Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
STAR Protoc ; 4(3): 102465, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37481728

RESUMO

Herein, we provide a protocol for visualizing active osteoclast cathepsin K (CatK) with the quenched-fluorescent-activity-based probe qTJK17. We describe steps for isolating peripheral blood mononuclear cells, their differentiation into osteoclasts, and TRAP staining using an acid phosphatase leukocyte kit. We then detail visualization of active CatK. The probe qTJK17 includes a reactive group, acyloxymethylketone, that binds to the CatK active site, recognition sequence, and fluorescence donor-acceptor pair. This protocol can determine the exact localization of active CatK in osteoclasts. For complete details on the use and execution of this protocol, please refer to Janiszewski et al. (2023).1.


Assuntos
Corantes Fluorescentes , Osteoclastos , Osteoclastos/metabolismo , Catepsina K/metabolismo , Corantes Fluorescentes/metabolismo , Fluorescência , Leucócitos Mononucleares/metabolismo
2.
Cell Chem Biol ; 30(2): 159-174.e8, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36696904

RESUMO

Cathepsin K (CatK) is a lysosomal cysteine protease whose highest expression is found in osteoclasts, which are the cells responsible for bone resorption. Investigations of the functions and physiological relevance of CatK have often relied on antibody-related techniques, which makes studying its activity patterns a challenging task. Hence, we developed a set of chemical tools for the investigation of CatK activity. We show that our probe is a valuable tool for monitoring the proteolytic activation of CatK during osteoclast formation. Moreover, we demonstrate that our inhibitor of CatK impedes osteoclastogenesis and bone resorption and that CatK is stored in its active form in osteoclasts within their lysosomal compartment and mainly in the ruffled borders of osteoclasts. Given that our probe recognizes active CatK within living cells without exhibiting any observed cytotoxicity in the several models tested, we expect that it would be well suited to theranostic applications in CatK-related diseases.


Assuntos
Reabsorção Óssea , Osteoclastos , Humanos , Osteoclastos/metabolismo , Osteogênese , Catepsina K/metabolismo , Reabsorção Óssea/metabolismo
3.
Mol Aspects Med ; 88: 101143, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36152458

RESUMO

Toll-like receptors (TLRs) are pattern recognition receptors (PRRs) that respond to pathogen-associated molecular patterns (PAMPs). The recognition of specific microbial ligands by TLRs triggers an innate immune response and also promotes adaptive immunity, which is necessary for the efficient elimination of invading pathogens. Successful pathogens have therefore evolved strategies to subvert and/or manipulate TLR signaling. Both the impairment and uncontrolled activation of TLR signaling can harm the host, causing tissue destruction and allowing pathogens to proliferate, thus favoring disease progression. In this context, microbial proteases are key virulence factors that modify components of the TLR signaling pathway. In this review, we discuss the role of bacterial and viral proteases in the manipulation of TLR signaling, highlighting the importance of these enzymes during the development of infectious diseases.


Assuntos
Doenças Transmissíveis , Receptores Toll-Like , Proteases Virais , Humanos , Doenças Transmissíveis/metabolismo , Doenças Transmissíveis/microbiologia , Imunidade Inata , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Proteases Virais/imunologia , Proteases Virais/metabolismo , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Peptídeo Hidrolases/imunologia , Peptídeo Hidrolases/metabolismo , Viroses/metabolismo , Infecções Bacterianas/metabolismo
4.
mBio ; 13(3): e0378721, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35491845

RESUMO

Porphyromonas gingivalis, a keystone pathogen in periodontitis (PD), produces cysteine proteases named gingipains (RgpA, RgpB, and Kgp), which strongly affect the host immune system. The range of action of gingipains is extended by their release as components of outer membrane vesicles, which efficiently diffuse into surrounding gingival tissues. However, away from the anaerobic environment of periodontal pockets, increased oxygen levels lead to oxidation of the catalytic cysteine residues of gingipains, inactivating their proteolytic activity. In this context, the influence of catalytically inactive gingipains on periodontal tissues is of significant interest. Here, we show that proteolytically inactive RgpA induced a proinflammatory response in both gingival keratinocytes and dendritic cells. Inactive RgpA is bound to the cell surface of gingival keratinocytes in the region of lipid rafts, and using affinity chromatography, we identified RgpA-interacting proteins, including epidermal growth factor receptor (EGFR). Next, we showed that EGFR interaction with inactive RgpA stimulated the expression of inflammatory cytokines. The response was mediated via the EGFR-phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway, which when activated in the gingival tissue rich in dendritic cells in the proximity of the alveolar bone, may significantly contribute to bone resorption and the progress of PD. Taken together, these findings broaden our understanding of the biological role of gingipains, which in acting as proinflammatory factors in the gingival tissue, create a favorable milieu for the growth of inflammophilic pathobionts. IMPORTANCE Gingipain cysteine proteases are essential virulence factors of Porphyromonas gingivalis, an oral bacterium implicated in development of periodontitis. Gingipains diffusing from anaerobic periodontal pockets lose proteolytic activity in the oxygenated environment of gingival tissues. We found that despite the loss of activity, gingipains still elicit a strong inflammatory response, which may contribute to the progression of periodontitis and bone resorption. Moreover, we identified the host molecules utilized by the pathogen as receptors for proteolytically inactivated gingipains. The broad distribution of those receptors in human tissue suggests their involvement in systemic diseases associated with periodontal pathogens.


Assuntos
Reabsorção Óssea , Periodontite , Adesinas Bacterianas/metabolismo , Cisteína Endopeptidases/metabolismo , Receptores ErbB/metabolismo , Cisteína Endopeptidases Gingipaínas , Humanos , Imunidade , Bolsa Periodontal , Periodontite/microbiologia , Fosfatidilinositol 3-Quinases/metabolismo , Porphyromonas gingivalis/fisiologia
5.
mBio ; 12(3): e0050221, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34182783

RESUMO

Periodontal disease (PD) is an inflammatory disease of the supporting tissues of the teeth that develops in response to formation of a dysbiotic biofilm on the subgingival tooth surface. Although exacerbated inflammation leads to alveolar bone destruction and may cause tooth loss, the molecular basis of PD initiation and progression remains elusive. Control over the inflammatory reaction and return to homeostasis can be efficiently restored by negative regulators of Toll-like receptor (TLR) signaling pathways such as monocyte chemoattractant protein-induced protein 1 (MCPIP-1), which is constitutively expressed in gingival keratinocytes and prevents hyperresponsiveness in the gingiva. Here, we found that inflammophilic periodontal species influence the stability of MCPIP-1, leading to an aggravated response of the epithelium to proinflammatory stimulation. Among enzymes secreted by periodontal species, gingipains-cysteine proteases from Porphyromonas gingivalis-are considered major contributors to the pathogenic potential of bacteria, strongly influencing the components of the innate and adaptive immune system. Gingipain proteolytic activity leads to a rapid degradation of MCPIP-1, exacerbating the inflammatory response induced by endotoxin. Collectively, these results establish a novel mechanism of corruption of inflammatory signaling by periodontal pathogens, indicating new possibilities for treatment of this chronic disease. IMPORTANCE Periodontitis is a highly prevalent disease caused by accumulation of a bacterial biofilm. Periodontal pathogens use a number of virulence strategies that are under intensive study to find optimal therapeutic approaches against bone loss. In our work, we present a novel mechanism utilized by the key periodontal pathogen Porphyromonas gingivalis, based on the selective degradation of the negative regulator of inflammation, MCPIP-1. We found that the diminished levels of MCPIP-1 in gingival keratinocytes-cells at the forefront of the fight against bacteria-cause sensitization to endotoxins produced by other oral species. This results in an enhanced inflammatory response, which promotes the growth of inflammophilic pathobionts and damage of tooth-supporting tissues. Our observation is relevant to understanding the molecular basis of periodontitis and the development of new methods for treatment.


Assuntos
Gengiva/citologia , Inflamação , Queratinócitos/imunologia , Lipopolissacarídeos/metabolismo , Porphyromonas gingivalis/imunologia , Porphyromonas gingivalis/metabolismo , Ribonucleases/metabolismo , Transdução de Sinais , Animais , Biofilmes/crescimento & desenvolvimento , Células Cultivadas , Feminino , Cisteína Endopeptidases Gingipaínas , Queratinócitos/metabolismo , Queratinócitos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Periodontite/microbiologia , Porphyromonas gingivalis/fisiologia , Ribonucleases/genética , Ribonucleases/imunologia , Organismos Livres de Patógenos Específicos
6.
Int J Mol Sci ; 21(6)2020 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-32183255

RESUMO

Candida albicans is a pathogenic fungus capable of switching its morphology between yeast-like cells and filamentous hyphae and can associate with bacteria to form mixed biofilms resistant to antibiotics. In these structures, the fungal milieu can play a protective function for bacteria as has recently been reported for C. albicans and a periodontal pathogen-Porphyromonas gingivalis. Our current study aimed to determine how this type of mutual microbe protection within the mixed biofilm affects the contacting host cells. To analyze C. albicans and P. gingivalis persistence and host infection, several models for host-biofilm interactions were developed, including microbial exposure to a representative monocyte cell line (THP1) and gingival fibroblasts isolated from periodontitis patients. For in vivo experiments, a mouse subcutaneous chamber model was utilized. The persistence of P. gingivalis cells was observed within mixed biofilm with C. albicans. This microbial co-existence influenced host immunity by attenuating macrophage and fibroblast responses. Cytokine and chemokine production decreased compared to pure bacterial infection. The fibroblasts isolated from patients with severe periodontitis were less susceptible to fungal colonization, indicating a modulation of the host environment by the dominating bacterial infection. The results obtained for the mouse model in which a sequential infection was initiated by the fungus showed that this host colonization induced a milder inflammation, leading to a significant reduction in mouse mortality. Moreover, high bacterial counts in animal organisms were noted on a longer time scale in the presence of C. albicans, suggesting the chronic nature of the dual-species infection.


Assuntos
Infecções por Bacteroidaceae/imunologia , Candida albicans/fisiologia , Gengiva/imunologia , Evasão da Resposta Imune/imunologia , Periodontite/imunologia , Porphyromonas gingivalis/imunologia , Animais , Infecções por Bacteroidaceae/microbiologia , Biofilmes/efeitos dos fármacos , Células Cultivadas , Coinfecção/imunologia , Coinfecção/microbiologia , Modelos Animais de Doenças , Feminino , Fibroblastos/imunologia , Gengiva/microbiologia , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Camundongos , Interações Microbianas , Periodontite/microbiologia
7.
PLoS Pathog ; 15(5): e1007773, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31107907

RESUMO

Neutrophil-derived networks of DNA-composed extracellular fibers covered with antimicrobial molecules, referred to as neutrophil extracellular traps (NETs), are recognized as a physiological microbicidal mechanism of innate immunity. The formation of NETs is also classified as a model of a cell death called NETosis. Despite intensive research on the NETs formation in response to pathogens, the role of specific bacteria-derived virulence factors in this process, although postulated, is still poorly understood. The aim of our study was to determine the role of gingipains, cysteine proteases responsible for the virulence of P. gingivalis, on the NETosis process induced by this major periodontopathogen. We showed that NETosis triggered by P. gingivalis is gingipain dependent since in the stark contrast to the wild-type strain (W83) the gingipain-null mutant strain only slightly induced the NETs formation. Furthermore, the direct effect of proteases on NETosis was documented using purified gingipains. Notably, the induction of NETosis was dependent on the catalytic activity of gingipains, since proteolytically inactive forms of enzymes showed reduced ability to trigger the NETs formation. Mechanistically, gingipain-induced NETosis was dependent on proteolytic activation of protease-activated receptor-2 (PAR-2). Intriguingly, both P. gingivalis and purified Arg-specific gingipains (Rgp) induced NETs that not only lacked bactericidal activity but instead stimulated the growth of bacteria species otherwise susceptible to killing in NETs. This protection was executed by proteolysis of bactericidal components of NETs. Taken together, gingipains play a dual role in NETosis: they are the potent direct inducers of NETs formation but in the same time, their activity prevents P. gingivalis entrapment and subsequent killing. This may explain a paradox that despite the massive accumulation of neutrophils and NETs formation in periodontal pockets periodontal pathogens and associated pathobionts thrive in this environment.


Assuntos
Adesinas Bacterianas/imunologia , Infecções por Bacteroidaceae/imunologia , Cisteína Endopeptidases/imunologia , Armadilhas Extracelulares/imunologia , Neutrófilos/imunologia , Peritonite/imunologia , Porphyromonas gingivalis/imunologia , Porphyromonas gingivalis/patogenicidade , Receptor PAR-2/metabolismo , Adesinas Bacterianas/metabolismo , Animais , Infecções por Bacteroidaceae/metabolismo , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/patologia , Células Cultivadas , Cisteína Endopeptidases/metabolismo , Armadilhas Extracelulares/microbiologia , Feminino , Cisteína Endopeptidases Gingipaínas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/microbiologia , Neutrófilos/patologia , Peritonite/metabolismo , Peritonite/microbiologia , Receptor PAR-2/imunologia , Transdução de Sinais
8.
Sci Rep ; 9(1): 4376, 2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30867500

RESUMO

The oral cavity contains different types of microbial species that colonize human host via extensive cell-to-cell interactions and biofilm formation. Candida albicans-a yeast-like fungus that inhabits mucosal surfaces-is also a significant colonizer of subgingival sites in patients with chronic periodontitis. It is notable however that one of the main infectious agents that causes periodontal disease is an anaerobic bacterium-Porphyromonas gingivalis. In our study, we evaluated the different strategies of both pathogens in the mutual colonization of an artificial surface and confirmed that a protective environment existed for P. gingivalis within developed fungal biofilm formed under oxic conditions where fungal cells grow mainly in their filamentous form i.e. hyphae. A direct physical contact between fungi and P. gingivalis was initiated via a modulation of gene expression for the major fungal cell surface adhesin Als3 and the aspartic proteases Sap6 and Sap9. Proteomic identification of the fungal surfaceome suggested also an involvement of the Mp65 adhesin and a "moonlighting" protein, enolase, as partners for the interaction with P. gingivalis. Using mutant strains of these bacteria that are defective in the production of the gingipains-the proteolytic enzymes that also harbor hemagglutinin domains-significant roles of these proteins in the formation of bacteria-protecting biofilm were clearly demonstrated.


Assuntos
Adesinas Bacterianas/metabolismo , Bactérias Anaeróbias/fisiologia , Biofilmes , Candida albicans/fisiologia , Interações Microbianas , Porphyromonas gingivalis/fisiologia , Adesinas Bacterianas/genética , Aderência Bacteriana , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Viabilidade Microbiana , Proteômica/métodos , Virulência
9.
Pathog Dis ; 76(4)2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29668945

RESUMO

Porphyromonas gingivalis, an anaerobic Gram-negative bacterium critically involved in the development of human periodontitis, belongs to the late colonizers of the oral cavity. The success of this pathogen in the host colonization and infection results from the presence of several virulence factors, including extracellular peptidylarginine deiminase (PPAD), an enzyme that converts protein arginine residues to citrullines. A common opportunistic fungal pathogen of humans, Candida albicans, is also frequently identified among microorganisms that reside at subgingival sites. The aim of the current work was to verify if protein citrullination can influence the formation of mixed biofilms by both microorganisms under hypoxic and normoxic conditions. Quantitative estimations of the bacterial adhesion to fungal cells demonstrated the importance of PPAD activity in this process, since the level of binding of P. gingivalis mutant strain deprived of PPAD was significantly lower than that observed for the wild-type strain. These results were consistent with mass spectrometric detection of the citrullination of selected surface-exposed C. albicans proteins. Furthermore, a viability of P. gingivalis cells under normoxia increased in the presence of fungal biofilm compared with the bacteria that formed single-species biofilm. These findings suggest a possible protection of these strict anaerobes under unfavorable aerobic conditions by C. albicans during mixed biofilm formation.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Candida albicans/patogenicidade , Porphyromonas gingivalis/patogenicidade , Desiminases de Arginina em Proteínas/metabolismo , Fatores de Virulência/metabolismo , Aderência Bacteriana , Proteínas de Bactérias/genética , Candida albicans/fisiologia , Expressão Gênica , Humanos , Interações Microbianas , Porphyromonas gingivalis/enzimologia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/crescimento & desenvolvimento , Desiminases de Arginina em Proteínas/genética , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA