Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Int J Mol Sci ; 25(15)2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39126017

RESUMO

Pseudomonas aeruginosa is an opportunistic pathogen that causes severe infections in compromised hosts. P. aeruginosa infections are difficult to treat because of the inherent ability of the bacteria to develop antibiotic resistance, secrete a variety of virulence factors, and form biofilms. The secreted aminopeptidase (PaAP) is an emerging virulence factor, key in providing essential low molecular weight nutrients and a cardinal modulator of biofilm development. PaAP is therefore a new potential target for therapy of P. aeruginosa infections. The present review summarizes the current knowledge of PaAP, with special emphasis on its biochemical and enzymatic properties, activation mechanism, biological roles, regulation, and structure. Recently developed specific inhibitors and their potential as adjuncts in the treatment of P. aeruginosa infections are also described.


Assuntos
Aminopeptidases , Pseudomonas aeruginosa , Fatores de Virulência , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/patogenicidade , Pseudomonas aeruginosa/metabolismo , Aminopeptidases/metabolismo , Humanos , Fatores de Virulência/metabolismo , Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/tratamento farmacológico , Animais
2.
Int J Mol Sci ; 20(20)2019 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-31658765

RESUMO

In this study, we characterize the interactions between the extracellular matrix protein, procollagen C-proteinase enhancer-1 (PCPE-1), and glycosaminoglycans (GAGs), which are linear anionic periodic polysaccharides. We applied molecular modeling approaches to build a structural model of full-length PCPE-1, which is not experimentally available, to predict GAG binding poses for various GAG lengths, types and sulfation patterns, and to determine the effect of calcium ions on the binding. The computational data are analyzed and discussed in the context of the experimental results previously obtained using surface plasmon resonance binding assays. We also provide experimental data on PCPE-1/GAG interactions obtained using inhibition assays with GAG oligosaccharides ranging from disaccharides to octadecasaccharides. Our results predict the localization of GAG-binding sites at the amino acid residue level onto PCPE-1 and is the first attempt to describe the effects of ions on protein-GAG binding using modeling approaches. In addition, this study allows us to get deeper insights into the in silico methodology challenges and limitations when applied to GAG-protein interactions.


Assuntos
Cálcio/química , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Íons , Modelos Moleculares , Simulação de Acoplamento Molecular , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas
3.
Clin Exp Rheumatol ; 34 Suppl 100(5): 23-29, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26886502

RESUMO

OBJECTIVES: γδ T cells of the Vγ9Vδ2 subtype secrete anti-fibrotic cytokines upon isopentenyl pyrophosphate (IPP) stimulation. In this study, we sought to compare IPP and Zoledronate, an up-regulator of IPP, effects on proliferation and cytokine secretion of Vγ9+ T cells from systemic sclerosis (SSc) patients and healthy controls (HCs). We also examined the effect of IPP-triggered peripheral blood mononuclear cells (PBMC) on fibroblast procolla- gen secretion. METHODS: PBMC from SSc patients and HCs were stimulated by increasing concentrations of Zoledronate, with or without IPP, and Vγ9+ T cell percentages were calculated using FACScan analysis. Subsequently, PBMC were cultured with IPP or toxic shock syndrome toxin-1 (TSST-1), and contents of the anti-fibrotic cytokines tumour necrosis factor (TNF)-α and interferon (IFN)-γ were measured by ELISA kits. Finally, supernatants of IPP-triggered Vγ9+ T cells from SSc patients were added to fibroblast cultures, and relative intensities of procollagen α1 chains were determined by densinometry. RESULTS: Higher concentrations of Zoledronate were required for maximal proliferation of Vγ9+ T cells in 9 SSc patients compared to 9 HCs, irrespective of exogenous IPP. When compared to stimulation by TSST-1, a non-Vγ9+ selective reagent, secretion of the anti-fibrotic cytokines TNF-α and IFN-γ in response to IPP was relatively diminished in SSc but not in HCs. Reduction of procollagen secretion by fibroblasts cultured with supernatants of IPP-stimulated PBMC was observed only in some SSc patients. CONCLUSIONS: Activated Vγ9+ T cells could act as anti-fibrotic mediators in SSc, although decreased responsiveness to IPP may play a role in the pathological fibrosis of this disease.


Assuntos
Ativação Linfocitária , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Escleroderma Sistêmico/imunologia , Subpopulações de Linfócitos T/imunologia , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Colágeno Tipo I/metabolismo , Difosfonatos/farmacologia , Relação Dose-Resposta a Droga , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibrose , Hemiterpenos/farmacologia , Humanos , Imidazóis/farmacologia , Interferon gama/imunologia , Interferon gama/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Compostos Organofosforados/farmacologia , Fenótipo , Pró-Colágeno/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Escleroderma Sistêmico/metabolismo , Escleroderma Sistêmico/patologia , Transdução de Sinais , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Ácido Zoledrônico
4.
Biochem J ; 457(1): 137-49, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24117177

RESUMO

PCPE-1 (procollagen C-proteinase enhancer-1) is an extracellular matrix glycoprotein that can stimulate procollagen processing by procollagen C-proteinases such as BMP-1 (bone morphogenetic protein 1). PCPE-1 interacts with several proteins in addition to procollagens and BMP-1, suggesting that it could be involved in biological processes other than collagen maturation. We thus searched for additional partners of PCPE-1 in the extracellular matrix, which could provide new insights into its biological roles. We identified 17 new partners of PCPE-1 by SPR (surface plasmon resonance) imaging. PCPE-1 forms a transient complex with the ß-amyloid peptide, whereas it forms high or very high affinity complexes with laminin-111 (KD=58.8 pM), collagen VI (KD=9.5 nM), TSP-1 (thrombospondin-1) (KD1=19.9 pM, KD2=14.5 nM), collagen IV (KD=49.4 nM) and endostatin, a fragment of collagen XVIII (KD1=0.30 nM, KD2=1.1 nM). Endostatin binds to the NTR (netrin-like) domain of PCPE-1 and decreases the degree of superstimulation of PCPE-1 enhancing activity by heparin. The analysis of the PCPE-1 interaction network based on Gene Ontology terms suggests that, besides its role in collagen deposition, PCPE-1 might be involved in tumour growth, neurodegenerative diseases and angiogenesis. In vitro assays have indeed shown that the CUB1CUB2 (where CUB is complement protein subcomponents C1r/C1s, urchin embryonic growth factor and BMP-1) fragment of PCPE-1 inhibits angiogenesis.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Mapas de Interação de Proteínas , Cálcio/farmacologia , Endostatinas/metabolismo , Proteínas da Matriz Extracelular/química , Ontologia Genética , Glicoproteínas/química , Células HEK293 , Heparina/metabolismo , Humanos , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Neovascularização Fisiológica , Ligação Proteica/efeitos dos fármacos , Ressonância de Plasmônio de Superfície
5.
Graefes Arch Clin Exp Ophthalmol ; 250(2): 223-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21947265

RESUMO

BACKGROUND: Therapy of S. aureus keratitis is increasingly challenging due to emerging resistant strains. Staphylolysin (LasA protease) is a staphylolytic endopeptidase secreted by Pseudomonas aeruginosa. The purpose of the current study was to study the effect of treatment with staphylolysin on experimental keratitis caused by various Staphylococcus aureus strains. METHODS: The therapeutic effect was studied in a keratitis model induced in rabbits by intrastromal injections of 10(3) S. aureus cells of three different methicillin-resistant S. aureus (MRSA) strains and one methicillin-susceptible S. aureus strain (MSSA). Topical treatment with either staphylolysin or bovine serum albumin (BSA; control) was applied every half hour for 5 h, starting at 4 h after infection. Corneas were removed for bacterial quantification. Histopathological analysis was performed on MSSA-infected rabbits, killed at either one or 84 h after completion of treatment and on uninfected eyes 1 h after treatment termination. RESULTS: The number of bacteria in the staphylolysin-treated corneas was significantly reduced in all infections with the four S. aureus strains studied as compared to controls: the staphylolysin-treated eyes infected with MRSA strains were either completely sterilized or showed a 3-4 orders of magnitude decrease in the number of cfu/cornea (p = 0.004 to 0.005); all of the staphylolysin-treated MSSA-infected eyes were sterile. Histopathological analysis of the methicillin-sensitive (MSSA) strain-infected eyes at 84 h after completion of treatment showed moderate inflammation in the staphylolysin-treated eyes as compared with extensive abscess formation in the control group. The uninfected corneas showed only mild stromal edema in both the staphylolysin and BSA-treated groups. CONCLUSIONS: Staphylolysin provided long-lasting protection against several strains of S. aureus, evident by both its strong anti-bacterial activity and beneficial histopathological results of treatment.


Assuntos
Úlcera da Córnea/tratamento farmacológico , Endotoxinas/uso terapêutico , Infecções Oculares Bacterianas/tratamento farmacológico , Staphylococcus aureus Resistente à Meticilina/fisiologia , Infecções Estafilocócicas/tratamento farmacológico , Administração Tópica , Animais , Contagem de Colônia Microbiana , Córnea/microbiologia , Úlcera da Córnea/microbiologia , Modelos Animais de Doenças , Endotoxinas/administração & dosagem , Infecções Oculares Bacterianas/microbiologia , Coelhos , Infecções Estafilocócicas/microbiologia , Resultado do Tratamento
6.
Biochem J ; 433(2): 371-81, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20979576

RESUMO

The predominant form of type V collagen is the [α1(V)]2α2(V) heterotrimer. Mutations in COL5A1 or COL5A2, encoding respectively the α1(V)- and α2(V)-collagen chain, cause classic EDS (Ehlers-Danlos syndrome), a heritable connective tissue disorder, characterized by fragile hyperextensible skin and joint hypermobility. Approximately half of the classic EDS cases remain unexplained. Type V collagen controls collagen fibrillogenesis through its conserved α1(V)-N-propeptide domain. To gain an insight into the role of this domain, a yeast two-hybrid screen among proteins expressed in human dermal fibroblasts was performed utilizing the N-propeptide as a bait. We identified 12 interacting proteins, including extracellular matrix proteins and proteins involved in collagen biosynthesis. Eleven interactions were confirmed by surface plasmon resonance and/or co-immunoprecipitation: α1(I)- and α2(I)-collagen chains, α1(VI)-, α2(VI)- and α3(VI)-collagen chains, tenascin-C, fibronectin, PCPE-1 (procollagen C-proteinase enhancer-1), TIMP-1 (tissue inhibitor of metalloproteinases-1), MMP-2 (matrix metalloproteinase 2) and TGF-ß1 (transforming growth factor ß1). Solid-phase binding assays confirmed the involvement of the α1(V)-N-propeptide in the interaction between native type V collagen and type VI collagen, suggesting a bridging function of this protein complex in the cell-matrix environment. Enzymatic studies showed that processing of the α1(V)-N-propeptide by BMP-1 (bone morphogenetic protein 1)/procollagen C-proteinase is enhanced by PCPE-1. These interactions are likely to be involved in extracellular matrix homoeostasis and their disruption could explain the pathogenetic mechanism in unresolved classic EDS cases.


Assuntos
Colágeno Tipo V/metabolismo , Síndrome de Ehlers-Danlos/metabolismo , Peptídeos/metabolismo , Pró-Colágeno/metabolismo , Células HEK293 , Homeostase , Humanos , Ligação Proteica
7.
Hum Mutat ; 32(6): 598-609, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21344539

RESUMO

Osteogenesis imperfecta (OI) is most often caused by mutations in the type I procollagen genes (COL1A1/COL1A2). We identified two children with substitutions in the type I procollagen C-propeptide cleavage site, which disrupt a unique processing step in collagen maturation and define a novel phenotype within OI. The patients have mild OI caused by mutations in COL1A1 (Patient 1: p.Asp1219Asn) or COL1A2 (Patient 2: p.Ala1119Thr), respectively. Patient 1 L1-L4 DXA Z-score was +3.9 and pQCT vBMD was+3.1; Patient 2 had L1-L4 DXA Z-score of 0.0 and pQCT vBMD of -1.8. Patient BMD contrasts with radiographic osteopenia and histomorphometry without osteosclerosis. Mutant procollagen processing is impaired in pericellular and in vitro assays. Patient dermal collagen fibrils have irregular borders. Incorporation of pC-collagen into matrix leads to increased bone mineralization. FTIR imaging confirms elevated mineral/matrix ratios in both patients, along with increased collagen maturation in trabecular bone, compared to normal or OI controls. Bone mineralization density distribution revealed a marked shift toward increased mineralization density for both patients. Patient 1 has areas of higher and lower bone mineralization than controls; Patient 2's bone matrix has a mineral content exceeding even classical OI bone. These patients define a new phenotype of high BMD OI and demonstrate that procollagen C-propeptide cleavage is crucial to normal bone mineralization.


Assuntos
Osso e Ossos/anormalidades , Osso e Ossos/patologia , Colágeno Tipo I/genética , Osteogênese Imperfeita/genética , Osteogênese Imperfeita/patologia , Fragmentos de Peptídeos/genética , Pró-Colágeno/genética , Adolescente , Sequência de Aminoácidos , Animais , Densidade Óssea/genética , Matriz Óssea , Calcificação Fisiológica/genética , Criança , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Feminino , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Mutação , Fragmentos de Peptídeos/metabolismo , Fenótipo , Pró-Colágeno/metabolismo
8.
J Biol Chem ; 285(44): 33867-74, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20729553

RESUMO

Procollagen C-proteinase enhancer-1 (PCPE-1) is an extracellular matrix (ECM) glycoprotein that can stimulate procollagen processing by procollagen C-proteinases (PCPs) such as bone morphogenetic protein-1 (BMP-1). The PCPs can process additional extracellular protein precursors and play fundamental roles in developmental processes and assembly of the ECM. The stimulatory activity of PCPE-1 is restricted to the processing of fibrillar procollagens, suggesting PCPE-1 is a specific regulator of collagen deposition. PCPE-1 consists of two CUB domains that bind to the procollagen C-propeptides and are required for PCP enhancing activity, and one NTR domain that binds heparin. To understand the biological role of the NTR domain, we performed surface plasmon resonance (SPR) binding assays, cell attachment assays as well as immunofluorescence and activity assays, all indicating that the NTR domain can mediate PCPE-1 binding to cell surface heparan sulfate proteoglycans (HSPGs). The SPR data revealed binding affinities to heparin/HSPGs in the high nanomolar range and dependence on calcium. Both 3T3 mouse fibroblasts and human embryonic kidney cells (HEK-293) attached to PCPE-1, an interaction that was inhibited by heparin. Cell attachment was also inhibited by an NTR-specific antibody and the NTR fragment. Immunofluorescence analysis revealed that PCPE-Flag binds to mouse fibroblasts and heparin competes for this binding. Cell-associated PCPE-Flag stimulated procollagen processing by BMP-1 several fold. Our data suggest that through interaction with cell surface HSPGs, the NTR domain can anchor PCPE-1 to the cell membrane, permitting pericellular enhancement of PCP activity. This points to the cell surface as a physiological site of PCPE-1 action.


Assuntos
Proteínas da Matriz Extracelular/química , Glicoproteínas/química , Heparina/química , Heparitina Sulfato/química , Células 3T3 , Animais , Linhagem Celular , Colágeno/química , Humanos , Cinética , Camundongos , Microscopia de Fluorescência/métodos , Pró-Colágeno/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
9.
J Biol Chem ; 285(21): 15950-9, 2010 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-20207734

RESUMO

The netrin-like (NTR) domain is a feature of several extracellular proteins, most notably the N-terminal domain of tissue inhibitors of metalloproteinases (TIMPs), where it functions as a strong inhibitor of matrix metalloproteinases and some other members of the metzincin superfamily. The presence of a C-terminal NTR domain in procollagen C-proteinase enhancers (PCPEs), proteins that stimulate the activity of astacin-like tolloid proteinases, raises the possibility that this might also have inhibitory activity. Here we show that both long and short forms of the PCPE-1 NTR domain, the latter beginning at the N-terminal cysteine known to be critical for TIMP activity, show no inhibition, at micromolar concentrations, of several members of the metzincin superfamily, including matrix metalloproteinase-2, bone morphogenetic protein-1 (a tolloid proteinase), and different ADAMTS (a disintegrin and a metalloproteinase with thrombospondin motifs) proteinases from the adamalysin family. In contrast, we report that the NTR domain within PCPE-1 leads to superstimulation of bone morphogenetic protein-1 activity in the presence of heparin and heparan sulfate. These observations point to a new mechanism whereby binding to cell surface-associated or extracellular heparin-like sulfated glycosaminoglycans might provide a means to accelerate procollagen processing in specific cellular and extracellular microenvironments.


Assuntos
Proteínas ADAM/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Metaloproteases Semelhantes a Toloide/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Linhagem Celular , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/genética , Glicoproteínas/química , Glicoproteínas/genética , Humanos , Pró-Colágeno/química , Pró-Colágeno/genética , Pró-Colágeno/metabolismo , Estrutura Terciária de Proteína , Inibidores Teciduais de Metaloproteinases/química , Inibidores Teciduais de Metaloproteinases/genética , Inibidores Teciduais de Metaloproteinases/metabolismo , Metaloproteases Semelhantes a Toloide/química , Metaloproteases Semelhantes a Toloide/genética
10.
PLoS One ; 16(6): e0252970, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34133429

RESUMO

Pseudomonas aeruginosa secretes several endopeptidases, including elastase, alkaline proteinase (Apr), a lysine-specific endopeptidase (LysC), and an aminopeptidase (PaAP), all of which are important virulence factors. Activation of the endopeptidases requires removal of an inhibitory N-terminal propeptide. Activation of pro-PaAP, in contrast, requires C-terminal processing. The activating proteases of pro-PaAP and their cleavage site(s) have not yet been defined. Studying pro-PaAP processing in a wild type P. aeruginosa strain and strains lacking either elastase or both elastase and Apr, we detected three processing variants, each ~56 kDa in size (AP56). Activity assays and N- and C-terminal sequence analyses of these variants pointed at LysC as the principal activating protease, cleaving a Lys512-Ala513 peptide bond at the C-terminal end of pro-PaAP. Elastase and/or Apr are required for activation of LysC, suggesting both are indirectly involved in activation of PaAP. To shed light on the function(s) of the N-terminal domain of AP56, we purified recombinant AP56 and generated from it the 28 kDa catalytic domain (AP28). The kinetic constants (Km and Kcat) for hydrolysis of Leu-, Lys-, Arg- and Met-p-nitroanilide (pNA) derivatives by AP56 and AP28 were then determined. The catalytic coefficients (Kcat/Km) for hydrolysis of all four substrates by AP28 and AP56 were comparable, indicating that the non-catalytic domain is not involved in hydrolysis of small substrates. It may, however, regulate hydrolysis of natural peptides/proteins. Lys-pNA was hydrolyzed 2 to 3-fold more rapidly than Leu-pNA and ~8-fold faster than Arg- or Met-pNA, indicating that Lys-pNA was the preferred substrate.


Assuntos
Aminopeptidases/metabolismo , Metaloendopeptidases/metabolismo , Elastase Pancreática/metabolismo , Proteólise , Pseudomonas aeruginosa/enzimologia , Sequência de Aminoácidos , Aminopeptidases/genética , Domínio Catalítico , Hidrólise , Cinética , Metaloendopeptidases/genética , Elastase Pancreática/genética , Domínios Proteicos , Pseudomonas aeruginosa/crescimento & desenvolvimento , Especificidade por Substrato
11.
Biol Chem ; 391(6): 705-16, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20370321

RESUMO

Elafin is a 6-kDa innate immune protein present at several epithelial surfaces including the pulmonary epithelium. It is a canonical protease inhibitor of two neutrophil serine proteases [neutrophil elastase (NE) and proteinase 3] with the capacity to covalently bind extracellular matrix proteins by transglutamination. In addition to these properties, elafin also possesses antimicrobial and immunomodulatory activities. The aim of the present study was to investigate the effect of Pseudomonas aeruginosa proteases on elafin function. We found that P. aeruginosa PAO1-conditioned medium and two purified Pseudomonas metalloproteases, pseudolysin (elastase) and aeruginolysin (alkaline protease), are able to cleave recombinant elafin. Pseudolysin was shown to inactivate the anti-NE activity of elafin by cleaving its protease-binding loop. Interestingly, antibacterial properties of elafin against PAO1 were found to be unaffected after pseudolysin treatment. In contrast to pseudolysin, aeruginolysin failed to inactivate the inhibitory properties of elafin against NE. Aeruginolysin cleaves elafin at the amino-terminal Lys6-Gly7 peptide bond, resulting in a decreased ability to covalently bind purified fibronectin following transglutaminase activity. In conclusion, this study provides evidence that elafin is susceptible to proteolytic cleavage at alternative sites by P. aeruginosa metalloproteinases, which can affect different biological functions of elafin.


Assuntos
Proteínas de Bactérias/metabolismo , Elafina/metabolismo , Metaloproteases/metabolismo , Pseudomonas aeruginosa/enzimologia , Western Blotting , Cromatografia Líquida de Alta Pressão , Eletroforese em Gel de Poliacrilamida , Fibronectinas/metabolismo , Humanos , Espectrometria de Massas
12.
J Biomed Biotechnol ; 2010: 376927, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20625483

RESUMO

Collagen V, a fibrillar collagen with important functions in tissues, assembles into distinct chain associations. The most abundant and ubiquitous molecular form is the heterotrimer [alpha1(V)](2)alpha2(V). In the attempt to produce high levels of recombinant collagen V heterotrimer for biomedical device uses, and to identify key factors that drive heterotrimeric chain association, several cell expression systems (yeast, insect, and mammalian cells) have been assayed by cotransfecting the human proalpha1(V) and proalpha2(V) chain cDNAs. Suprisingly, in all recombinant expression systems, the formation of [alpha1(V)](3) homotrimers was considerably favored over the heterotrimer. In addition, pepsin-sensitive proalpha2(V) chains were found in HEK-293 cell media indicating that these cells lack quality control proteins preventing collagen monomer secretion. Additional transfection with Hsp47 cDNA, encoding the collagen-specific chaperone Hsp47, did not increase heterotrimer production. Double immunofluorescence with antibodies against collagen V alpha-chains showed that, contrary to fibroblasts, collagen V alpha-chains did not colocalized intracellularly in transfected cells. Monensin treatment had no effect on the heterotrimer production. The heterotrimer production seems to require specific machinery proteins, which are not endogenously expressed in the expression systems. The different constructs and transfected cells we have generated represent useful tools to further investigate the mechanisms of collagen trimer assembly.


Assuntos
Colágeno Tipo V/biossíntese , Multimerização Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Animais , Linhagem Celular , Células Clonais , Colágeno Tipo V/química , Imunofluorescência , Proteínas de Choque Térmico HSP47/metabolismo , Humanos , Insetos/citologia , Pichia/metabolismo
13.
Anat Rec (Hoboken) ; 303(6): 1670-1679, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31136096

RESUMO

Removal of the C-propeptide from fibrillar procollagens is essential for collagen fibril assembly. The reaction is catalyzed by bone morphogenetic protein-1/tolloid-like proteinases and is accelerated by procollagen C-proteinase enhancer 1 (PCPE-1), an extracellular matrix glycoprotein that binds to the procollagen C-propeptide and its expression overlaps that of collagen. Ascorbic acid (Asc) is vital for collagen hydroxylation, folding, and secretion. It also increases collagen gene expression. The role of Asc as a regulator of PCPE-1 expression is debatable. To shed further light on this matter, herein, we studied the effects of Asc on PCPE-1 expression, secretion, and cellular localization in Rat2 and/or mouse 3T3 fibroblasts. Asc increased PCPE-1 expression at the translational and transcriptional levels about two-fold. It also increased the rate of PCPE-1 secretion approximately six-fold. Endogenous PCPE-1 was found to be cell associated, and Asc increased the amount of PCPE-1 on the cell surface. In the absence of PCPE-1 hydroxylation, we propose that the dependence of PCPE-1 secretion on Asc may be related to its role in procollagen secretion. Localization of PCPE-1 to the cell membrane favors the cell-surface as a physiological site of PCPE-1 action. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.


Assuntos
Ácido Ascórbico/farmacologia , Membrana Celular/efeitos dos fármacos , Colágeno/biossíntese , Proteínas da Matriz Extracelular/metabolismo , Animais , Membrana Celular/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Camundongos , Ratos
14.
Graefes Arch Clin Exp Ophthalmol ; 247(7): 913-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19280208

RESUMO

BACKGROUND: Therapy of S. aureus ocular infections is increasingly challenging due to emerging resistant strains. Staphylolysin (also called LasA protease) is a staphylolytic endopeptidase secreted by Pseudomonas aeruginosa. The purpose of this study was to evaluate the efficacy of staphylolysin as a therapy for experimental methicillin-resistant Staphylococcus aureus (MRSA) endophthalmitis, focusing on its bactericidal activity. METHODS: Endophthalmitis was induced in the right eyes of 46 rats by an intravitreal injection of 50-160 MRSA cells. Two therapeutic regimens were evaluated: (i) an intravitreal injection of staphylolysin at 6 hours post-infection; (ii) two successive intravitreal injections of staphylolysin given at 6 and 30 hours post-infection. Control eyes were injected with vehicle alone at the same times. The rats were sacrificed 48 hours after infection, and the vitreous was withdrawn for determination of colony forming units (CFU). Potential adverse effects of intravitreal staphylolysin injection were assessed histopathologically in four uninfected eyes, enucleated from rats sacrificed 1 month after intravitreal staphylolysin injection. RESULTS: In eyes treated by the single-injection regimen, staphylolysin reduced the mean CFU value per vitreous threefold as compared to control (2,055 +/- 3,144 and 6,432 +/- 6,389 CFU/vitreous, respectively; P = 0.02). The repeated injection protocol was more effective, reducing the mean CFU value per vitreous by two orders of magnitude as compared to control (1,148 +/- 3,096 and 143,519 +/- 151,358 CFU/vitreous, respectively; P = 0.0005). Histopathological analysis showed no structural damage in eyes injected intravitreally with staphylolysin. CONCLUSIONS: Staphylolysin is effective in the treatment of experimental MRSA-induced endophthalmitis in rats, and causes no morphological adverse effects to ocular tissues. Staphylolysin may be beneficial in the treatment of S. aureus endophthalmitis in humans.


Assuntos
Endoftalmite/microbiologia , Endotoxinas/metabolismo , Endotoxinas/uso terapêutico , Staphylococcus aureus Resistente à Meticilina , Pseudomonas aeruginosa/enzimologia , Infecções Estafilocócicas/tratamento farmacológico , Animais , Contagem de Colônia Microbiana , Esquema de Medicação , Endotoxinas/administração & dosagem , Injeções , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Ratos , Ratos Wistar , Resultado do Tratamento , Corpo Vítreo/microbiologia
15.
Methods Mol Biol ; 1944: 189-201, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30840244

RESUMO

Fibrosis is characterized by excessive deposition of collagen and additional extracellular matrix (ECM) components in response to chronic injuries. Liver fibrosis often results from chronic hepatitis C virus infection and alcohol abuse that can deteriorate to cirrhosis and liver failure. Current noninvasive diagnostic methods of liver fibrosis are limited in their ability to detect and differentiate between early and intermediate stages of fibrosis. New biomarkers of fibrosis that reflect ECM turnover are therefore badly needed. Procollagen C-proteinase enhancer 1 (PCPE-1), a connective tissue glycoprotein that functions as a positive regulator of C-terminal procollagen processing and subsequent collagen fibril assembly, is a promising candidate. Its tissue distribution and expression profile overlap those of collagen, and its expression in fibrosis is upregulated in parallel to the increase in collagen expression. The potential of PCPE-1 as a biomarker of liver fibrosis was recently established using a CCl4 mouse model of liver fibrosis by showing that the increase in collagen and PCPE-1 content in the fibrotic mouse liver was reflected by elevated plasma levels of PCPE-1. This was achieved using a newly developed highly sensitive, specific, accurate, and reproducible ELISA for mouse PCPE-1, which is based on commercially available antibodies and is offered as a new research tool in the field. A similar ELISA test was developed for human PCPE-1, and preliminary results with plasma from liver fibrosis patients revealed increased plasma concentrations of PCPE-1 in some patients. The protocols of both ELISA tests are outlined herein in great detail to permit their application by any laboratory with similar interests.


Assuntos
Ensaio de Imunoadsorção Enzimática/métodos , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Glicoproteínas/metabolismo , Cirrose Hepática/diagnóstico , Cirrose Hepática/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Humanos , Camundongos
16.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2210-2223, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31055083

RESUMO

Mutations in the type I procollagen C-propeptide occur in ~6.5% of Osteogenesis Imperfecta (OI) patients. They are of special interest because this region of procollagen is involved in α chain selection and folding, but is processed prior to fibril assembly and is absent in mature collagen fibrils in tissue. We investigated the consequences of seven COL1A1 C-propeptide mutations for collagen biochemistry in comparison to three probands with classical glycine substitutions in the collagen helix near the C-propeptide and a normal control. Procollagens with C-propeptide defects showed the expected delayed chain incorporation, slow folding and overmodification. Immunofluorescence microscopy indicated that procollagen with C-propeptide defects was mislocalized to the ER lumen, in contrast to the ER membrane localization of normal procollagen and procollagen with helical substitutions. Notably, pericellular processing of procollagen with C-propeptide mutations was defective, with accumulation of pC-collagen and/or reduced production of mature collagen. In vitro cleavage assays with BMP-1 ±â€¯PCPE-1 confirmed impaired C-propeptide processing of procollagens containing mutant proα1(I) chains. Overmodified collagens were incorporated into the matrix in culture. Dermal fibrils showed alterations in average diameter and diameter variability and bone fibrils were disorganized. Altered ER-localization and reduced pericellular processing of defective C-propeptides are expected to contribute to abnormal osteoblast differentiation and matrix function, respectively.


Assuntos
Colágeno Tipo I/genética , Retículo Endoplasmático/metabolismo , Pró-Colágeno/metabolismo , Varredura Diferencial de Calorimetria , Células Cultivadas , Colágeno Tipo I/química , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Microscopia de Fluorescência , Mutação de Sentido Incorreto , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Estrutura Terciária de Proteína
17.
Data Brief ; 14: 777-781, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28948200

RESUMO

This article provides a protocol for determination of human procollagen C-proteinase enhancer 1 (PCPE-1) concentrations by ELISA. The inter-assay and intra-assay coefficients of variability are given and so are the average plasma concentrations of PCPE-1 in healthy (control) individuals and liver fibrosis patients.

18.
Int J Biochem Cell Biol ; 38(3): 358-65, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16300990

RESUMO

Excessive collagen deposition is a common complication of myocardial infarction that causes progressive heart disease. Several pro-fibrotic cytokines and hormones, including aldosterone, control this process. Procollagen processing by procollagen C-proteinase(s) is critical for collagen deposition and is potentiated by procollagen C-proteinase enhancer proteins (PCPEs). We have shown previously that, in addition to stimulation of collagen I expression, aldosterone increases PCPE-1 expression in cultured heart fibroblasts. The present study was designed to examine whether aldosterone acts similarly in vivo. Rats underwent coronary artery ligation to induce myocardial infarction. They were then left either untreated (control) or treated with spironolactone (an aldosterone receptor antagonist) for 5 weeks when they were sacrificed and their hearts removed for analysis. In situ hybridization co-localized PCPE-1 and collagen I mRNAs to fibroblasts surrounding the scar region and adjacent blood vessels. The levels of both transcripts in the remodeling myocardium of untreated rats increased twofold as compared to sham-operated controls, an increase greatly reduced by spironolactone. Correspondingly, a 2-5 fold increase in PCPE-1 and collagen I was observed in the hearts of untreated rats as compared to both the spironolactone-treated and sham-operated controls. The results establish aldosterone as a physiological stimulator of PCPE-1 expression in the remodeling myocardium after infarction. Since PCPE-1 itself is a positive regulator of collagen deposition, this finding suggests PCPE-1 as a new potential target for intervention with cardiac fibrosis.


Assuntos
Aldosterona/farmacologia , Glicoproteínas/metabolismo , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Remodelação Ventricular/fisiologia , Animais , Peso Corporal , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Glicoproteínas/genética , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Tamanho do Órgão , Distribuição Aleatória , Ratos , Ratos Wistar , Espironolactona/farmacologia
19.
Data Brief ; 9: 883-887, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27872885

RESUMO

This article provides kinetic constants for C-terminal processing of procollagen type I by bone morphogenetic protein 1 (BMP-1; the major procollagen C-proteinase), a reaction stimulated by the connective tissue glycoprotein procollagen C-proteinase enhancer 1 (PCPE-1). Reported are Km , Vmax , Kcat and Kcat /Km (catalytic coefficient) values for BMP-1 alone, BMP-1 with intact PCPE-1, BMP-1 with the CUB (Complement C1r/C1s, Uegf, BMP-1) domains fragment of PCPE-1 as well as its NTR (netrin-like) domain.

20.
PLoS One ; 11(9): e0162747, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27598301

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0159606.].

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa