Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Appl Microbiol ; 132(5): 3853-3869, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35262250

RESUMO

AIMS: We evaluated two species of human oral commensal streptococci in protection against dental caries induced by Streptococcus mutans. METHODS AND RESULTS: Candidate probiotics, Streptococcus sp. A12, Streptococcus sanguinis BCC23 and an arginine deiminase mutant of BCC23 (∆arcADS) were tested for their ability to reduce S. mutans-induced caries in an established mouse model. Mice were colonized with a probiotic, challenged with S. mutans, then intermittently reinoculated with a probiotic strain. Oral colonization of each strain and autochthonous bacteria was assessed by quantitative polymerase chain reaction. Both BCC23 strains, but not A12, were associated with markedly reduced sulcal caries, persistently colonized mucosal and dental biofilms, and significantly lowered S. mutans counts. All three strains enhanced mucosal colonization of autochthonous bacteria. In a follow-up experiment, when S. mutans was established first, dental and mucosal colonization of S. mutans was unaltered by a subsequent challenge with either BCC23 strain. Results between BCC23 and BCC23 ∆arcADS were equivalent. CONCLUSIONS: BCC23 is a potential probiotic to treat patients at high caries risk. Its effectiveness is independent of ADS activity, but initial dental cleaning to enhance establishment in dental biofilms may be required. SIGNIFICANCE AND IMPACT OF THE STUDY: In vivo testing of candidate probiotics is highly informative, as effectiveness is not always reflected by genotype or in vitro behaviours.


Assuntos
Cárie Dentária , Probióticos , Animais , Biofilmes , Cárie Dentária/prevenção & controle , Humanos , Camundongos , Probióticos/farmacologia , Streptococcus/genética , Streptococcus mutans/genética , Streptococcus sanguis
2.
Mol Microbiol ; 117(2): 375-393, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34862691

RESUMO

Streptococcus sanguinis is an oral commensal and an etiological agent of infective endocarditis. Previous studies have identified the SsaACB manganese transporter as essential for endocarditis virulence; however, the significance of SsaACB in the oral environment has never been examined. Here we report that a ΔssaACB deletion mutant of strain SK36 exhibits reduced growth and manganese uptake under acidic conditions. Further studies revealed that these deficits resulted from the decreased activity of TmpA, shown in the accompanying paper to function as a ZIP-family manganese transporter. Transcriptomic analysis of fermentor-grown cultures of SK36 WT and ΔssaACB strains identified pH-dependent changes related to carbon catabolite repression in both strains, though their magnitude was generally greater in the mutant. In strain VMC66, which possesses a MntH transporter, loss of SsaACB did not significantly alter growth or cellular manganese levels under the same conditions. Interestingly, there were only modest differences between SK36 and its ΔssaACB mutant in competition with Streptococcus mutans in vitro and in a murine oral colonization model. Our results suggest that the heterogeneity of the oral environment may provide a rationale for the variety of manganese transporters found in S. sanguinis.


Assuntos
Endocardite Bacteriana , Streptococcus sanguis , Animais , Manganês , Camundongos , Streptococcus mutans , Virulência
3.
Front Physiol ; 12: 699104, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34276419

RESUMO

Saliva protects dental surfaces against cavities (i. e., dental caries), a highly prevalent infectious disease frequently associated with acidogenic Streptococcus mutans. Substantial in vitro evidence supports amylase, a major constituent of saliva, as either protective against caries or supporting caries. We therefore produced mice with targeted deletion of salivary amylase (Amy1) and determined the impact on caries in mice challenged with S. mutans and fed a diet rich in sucrose to promote caries. Total smooth surface and sulcal caries were 2.35-fold and 1.79-fold greater in knockout mice, respectively, plus caries severities were twofold or greater on sulcal and smooth surfaces. In in vitro experiments with samples of whole stimulated saliva, amylase expression did not affect the adherence of S. mutans to saliva-coated hydroxyapatite and slightly increased its aggregation in solution (i.e., oral clearance). Conversely, S. mutans in biofilms formed in saliva with 1% glucose displayed no differences when cultured on polystyrene, but on hydroxyapatite was 40% less with amylase expression, suggesting that recognition by S. mutans of amylase bound to hydroxyapatite suppresses growth. However, this effect was overshadowed in vivo, as the recoveries of S. mutans from dental plaque were similar between both groups of mice, suggesting that amylase expression helps decrease plaque acids from S. mutans that dissolve dental enamel. With amylase deletion, commensal streptococcal species increased from ~75 to 90% of the total oral microbiota, suggesting that amylase may promote higher plaque pH by supporting colonization by base-producing oral commensals. Importantly, collective results indicate that amylase may serve as a biomarker of caries risk.

4.
Appl Environ Microbiol ; 87(4)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33277269

RESUMO

A collection of 113 Streptococcus strains from supragingival dental plaque of caries-free individuals were recently tested in vitro for direct antagonism of the dental caries pathogen Streptococcus mutans, and for their capacity for arginine catabolism via the arginine deiminase system (ADS). To advance their evaluation as potential probiotics, twelve strains of commensal oral streptococci with various antagonistic and ADS potentials were assessed in a mouse model for oral (i.e., oral mucosal pellicles and saliva) and dental colonization under four diets (healthy or high-sucrose, with or without prebiotic arginine). Colonization by autochthonous bacteria was also monitored. One strain failed to colonize, whereas oral colonization by the other eleven strains varied by 3 log units. Dental colonization was high for five strains regardless of diet, six strains increased colonization with at least one high-sucrose diet, and added dietary arginine decreased dental colonization of two strains. Streptococcus sp. A12 (high in vitro ADS activity and antagonism) and two engineered mutants lacking the ADS (ΔarcADS) or pyruvate oxidase-mediated H2O2 production (ΔspxB) were tested for competition against S. mutans UA159. A12 wild type and ΔarcADS colonized only transiently, whereas ΔspxB persisted, but without altering oral or dental colonization by S. mutans In testing four additional candidates, S. sanguinis BCC23 markedly attenuated S. mutans' oral and dental colonization, enhanced colonization of autochthonous bacteria, and decreased severity of smooth surface caries under highly cariogenic conditions. Results demonstrate the utility of the mouse model to evaluate potential probiotics, revealing little correlation between in vitro antagonism and competitiveness against S. mutans in vivo IMPORTANCE Our results demonstrate in vivo testing of potential oral probiotics can be accomplished and can yield information to facilitate the ultimate design and optimization of novel anti-caries probiotics. We show human oral commensals associated with dental health are an important source of potential probiotics that may be used to colonize patients under dietary conditions of highly varying cariogenicity. Assessment of competitiveness against dental caries pathogen Streptococcus mutans and impact on caries identified strains or genetic elements for further study. Results also uncovered strains that enhanced oral and dental colonization by autochthonous bacteria when challenged with S. mutans, suggesting cooperative interactions for future elucidation. Distinguishing a rare strain that effectively compete with S. mutans under conditions that promote caries further validates our systematic approach to more critically evaluate probiotics for use in humans.

5.
Pflugers Arch ; 472(3): 385-403, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31932898

RESUMO

Mucin secretion by salivary mucous glands is mediated predominantly by parasympathetic acetylcholine activation of cholinergic muscarinic receptors via increased intracellular free calcium ([Ca2+]i) and activation of conventional protein kinase C isozymes (cPKC). However, the parasympathetic co-neurotransmitter, vasoactive intestinal peptide (VIP), also initiates secretion, but to a lesser extent. In the present study, cross talk between VIP- and muscarinic-induced mucin secretion was investigated using isolated rat sublingual tubuloacini. VIP-induced secretion is mediated by cAMP-activated protein kinase A (PKA), independently of increased [Ca2+]i. Synergistic secretion between VIP and the muscarinic agonist, carbachol, was demonstrated but only with submaximal carbachol. Carbachol has no effect on cAMP ± VIP. Instead, PKA activated by VIP releases Ca2+ from an intracellular pool maintained by the sarco/endoplasmic reticulum Ca2+-ATPase pump. Calcium release was independent of phospholipase C activity. The resultant sustained [Ca2+]i increase is additive to submaximal, but not maximal carbachol-induced [Ca2+]i. Synergistic mucin secretion was mimicked by VIP plus either phorbol 12-myristate 13-acetate or 0.01 µM thapsigargin, and blocked by the PKC inhibitor, Gö6976. VIP-induced Ca2+ release also promoted store-operated Ca2+ entry. Synergism is therefore driven by VIP-mediated [Ca2+]i augmenting cPKC activity to enhance muscarinic mucin secretion. Additional data suggest ryanodine receptors control VIP/PKA-mediated Ca2+ release from a Ca2+ pool also responsive to maximal carbachol. A working model of muscarinic and VIP control of mucous cell exocrine secretion is presented. Results are discussed in relation to synergistic mechanisms in other secretory cells, and the physiological and therapeutic significance of VIP/muscarinic synergism controlling salivary mucous cell exocrine secretion.


Assuntos
Secreções Corporais/metabolismo , Cálcio/metabolismo , Colinérgicos/farmacologia , Mucinas/metabolismo , Proteína Quinase C/metabolismo , Glândulas Salivares/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/farmacologia , Adenosina Trifosfatases/metabolismo , Animais , Isoenzimas/metabolismo , Masculino , Agonistas Muscarínicos/farmacologia , Ésteres de Forbol/farmacologia , Ratos , Ratos Wistar , Receptores Muscarínicos/metabolismo , Glândulas Salivares/metabolismo , Tapsigargina/farmacologia
6.
Oral Dis ; 25(5): 1325-1334, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30920100

RESUMO

OBJECTIVES: To determine expression and localization of membrane-associated mucins within human keratinized and non-keratinized oral epithelia, and to explore transcriptional changes associated with primary Sjögren's syndrome. SUBJECTS AND METHODS: Mucin transcripts and glycoproteins were determined by RT-PCR and immunohistochemistry, respectively, in oral keratinized (hard palate) and non-keratinized (buccal) epithelia obtained from three cadavers. Mucin transcripts assessed by quantitative PCR were compared between cells harvested by brushing buccal and palatal epithelia of 25 female primary Sjögren's syndrome patients vs 25 healthy age-matched female control subjects. RESULTS: In hard palate, MUC4 is absent and MUC1 localized to deeper cell layers. Both mucins are within the apical layers of buccal epithelium. MUC15 is localized throughout all palatal cell layers and in all but the basal layer of buccal epithelia. MUC16, MUC20, and MUC21 glycoproteins are localized within all but the basal cell layer of both tissue types. In buccal cells of primary Sjögren's patients, MUC21 transcripts are down-regulated 3.4-fold and MUC20 2.6-fold. Dysregulation of select epithelial mucins may therefore contribute to xerostomia. CONCLUSIONS: Differential expression of multiple mucins and down-regulation in Sjögren's syndrome support further study of oral epithelial mucin physiology and pathophysiology, including their functions in hydration and lubrication of the oral mucosal pellicle.


Assuntos
Mucosa Bucal/metabolismo , Mucosa Bucal/patologia , Mucinas/metabolismo , Síndrome de Sjogren/metabolismo , Síndrome de Sjogren/patologia , Adulto , Idoso , Estudos de Casos e Controles , Película Dentária , Epitélio , Feminino , Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Mucinas/genética , Síndrome de Sjogren/genética
7.
mSphere ; 3(1)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29435491

RESUMO

Transposon mutagenesis coupled with next-generation DNA sequencing (Tn-seq) is a powerful tool for discovering regions of the genome that are required for the survival of bacteria in different environments. We adapted this technique to the dental caries pathogen Streptococcus mutans UA159 and identified 11% of the genome as essential, with many genes encoding products required for replication, translation, lipid metabolism, and cell wall biogenesis. Comparison of the essential genome of S. mutans UA159 with those of selected other streptococci for which such information is available revealed several metabolic pathways and genes that are required in S. mutans, but not in some Streptococcus spp. We further identified genes that are essential for sustained growth in rich or defined medium, as well as for persistence in vivo in a rodent model of oral infection. Collectively, our results provide a novel and comprehensive view of the genes required for essential processes of S. mutans, many of which could represent potential targets for therapeutics. IMPORTANCE Tooth decay (dental caries) is a common cause of pain, impaired quality of life, and tooth loss in children and adults. It begins because of a compositional change in the microorganisms that colonize the tooth surface driven by repeated and sustained carbohydrate intake. Although several bacterial species are associated with tooth decay, Streptococcus mutans is the most common cause. Therefore, it is important to identify biological processes that contribute to the survival of S. mutans in the human mouth, with the aim of disrupting the processes with antimicrobial agents. We successfully applied Tn-seq to S. mutans, discovering genes that are required for survival, growth, and persistence, both in laboratory environments and in a mouse model of tooth decay. This work highlights new avenues for the control of an important human pathogen.

8.
J Biol Chem ; 290(5): 2993-3008, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25512380

RESUMO

Saliva functions in innate immunity of the oral cavity, protecting against demineralization of teeth (i.e. dental caries), a highly prevalent infectious disease associated with Streptococcus mutans, a pathogen also linked to endocarditis and atheromatous plaques. Gel-forming mucins are a major constituent of saliva. Because Muc19 is the dominant salivary gel-forming mucin in mice, we studied Muc19(-/-) mice for changes in innate immune functions of saliva in interactions with S. mutans. When challenged with S. mutans and a cariogenic diet, total smooth and sulcal surface lesions are more than 2- and 1.6-fold higher in Muc19(-/-) mice compared with wild type, whereas the severity of lesions are up to 6- and 10-fold higher, respectively. Furthermore, the oral microbiota of Muc19(-/-) mice display higher levels of indigenous streptococci. Results emphasize the importance of a single salivary constituent in the innate immune functions of saliva. In vitro studies of S. mutans and Muc19 interactions (i.e. adherence, aggregation, and biofilm formation) demonstrate Muc19 poorly aggregates S. mutans. Nonetheless, aggregation is enhanced upon adding Muc19 to saliva from Muc19(-/-) mice, indicating Muc19 assists in bacterial clearance through formation of heterotypic complexes with salivary constituents that bind S. mutans, thus representing a novel innate immune function for salivary gel-forming mucins. In humans, expression of salivary MUC19 is unclear. We find MUC19 transcripts in salivary glands of seven subjects and demonstrate MUC19 glycoproteins in glandular mucous cells and saliva. Similarities and differences between mice and humans in the expression and functions of salivary gel-forming mucins are discussed.


Assuntos
Cárie Dentária/metabolismo , Cárie Dentária/microbiologia , Imunidade Inata/fisiologia , Mucinas/metabolismo , Saliva/metabolismo , Streptococcus mutans/patogenicidade , Adulto , Animais , Cárie Dentária/imunologia , Feminino , Humanos , Imunidade Inata/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Mucinas/genética
9.
J Biol Chem ; 288(21): 14742-55, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23580649

RESUMO

The autosomal recessive mutation, sld, attenuates mucous cell expression in murine sublingual glands with corresponding effects on mucin 19 (Muc19). We conducted a systematic study including genetic mapping, sequencing, and functional analyses to elucidate a mutation to explain the sld phenotype in neonatal mice. Genetic mapping and gene expression analyses localized the sld mutation within the gene Muc19/Smgc, specifically attenuating Muc19 transcripts, and Muc19 knock-out mice mimic the sld phenotype in neonates. Muc19 transcription is unaffected in sld mice, whereas mRNA stability is markedly decreased. Decreased mRNA stability is not due to a defect in 3'-end processing nor to sequence differences in Muc19 transcripts. Comparative sequencing of the Muc19/Smgc gene identified four candidate intronic mutations within the Muc19 coding region. Minigene splicing assays revealed a novel splicing event in which insertion of two additional repeats within a CA repeat region of intron 53 of the sld genome enhances retention of intron 54, decreasing the levels of correctly spliced transcripts. Moreover, pateamine A, an inhibitor of nonsense-mediated mRNA decay, inhibits degradation of aberrant Muc19 transcripts. The mutation in intron 53 thus enhances aberrant splicing leading to degradation of aberrant transcripts and decreased Muc19 message stability, consistent with the sld phenotype. We propose a working model of the unique splicing event enhanced by the mutation, as well as putative explanations for the gradual but limited increase in Muc19 glycoprotein expression and its restricted localization to subpopulations of mucous cells in sld mice during postnatal gland development.


Assuntos
Íntrons/fisiologia , Modelos Biológicos , Mucinas/biossíntese , Mutação , Estabilidade de RNA/fisiologia , RNA Mensageiro/metabolismo , Glândula Sublingual/metabolismo , Processamento Alternativo/fisiologia , Animais , Regulação da Expressão Gênica/fisiologia , Camundongos , Camundongos Knockout , Mucinas/genética , Fases de Leitura Aberta/fisiologia , RNA Mensageiro/genética , Glândula Sublingual/citologia , Glândula Sublingual/crescimento & desenvolvimento
10.
Biochim Biophys Acta ; 1812(12): 1567-76, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21945428

RESUMO

Carbonic anhydrase VI (CA VI), encoded by type A transcripts of the gene Car6, is a secretory product of salivary glands and is found in the enamel pellicle. Because higher caries prevalence is associated with lower salivary concentrations of CA VI in humans, we tested whether CA VI protects enamel surfaces from caries induced by Streptococcus mutans, using Car6(-/-) mice, in which salivary CA VI expression is absent. We detected aberrant Car6 type A transcripts in Car6(-/-) mice, likely targets for nonsense-mediated mRNA decay. Expression of the intracellular stress-induced isoform of CA VI encoded by type B transcripts was restricted to parotid and submandibular glands of wild type mice. The salivary function of Car6(-/-) mice was normal as assessed by the histology and protein/glycoprotein profiles of glands, salivary flow rates and protein/glycoprotein compositions of saliva. Surprisingly, total smooth surface caries and sulcal caries in Car6(-/-) mice were more than 6-fold and 2-fold lower than in wild type mice after infection with S. mutans strain UA159. Recoveries of S. mutans and total microbiota from molars were also lower in Car6(-/-) mice. To explore possible mechanisms for increased caries susceptibility, we found no differences in S. mutans adherence to salivary pellicles, in vitro. Interestingly, higher levels of Lactobacillus murinus and an unidentified Streptococcus species were cultivated from the oral microbiota of Car6(-/-) mice. Collective results suggest salivary CA VI may promote caries by modulating the oral microbiota to favor S. mutans colonization and/or by the enzymatic production of acid within plaque.


Assuntos
Anidrases Carbônicas/genética , Cárie Dentária/microbiologia , Placa Dentária/microbiologia , Saliva/enzimologia , Infecções Estreptocócicas/microbiologia , Streptococcus mutans/isolamento & purificação , Animais , Aderência Bacteriana , Anidrases Carbônicas/metabolismo , Cárie Dentária/patologia , Durapatita , Feminino , Deleção de Genes , Masculino , Metagenoma , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dente Molar/microbiologia , Dente Molar/patologia , RNA Ribossômico 16S/genética , Glândulas Salivares/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus mutans/genética , Transcrição Gênica
11.
J Histochem Cytochem ; 58(2): 141-56, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19826070

RESUMO

The recently identified gene Muc19/Smgc encodes two diverse splice variants, Smgc (submandibular gland protein C) and Muc19 (mucin 19). Muc19 is a member of the large gel-forming mucin family and is an exocrine product of sublingual mucous salivary glands in mice. SMGC is a transiently expressed secretion product of developing rodent submandibular and sublingual glands. Little is known about the expression of Muc19/Smgc gene products in other murine salivary and non-salivary tissues containing the mucous cell phenotype. Muc19 expression was therefore initially assessed by RT-PCR and immunohistochemistry. As a complementary approach, we developed a knockin mouse model, Muc19-EGFP, in which mice express a fusion protein containing the first 69 residues of Muc19 followed by enhanced green fluorescent protein (EGFP) as a marker of Muc19 expression. Results from both approaches are consistent, with preferential Muc19 expression in salivary major and minor mucous glands as well as submucosal glands of the tracheolarynx and bulbourethral glands. Evidence also indicates that individual mucous cells of minor salivary and bulbourethral glands produce another gel-forming mucin in addition to Muc19. We further find tissue expression of full-length Smgc transcripts, which encode for SMGC, and are restricted to neonatal tracheolarynx and all salivary tissues.


Assuntos
Mucinas/genética , Mucinas/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Homozigoto , Masculino , Camundongos , Dados de Sequência Molecular , Mucinas/análise , Mucinas/química , Especificidade de Órgãos , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Glândulas Salivares/metabolismo
12.
J Histochem Cytochem ; 57(4): 383-96, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19110483

RESUMO

Muc19/Smgc expresses two splice variants, Smgc (submandibular gland protein C) and Muc19 (mucin 19), the latter a major exocrine product of differentiated murine sublingual mucous cells. Transcripts for Smgc were detected recently in neonatal sublingual glands, suggesting that SMGC proteins are expressed during initial salivary mucous cell cytodifferentiation. We therefore compared developmental expression of transcripts and translation products of Smgc and Muc19 in sublingual glands. We find abundant expression of SMGC within the initial terminal bulbs, with a subsequent decrease as Muc19 expression increases. During postnatal gland expansion, SMGC is found in presumptive newly formed acinar cells and then persists in putative acinar stem cells. Mucin levels increase 7-fold during the first 3 weeks of life, with little change in transcript levels, whereas between postnatal days 21 and 28, there is a 3-fold increase in Muc19 mRNA and heteronuclear RNA. Our collective results demonstrate the direct transition from SMGC to Muc19 expression during early mucous cell cytodifferentiation and further indicate developmentally regulated changes in Muc19/Smgc transcription, alternative splicing, and translation. These changes in Muc19/Smgc gene expression delineate multiple stages of salivary mucous cell cytodifferentiation and subsequent maturation during embryonic gland development through the first 4 weeks of postnatal life.


Assuntos
Mucinas/biossíntese , Glândula Sublingual/metabolismo , Processamento Alternativo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Mucinas/genética , RNA Mensageiro/biossíntese , Fatores Sexuais , Glândula Sublingual/embriologia , Glândula Sublingual/crescimento & desenvolvimento , Fatores de Tempo , Transcrição Gênica
13.
J Histochem Cytochem ; 52(5): 671-81, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15100244

RESUMO

Human fractalkine (CX3CL1), a delta-chemokine, is implicated in the mediation of multiple cell functions. In addition to serving as a chemotactic factor for mononuclear cell subtypes, membrane-bound fractalkine may promote viral infection by interacting with virions that encode putative fractalkine-binding proteins. Fractalkine expression in normal epithelial tissues studied to date is either constitutive or is upregulated with inflammation. In salivary glands, the expression of fractalkine is unknown. Moreover, salivary glands are a major site for the persistent and productive infection by human herpesvirus (HHV)-7, which encodes two putative fractalkine-binding gene products, U12 and U51. Surprisingly, the cellular distribution of HHV-7 in major salivary glands has not been explored. We therefore determined by immunohistochemistry the cellular localization of fractalkine in three different salivary glands: parotid, submandibular, and labial glands. Fractalkine expression was highly variable, ranging from high to undetectable levels. We further examined the association of fractalkine with inflammatory cell infiltration or HHV-7 infection of salivary epithelial cells. Inflammatory cells were always adjacent to epithelial cells expressing fractalkine, consistent with a function of fractalkine in inflammatory cell recruitment and/or retention in salivary glands. In contrast, HHV-7-infected epithelial cells did not always express fractalkine, suggesting that fractalkine may not be an absolute requirement for viral entry.


Assuntos
Quimiocinas CX3C/metabolismo , Herpesvirus Humano 7/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Proteínas de Membrana/metabolismo , Glândulas Salivares/metabolismo , Adulto , Idoso , Complexo CD3/metabolismo , Quimiocina CX3CL1 , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Feminino , Herpesvirus Humano 7/imunologia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Inflamação/patologia , Masculino , Pessoa de Meia-Idade , Glândula Parótida/metabolismo , Glândula Parótida/patologia , Glândula Parótida/virologia , Glândulas Salivares/patologia , Glândulas Salivares/virologia , Glândula Submandibular/metabolismo , Glândula Submandibular/patologia , Glândula Submandibular/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...