Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Appl Mater Interfaces ; 14(1): 214-224, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-34935338

RESUMO

Smart hydrogels with versatile properties, including a tunable gelation time, nonswelling attributes, and biocompatibility, are in great need in the biomedical field. To meet this urgent demand, we explored novel biomaterials with the desired properties from sessile marine organisms. To this end, a novel protein, Sbp9, derived from scallop byssus was extensively investigated, which features typical epidermal growth factor-like (EGFL) multiple repetitive motifs. Our current work demonstrated that the key fragment of Sbp9 (calcium-binding domain (CBD) and 4 EGFL repeats (CE4)) was able to form a smart hydrogel driven by noncovalent interactions and facilitated by disulfide bonds. More importantly, this smart hydrogel demonstrates several desirable and beneficial features, which could offset the drawbacks of typical protein-based hydrogels, including (1) a redox-responsive gelation time (from <1 to 60 min); (2) tunable mechanical properties, nonswelling abilities, and an appropriate microstructure; and (3) good biocompatibility and degradability. Furthermore, proof-of-concept demonstrations showed that the newly discovered hydrogel could be used for anticancer drug delivery and cell encapsulation. Taken together, a smart hydrogel inspired by marine sessile organisms with desirable properties was generated and characterized and demonstrated to have extensive applicability potential in biomedical applications, including tissue engineering and drug release.


Assuntos
Proteínas de Ligação ao Cálcio/química , Encapsulamento de Células/métodos , Portadores de Fármacos/química , Hidrogéis/química , Pectinidae/química , Materiais Inteligentes/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Proteínas de Ligação ao Cálcio/toxicidade , Linhagem Celular Tumoral , Doxorrubicina/química , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Humanos , Hidrogéis/toxicidade , Peróxido de Hidrogênio/química , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Oxirredução , Porosidade , Domínios Proteicos , Ratos Sprague-Dawley , Materiais Inteligentes/toxicidade
2.
Int J Biol Macromol ; 146: 1015-1023, 2020 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-31726124

RESUMO

Necrotic enteritis (NE) is a multifactorial disease in broiler that is caused by colonization of Clostridium perfringens in their gastrointestinal tract. Recently several immunogenic proteins from virulent C. perfringens have been considered as vaccines to provide protection against NE. In this study, a novel trivalent fusion protein including immunogenic epitopes of three virulence factors of, NetB, alpha toxin and a metallopeptidase protein (NAM) was designed using in silico studies. Circular dichroism spectra was applied for determination of secondary structure and folding properties of the purified recombinant NAM (rNAM) expressed in E. coli. The antigenicity of rNAM was confirmed by induction of immune response in rabbit and neutralization experiments of the toxins in cell culture studies. To this end, anti-rNAM antisera neutralized the crude toxins produced by a wild type virulent C. perfringens strain using chicken hepatocellular carcinoma (LMH) cell lines. The cells were exposed to a mixture of anti-rNAM antisera and 2 × LD50 doses of the toxins. The result showed 94% viability of the cells against the crude toxins, in the presence of anti-rNAM antisera. Our study suggests that combination of metallopeptidase protein along with alpha toxin and NetB toxins is a potent immunogen which is able to neutralize the toxicity of crude extracellular toxins. The recombinant chimeric NAM could be a suitable and effective subunit vaccine candidate to prevent NE disease caused by C. perfringens.


Assuntos
Vacinas Bacterianas/imunologia , Clostridium perfringens/imunologia , Simulação por Computador , Proteínas Recombinantes de Fusão/imunologia , Fatores de Virulência/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/toxicidade , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Galinhas , Epitopos de Linfócito B/imunologia , Metaloproteases/metabolismo , Testes de Neutralização , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA Mensageiro/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Reprodutibilidade dos Testes , Fosfolipases Tipo C/toxicidade
3.
Sci Rep ; 7(1): 5217, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701754

RESUMO

Clostridium perfringens α-toxin induces hemolysis of erythrocytes from various species, but it has not been elucidated whether the toxin affects erythropoiesis. In this study, we treated bone marrow cells (BMCs) from mice with purified α-toxin and found that TER119+ erythroblasts were greatly decreased by the treatment. A variant α-toxin defective in enzymatic activities, phospholipase C and sphingomyelinase, had no effect on the population of erythroblasts, demonstrating that the decrease in erythroblasts was dependent of its enzymatic activities. α-Toxin reduced the CD71+TER119+ and CD71-TER119+ cell populations but not the CD71+TER119- cell population. In addition, α-toxin decreased the number of colony-forming unit erythroid colonies but not burst-forming unit erythroid colonies, indicating that α-toxin preferentially reduced mature erythroid cells compared with immature cells. α-Toxin slightly increased annexinV+ cells in TER119+ cells. Additionally, simultaneous treatment of BMCs with α-toxin and erythropoietin greatly attenuated the reduction of TER119+ erythroblasts by α-toxin. Furthermore, hemin-induced differentiation of human K562 erythroleukemia cells was impaired by α-toxin, whereas the treatment exhibited no apparent cytotoxicity. These results suggested that α-toxin mainly inhibited erythroid differentiation. Together, our results provide new insights into the biological activities of α-toxin, which might be important to understand the pathogenesis of C. perfringens infection.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/toxicidade , Diferenciação Celular/efeitos dos fármacos , Células Precursoras Eritroides/patologia , Eritropoese/efeitos dos fármacos , Fosfolipases Tipo C/toxicidade , Animais , Antígenos CD/metabolismo , Antígenos de Grupos Sanguíneos/metabolismo , Células Cultivadas , Células Precursoras Eritroides/efeitos dos fármacos , Humanos , Células K562 , Camundongos , Camundongos Endogâmicos C57BL , Receptores da Transferrina/metabolismo
4.
Chem Phys Lipids ; 203: 54-70, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28104376

RESUMO

Clostridium perfringens alpha-toxin (ATX) is considered as a prototype of cytotoxic bacterial phospholipases C, and is the major virulence factor in C. perfringens-induced gas gangrene. It is known that, depending on the dose, ATX causes membrane disruption and cytolysis or only limited hydrolysis of its substrates. In the latter case, toxin activity leads to the unregulated generation of bioactive lipids that can ultimately induce cell death. We have characterized apoptosis and necrosis in highly ATX-sensitive, ganglioside-deficient cells exposed to different concentrations of ATX and we have studied the lipidomic profile of cells treated with ATX as compared to native cells to detect the main changes in the lipidomic profile and the possible involvement of lipid signals in cell death. ATX causes both apoptosis and necrosis, depending on dose and time. ATX activates cell death, stimulating the release of cytochrome C from mitochondria and the consequent activation of caspases-3. Moreover GM95 cells treated with ATX showed important lipidomic alterations, among them we detected a general decrease in several phospholipid species and important changes in lipids involved in programmed cell death e.g. ceramide. The data suggest two different mechanisms of cell death caused by ATX, one leading to (mainly saturated) glycerophospholipid hydrolysis related to an increase in diacylglycerols and associated to membrane damage and necrosis, and a second mechanism involving chiefly sphingomyelin hydrolysis and generation of proapoptotic lipidic mediators such as ceramide, N-acylethanolamine and saturated non-esterified fatty acids.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/toxicidade , Lipídeos/química , Fosfolipases Tipo C/toxicidade , Animais , Toxinas Bacterianas/química , Proteínas de Ligação ao Cálcio/química , Morte Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Camundongos , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Fosfolipases Tipo C/química
5.
Sci Rep ; 6: 28192, 2016 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-27306065

RESUMO

Although granulopoiesis is accelerated to suppress bacteria during infection, some bacteria can still cause life-threatening infections, but the mechanism behind this remains unclear. In this study, we found that mature neutrophils in bone marrow cells (BMCs) were decreased in C. perfringens-infected mice and also after injection of virulence factor α-toxin. C. perfringens infection interfered with the replenishment of mature neutrophils in the peripheral circulation and the accumulation of neutrophils at C. perfringens-infected sites in an α-toxin-dependent manner. Measurements of bacterial colony-forming units in C. perfringens-infected muscle revealed that α-toxin inhibited a reduction in the load of C. perfringens. In vitro treatment of isolated BMCs with α-toxin (phospholipase C) revealed that α-toxin directly decreased mature neutrophils. α-Toxin did not influence the viability of isolated mature neutrophils, while simultaneous treatment of BMCs with granulocyte colony-stimulating factor attenuated the reduction of mature neutrophils by α-toxin. Together, our results illustrate that impairment of the innate immune system by the inhibition of neutrophil differentiation is crucial for the pathogenesis of C. perfringens to promote disease to a life-threatening infection, which provides new insight to understand how pathogenic bacteria evade the host immune system.


Assuntos
Toxinas Bacterianas/toxicidade , Células da Medula Óssea/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/toxicidade , Clostridium perfringens/patogenicidade , Imunidade Inata/imunologia , Neutrófilos/imunologia , Fosfolipases Tipo C/toxicidade , Fatores de Virulência/toxicidade , Animais , Bacillus subtilis/genética , Bacillus subtilis/patogenicidade , Toxinas Bacterianas/genética , Proteínas de Ligação ao Cálcio/genética , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Infecções por Clostridium/patologia , Clostridium perfringens/genética , Fator Estimulador de Colônias de Granulócitos/farmacologia , Imunidade Inata/efeitos dos fármacos , Contagem de Leucócitos , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/citologia , Fosfolipases Tipo C/genética , Fatores de Virulência/genética
6.
Avian Pathol ; 44(2): 81-91, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25584964

RESUMO

The causative pathogen of necrotic enteritis is the Gram-positive bacterium Clostridium perfringens. Its main cell wall component, peptidoglycan (PGN), can be recognized by Toll-like receptor 2 and nucleotide-binding oligomerization domain (NOD). Consequently, the immune response is initiated via activation of nuclear factor kappa B (NF-κB) signalling pathway. An in vitro study was conducted to investigate chicken intestinal inflammatory responses to C. perfringens type A and one of its virulence factors, α-toxin. In primary intestinal epithelial cells, C. perfringens as well as commercially available PGN and α-toxin challenge upregulated mRNA expression of interleukin (IL)-6, IL-8 and inducible nitric oxide synthase (iNOS) with a dosage-dependent manner at 3 h post infection (p.i.; P ≤ 0.001). Time-course effects of three stimulators at high concentration were further examined. C. perfringens infection elevated IL-6, IL-8 and iNOS levels from 1 h to 9 h p.i., while PGN treatment increased IL-6 and IL-8 expression at 1 h and 3 h p.i. (P < 0.05). Bacterial and PGN treatments induced NOD1 expression at 6 h p.i. and only bacterial infection boosted NF-κB p65 expression at 6 h and 9 h p.i. (P < 0.05). α-Toxin treatment upregulated IL-6 and IL-8 expression throughout infection, as well as iNOS, TNF-α and NF-κB p65 expression at later hours p.i. (P < 0.05). In conclusion, both C. perfringens and α-toxin challenge induced intense cytokine expression associated with NF-κB activation in chicken intestinal epithelial cells. The receptors for the recognition of PGN component of C. perfringens need further investigation.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/toxicidade , Clostridium perfringens/imunologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Regulação da Expressão Gênica/imunologia , Mucosa Intestinal/citologia , Transdução de Sinais/imunologia , Fosfolipases Tipo C/toxicidade , Animais , Embrião de Galinha , Células Epiteliais/microbiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Interleucina-6/metabolismo , Interleucina-8/metabolismo , NF-kappa B/imunologia , Óxido Nítrico Sintase Tipo II/metabolismo
7.
Microbes Infect ; 11(3): 413-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19284973

RESUMO

Clostridium perfringens and Clostridium septicum are the most common causes of clostridial myonecrosis or gas gangrene. Although they mediate a similar disease pathology, they elaborate functionally very different alpha-toxins. We used a reciprocal complementation approach to assess the contribution of the primary toxin of each species to disease and found that C. perfringens alpha-toxin (PLC) was able to mediate the gross pathology of myonecrosis even in a C. septicum background, although it could not induce vascular leukostasis. Conversely, while C. septicum alpha-toxin restored some virulence to a C. perfringens plc mutant, it was less active than in its native background.


Assuntos
Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/toxicidade , Clostridium perfringens/patogenicidade , Clostridium septicum/patogenicidade , Gangrena Gasosa/microbiologia , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/toxicidade , Animais , Clostridium perfringens/genética , Clostridium septicum/genética , Feminino , Teste de Complementação Genética , Camundongos , Camundongos Endogâmicos BALB C
8.
Poult Sci ; 88(1): 199-204, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19096074

RESUMO

The present report demonstrates the effect of alpha toxin from Clostridium perfringens on electrophysiological indexes of jejunal mucosa from laying hens pretreated with inulin and N-acetyl-l-cysteine (ACC), a mucolytic agent. In a first set of experiments, the effect of alpha toxin with or without pretreatment with ACC on the electrophysiological parameters was determined when jejunal tissues from laying hens were mounted in Ussing chambers. The short-circuit current remained unchanged when alpha toxin was added mucosally in the tissues whether pretreated with ACC or not. The change in the transmural tissue conductance (DeltaGt) was higher (P = 0.18) after 90 min exposure of toxin independent of pretreament with ACC. The effect of alpha toxin on DeltaGt became significant (P < or = 0.05) after 120 min of incubation. In the second set of experiments, the effect of alpha toxin on the jejunal tissues preincubated with inulin (0.1%) was investigated. The effect of toxin was also time dependent, and DeltaGt became significantly higher (P < or = 0.05) after 120 min of incubation independent of preinubation with inulin. Inulin did not influence the DeltaGt during the experimental period when compared with control tissues. In conclusion, alpha toxin from C. perfringens can impair the intestinal mucosal barrier. The effect is obviously not dependent on the presence of a mucolytic agent nor can it be affected by direct addition of inulin under in vitro conditions. Whether there is an effect of inulin after long-term supplementation in feeding trials or it is due to fermentation bacterial metabolites remains an open question.


Assuntos
Acetilcisteína/farmacologia , Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/toxicidade , Galinhas , Inulina/farmacologia , Jejuno/efeitos dos fármacos , Fosfolipases Tipo C/toxicidade , Animais , Eletrofisiologia , Feminino , Mucosa Intestinal/efeitos dos fármacos , Oviposição
9.
Infect Immun ; 74(5): 2876-86, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16622226

RESUMO

Clostridium perfringens alpha-toxin induces the generation of superoxide anion (O2(-)) via production of 1,2-diacylglycerol (DG) in rabbit neutrophils. The mechanism of the generation, however, remains poorly understood. Here we report a novel mechanism for the toxin-induced production of O2(-) in rabbit neutrophils. Treatment of the cells with the toxin resulted in tyrosine phosphorylation of a protein of about 140 kDa. The protein reacted with anti-TrkA (nerve growth factor high-affinity receptor) antibody and bound nerve growth factor. Anti-TrkA antibody inhibited the production of O2(-) and binding of the toxin to the protein. The toxin induced phosphorylation of 3-phosphoinositide-dependent protein kinase 1 (PDK1). K252a, an inhibitor of TrkA receptor, and LY294002, an inhibitor of phosphatidylinositol 3-kinase (PI3K), reduced the toxin-induced production of O2(-) and phosphorylation of PDK1, but not the formation of DG. These inhibitors inhibited the toxin-induced phosphorylation of protein kinase C theta (PKCtheta). U73122, a phospholipase C (PLC) inhibitor, and pertussis toxin inhibited the toxin-induced generation of O2(-) and formation of DG, but not the phosphorylation of PDK1. These observations show that the toxin independently induces production of DG through activation of endogenous PLC and phosphorylation of PDK1 via the TrkA receptor signaling pathway and that these events synergistically activate PKCtheta in stimulating an increase in O2(-). In addition, we show the participation of mitogen-activated protein kinase-associated signaling events via activation of PKCtheta in the toxin-induced generation of O2(-).


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/toxicidade , Neutrófilos/efeitos dos fármacos , Transdução de Sinais/fisiologia , Superóxidos/metabolismo , Fosfolipases Tipo C/toxicidade , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Diglicerídeos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Proteína Quinase C/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Coelhos , Receptor trkA/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
10.
Infect Immun ; 72(9): 5204-15, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15322015

RESUMO

Clostridium perfringens is the most common cause of clostridial myonecrosis (gas gangrene). Polymorphonuclear cells (PMNs) appear to play only a minor role in preventing the onset of myonecrosis in a mouse animal model of the disease (unpublished results). However, the importance of macrophages in the host defense against C. perfringens infections is still unknown. Two membrane-active toxins produced by the anaerobic C. perfringens, alpha-toxin (PLC) and perfringolysin O (PFO), are thought to be important in the pathogenesis of gas gangrene and the lack of phagocytic cells at the site of infection. Therefore, C. perfringens mutants lacking PFO and PLC were examined for their relative cytotoxic effects on macrophages, their ability to escape the phagosome of macrophages, and their persistence in mouse tissues. C. perfringens survival in the presence of mouse peritoneal macrophages was dependent on both PFO and PLC. PFO was shown to be the primary mediator of C. perfringens-dependent cytotoxicity to macrophages. Escape of C. perfringens cells from phagosomes of macrophage-like J774-33 cells and mouse peritoneal macrophages was mediated by either PFO or PLC, although PFO seemed to play a more important role in escape from the phagosome in peritoneal macrophages. At lethal doses (10(9)) of bacteria only PLC was necessary for the onset of myonecrosis, while at sublethal doses (10(6)) both PFO and PLC were necessary for survival of C. perfringens in mouse muscle tissue. These results suggest PFO-mediated cytotoxicity toward macrophages and the ability to escape macrophage phagosomes may be important factors in the ability of C. perfringens to survive in host tissues when bacterial numbers are low relative to those of phagocytic cells, e.g., early in an infection.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/toxicidade , Clostridium perfringens/patogenicidade , Macrófagos/microbiologia , Macrófagos/patologia , Fosfolipases Tipo C/toxicidade , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular , Células Cultivadas , Infecções por Clostridium/imunologia , Infecções por Clostridium/patologia , Clostridium perfringens/crescimento & desenvolvimento , Feminino , Proteínas Hemolisinas , Macrófagos Peritoneais/microbiologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Fagossomos/microbiologia , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo
11.
Blood ; 94(7): 2461-8, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10498619

RESUMO

Several angiogenesis inhibitors are fragments of larger proteins that are themselves not active as angiogenesis inhibitors. Vasostatin, the N-terminal domain of calreticulin inclusive of amino acids 1-180, is an angiogenesis inhibitor that exerts antitumor effects in vivo. In the present study, we examined whether the full-length calreticulin molecule shares the antiangiogenic and antitumor activities of vasostatin. Similar to vasostatin, calreticulin selectively inhibited endothelial cell proliferation in vitro, but not cells of other lineages, and suppressed angiogenesis in vivo. When inoculated into athymic mice, calreticulin inhibited Burkitt tumor growth comparably with vasostatin. Calreticulin lacking the N-terminal 1-120 amino acids inhibited endothelial cell proliferation in vitro and Burkitt tumor growth in vivo comparably with vasostatin. An internal calreticulin fragment encompassing amino acids 120-180 also inhibited endothelial cell proliferation in vitro and angiogenesis in vivo comparably with calreticulin and vasostatin. These results suggest that the antiangiogenic activities of vasostatin reside in a domain that is accessible from the full-length calreticulin molecule and localize to calreticulin N-terminal amino acids 120-180. Thus, calreticulin and calreticulin fragments are inhibitors of angiogenesis that directly target endothelial cells, inhibit angiogenesis, and suppress tumor growth. This information may be critical in designing targeted inhibitors of pathological angiogenesis that underlies cancer and other diseases.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/uso terapêutico , Linfoma de Burkitt/patologia , Proteínas de Ligação ao Cálcio/farmacologia , Proteínas de Ligação ao Cálcio/toxicidade , Endotélio Vascular/fisiologia , Neovascularização Fisiológica/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/toxicidade , Ribonucleoproteínas/farmacologia , Ribonucleoproteínas/toxicidade , Animais , Antineoplásicos/farmacologia , Linfoma de Burkitt/tratamento farmacológico , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/uso terapêutico , Calreticulina , Bovinos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Feminino , Coração , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/patologia , Neovascularização Patológica/prevenção & controle , Fragmentos de Peptídeos/uso terapêutico , Proteínas Recombinantes/farmacologia , Ribonucleoproteínas/química , Ribonucleoproteínas/uso terapêutico , Transplante Heterólogo , Células Tumorais Cultivadas
12.
Invest Ophthalmol Vis Sci ; 35(8): 3342-5, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8045724

RESUMO

PURPOSE: Recoverin, a calcium-binding protein that selectively localizes to the retina and pineal gland, has been identified as the target for the putative pathogenic autoimmune process of cancer-associated retinopathy (CAR). The present study was aimed at testing the capacity of recoverin to induce experimental autoimmune uveoretinitis and pinealitis in Lewis rats. METHODS: Lewis rats were immunized against recombinant myristoylated recoverin by a single footpad injection of the protein, at various doses, emulsified in complete Freund's adjuvant. Development of uveoretinitis was monitored by clinical and histologic examinations, whereas pinealitis was detected by histologic examination. RESULTS: Immunization with recoverin induced severe panuveitic changes that closely resemble those induced by S-antigen (arrestin). The effect was dose-dependent, with 10 micrograms/rat the lowest immunopathogenic dose. Rats immunized with recoverin also developed pineal inflammation. CONCLUSION: Recoverin is highly immunopathogenic in Lewis rats. Although the ocular changes induced in rats differ from those seen in CAR, the data recorded here are in line with the concept that recoverin can initiate pathogenic autoimmune processes in the eye.


Assuntos
Doenças Autoimunes/imunologia , Proteínas de Ligação ao Cálcio/toxicidade , Proteínas do Olho/toxicidade , Lipoproteínas , Proteínas do Tecido Nervoso , Retinite/imunologia , Uveíte/imunologia , Animais , Doenças Autoimunes/patologia , Relação Dose-Resposta a Droga , Feminino , Hipocalcina , Masculino , Glândula Pineal/patologia , Ratos , Ratos Endogâmicos Lew , Recoverina , Retinite/patologia , Uveíte/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA