Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Vet Res ; 55(1): 76, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38867337

RESUMO

Bovine mastitis remains a major disease in cattle world-wide. In the mammary gland, mammary epithelial cells (MEC) are sentinels equipped with receptors allowing them to detect and respond to the invasion by bacterial pathogens, in particular Escherichia coli. Lipopolysaccharide (LPS) is the major E. coli motif recognized by MEC through its interaction with the TLR4 receptor and the CD14 co-receptor. Previous studies have highlighted the role of soluble CD14 (sCD14) in the efficient recognition of LPS molecules possessing a full-length O-antigen (LPSS). We demonstrate here that MEC are able to secrete CD14 and are likely to contribute to the presence of sCD14 in milk. We then investigated how sCD14 modulates and is required for the response of MEC to LPSS. This study highlights the key role of sCD14 for the full activation of the Myd88-independent pathway by LPSS. We also identified several lncRNA that are activated in MEC in response to LPS, including one lncRNA showing homologies with the mir-99a-let-7c gene (MIR99AHG). Altogether, our results show that a full response to LPS by mammary epithelial cells requires sCD14 and provide detailed information on how milk sCD14 can contribute to an efficient recognition of LPS from coliform pathogens.


Assuntos
Células Epiteliais , Receptores de Lipopolissacarídeos , Lipopolissacarídeos , Glândulas Mamárias Animais , Animais , Receptores de Lipopolissacarídeos/metabolismo , Receptores de Lipopolissacarídeos/genética , Bovinos , Células Epiteliais/metabolismo , Lipopolissacarídeos/farmacologia , Feminino , Glândulas Mamárias Animais/metabolismo , Mastite Bovina/microbiologia , Mastite Bovina/imunologia , Mastite Bovina/metabolismo , Leite
2.
PLoS One ; 13(8): e0202664, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30142177

RESUMO

Escherichia coli is one of the major pathogens causing mastitis in dairy cattle. Yet, the factors which mediate the ability for E. coli to develop in the bovine mammary gland remain poorly elucidated. In a mouse model, infections induced by the reference mastitis E. coli P4 showed a strong colonisation of the mammary gland, while this strain had a low stimulating power on cells of the PS bovine mammary epithelial cell line. In order to understand if such a reduced response contributes to the severity of infection, a library of random mutants of P4 strain was screened to identify mutants inducing stronger response of PS cells. Among hyper-stimulating mutants, six were altered in genes involved in biosynthesis of lipopolysaccharide (LPS) and had lost their O-polysaccharide region, suggesting that the presence of O-antigen impairs the response of PS cells to LPS. Using purified smooth (S) and rough (R) fractions of LPS, we showed that the R-LPS fraction induced a stronger response from PS cells than the smooth LPS fraction. Biological activity of the S-LPS fraction could be restored by the addition of recombinant bovine CD14 (rbCD14), indicating a crucial role of CD14 in the recognition of S-LPS by Mammary Epithelial Cells (MEC). When S-LPS and R-LPS were injected in udder quarters of healthy lactating cows, an inflammation developed in all infused quarters, but the S-LPS induced a more intense pro-inflammatory response, possibly in relation to sizeable concentrations of CD14 in milk. Altogether, our results demonstrate that the O-antigen modulates the pro-inflammatory response of MEC to LPS, that S-LPS and R-LPS trigger different responses of MEC and that these responses depend on the presence of CD14.


Assuntos
Escherichia coli/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/imunologia , Antígenos O/metabolismo , Animais , Bovinos , Linhagem Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Elementos de DNA Transponíveis/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/isolamento & purificação , Feminino , Células HEK293 , Humanos , Receptores de Lipopolissacarídeos/química , Receptores de Lipopolissacarídeos/genética , Lipopolissacarídeos/análise , Lipopolissacarídeos/metabolismo , Mastite Bovina/imunologia , Mastite Bovina/microbiologia , Mastite Bovina/patologia , Leite/metabolismo , Leite/microbiologia , Mutagênese , Antígenos O/química , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
3.
PLoS One ; 11(8): e0161573, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27561012

RESUMO

Defensins are frontline peptides of mucosal immunity in the animal kingdom, including birds. Their resistance to proteolysis and their ensuing ability to maintain antimicrobial potential remains questionable and was therefore investigated. We have shown by bottom-up mass spectrometry analysis of protein extracts that both avian beta-defensins AvBD2 and AvBD7 were ubiquitously distributed along the chicken gut. Cathepsin B was found by immunoblotting in jejunum, ileum, caecum, and caecal tonsils, while cathepsins K, L, and S were merely identified in caecal tonsils. Hydrolysis product of AvBD2 and AvBD7 incubated with a panel of proteases was analysed by RP-HPLC, mass spectrometry and antimicrobial assays. AvBD2 and AvBD7 were resistant to serine proteases and to cathepsins D and H. Conversely cysteine cathepsins B, K, L, and S degraded AvBD2 and abolished its antibacterial activity. Only cathepsin K cleaved AvBD7 and released Ile4-AvBD7, a N-terminal truncated natural peptidoform of AvBD7 that displayed antibacterial activity. Besides the 3-stranded antiparallel beta-sheet typical of beta-defensins, structural analysis of AvBD7 by two-dimensional NMR spectroscopy highlighted the restricted accessibility of the C-terminus embedded by the N-terminal region and gave a formal evidence of a salt bridge (Asp9-Arg12) that could account for proteolysis resistance. The differential susceptibility of avian defensins to proteolysis opens intriguing questions about a distinctive role in the mucosal immunity against pathogen invasion.


Assuntos
Galinhas/imunologia , Peptídeo Hidrolases/metabolismo , beta-Defensinas/metabolismo , Animais , Catepsina B/metabolismo , Catepsina D/metabolismo , Catepsina K/metabolismo , Catepsina L/metabolismo , Catepsinas/metabolismo , Quimotripsina/química , Hidrólise , Mucosa Intestinal/metabolismo , Elastase de Leucócito/metabolismo , Espectrometria de Massas , Conformação Molecular , Tonsila Palatina/metabolismo , Proteólise , Tripsina/química
4.
Vet Res ; 46: 56, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-26062913

RESUMO

Mastitis remains a major disease of cattle with a strong impact on the dairy industry. There is a growing interest in understanding how cell mediated immunity contributes to the defence of the mammary gland against invading mastitis causing bacteria. Cytokines belonging to the IL-17 family, and the cells that produce them, have been described as important modulators of the innate immunity, in particular that of epithelial cells. We report here that expression of IL-17A and IL-17F genes, encoding two members of the IL-17 family, are induced in udder tissues of cows experimentally infected with Escherichia coli. The impact of IL-17A on the innate response of bovine mammary epithelial cells was investigated using a newly isolated cell line, the PS cell line. We first showed that PS cells, similar to primary bovine mammary epithelial cells, were able to respond to agonists of TLR2 and to LPS, provided CD14 was added to the culture medium. We then showed that secretion of CXCL8 and transcription of innate immunity related-genes by PS cells were increased by IL-17A, in particular when these cells were stimulated with live E. coli bacteria. Together with data from the literature, these results support the hypothesis that IL-17A and IL-17 F could play an important role in mediating of host-pathogen interactions during mastitis.


Assuntos
Infecções por Escherichia coli/veterinária , Escherichia coli/fisiologia , Regulação da Expressão Gênica , Imunidade Inata , Interleucina-17/genética , Mastite Bovina/genética , Mastite Bovina/imunologia , Animais , Bovinos , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Feminino , Interleucina-17/metabolismo , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/microbiologia , Mastite Bovina/microbiologia
5.
Eukaryot Cell ; 13(7): 884-95, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24839124

RESUMO

Aminopeptidases N are metalloproteases of the M1 family that have been reported in numerous apicomplexan parasites, including Plasmodium, Toxoplasma, Cryptosporidium, and Eimeria. While investigating the potency of aminopeptidases as therapeutic targets against coccidiosis, one of the most important avian diseases caused by the genus Eimeria, we identified and characterized Eimeria tenella aminopeptidase N1 (EtAPN1). Its inhibition by bestatin and amastatin, as well as its reactivation by divalent ions, is typical of zinc-dependent metalloproteases. EtAPN1 shared a similar sequence, three-dimensional structure, and substrate specificity and similar kinetic parameters with A-M1 from Plasmodium falciparum (PfA-M1), a validated target in the treatment of malaria. EtAPN1 is synthesized as a 120-kDa precursor and cleaved into 96-, 68-, and 38-kDa forms during sporulation. Further, immunolocalization assays revealed that, similar to PfA-M1, EtAPN1 is present during the intracellular life cycle stages in both the parasite cytoplasm and the parasite nucleus. The present results support the hypothesis of a conserved role between the two aminopeptidases, and we suggest that EtAPN1 might be a valuable target for anticoccidiosis drugs.


Assuntos
Aminopeptidases/metabolismo , Eimeria tenella/enzimologia , Metaloproteases/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Aminopeptidases/química , Aminopeptidases/genética , Antiprotozoários/farmacologia , Eimeria tenella/efeitos dos fármacos , Eimeria tenella/crescimento & desenvolvimento , Leucina/análogos & derivados , Leucina/farmacologia , Metaloproteases/química , Metaloproteases/genética , Dados de Sequência Molecular , Peptídeos/farmacologia , Filogenia , Precursores de Proteínas/metabolismo , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Esporos de Protozoários/crescimento & desenvolvimento , Esporos de Protozoários/metabolismo , Especificidade por Substrato
6.
Vet Res ; 44: 40, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23758654

RESUMO

Mastitis caused by Escherichia coli and Staphylococcus aureus is a major pathology of dairy cows. To better understand the differential response of the mammary gland to these two pathogens, we stimulated bovine mammary epithelial cells (bMEC) with either E. coli crude lipopolysaccharide (LPS) or with S. aureus culture supernatant (SaS) to compare the transcriptomic profiles of the initial bMEC response. By using HEK 293 reporter cells for pattern recognition receptors, the LPS preparation was found to stimulate TLR2 and TLR4 but not TLR5, Nod1 or Nod2, whereas SaS stimulated TLR2. Biochemical analysis revealed that lipoteichoic acid, protein A and α-hemolysin were all present in SaS, and bMEC were found to be responsive to each of these molecules. Transcriptome profiling revealed a core innate immune response partly shared by LPS and SaS. However, LPS induced expression of a significant higher number of genes and the fold changes were of greater magnitude than those induced by SaS. Microarray data analysis suggests that the activation pathways and the early chemokine and cytokine production preceded the defense and stress responses. A major differential response was the activation of the type I IFN pathway by LPS but not by SaS. The higher upregulation of chemokines (Cxcl10, Ccl2, Ccl5 and Ccl20) that target mononuclear leucocytes by LPS than by SaS is likely to be related to the differential activation of the type I IFN pathway, and could induce a different profile of the initial recruitment of leucocytes. The MEC responses to the two stimuli were different, as LPS was associated with NF-κB and Fas signaling pathways, whereas SaS was associated with AP-1 and IL-17A signaling pathways. It is noteworthy that at the protein level secretion of TNF-α and IL-1ß was not induced by either stimulus. These results suggest that the response of MEC to diffusible stimuli from E. coli and S. aureus contributes to the onset of the response with differential leucocyte recruitment and distinct inflammatory and innate immune reactions of the mammary gland to infection.


Assuntos
Infecções por Escherichia coli/imunologia , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intracelular/genética , Glândulas Mamárias Animais/imunologia , Mastite Bovina/imunologia , Infecções Estafilocócicas/imunologia , Receptores Toll-Like/genética , Animais , Bovinos , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática/veterinária , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Escherichia coli/fisiologia , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Feminino , Perfilação da Expressão Gênica/veterinária , Regulação da Expressão Gênica , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/fisiologia , Glândulas Mamárias Animais/microbiologia , Mastite Bovina/genética , Mastite Bovina/microbiologia , Proteínas Adaptadoras de Sinalização NOD/genética , Proteínas Adaptadoras de Sinalização NOD/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Transdução de Sinais , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Receptores Toll-Like/metabolismo
7.
Physiol Genomics ; 44(7): 403-16, 2012 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-22337903

RESUMO

Staphylococcus aureus is a prevalent pathogen for mastitis in dairy ruminants and is responsible for both clinical and subclinical mastitis. Mammary epithelial cells (MEC) represent not only a physical barrier against bacterial invasion but are also active players of the innate immune response permitting infection clearance. To decipher their functions in general and in animals showing different levels of genetic predisposition to Staphylococcus in particular, MEC from ewes undergoing a divergent selection on milk somatic cell count were stimulated by S. aureus. MEC response was also studied according to the stimulation condition with live bacteria or culture supernatant. The early MEC response was studied during a 5 h time course by microarray to identify differentially expressed genes with regard to the host genetic background and as a function of the conditions of stimulation. In both conditions of stimulation, metabolic processes were altered, the apoptosis-associated pathways were considerably modified, and inflammatory and immune responses were enhanced with the upregulation of il1a, il1b, and tnfa and several chemokines known to enhance neutrophil (cxcl8) or mononuclear leukocyte (ccl20) recruitment. Genes associated with oxidative stress were increased after live bacteria stimulation, whereas immune response-related genes were higher after supernatant stimulation in the early phase. Only 20 genes were differentially expressed between Staphylococcus spp-mastitis resistant and susceptible animals without any clearly defined role on the control of infection. To conclude, this suggests that MEC may not represent the cell type at the origin of the difference of mastitis susceptibility, at least as demonstrated in our genetic model. Supernatant or heat-killed S. aureus produce biological effects that are essentially different from those induced by live bacteria.


Assuntos
Células Epiteliais/metabolismo , Perfilação da Expressão Gênica , Predisposição Genética para Doença , Glândulas Mamárias Animais/patologia , Mastite/veterinária , Ovinos/genética , Staphylococcus aureus/fisiologia , Animais , Análise por Conglomerados , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Feminino , Redes Reguladoras de Genes/genética , Glândulas Mamárias Animais/microbiologia , Mastite/genética , Mastite/microbiologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ovinos/microbiologia , Frações Subcelulares/metabolismo
8.
Vet Res ; 43: 14, 2012 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-22330199

RESUMO

Escherichia coli is a frequent cause of clinical mastitis in dairy cows. It has been shown that a prompt response of the mammary gland after E. coli entry into the lumen of the gland is required to control the infection, which means that the early detection of bacteria is of prime importance. Yet, apart from lipopolysaccharide (LPS), little is known of the bacterial components which are detected by the mammary innate immune system. We investigated the repertoire of potential bacterial agonists sensed by the udder and bovine mammary epithelial cells (bMEC) during E. coli mastitis by using purified or synthetic molecular surrogates of bacterial agonists of identified pattern-recognition receptors (PRRs). The production of CXCL8 and the influx of leucocytes in milk were the readouts of reactivity of stimulated cultured bMEC and challenged udders, respectively. Quantitative PCR revealed that bMEC in culture expressed the nucleotide oligomerization domain receptors NOD1 and NOD2, along with the Toll-like receptors TLR1, TLR2, TLR4, and TLR6, but hardly TLR5. In line with expression data, bMEC proved to react to the cognate agonists C12-iE-DAP (NOD1), Pam3CSK4 (TLR1/2), Pam2CSK4 (TLR2/6), pure LPS (TLR4), but not to flagellin (TLR5). As the udder reactivity to NOD1 and TLR5 agonists has never been reported, we tested whether the mammary gland reacted to intramammary infusion of C12-iE-DAP or flagellin. The udder reacted to C12-iE-DAP, but not to flagellin, in line with the reactivity of bMEC. These results extend our knowledge of the reactivity of the bovine mammary gland to bacterial agonists of the innate immune system, and suggest that E. coli can be recognized by several PRRs including NOD1, but unexpectedly not by TLR5. The way the mammary gland senses E. coli is likely to shape the innate immune response and finally the outcome of E. coli mastitis.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/genética , Células Epiteliais/imunologia , Infecções por Escherichia coli/veterinária , Proteínas de Escherichia coli/metabolismo , Glândulas Mamárias Animais/imunologia , Mastite Bovina/imunologia , Receptores Toll-Like/genética , Animais , Proteínas Adaptadoras de Sinalização CARD/agonistas , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Bovinos , Células Cultivadas , Células Epiteliais/microbiologia , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Feminino , Imunidade Inata , Glândulas Mamárias Animais/microbiologia , Mastite Bovina/microbiologia , Reação em Cadeia da Polimerase/veterinária , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
9.
Cytokine ; 56(3): 749-59, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22004923

RESUMO

Interleukin-17A (IL-17A) and IL-17F have been shown to mediate a crucial crosstalk between the immune system and various epithelial tissues, stimulating various defensive mechanisms to bacterial infections. A number of studies have characterized the response to IL-17A and IL-17F of epithelial cells from airways, intestine, and skin, but not from the mammary gland. To evaluate the potential contribution of IL-17 to the immune defense of the mammary gland, we analyzed the effects of recombinant bovine IL-17A and IL-17F on primary bovine mammary epithelial cells (MEC) by quantitative PCR and ELISA. We found expression (mRNA) of the two components of the IL-17 receptor complex, IL-17RA and IL-17RC, in mammary tissue and MEC in vitro. The expression of a number of genes encoding cytokines, chemokines and proteins endowed with antibacterial activities was increased by IL-17A, and to a lesser extent by IL-17F, but the magnitude of responses was modest. As expected, responses were augmented by the combination of IL-17A or IL-17F with TNF-α. Interestingly, responses of a few of the tested genes, such as IL8, CCL20, iNOS, and CfB, were augmented by the combination of IL-17A with staphylococcal lipoteichoic acid or muramyl dipeptide, bacterial agonists of the innate immune system. This can be interpreted as indicating that IL-17A and IL-17F are tailored to exert their full potential in a septic environment. MEC responses were characterized by the expression of chemokines targeting not only neutrophils (CXCL3 and CXCL8) but also mononuclear leucocytes (CCL2, CCL20). Production of IL-6 was low and the inflammatory cytokines TNF-α and IL-1ß were expressed (mRNA) but proteins were not secreted. Altogether, our results suggest that IL-17A and IL-17F have a potential to modulate the mammary gland immune response to mastitis-causing pathogens.


Assuntos
Células Epiteliais/microbiologia , Regulação da Expressão Gênica/imunologia , Imunidade/genética , Interleucina-17/farmacologia , Glândulas Mamárias Animais/citologia , Receptores de Reconhecimento de Padrão/imunologia , Staphylococcus/imunologia , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Animais , Bovinos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunidade/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Interleucina-8/metabolismo , Lipopolissacarídeos/farmacologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/imunologia , Staphylococcus/efeitos dos fármacos , Ácidos Teicoicos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
10.
Microb Pathog ; 51(6): 396-401, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21951578

RESUMO

An early recruitment of neutrophils in mammary tissue and milk is considered an important component of the defense of the mammary gland against Staphylococcus aureus. We investigated whether the leukotoxin LukM/F', which is produced by a proportion of mastitis-causing strains of S. aureus, would be able to trigger inflammation in the udder. Infusion of purified LukM/F' toxin in lactating mammary glands did not cause neutrophil influx in milk, showing that the toxin was not able to cause mastitis on its own. Purified LukM/F' did not kill or stimulate mammary epithelial cells in culture. As expected, LukM bound to mammary macrophages and the complete LukM/F' toxin killed these cells, but subcytotoxic LukM/F' concentrations did not induce secretion of IL-8, TNF-α, IL-1ß or IL-6 by macrophages. On the contrary, the production of these pro-inflammatory mediators by adhesion-stimulated macrophages was reduced. Overall, these results indicate that purified leukotoxin LukM/F' is not likely to contribute to the initiation of the inflammatory response and could even play an anti-inflammatory role in the mammary gland by inactivating macrophages.


Assuntos
Proteínas de Bactérias/metabolismo , Leucocidinas/metabolismo , Glândulas Mamárias Animais/microbiologia , Glândulas Mamárias Animais/patologia , Staphylococcus aureus/patogenicidade , Fatores de Virulência/metabolismo , Animais , Bovinos , Sobrevivência Celular , Células Cultivadas , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Feminino , Inflamação/microbiologia , Inflamação/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Neutrófilos/imunologia
11.
Clin Vaccine Immunol ; 17(11): 1797-809, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20826612

RESUMO

Staphylococcus aureus, a major pathogen for the mammary gland of dairy ruminants, elicits the recruitment of neutrophils into milk during mastitis, but the mechanisms are incompletely understood. We investigated the response of the bovine mammary gland to muramyl dipeptide (MDP), an elementary constituent of the bacterial peptidoglycan, alone or in combination with lipoteichoic acid (LTA), another staphylococcal microbial-associated molecular pattern (MAMP). MDP induced a prompt and marked influx of neutrophils in milk, and its combination with LTA elicited a more intense and prolonged influx than the responses to either stimulus alone. The concentrations of several chemoattractants for neutrophils (CXCL1, CXCL2, CXCL3, CXCL8, and C5a) increased in milk after challenge, and the highest increases followed challenge with the combination of MDP and LTA. MDP and LTA were also synergistic in inducing in vitro chemokine production by bovine mammary epithelial cells (bMEpC). Nucleotide-binding oligomerization domain 2 (NOD2), a major sensor of MDP, was expressed (mRNA) in bovine mammary tissue and by bMEpC in culture. The production of interleukin-8 (IL-8) following the stimulation of bMEpC by LTA and MDP was dependent on the activation of NF-κB. LTA-induced IL-8 production did not depend on platelet-activating factor receptor (PAFR), as the PAFR antagonist WEB2086 was without effect. In contrast, bMEpC and mammary tissue are known to express Toll-like receptor 2 (TLR2) and to respond to TLR2 agonists. Although the levels of expression of the inflammatory cytokines tumor necrosis factor alpha (TNF-α) and IL-1ß were increased by LTA and MDP at the mRNA level, no protein could be detected in the bMEpC culture supernatant. The level of induction of IL-6 was low at both the mRNA and protein levels. These results indicate that MDP and LTA exert synergistic effects to induce neutrophilic inflammation in the mammary gland. These results also show that bMEpC could contribute to the inflammatory response by recognizing LTA and MDP and secreting chemokines but not proinflammatory cytokines. Overall, this study indicates that the TLR2 and NOD2 pathways could cooperate to trigger an innate immune response to S. aureus mastitis.


Assuntos
Acetilmuramil-Alanil-Isoglutamina/imunologia , Lipopolissacarídeos/imunologia , Glândulas Mamárias Animais/imunologia , Mastite Bovina/microbiologia , Neutrófilos/imunologia , Infecções Estafilocócicas/veterinária , Staphylococcus aureus/patogenicidade , Ácidos Teicoicos/imunologia , Acetilmuramil-Alanil-Isoglutamina/toxicidade , Animais , Bovinos , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Lipopolissacarídeos/toxicidade , Glândulas Mamárias Animais/patologia , Mastite Bovina/patologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/imunologia , Ácidos Teicoicos/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA