1 release from cells resulting in greatly enhanced inhibition of TxB(2) generation. GCs also stimulated phosphatase accumulation at the plasma membrane of U937 cells. Both cromoglycate and nedocromil inhibited this enzymatic activity as well as that of a highly purified PP2A phosphatase preparation. We conclude that stimulation by the cromoglycate-like drugs of intracellular Anx-A1 trafficking and release (hence inhibition of eicosanoid release) is secondary to inhibition of a phosphatase PP2A (phosphoprotein phosphatase; EC 3.1.3.16), which probably forms part of a control loop to limit Anx-A1 release. These experiments provide a basis for a novel mechanism of action for the cromolyns, a group of drugs that have long puzzled investigators.


Assuntos
Anexina A1/efeitos dos fármacos , Cromolina Sódica/farmacologia , Eicosanoides/antagonistas & inibidores , Anexina A1/metabolismo , Antiasmáticos/farmacologia , Quimioterapia Combinada , Glucocorticoides/farmacologia , Humanos , Nedocromil/farmacologia , Proteína Quinase C/metabolismo , Proteína Fosfatase 2/metabolismo , Transporte Proteico , Células U937
7.
J Neuroendocrinol ; 18(12): 949-59, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17076770

RESUMO

Perinatal glucocorticoid (GC) treatment is increasingly associated with long-term disturbances in hypothalamo-pituitary-adrenocortical function. In the male rat, such treatment induces profound molecular, morphological and functional changes in the anterior pituitary gland at adulthood. To determine whether these effects are sex-specific, we have examined the effects of perinatal dexamethasone treatment on the female pituitary gland, focusing on (i) the integrity of the annexin 1 (ANXA1) dependent regulatory effects of GCs on adrenocorticotrophic hormone (ACTH) release and (ii) corticotroph and folliculo-stellate (FS) cell morphology. Dexamethasone was given to pregnant (gestational days 16-19) or lactating (days 1-7 post partum) rats via the drinking water (1 microg/ml); controls received normal drinking water. Pituitary tissue from the female offspring was examined ex vivo at adulthood (60-90 days). Both treatment regimes reduced the intracellular and cell surface ANXA1 expression, as determined by western blot analysis and quantitative immunogold electron microscopic histochemistry. In addition, they compromised the ability of dexamethasone to suppress the evoked release of ACTH from the excised tissue in vitro, a process which requires the translocation of ANXA1 from the cytoplasm to the cell surface of FS cells. Although neither treatment regime affected the number of FS cells or corticotrophs, both altered the subcellular morphology of these cells. Thus, prenatal dexamethasone treatment increased while neonatal treatment decreased FS cell size and cytoplasmic area. By contrast, corticotroph size was unaffected by either treatment, as also was the size of the secretory granules. Corticotroph granule density and margination were, however, increased markedly by the prenatal treatment, while the neonatal treatment had no effect on granule density but decreased granule margination. Thus, perinatal dexamethasone treatment exerts long-term effects on the female pituitary gland, altering gene expression, cell morphology and the ANXA1-dependent GC regulation of ACTH secretion. The changes are similar but not identical to those reported in the male.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Anexina A1/metabolismo , Glucocorticoides/fisiologia , Adeno-Hipófise/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Fatores Etários , Animais , Anexina A1/efeitos dos fármacos , Corticotrofos/efeitos dos fármacos , Corticotrofos/ultraestrutura , Dexametasona/farmacologia , Retroalimentação Fisiológica/fisiologia , Feminino , Glucocorticoides/farmacologia , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/ultraestrutura , Gravidez , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Fatores de Tempo
8.
J Leukoc Biol ; 78(3): 639-46, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16000391

RESUMO

The role of the endogenous anti-inflammatory mediator annexin 1 (AnxA1) in controlling polymorphonuclear leukocyte (PMN) trafficking and activation was addressed using the recently generated AnxA1 null mouse. In the zymosan peritonitis model, AnxA1 null mice displayed a higher degree (50-70%) of PMN recruitment compared with wild-type littermate mice, and this was associated with reduced numbers of F4/80+ cells. Intravital microscopy analysis of the cremaster microcirculation inflamed by zymosan (6 h time-point) indicated a greater extent of leukocyte emigration, but not rolling or adhesion, in AnxA1 null mice. Real-time analysis of the cremaster microcirculation did not show spontaneous activation in the absence of AnxA1; however, superfusion with a direct-acting PMN activator (1 nM platelet-activating factor) revealed a subtle yet significant increase in leukocyte emigration, but not rolling or adhesion, in this genotype. Changes in the microcirculation were not secondary to alterations in hemodynamic parameters. The phenotype of the AnxA1 null PMN was investigated in two in vitro assays of cell activation (CD11b membrane expression and chemotaxis): the data obtained indicated a higher degree of cellular responses irrespective of the stimulus used. In conclusion, we have used a combination of inflammatory protocols and in vitro assays to address the specific counter-regulatory role of endogenous AnxA1, demonstrating its inhibitory control on PMN activation and the consequent impact on the inflamed microcirculation.


Assuntos
Anexina A1/genética , Quimiotaxia/fisiologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Animais , Anexina A1/efeitos dos fármacos , Anexina A1/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/genética , Modelos Animais de Doenças , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação/efeitos dos fármacos , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/efeitos dos fármacos , Fator de Ativação de Plaquetas/administração & dosagem , Fator de Ativação de Plaquetas/farmacologia , Fatores de Tempo , Zimosan/administração & dosagem , Zimosan/farmacologia
9.
Mediators Inflamm ; 12(6): 329-37, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14668092

RESUMO

AIMS: Annexin 1 (ANXA1), a member of the annexin family of calcium-binding and phospholipid-binding proteins, is a key mediator of the anti-inflammatory actions of steroid hormones. We have previously demonstrated that, in the human lymphoblastic CCRF-CEM cell line, both the synthetic glucocorticoid hormone, dexamethasone (Dex), and the estrogen hormone, 17beta-estradiol (E2beta), induce the synthesis of ANXA1, by a mechanism independent of the activation of their nuclear receptors. Recently, it was reported that the gene coding for ANXA1 contains acAMP-responsive element (CRE). In this work, we investigated whether Dex and E2beta were able to induce the activation of CRE binding proteins (CREB) in the CCRF-CEM cells. Moreover, we studied the intracellular signalling pathways involved in CREB activation and ANXA1 synthesis in response to Dex and E2beta; namely, the role of cAMP and the p38 mitogen activated protein kinase (MAPK). RESULTS: The results show that Dex and E2beta were as effective as the cAMP analogue, dBcAMP, in inducing CREB activation. On the contrary, dBcAMP induced ANXA1 synthesis as effectively as these steroid hormones. Furthermore, the cAMP antagonist, Rp-8-Br-cAMPS, and the specific p38 MAPK inhibitor,SB203580, effectively prevented both Dex-induced, E2beta-induced and dBcAMP-induced CREB activation and ANXA1 synthesis. CONCLUSIONS: Taken together, our results suggest that,in CCRF-CEM cells, Dex-induced and E2beta-inducedANXA1 expression requires the activation of the transcription factor CREB, which in turn seems to be mediated by cAMP and the p38 MAPK. These findings also suggest that, besides the nuclear steroid hormone receptors, other transcription factors, namely CREB, may play important roles in mediating the anti-inflammatory actions of glucocorticoids and oestrogen hormones.


Assuntos
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Anexina A1/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Dexametasona/farmacologia , Estradiol/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Anexina A1/efeitos dos fármacos , Bucladesina/farmacologia , Células Cultivadas , AMP Cíclico/antagonistas & inibidores , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Imidazóis/farmacologia , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Piridinas/farmacologia , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética , Transdução de Sinais , Tionucleotídeos/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
10.
J Neuroendocrinol ; 14(8): 621-8, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12153464

RESUMO

In the pituitary gland, annexin-1 (lipocortin-1) located in folliculo-stellate (FS) cells has been advocated as one of the candidates for paracrine agents produced by FS cells that modulate the release of pituitary hormones. However, the expression and distribution pattern of annexin-1 in FS cells under different circulating corticosteroid conditions has not been examined. Thus, by means of pre-embedding immunoelectron microscopy, we investigated the expression of annexin-1 in FS cells under different corticosteroid conditions. Annexin-1-immunoreactivity was observed in the cytoplasm; especially intense immunoreactivity was detected in the follicle surface of FS cells under control conditions. After adrenalectomy, annexin-1-immunoreactivity almost disappeared, but the immunoreactivity recovered with corticosterone replacement. The expression of glucocorticoid receptor immunoreactivity in the nucleus of FS cells also showed a similar pattern to annexin-1 associated with the changes in the corticosteroid conditions. However, S-100 immunoreactivity, a marker for FS cells, was not changed whatever the corticosteroid conditions. These results confirm that glucocorticoids regulate the annexin-1 expression and demonstrate the translocation of annexin-1 from intracellular to pericellular sites in the FS cells of the rat anterior pituitary gland.


Assuntos
Anexina A1/análise , Adeno-Hipófise/química , Adrenalectomia , Animais , Anexina A1/efeitos dos fármacos , Corticosterona/farmacologia , Imuno-Histoquímica , Masculino , Microscopia Imunoeletrônica , Adeno-Hipófise/citologia , Adeno-Hipófise/ultraestrutura , Ratos , Ratos Wistar , Receptores de Glucocorticoides/análise , Proteínas S100/análise
11.
Eur J Biochem ; 238(3): 777-84, 1996 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8706680

RESUMO

Annexin I belongs to a family of calcium-dependent phospholipid-binding and membrane-binding proteins. Although many of the biochemical properties and the three-dimensional structure of this protein are known, its true physiological roles have yet to be thoroughly defined. Its putative functions include participation in the regulation of actin microfilaments dynamics, proposed after the discovery of an interaction with actin. In accordance with this hypothesis, we found that annexin I can also interact with profilin. We used different methods, overlay and surface plasmon resonance (BIAcore), to measure the parameters of the association equilibrium, i.e. k(on), k(off) and k(d). The affinity of annexin I for profilin was between 10(7) M and 10(8) M. High concentrations of KCl did not prevent the interaction, although a slight decrease in affinity was observed. Calcium, a modulator of annexin I functions interfered only marginally with the association, in a manner comparable to magnesium. Proteins or compounds known to interact with annexin I or profilin were found to inhibit the annexin-I--profilin interaction when added in the reaction medium. Recombinant profilin exhibited a slightly lower affinity than natural platelet protein when measured with BIAcore. Due to the submembrane localisation of annexin I and the regulatory activity of profilin on the cytoskeleton, an interaction between annexin I and profilin may therefore be implicated in the regulation of some cellular functions, particularly those governing membrane-cytoskeleton dynamic organization.


Assuntos
Anexina A1/metabolismo , Proteínas Contráteis , Proteínas dos Microfilamentos/metabolismo , Actinas/metabolismo , Anexina A1/efeitos dos fármacos , Ligação Competitiva , Bioensaio/métodos , Técnicas Biossensoriais , Eletroforese em Gel de Poliacrilamida/métodos , Proteínas dos Microfilamentos/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/farmacologia , Profilinas , Prometazina/farmacologia , Desnaturação Proteica
12.
Aliment Pharmacol Ther ; 9(5): 541-5, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8580275

RESUMO

BACKGROUND: Corticosteroids are widely used to treat children with inflammatory bowel disease although the response is variable, side-effects are common, and many patients develop a partial or complete steroid resistance. The mechanism underlying these phenomena are unclear. Corticosteroids mediate some of their actions through lipocortin-1, and the induction of autoantibodies to lipocortin has been proposed as a possible mechanism by which steroid efficacy is suboptimal in vivo. PATIENTS AND METHODS: We have measured serum lipocortin-1 antibody concentration by ELISA in 38 children with Crohn's disease, 12 with ulcerative colitis and in 15 controls. RESULTS: IgG and IgA anti-lipocortin-1 antibody levels were higher in the Crohn's group than in the ulcerative colitis or control groups. Elevated concentrations did not relate to disease activity, history of steroid therapy or steroid-responsiveness. Lipocortin IgM antibody status was similar in all three groups. CONCLUSION: It is therefore unlikely that serum antibodies to lipocortin-1 have a role in the development of steroid-resistance in children with inflammatory bowel disease.


Assuntos
Corticosteroides/efeitos adversos , Anexina A1/imunologia , Autoanticorpos/sangue , Doenças Inflamatórias Intestinais/imunologia , Adolescente , Corticosteroides/uso terapêutico , Anexina A1/efeitos dos fármacos , Autoanticorpos/efeitos dos fármacos , Criança , Colite Ulcerativa/imunologia , Doença de Crohn/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Doenças Inflamatórias Intestinais/tratamento farmacológico , Masculino
13.
Eur J Pharmacol ; 283(1-3): 9-18, 1995 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-7498325

RESUMO

Platelet-activating factor (PAF; 100 ng i.v.) transiently modified the number of circulating neutrophils in the mouse, inducing a fast neutropenia (2 min) followed by a late onset neutrophilia (2 h). The potential involvement in PAF-induced neutrophilia of granulocytotic agents such as interleukin-1 and tumor necrosis factor-alpha could be excluded on the basis of the ineffectiveness of interleukin-1 receptor antagonist and of a specific monoclonal antibody anti-murine tumor necrosis factor-alpha. PAF granulocytosis was preceded by a significant rise in plasma corticosterone at 20 min. The involvement of endogenous corticosteroids was confirmed by the experiments with adrenalectomized mice and in animals pretreated with the steroid antagonist RU486 (11 beta-(4-dimethyl amino-phenyl) 17 beta-hydroxy, 17 alpha(prop-1-ynyl) estra 4,9-dien-3-one), where PAF-induced neutrophilia was greatly reduced (approximately 50%). Moreover, sustained increase in plasma corticosterone by administration of adrenocorticotropic hormone was paralleled by an intense neutrophilia. We show evidence that endogenous corticosterone acts through the glucocorticoid-inducible protein lipocortin 1.


Assuntos
Corticosteroides/farmacologia , Neutrófilos/efeitos dos fármacos , Fator de Ativação de Plaquetas/farmacologia , Corticosteroides/sangue , Adrenalectomia , Hormônio Adrenocorticotrópico/farmacologia , Animais , Anexina A1/efeitos dos fármacos , Cinética , Leucócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Mifepristona/farmacologia , Radioimunoensaio , Fatores de Tempo
14.
Biochim Biophys Acta ; 1205(2): 215-22, 1994 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-8155700

RESUMO

We have mutated the lysine 128 of domain II of annexin I, which flanks a putative calcium-binding loop, into a glutamic acid residue. The properties of the mutated recombinant protein were compared to those of the wild-type recombinant protein. A change in the isotherm of calcium binding in the presence of lipids was observed. A slight decrease in the affinity for lipids was evident. When tested for the vesicle aggregation property, the mutation induced a change in lipid specificity; unlike the wild-type protein, the mutant protein aggregates vesicles containing phosphatidylserine plus phosphatidylethanolamine better than vesicles containing only phosphatidylserine. These experiments are in agreement with a model which suggests that a lipid molecule is inserted into the calcium-binding loop of annexin I and that the conserved lysine residue is involved in the specificity of annexins for anionic phospholipids.


Assuntos
Anexina A1/metabolismo , Cálcio/metabolismo , Fosfolipídeos/metabolismo , Sequência de Aminoácidos , Anexina A1/efeitos dos fármacos , Anexina A1/genética , Análise Mutacional de DNA , Lipossomos/metabolismo , Lisina/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosfatidiletanolaminas/farmacologia , Fosfatidilserinas/farmacologia , Ligação Proteica , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA