Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 13(12)2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-38136650

RESUMO

DAF-FM DA is widely used as a live staining compound to show the presence of nitric oxide (NO) in cells. Applying this stain to live zebrafish embryos is known to indicate early centers of bone formation, but the precise (cellular) location of the signal has hitherto not been revealed. Using sections of zebrafish embryos live-stained with DAF-FM DA, we could confirm that the fluorescent signals were predominantly located in areas of ongoing bone formation. Signals were observed in the bone and tooth matrix, in the notochord sheath, as well as in the bulbus arteriosus. Surprisingly, however, they were exclusively extracellular, even after very short staining times. Von Kossa and Alizarin red S staining to reveal mineral deposits showed that DAF-FM DA stains both the mineralized and non-mineralized bone matrix (osteoid), excluding that DAF-FM DA binds non-specifically to calcified structures. The importance of NO in bone formation by osteoblasts is nevertheless undisputed, as shown by the absence of bone structures after the inhibition of NOS enzymes that catalyze the formation of NO. In conclusion, in zebrafish skeletal biology, DAF-FM DA is appropriate to reveal bone formation in vivo, independent of mineralization of the bone matrix, but it does not demonstrate intracellular NO.


Assuntos
Osteogênese , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Óxido Nítrico/metabolismo , Osso e Ossos/metabolismo , Corantes/metabolismo , Coloração e Rotulagem
2.
Free Radic Biol Med ; 164: 399-409, 2021 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-33476796

RESUMO

Superoxide dismutase 3 (SOD3) is an extracellular protein with the capacity to convert superoxide into hydrogen peroxide, an important secondary messenger in redox regulation. To investigate the utility of zebrafish in functional studies of SOD3 and its relevance for redox regulation, we have characterized the zebrafish orthologues; Sod3a and Sod3b. Our analyses show that both recombinant Sod3a and Sod3b express SOD activity, however, only Sod3b is able to bind heparin. Furthermore, RT-PCR analyses reveal that sod3a and sod3b are expressed in zebrafish embryos and are present primarily in separate organs in adult zebrafish, suggesting distinct functions in vivo. Surprisingly, both RT-PCR and whole mount in situ hybridization showed specific expression of sod3b in skeletal tissue. To further investigate this observation, we compared femoral bone obtained from wild-type and SOD3-/- mice to determine whether a functional difference was apparent in healthy adult mice. Here we report, that bone from SOD3-/- mice is less mineralized and characterized by significant reduction of cortical and trabecular thickness in addition to reduced mechanical strength. These analyses show that SOD3 plays a hitherto unappreciated role in bone development and homeostasis.


Assuntos
Superóxido Dismutase , Peixe-Zebra , Animais , Osso e Ossos/metabolismo , Homeostase , Camundongos , Camundongos Knockout , Oxirredução , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
3.
Redox Biol ; 26: 101268, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31326693

RESUMO

Superoxide dismutase 3 (SOD3) is an extracellular enzyme with the capacity to modulate extracellular redox conditions by catalyzing the dismutation of superoxide to hydrogen peroxide. In addition to synthesis and release of this extracellular protein via the secretory pathway, several studies have shown that the protein also localizes to intracellular compartments in neutrophils and macrophages. Here we show that human macrophages release SOD3 from an intracellular compartment within 30 min following LPS stimulation. This release acutely increases the level of SOD3 on the cell surface as well as in the extracellular environment. Generation of the intracellular compartment in macrophages is supported by endocytosis of extracellular SOD3 via the LDL receptor-related protein 1 (LRP1). Using bone marrow-derived macrophages established from wild-type and SOD3-/- mice, we further show that the pro-inflammatory profile established in LPS-stimulated cells is altered in the absence of SOD3, suggesting that the active release of this protein affects the inflammatory response. The internalization and acute release from stimulated macrophages indicates that SOD3 not only functions as a passive antioxidant in the extracellular environment, but also plays an active role in modulating redox signaling to support biological responses.


Assuntos
Inflamação/etiologia , Inflamação/metabolismo , Macrófagos/metabolismo , Oxirredução , Superóxido Dismutase/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Endocitose , Humanos , Inflamação/patologia , Espaço Intracelular , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout
4.
Free Radic Biol Med ; 97: 478-488, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27394172

RESUMO

Extracellular superoxide dismutase (EC-SOD) is an antioxidant enzyme present in the extracellular matrix (ECM), where it provides protection against oxidative degradation of matrix constituents including type I collagen and hyaluronan. The enzyme is known to associate with macrophages and polymorphonuclear leukocytes (neutrophils) and increasing evidence supports a role for EC-SOD in the development of an inflammatory response. Here we show that human EC-SOD is present at the cell surface of isolated neutrophils as well as stored within secretory vesicles. Interestingly, we find that EC-SOD mRNA is absent throughout neutrophil maturation indicating that the protein is synthesized by other cells and subsequently endocytosed by the neutrophil. When secretory vesicles were mobilized by neutrophil stimulation using formyl-methionyl-leucyl-phenylalanine (fMLF) or phorbol 12-myristate 13-acetate (PMA), the protein was released into the extracellular space and found to associate with DNA released from stimulated cells. The functional consequences were evaluated by the use of neutrophils isolated from wild-type and EC-SOD KO mice, and showed that EC-SOD release significantly reduce the level of superoxide in the extracellular space, but does not affect the capacity to generate neutrophil extracellular traps (NETs). Consequently, our data signifies that EC-SOD released from activated neutrophils affects the redox conditions of the extracellular space and may offer protection against highly reactive oxygen species such as hydroxyl radicals otherwise generated as a result of respiratory burst activity of activated neutrophils.


Assuntos
Neutrófilos/enzimologia , Vesículas Secretórias/enzimologia , Superóxido Dismutase/metabolismo , Animais , Células Cultivadas , Espaço Extracelular/enzimologia , Armadilhas Extracelulares/metabolismo , Expressão Gênica , Humanos , Camundongos , Ativação de Neutrófilo , Neutrófilos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Explosão Respiratória , Superóxido Dismutase/genética
5.
Free Radic Biol Med ; 81: 38-46, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25582887

RESUMO

Extracellular superoxide dismutase (EC-SOD) is expressed by both macrophages and neutrophils and is known to influence the inflammatory response. Upon activation, neutrophils generate hypochlorous acid (HOCl) and secrete proteases to combat invading microorganisms. This produces a hostile environment in which enzymatic activity in general is challenged. In this study, we show that EC-SOD exposed to physiologically relevant concentrations of HOCl remains enzymatically active and retains the heparin-binding capacity, although HOCl exposure established oxidative modification of the N-terminal region (Met32) and the formation of an intermolecular cross-link in a fraction of the molecules. The cross-linking was also induced by activated neutrophils. Moreover, we show that the neutrophil-derived proteases human neutrophil elastase and cathepsin G cleaved the N-terminal region of EC-SOD irrespective of HOCl oxidation. Although the cleavage by elastase did not affect the quaternary structure, the cleavage by cathepsin G dissociated the molecule to produce EC-SOD monomers. The present data suggest that EC-SOD is stable and active at the site of inflammation and that neutrophils have the capacity to modulate the biodistribution of the protein by generating EC-SOD monomers that can diffuse into tissue.


Assuntos
Catepsina G/química , Ácido Hipocloroso/farmacologia , Elastase de Leucócito/química , Macrófagos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Superóxido Dismutase/química , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/enzimologia , Catepsina G/metabolismo , Bovinos , Espaço Extracelular/química , Espaço Extracelular/enzimologia , Heparina/química , Humanos , Ácido Hipocloroso/metabolismo , Elastase de Leucócito/metabolismo , Macrófagos/citologia , Macrófagos/enzimologia , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/enzimologia , Oxirredução , Cultura Primária de Células , Ligação Proteica , Estrutura Quaternária de Proteína/efeitos dos fármacos , Superóxido Dismutase/metabolismo
6.
Free Radic Biol Med ; 69: 348-56, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24512907

RESUMO

Extracellular superoxide dismutase (EC-SOD) is responsible for the dismutation of the superoxide radical produced in the extracellular space and known to be expressed by inflammatory cells, including macrophages and neutrophils. Here we show that EC-SOD is produced by resting macrophages and associated with the cell surface via the extracellular matrix (ECM)-binding region. Upon cellular activation induced by lipopolysaccharide, EC-SOD is relocated and detected both in the cell culture medium and in lipid raft structures. Although the secreted material presented a significantly reduced ligand-binding capacity, this could not be correlated to proteolytic removal of the ECM-binding region, because the integrity of the material recovered from the medium was comparable to that of the cell surface-associated protein. The naturally occurring R213G amino acid substitution located in the ECM-binding region of EC-SOD is known to affect the binding characteristics of the protein. However, the analysis of macrophages expressing R213G EC-SOD did not present evidence of an altered cellular distribution. Our results suggest that EC-SOD plays a dynamic role in the inflammatory response mounted by activated macrophages.


Assuntos
Inflamação/genética , Macrófagos/enzimologia , Neutrófilos/enzimologia , Superóxido Dismutase/genética , Substituição de Aminoácidos/genética , Animais , Matriz Extracelular/efeitos dos fármacos , Espaço Extracelular/enzimologia , Humanos , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Macrófagos/ultraestrutura , Camundongos , Mutação , Neutrófilos/ultraestrutura , Ligação Proteica/efeitos dos fármacos , Superóxido Dismutase/química , Superóxido Dismutase/ultraestrutura , Superóxidos/metabolismo
7.
Redox Biol ; 1: 24-31, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24024135

RESUMO

Superoxide dismutase (EC-SOD) controls the level of superoxide in the extracellular space by catalyzing the dismutation of superoxide into hydrogen peroxide and molecular oxygen. In addition, the enzyme reacts with hydrogen peroxide in a peroxidase reaction which is known to disrupt enzymatic activity. Here, we show that the peroxidase reaction supports a site-specific bond cleavage. Analyses by peptide mapping and mass spectrometry shows that oxidation of Pro112 supports the cleavage of the Pro112-His113 peptide bond. Substitution of Ala for Pro112 did not inhibit fragmentation, indicating that the oxidative fragmentation at this position is dictated by spatial organization and not by side-chain specificity. The major part of EC-SOD inhibited by the peroxidase reaction was not fragmented but found to encompass oxidations of histidine residues involved in the coordination of copper (His98 and His163). These oxidations are likely to support the dissociation of copper from the active site and thus loss of enzymatic activity. Homologous modifications have also been described for the intracellular isozyme, Cu/Zn-SOD, reflecting the almost identical structures of the active site within these enzymes. We speculate that the inactivation of EC-SOD by peroxidase activity plays a role in regulating SOD activity in vivo, as even low levels of superoxide will allow for the peroxidase reaction to occur.


Assuntos
Peróxido de Hidrogênio/metabolismo , Peroxidase/metabolismo , Superóxido Dismutase/antagonistas & inibidores , Superóxido Dismutase/metabolismo , Domínio Catalítico , Cobre/metabolismo , Histidina , Humanos , Espectrometria de Massas , Modelos Moleculares , Oxirredução , Mapeamento de Peptídeos , Superóxido Dismutase/química , Zinco/metabolismo
8.
Free Radic Biol Med ; 52(1): 191-7, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22062630

RESUMO

The antioxidant protein extracellular superoxide dismutase (EC-SOD) encompasses a C-terminal region that mediates interactions with a number of ligands in the extracellular matrix (ECM). This ECM-binding region can be removed by limited proteolysis before secretion, thus supporting the formation of EC-SOD tetramers with variable binding capacity. The ECM-binding region contains a cysteine residue (Cys219) that is known to be involved in an intersubunit disulfide bridge. We have determined the redox potential of this disulfide bridge and show that both EC-SOD dimers and EC-SOD monomers are present within the intracellular space. The proteolytic processing of the ECM-binding region in vitro was modulated by the redox status of Cys219, allowing cleavage under reducing conditions only. When wild-type EC-SOD or the monomeric variant Cys219Ser was expressed in mammalian cells proteolysis did not occur. However, when cells were exposed to oxidative stress conditions, proteolytic processing was observed for wild-type EC-SOD but not for the Cys219Ser variant. Although the cellular response to oxidative stress is complex, our data suggest that proteolytic removal of the ECM-binding region is regulated by the intracellular generation of an EC-SOD monomer and that Cys219 plays an important role as a redox switch allowing the cellular machinery to secrete cleaved EC-SOD.


Assuntos
Aorta/enzimologia , Cisteína/metabolismo , Estresse Oxidativo , Subunidades Proteicas/metabolismo , Proteólise , Superóxido Dismutase/metabolismo , Sequência de Aminoácidos , Aorta/citologia , Sítios de Ligação , Cromatografia de Afinidade , Dimerização , Dissulfetos/metabolismo , Matriz Extracelular/metabolismo , Espaço Extracelular/metabolismo , Células HEK293 , Humanos , Dados de Sequência Molecular , Oxirredução , Ligação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/genética , Superóxido Dismutase/química , Superóxido Dismutase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA