Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Molecules ; 28(13)2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37446705

RESUMO

The signal transduction paradigm in bacteria involves two-component systems (TCSs). Asgardarchaeota are archaea that may have originated the current eukaryotic lifeforms. Most research on these archaea has focused on eukaryotic-like features, such as genes involved in phagocytosis, cytoskeleton structure, and vesicle trafficking. However, little attention has been given to specific prokaryotic features. Here, the sequence and predicted structural features of TCS sensor kinases analyzed from two metagenome assemblies and a genomic assembly from cultured Asgardian archaea are presented. The homology of the sensor kinases suggests the grouping of Lokiarchaeum closer to bacterial homologs. In contrast, one group from a Lokiarchaeum and a meta-genome assembly from Candidatus Heimdallarchaeum suggest the presence of a set of kinases separated from the typical bacterial TCS sensor kinases. AtoS and ArcB homologs were found in meta-genome assemblies along with defined domains for other well-characterized sensor kinases, suggesting the close link between these organisms and bacteria that may have resulted in the metabolic link to the establishment of symbiosis. Several kinases are predicted to be cytoplasmic; some contain several PAS domains. The data shown here suggest that TCS kinases in Asgardian bacteria are witnesses to the transition from bacteria to eukaryotic organisms.


Assuntos
Archaea , Células Eucarióticas , Archaea/genética , Archaea/metabolismo , Bactérias/genética , Eucariotos/genética , Células Procarióticas , Evolução Molecular , Filogenia
3.
Parasitol Res ; 121(12): 3503-3512, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36171407

RESUMO

The parasite Trichomonas vaginalis is the aetiologic agent of trichomoniasis, the most common non-viral sexually transmitted disease worldwide. This infection often remains asymptomatic and is related to several health complications. The traditional treatment for trichomoniasis uses drugs of the 5-nitroimidazole family, such as metronidazole; however, scientific reports indicate an increasing number of drug-resistant strains. Antimicrobial peptides could be an alternative or complementary treatment. In this sense, one attractive candidate is the human cathelicidin, being LL-37 its active form. LL-37 possesses microbicidal activity against many microorganisms such as bacteria, Candida albicans, and Entamoeba histolytica. Shorter sequences derived from this peptide, such as KR-20, FK-13 and KR-12, have been shown to possess a higher microbicidal effect than LL-37. In this study, we determined the activity of LL-37 and its derivatives against T. vaginalis, which was unknown. The results showed that the four peptides (LL-37, KR-20, FK-13-NH2 and KR-12) decreased the viability of T. vaginalis on a 5-nitroimidazole-sensitive and a 5-nitroimidazole-resistant strain; however, KR-20 was the most effective peptide, followed by FK-13-NH2. Low concentrations of all peptides showed a better effect when combined with metronidazole in the sensitive and resistant T. vaginalis strains. These results are promising for potential future therapeutic uses.


Assuntos
Antiprotozoários , Tricomoníase , Trichomonas vaginalis , Humanos , Metronidazol/farmacologia , Peptídeos Antimicrobianos , Resistência a Medicamentos , Antiprotozoários/farmacologia
4.
Front Vet Sci ; 7: 390, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32793642

RESUMO

Defensins are an important group of host defense peptides. They have immunomodulatory properties, which have been mainly described for mammal defensins, but similar effects for plant defensins remain unknown. Previously, we showed that the defensin γ-thionin (Capsicum chinense) reduces Staphylococcus aureus internalization into bovine mammary epithelial cells (bMECs) while inducing Toll-like receptor 2 (TLR2), modulating the inflammatory response. Here, we analyze the effect of γ-thionin on the TLR2 pathway in bMECs infected with S. aureus and determine if it modulates epigenetic marks. Pre-treated bMECs with γ-thionin (100 ng/ml) reduced the basal activation of p38 and ERK1/2 (~3-fold), but JNK was increased (~1.5-fold). Also, infected bMECs induced p38, but this effect was reversed by γ-thionin, whereas ERK1/2 was reduced by infection but stimulated by γ-thionin. Likewise, γ-thionin reduced the activation of Akt kinase ~50%. Furthermore, γ-thionin induced the activation of transcriptional factors of inflammatory response, highlighting EGR, E2F-1, AP-1, and MEF, which were turned off by bacteria. Also, γ-thionin induced the activation of histone deacetylases (HDACs, ~4-fold) at 24 h in infected bMECs and reduced LSD1 demethylase (HDMs, ~30%) activity. Altogether, these results demonstrated the first time that a plant defensin interferes with inflammatory signaling pathways in mammalian cells.

5.
J Immunol Res ; 2019: 7083491, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31612151

RESUMO

Bovine mammary epithelial cells (bMECs) are capable of initiating an innate immune response (IIR) to invading bacteria. Staphylococcus aureus is not classically an intracellular pathogen, although it has been shown to be internalized into bMECs. S. aureus internalizes into nonprofessional phagocytes, which allows the evasion of the IIR and turns antimicrobial therapy unsuccessful. An alternative treatment to control this pathogen is the modulation of the innate immune response of the host. The Mexican avocado (Persea americana var. drymifolia) is a source of molecules with anti-inflammatory and immunomodulatory properties. Hence, we analyze the effect of a lipid-rich extract from avocado seed (LEAS) on S. aureus internalization into bMECs and their innate immunity response. The effects of LEAS (1-500 ng/ml) on the S. aureus growth and bMEC viability were assessed by turbidimetry and MTT assays, respectively. LEAS did not show neither antimicrobial nor cytotoxic effects. S. aureus internalization into bMECs was analyzed by gentamicin protection assays. Interestingly, LEAS (1-200 ng/ml) decreased bacterial internalization (60-80%) into bMECs. This effect correlated with NO production and the induction of the gene expression of IL-10, while the expression of the proinflammatory cytokine TNF-α was reduced. These effects could be related to the inhibition of MAPK p38 (∼60%) activation by LEAS. In conclusion, our results showed that LEAS inhibits the S. aureus internalization into bMECs and modulates the IIR, which indicates that avocado is a source of metabolites for control of mastitis pathogens.


Assuntos
Anti-Infecciosos/farmacologia , Extratos Vegetais/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/fisiologia , Animais , Bovinos , Linhagem Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/efeitos dos fármacos , Mastite Bovina/tratamento farmacológico , Persea , Sementes , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Microb Pathog ; 131: 15-21, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30930221

RESUMO

Staphylococcus aureus is a major pathogen of subclinical bovine mastitis that usually is chronic and recurrent, which has been related to its ability to internalize into bovine mammary epithelial cells (bMECs). Previously, we reported that short and medium fatty acids and cholecalciferol reduce S. aureus internalization into pretreated-bMECs with these molecules suggesting a role as immunomodulatory agents. Hence, we assessed the role of sodium butyrate (NaB), sodium octanoate (NaO) and cholecalciferol on S. aureus adhesin expression and its internalization into bMECs. S. aureus pre-treated 2 h with 0.5 mM or 2 mM NaB showed a reduction in internalization into bMECs (∼35% and ∼55%; respectively), which coincided with a down-regulated expression of clumping factor B (ClfB). Also, the S. aureus internalization reduction by 2 mM NaB (2 h) agreed with a down-regulated expression of sdrC. Moreover, the 2 mM NaB (24 h) pre-treatment induced bacterial internalization (∼3-fold), which was related with an up-regulation of spa, clfB and sdrC genes. Also, NaO (0.25 mM and 1 mM) only reduced S. aureus internalization when bacteria were grown 2 h with this molecule but there was no relationship with adhesin expression. In addition, cholecalciferol (50 nM) reduced bacteria internalization at similar levels (∼50%) when bacteria were grown 2 and 24 h in broth supplemented with this compound, which correlated with spa and sdrC mRNA expression down-regulated at 2 h, and fnba and clfB mRNA expression decreased at 24 h. In conclusion, our data support the fact that fatty acids and cholecalciferol regulate adhesin gene expression as well as bacteria internalization in nonprofessional phagocytic cells, which may lead to development of anti-virulence agents for control of pathogens.


Assuntos
Adesinas Bacterianas/genética , Células Epiteliais/imunologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Glândulas Mamárias Animais/imunologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Adesinas Bacterianas/efeitos dos fármacos , Animais , Proteínas de Bactérias/genética , Ácido Butírico , Caprilatos/farmacologia , Bovinos , Linhagem Celular , Colecalciferol/farmacologia , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/microbiologia , Ácidos Graxos/farmacologia , Feminino , Gentamicinas/farmacologia , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Imunomodulação , Glândulas Mamárias Animais/microbiologia , Mastite Bovina/imunologia , Mastite Bovina/microbiologia , Mastite Bovina/prevenção & controle , RNA Mensageiro/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Fatores de Virulência/genética
7.
J Immunol Res ; 2018: 6098961, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30246035

RESUMO

The innate immune system can function under hormonal control. 17ß-Estradiol (E2) is an important sexual hormone for the reproductive cycle of mammals, and it has immunomodulatory effects on epithelial cells, which are the first line of defense against incoming bacteria. E2 regulates various pathophysiological processes, including the response to infection in epithelial cells, and its effects involve the regulation of innate immune signaling pathways, which are mediated through estrogen receptors (ERs). E2 modulates the expression of inflammatory and antimicrobial elements such as cytokines and antimicrobial peptides. The E2 effects on epithelial cells during bacterial infections are characterized by an increase in the production of antimicrobial peptides and by the diminution of the inflammatory response to abrogate proinflammatory cytokine induction by bacteria. Here, we review several novel molecular mechanisms through which E2 regulates the innate immune response of epithelial cells against bacterial infections.


Assuntos
Infecções Bacterianas/tratamento farmacológico , Células Epiteliais/efeitos dos fármacos , Estradiol/farmacologia , Fatores Imunológicos/farmacologia , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Citocinas/metabolismo , Células Epiteliais/imunologia , Humanos , Imunidade Inata , Receptores de Estrogênio/metabolismo , Transdução de Sinais
8.
Artigo em Inglês | MEDLINE | ID: mdl-28361042

RESUMO

Bovine mammary epithelial cells (bMECs) contribute to mammary gland defense against invading pathogens, such as Staphylococcus aureus (intracellular facultative), which is recognized by TLR2. In a previous report, we showed that sodium octanoate [NaO, a medium chain fatty acid (C8)] induces (0.25 mM) or inhibits (1 mM) S. aureus internalization into bMECs and differentially regulates the innate immune response (IIR). However, the molecular mechanisms have not been described, which was the aim of this study. The results showed that α5ß1 integrin membrane abundance (MA) was increased in 0.25 mM NaO-treated cells, but TLR2 or CD36 MA was not modified. When these receptors were blocked individually, 0.25 mM NaO-increased S. aureus internalization was notably reduced. Interestingly, in this condition, the IIR of the bMECs was impaired because MAPK (p38, JNK, and ERK1/2) phosphorylation and the activation of transcription factors related to these pathways were decreased. In addition, the 1 mM NaO treatment induced TLR2 MA, but neither the integrin nor CD36 MA was modified. The reduction in S. aureus internalization induced by 1 mM NaO was increased further when TLR2 was blocked. In addition, the phosphorylation levels of the MAPKs increased, and 13 transcriptional factors related to the IIR were slightly activated (CBF, CDP, c-Myb, AP-1, Ets-1/Pea-3, FAST-1, GAS/ISRE, AP-2, NFAT-1, OCT-1, RAR/DR-5, RXR/DR-1, and Stat-3). Moreover, the 1 mM NaO treatment up-regulated gene expression of IL-8 and RANTES and secretion of IL-1ß. Notably, when 1 mM NaO-treated bMECs were challenged with S. aureus, the gene expression of IL-8 and IL-10 increased, while IL-1ß secretion was reduced. In conclusion, our results showed that α5ß1 integrin, TLR2 and CD36 are involved in 0.25 mM NaO-increased S. aureus internalization in bMECs. In addition, 1 mM NaO activates bMECs via the TLR2 signaling pathways (p38, JNK, and ERK1/2), which improves IIR before S. aureus invasion. Additionally, NaO (1 mM) might exert anti-inflammatory effects after bacterial internalization.


Assuntos
Caprilatos/metabolismo , Endocitose , Células Epiteliais/microbiologia , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/metabolismo , Sistema de Sinalização das MAP Quinases , Staphylococcus aureus/fisiologia , Animais , Bovinos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Staphylococcus aureus/imunologia
9.
Mol Immunol ; 68(2 Pt B): 445-55, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26471700

RESUMO

Staphylococcus aureus is an etiological agent of human and animal diseases, and it is able to internalize into non-professional phagocytic cells (i.e. bovine mammary epithelial cells, bMECs), which is an event that is related to chronic and recurrent infections. bMECs contribute to host innate immune responses (IIR) through TLR pathogen recognition, whereby TLR2 is the most relevant for S. aureus. In a previous report, we showed that sodium butyrate (NaB, 0.5mM), which is a short chain fatty acid (SCFA), reduced S. aureus internalization into bMECs by modulating their IIR. However, the molecular mechanism of this process has not been described, which was the aim of this study. The results showed that the TLR2 membrane abundance (MA) and mRNA expression were induced by 0.5mM NaB ∼1.6-fold and ∼1.7-fold, respectively. Additionally, 0.5mM NaB induced p38 phosphorylation, but not JNK1/2 or ERK1/2 phosphorylation in bMECs, which reached the baseline when the bMECs were S. aureus-challenged. Additionally, bMECs that were treated with 0.5mM NaB (24h) showed activation of 8 transcriptional factors (AP-1, E2F-1, FAST-1, MEF-1, EGR, PPAR, ER and CBF), which were partially reverted when the bMECs were S. aureus-challenged. Additionally, 0.5mM NaB (24h) up-regulated mRNA expression of the antimicrobial peptides, TAP (∼4.8-fold), BNBD5 (∼3.2-fold) and BNBD10 (∼2.6-fold). Notably, NaB-treated and S. aureus-challenged bMECs increased the mRNA expression of all of the antimicrobial peptides that were evaluated, and this was evident for LAP and BNBD5. In the NaB-treated bMECs, we did not detect significant expression changes for IL-1ß and IL-6 and only TNF-α, IL-10 and IL-8 were induced. Interestingly, the NaB-treated and S. aureus-challenged bMECs maintained the anti-inflammatory response that was induced by this SCFA. In conclusion, our results suggest that 0.5mM NaB activates bMECs via TLR2/p38, which leads to improved antimicrobial defense before/after pathogen invasion, and NaB may exert anti-inflammatory effects during infection.


Assuntos
Ácido Butírico/farmacologia , Glândulas Mamárias Animais/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Receptor 2 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Transporte Biológico/efeitos dos fármacos , Antígenos CD36/metabolismo , Bovinos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Interleucina-10/biossíntese , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Interleucina-8/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Oligopeptídeos/biossíntese , Oligopeptídeos/genética , Fosforilação , RNA Mensageiro/biossíntese , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , beta-Defensinas/biossíntese , beta-Defensinas/genética
10.
Microb Pathog ; 89: 43-53, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26341952

RESUMO

Staphylococcus aureus has the ability to invade mammary epithelial cells (bMECs) causing mastitis. This event depends primarily on the α5ß1 integrin in the host cell. In addition, bMECs are a target for the hormone prolactin (PRL), which can regulate ß1 integrin-dependent actions related to differentiation and lactation. Previously, we demonstrated that bovine PRL (bPRL, 5 ng/ml) stimulates S. aureus internalization into bMECs. TLR2 is important during S. aureus infections, but its activation by PRL has not yet been established. The objective of this study was to determine the role of α5ß1 integrin and TLR2 during S. aureus internalization into bMECs stimulated with bPRL. We demonstrated that the prolactin-stimulated internalization of S. aureus decreases in response to the blockage of α5ß1 integrin (∼ 80%) and TLR2 (∼ 80%). bPRL increases the membrane abundance (MA) of α5ß1 integrin (∼ 20%) and induces TLR2 MA (∼ 2-fold). S. aureus reduces the α5ß1 integrin MA in bMECs treated with bPRL (∼ 75%) but induces TLR2 MA in bMECs (∼ 3-fold). Bacteria and bPRL did not modify TLR2 MA compared with the hormone alone. S. aureus induces the activation of the transcription factor AP-1, which was inhibited in bMECs treated with bPRL and infected. In general, bPRL induces both pro- and anti-inflammatory responses in bMECs, which are abated in response to bacterial challenge. Interestingly, the canonical Stat-5 transcription factor was not activated in the challenged bMECs and/or treated with bPRL. Taken together, these results support novel functions of prolactin as a modulator of the innate immune response that do not involve the classical prolactin pathway.


Assuntos
Endocitose , Células Epiteliais/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/metabolismo , Prolactina/metabolismo , Staphylococcus aureus/fisiologia , Receptor 2 Toll-Like/metabolismo , Animais , Bovinos , Células Cultivadas , Células Epiteliais/imunologia , Integrina alfa5beta1/metabolismo
11.
Microbiology (Reading) ; 161(7): 1516-23, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25918254

RESUMO

The Pseudomonas aeruginosa plasmid pUM505 contains the umuDC operon that encodes proteins similar to error-prone repair DNA polymerase V. The umuC gene appears to be truncated and its product is probably not functional. The umuD gene, renamed umuDpR, possesses an SOS box overlapped with a Sigma factor 70 type promoter; accordingly, transcriptional fusions revealed that the umuDpR gene promoter is activated by mitomycin C. The predicted sequence of the UmuDpR protein displays 23 % identity with the Ps. aeruginosa SOS-response LexA repressor. The umuDpR gene caused increased MMC sensitivity when transferred to the Ps. aeruginosa PAO1 strain. As expected, PAO1-derived knockout lexA- mutant PW6037 showed resistance to MMC; however, when the umuDpR gene was transferred to PW6037, MMC resistance level was reduced. These data suggested that UmuDpR represses the expression of SOS genes, as LexA does. To test whether UmuDpR exerts regulatory functions, expression of PAO1 SOS genes was evaluated by reverse transcription quantitative PCR assays in the lexA- mutant with or without the pUC_umuD recombinant plasmid. Expression of lexA, imuA and recA genes increased 3.4-5.3 times in the lexA- mutant, relative to transcription of the corresponding genes in the lexA+ strain, but decreased significantly in the lexA- /umuDpR transformant. These results confirmed that the UmuDpR protein is a repressor of Ps. aeruginosa SOS genes controlled by LexA. Electrophoretic mobility shift assays, however, did not show binding of UmuDpR to 5' regions of SOS genes, suggesting an indirect mechanism of regulation.


Assuntos
Regulação Bacteriana da Expressão Gênica , Plasmídeos , Pseudomonas aeruginosa/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Resposta SOS em Genética , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Perfilação da Expressão Gênica , Dados de Sequência Molecular , Óperon , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
12.
Microb Pathog ; 77: 24-30, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25457796

RESUMO

Vitamin D is an immunomodulator that exerts anti-inflammatory effects. In this work, the effects of cholecalciferol, a vitamin D precursor, on the inflammatory response of bovine mammary epithelial cells (bMECs) during the internalization of Staphylococcus aureus were analyzed. Cholecalciferol and S. aureus inhibited TLR2 mRNA expression, but cholecalciferol differentially modulated the TLR2 membrane abundance. In fact, 50 nM cholecalciferol inhibited the TLR2 membrane abundance in bMECs infected with S. aureus, and this concentration also exerted the highest inhibitory effect on internalization. Cholecalciferol down-regulated the mRNA expression of TNF-α and IL-1ß and up-regulated that of RANTES and IL-10 but did not modify IL-6 and IL-8 expression. S. aureus strongly induced the mRNA expression of TNF-α, RANTES and IL-10 and inhibited IL-8 expression. Interestingly, cholecalciferol pre-treatments inhibited the bacterial-induced expression of TNF-α, IL-1ß, RANTES and IL-10. In conclusion, cholecalciferol differentially regulates the inflammatory response of bMECs during S. aureus internalization and may be an effective innate immunity modulator in mammary gland tissues.


Assuntos
Colecalciferol/metabolismo , Endocitose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Fatores Imunológicos/metabolismo , Staphylococcus aureus/imunologia , Staphylococcus aureus/fisiologia , Animais , Bovinos , Células Cultivadas , Citocinas/biossíntese , Células Epiteliais/imunologia , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Receptor 2 Toll-Like/análise , Receptor 2 Toll-Like/antagonistas & inibidores
13.
Biomed Res Int ; 2014: 538546, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24826382

RESUMO

Staphylococcus aureus is a successful human and animal pathogen. The majority of infections caused by this pathogen are life threatening, primarily because S. aureus has developed multiple evasion strategies, possesses intracellular persistence for long periods, and targets the skin and soft tissues. Therefore, it is very important to understand the mechanisms employed by S. aureus to colonize and proliferate in these cells. The aim of this review is to describe the recent discoveries concerning the host receptors of nonprofessional phagocytes involved in S. aureus internalization. Most of the knowledge related to the interaction of S. aureus with its host cells has been described in professional phagocytic cells such as macrophages. Here, we showed that in nonprofessional phagocytes the α 5 ß 1 integrin host receptor, chaperons, and the scavenger receptor CD36 are the main receptors employed during S. aureus internalization. The characterization and identification of new bacterial effectors and the host cell receptors involved will undoubtedly lead to new discoveries with beneficial purposes.


Assuntos
Fagocitose , Receptores de Superfície Celular/metabolismo , Staphylococcus aureus/fisiologia , Animais , Aderência Bacteriana , Humanos , Ligação Proteica , Staphylococcus aureus/patogenicidade , Fatores de Virulência/metabolismo
14.
Biomed Res Int ; 2013: 927643, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23509807

RESUMO

Bovine mammary epithelial cells (bMECs) are capable of initiating an innate immune response to invading bacteria. Short chain fatty acids can reduce Staphylococcus aureus internalization into bMEC, but it has not been evaluated if octanoic acid (sodium octanoate, NaO), a medium chain fatty acid (MCFA), has similar effects. In this study we determined the effect of NaO on S. aureus internalization into bMEC and on the modulation of innate immune elements. NaO (0.25-2 mM) did not affect S. aureus growth and bMEC viability, but it differentially modulated bacterial internalization into bMEC, which was induced at 0.25-0.5 mM (~60%) but inhibited at 1-2 mM (~40%). Also, bMEC showed a basal expression of all the innate immune genes evaluated, which were induced by S. aureus. NaO induced BNBD4, LAP, and BNBD10 mRNA expression, but BNBD5 and TNF-α were inhibited. Additionally, the pretreatment of bMEC with NaO inhibited the mRNA expression induction generated by bacteria which coincides with the increase in internalization; only TAP and BNDB10 showed an increase in their expression; it coincides with the greatest effect on the reduction of bacterial internalization. In conclusion, NaO exerts a dual effect on S. aureus internalization in bMEC and modulates elements of innate immune response.


Assuntos
Caprilatos/farmacologia , Células Epiteliais/imunologia , Imunidade Inata/efeitos dos fármacos , Glândulas Mamárias Animais/citologia , Staphylococcus aureus/crescimento & desenvolvimento , Animais , Bovinos , Sobrevivência Celular , Células Cultivadas , Células Epiteliais/microbiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Fator de Necrose Tumoral alfa/metabolismo
15.
Vet Microbiol ; 160(1-2): 91-8, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22655972

RESUMO

Vitamin D has immunomodulatory functions regulating the expression of host defense genes. The aim of this study was to determine the effect of cholecalciferol (vitamin D3) on S. aureus internalization into bovine mammary epithelial cells (bMEC) and antimicrobial peptide (AP) mRNA expression. Cholecalciferol (1-200 nM) did not affect S. aureus growth and bMEC viability; but it reduced bacterial internalization into bMEC (15-74%). Also, bMEC showed a basal expression of all AP genes evaluated, which were induced by S. aureus. Cholecalciferol alone or together with bacteria diminished tracheal antimicrobial peptide (TAP) and bovine neutrophil ß-defensin (BNBD) 5 mRNA expression; while alone induced the expression of lingual antimicrobial peptide (LAP), bovine ß-defensin 1 (DEFB1) and bovine psoriasin (S100A7), which was inhibited in the presence of S. aureus. This compound (50 nM) increased BNBD10 mRNA expression coinciding with the greatest reduction in S. aureus internalization. Genes of vitamin D pathway (25-hydroxylase and 1 α-hydroxylase) show basal expression, which was induced by cholecalciferol or bacteria. S. aureus induced vitamin D receptor (VDR) mRNA expression, but not in the presence of cholecalciferol. In conclusion, cholecalciferol can reduce S. aureus internalization and differentially regulates AP expression in bMEC. Thus, vitamin D could be an effective innate immunity modulator in mammary gland, which leads to a better defense against bacterial infection.


Assuntos
Colecalciferol/farmacologia , Glândulas Mamárias Animais/microbiologia , Mastite Bovina/microbiologia , Infecções Estafilocócicas/veterinária , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , beta-Defensinas/biossíntese , Animais , Anti-Infecciosos/metabolismo , Anti-Infecciosos/farmacologia , Bovinos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/metabolismo , Mastite Bovina/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , beta-Defensinas/genética , beta-Defensinas/imunologia
16.
Cell Immunol ; 280(1): 108-12, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23298865

RESUMO

In this work, we explore if Gram-positive bacteria as Staphylococcus aureus or Gram-negative bacteria components as LPS, can induce the expression of seven antimicrobial peptides (AP) in an immortalized bovine umbilical vein endothelial cell line (BUVEC). By qPCR we determined the constitutive expression of all the AP evaluated. The stimulation with S. aureus or LPS induced the expression of lingual antimicrobial peptide (LAP), bovine ß-defensin 1 (DEFB1) and bovine neutrophil ß-defensin 4 (BNBD4). This expression was regulated by the autocrine production of tumor necrosis factor-α (TNF-α), indicating that bovine endothelial cells (EC) can play a more active role during infection.


Assuntos
Células Endoteliais/metabolismo , beta-Defensinas/biossíntese , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Comunicação Autócrina , Bovinos , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/metabolismo , Células Endoteliais/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Lipopolissacarídeos/farmacologia , Mastite Bovina/microbiologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/isolamento & purificação , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Veias Umbilicais , beta-Defensinas/genética
17.
Vet Microbiol ; 155(2-4): 324-31, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-21930351

RESUMO

Short chain fatty acids (SCFAs) are critical nutrients for ruminants and are mainly obtained from bacterial fermentation of carbohydrates. In addition to their nutrimental function, SCFAs have antimicrobial and anti-inflammatory properties, as well as immunomodulatory roles. It has been reported that sodium butyrate reduces Staphylococcus aureus internalization into bovine mammary epithelial cells (bMEC) and modulates antimicrobial peptide mRNA expression. Nevertheless, it has not been evaluated if sodium propionate (NaP) and sodium hexanoate (NaH) have similar actions. Since they are present in milk, the aim of this study was to determinate the effect of both SCFAs on S. aureus internalization into bMEC and to evaluate their effects on modulation of innate immunity elements. Our data showed that both SCFAs (0.25-5mM) did not affect S. aureus growth and bMEC viability. By gentamicin protection assay (MOI 30:1) we showed that NaP and NaH reduced bacterial internalization into bMEC, which ranged 27-55% and 39-65%, respectively, in relation to non treated controls. Also, both SCFAs up-regulate tracheal antimicrobial peptide (TAP) mRNA expression; however, bovine neutrophil ß-defensin 5 (BNBD5) mRNA expression was not modified or was down-regulated. In addition, TAP and BNBD5 expression was up-regulated by S. aureus. Finally, the decrease in bacterial internalization under SCFA treatments is not related to nitric oxide production. In conclusion, NaP and NaH decrease S. aureus internalization into bMEC and modulate TAP gene expression, which may be related to the reduction in bacterial internalization.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Caproatos/farmacologia , Células Epiteliais/microbiologia , Glândulas Mamárias Animais/microbiologia , Propionatos/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Bovinos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Óxido Nítrico/biossíntese , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento , beta-Defensinas/biossíntese , beta-Defensinas/genética
18.
Vet Microbiol ; 138(1-2): 179-83, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-19359107

RESUMO

Antimicrobial therapy is a useful tool to control bovine mastitis caused by Staphylococcus aureus, as consequence an increase in staphylococci resistant cases has been registered. Alternative strategies are desirable and bacteriocins represent attractive control agents to prevent bovine mastitis. The aim of this work was to evaluate the activity of five bacteriocins synthesized by Bacillus thuringiensis against S. aureus isolates associated to bovine mastitis. Fifty S. aureus isolates were recovered from milk composite samples of 26 Holstein lactating cows from one herd during September 2007 to February 2008 in México and susceptibility of those isolates to 12 antibiotics and 5 bacteriocins from B. thuringiensis was evaluated. S. aureus isolates were mainly resistant to penicillin (92%), dicloxacillin (86%), ampicillin (74%) and erythromycin (74%); whereas susceptibility to gentamicin, trimethoprim and tetracycline was detected at, respectively, 92%, 88%, and 72%. All S. aureus isolates showed susceptibility to the five bacteriocins synthesized by B. thuringiensis, mainly to morricin 269 and kurstacin 287 followed by kenyacin 404, entomocin 420 and tolworthcin 524. Our results showed that S. aureus isolates had differences in the antimicrobial resistance patterns and were susceptible to bacteriocins produced by B. thuringiensis, which could be useful as an alternative method to control bovine mastitis.


Assuntos
Bacillus thuringiensis/fisiologia , Bacteriocinas/farmacologia , Mastite Bovina/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Bacteriocinas/biossíntese , Bacteriocinas/isolamento & purificação , Bovinos , Feminino , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/veterinária , Staphylococcus aureus/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA