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.
Front Cell Infect Microbiol ; 13: 1285193, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38094743

RESUMO

Bacterial infections and the concurrent inflammation have been associated with increased long-term cardiovascular (CV) risk. In patients receiving peritoneal dialysis (PD), bacterial peritonitis is a common occurrence, and each episode further increases late CV mortality risk. However, the underlying mechanism(s) remains to be elucidated before safe and efficient anti-inflammatory interventions can be developed. Damage-Associated Molecular Patterns (DAMPs) have been shown to contribute to the acute inflammatory response to infections, but a potential role for DAMPs in mediating long-term vascular inflammation and CV risk following infection resolution in PD, has not been investigated. We found that bacterial peritonitis in mice that resolved within 24h led to CV disease-promoting systemic and vascular immune-mediated inflammatory responses that were maintained up to 28 days. These included higher blood proportions of inflammatory leukocytes displaying increased adhesion molecule expression, higher plasma cytokines levels, and increased aortic inflammatory and atherosclerosis-associated gene expression. These effects were also observed in infected nephropathic mice and amplified in mice routinely exposed to PD fluids. A peritonitis episode resulted in elevated plasma levels of the DAMP Calprotectin, both in PD patients and mice, here the increase was maintained up to 28 days. In vitro, the ability of culture supernatants from infected cells to promote key inflammatory and atherosclerosis-associated cellular responses, such as monocyte chemotaxis, and foam cell formation, was Calprotectin-dependent. In vivo, Calprotectin blockade robustly inhibited the short and long-term peripheral and vascular consequences of peritonitis, thereby demonstrating that targeting of the DAMP Calprotectin is a promising therapeutic strategy to reduce the long-lasting vascular inflammatory aftermath of an infection, notably PD-associated peritonitis, ultimately lowering CV risk.


Assuntos
Aterosclerose , Infecções Bacterianas , Diálise Peritoneal , Peritonite , Humanos , Camundongos , Animais , Diálise Peritoneal/efeitos adversos , Diálise Peritoneal/métodos , Inflamação/complicações , Infecções Bacterianas/complicações , Aterosclerose/complicações
2.
Front Immunol ; 14: 1240679, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37849759

RESUMO

Chronic Kidney Disease (CKD) is associated with markedly increased cardiovascular (CV) morbidity and mortality. Chronic inflammation, a hallmark of both CKD and CV diseases (CVD), is believed to drive this association. Pro-inflammatory endogenous TLR agonists, Damage-Associated Molecular Patterns (DAMPs), have been found elevated in CKD patients' plasma and suggested to promote CVD, however, confirmation of their involvement, the underlying mechanism(s), the extent to which individual DAMPs contribute to vascular pathology in CKD and the evaluation of potential therapeutic strategies, have remained largely undescribed. A multi-TLR inhibitor, soluble TLR2, abrogated chronic vascular inflammatory responses and the increased aortic atherosclerosis-associated gene expression observed in nephropathic mice, without compromising infection clearance. Mechanistically, we confirmed elevation of 4 TLR DAMPs in CKD patients' plasma, namely Hsp70, Hyaluronic acid, HMGB-1 and Calprotectin, which displayed different abilities to promote key cellular responses associated with vascular inflammation and progression of atherosclerosis in a TLR-dependent manner. These included loss of trans-endothelial resistance, enhanced monocyte migration, increased cytokine production, and foam cell formation by macrophages, the latter via cholesterol efflux inhibition. Calprotectin and Hsp70 most consistently affected these functions. Calprotectin was further elevated in CVD-diagnosed CKD patients and strongly correlated with the predictor of CV events CRP. In nephropathic mice, Calprotectin blockade robustly reduced vascular chronic inflammatory responses and pro-atherosclerotic gene expression in the blood and aorta. Taken together, these findings demonstrated the critical extent to which the DAMP-TLR pathway contributes to vascular inflammatory and atherogenic responses in CKD, revealed the mechanistic contribution of specific DAMPs and described two alternatives therapeutic approaches to reduce chronic vascular inflammation and lower CV pathology in CKD.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Insuficiência Renal Crônica , Humanos , Animais , Camundongos , Insuficiência Renal Crônica/patologia , Alarminas , Aterosclerose/tratamento farmacológico , Inflamação/metabolismo , Doenças Cardiovasculares/complicações , Complexo Antígeno L1 Leucocitário
3.
Front Physiol ; 9: 1692, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30538643

RESUMO

Peritoneal dialysis (PD) is an essential daily life-saving treatment for end-stage renal failure. PD therapy is limited by peritoneal inflammation, which leads to peritoneal membrane failure as a result of progressive fibrosis. Peritoneal infections, with the concomitant acute inflammatory response and membrane fibrosis development, worsen PD patient outcomes. Patients who remain infection-free, however, also show evidence of inflammation-induced membrane damage and fibrosis, leading to PD cessation. In this case, uraemia, prolonged exposure to bio-incompatible PD solutions and surgical catheter insertion have been reported to induce sterile peritoneal inflammation and fibrosis as a result of cellular stress or tissue injury. Attempts to reduce inflammation (either infection-induced or sterile) and, thus, minimize fibrosis development in PD have been hampered because the immunological mechanisms underlying this PD-associated pathology remain to be fully defined. Toll-like receptors (TLRs) are central to mediating inflammatory responses by recognizing a wide variety of microorganisms and endogenous components released following cellular stress or generated as a consequence of extracellular matrix degradation during tissue injury. Given the close link between inflammation and fibrosis, recent investigations have evaluated the role that TLRs play in infection-induced and sterile peritoneal fibrosis development during PD. Here, we review the findings and discuss the potential of reducing peritoneal TLR activity by using a TLR inhibitor, soluble TLR2, as a therapeutic strategy to prevent PD-associated peritoneal fibrosis.

4.
Kidney Int ; 94(2): 346-362, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29861057

RESUMO

Peritoneal membrane failure due to fibrosis limits the use of peritoneal dialysis (PD). Peritoneal fibrosis may potentially be induced by sterile inflammation caused by ongoing cellular stress due to prolonged exposure to PD solutions (PDS). Effective therapies to prevent this process remain to be developed. Toll-like receptors (TLRs) mediate sterile inflammation by recognizing damage-associated molecular patterns (DAMPs) released by cellular stress. We evaluated the involvement of TLRs and DAMPs in PDS-induced fibrosis models and the therapeutic potential of TLR-DAMP targeting for preventing fibrosis. A range of PDS elicited pro-inflammatory and fibrotic responses from PD patient peritoneal leukocytes, mesothelial cells and mouse peritoneal leukocytes. TLR2/4 blockade of human peritoneal cells or TLR2/4 knockouts inhibited these effects. PDS did not induce rapid ERK phosphorylation or IκB-α degradation, suggesting that they do not contain components capable of direct TLR activation. However, PDS increased the release of Hsp70 and hyaluronan, both TLR2/4 DAMP ligands, by human and mouse peritoneal cells, and their blockade decreased PDS-driven inflammation. Soluble TLR2, a TLR inhibitor, reduced PDS-induced pro-inflammatory and fibrotic cytokine release ex vivo. Daily catheter infusion of PDS in mice caused peritoneal fibrosis, but co-administration of soluble TLR2 prevented fibrosis, suppressed pro-fibrotic gene expression and pro-inflammatory cytokine production, reduced leukocyte/neutrophil recruitment, recovered Treg cell levels and increased the Treg:Th17 ratio. Thus, TLR2/4, Hsp70 and hyaluronan showed major roles in PDS-induced peritoneal inflammation and fibrosis. The study demonstrates the therapeutic potential of a TLR-DAMP targeting strategy to prevent PDS-induced fibrosis.


Assuntos
Soluções para Diálise/toxicidade , Inflamação/prevenção & controle , Fibrose Peritoneal/prevenção & controle , Receptor 2 Toll-Like/administração & dosagem , Receptores Toll-Like/antagonistas & inibidores , Alarminas/antagonistas & inibidores , Alarminas/imunologia , Alarminas/metabolismo , Animais , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Voluntários Saudáveis , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/patologia , Falência Renal Crônica/terapia , Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Diálise Peritoneal/efeitos adversos , Diálise Peritoneal/métodos , Fibrose Peritoneal/induzido quimicamente , Fibrose Peritoneal/imunologia , Fibrose Peritoneal/patologia , Peritônio/citologia , Peritônio/patologia , Cultura Primária de Células , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
5.
Intensive Care Med Exp ; 5(1): 2, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28092080

RESUMO

Soluble TLR2 levels are elevated in infective and inflammatory conditions, but its diagnostic value with sepsis-induced multi-organ failure has not been evaluated. 37 patients with a diagnosis of severe sepsis/septic shock (sepsis) and 27 patients with organ failure without infection (SIRS) were studied. Median (IQR) plasma sTLR2 levels were 2.7 ng/ml (1.4-6.1) in sepsis and 0.6 ng/ml (0.4-0.9) in SIRS p < 0.001. sTLR2 showed good diagnostic value for sepsis at cut-off of 1.0 ng/ml, AUC:0.959. We report the ability of sTLR2 levels to discriminate between sepsis and SIRS within 12 h of ICU admission in patients with multi-organ failure.

6.
J Am Soc Nephrol ; 28(2): 461-478, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27432741

RESUMO

Peritoneal dialysis (PD) remains limited by dialysis failure due to peritoneal membrane fibrosis driven by inflammation caused by infections or sterile cellular stress. Given the fundamental role of Toll-like receptors (TLRs) and complement in inflammation, we assessed the potential of peritoneal TLR2, TLR4 and C5a receptors, C5aR and C5L2, as therapeutic targets in PD-associated fibrosis. We detected TLR2-, TLR4-, and C5aR-mediated proinflammatory and fibrotic responses to bacteria that were consistent with the expression of these receptors in peritoneal macrophages (TLR2/4, C5aR) and mesothelial cells (TLR2, C5aR). Experiments in knockout mice revealed a major role for TLR2, a lesser role for TLR4, a supplementary role for C5aR, and no apparent activity of C5L2 in infection-induced peritoneal fibrosis. Similarly, antibody blockade of TLR2, TLR4, or C5aR differentially inhibited bacteria-induced profibrotic and inflammatory mediator production by peritoneal leukocytes isolated from the peritoneal dialysis effluent (PDE) of noninfected uremic patients. Additionally, antibodies against TLR2, TLR4, or the coreceptor CD14 reduced the profibrotic responses of uremic leukocytes to endogenous components present in the PDE of noninfected patients. Enhancing TLR2-mediated inflammation increased fibrosis in vivo Furthermore, soluble TLR2 (sTLR2), a negative modulator of TLRs that we detected in PDE, inhibited PDE-induced, TLR2- or TLR4-mediated profibrotic responses. Notably, sTLR2 treatment markedly reduced Gram-positive and -negative bacteria-induced fibrosis in vivo, inhibiting proinflammatory and fibrotic genes without affecting infection clearance. These findings reveal the influence of peritoneal TLR2 and TLR4 on PD-associated fibrosis and describe a therapeutic strategy against fibrosis.


Assuntos
Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/tratamento farmacológico , Fibrose Peritoneal/etiologia , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Humanos , Camundongos , Camundongos Knockout
7.
Curr Pharm Biotechnol ; 17(5): 414-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26778454

RESUMO

The Toll-like family of immune receptors (TLRs) are critical for an efficient immune response to a variety of microorganisms and other antigens that may cause pathology. Modulating immune responses by targeting TLRs therefore has substantial therapeutic potential, and a number of TLR-based therapeutic strategies have been developed. Minimizing the adverse effects that may result from the therapeutic manipulation of these signalling receptors nevertheless remains a major challenge. Efficient responses via TLRs require the activity of the co-receptor CD14, which enhances TLR responses. In an attempt to boost the immune response for therapeutic purposes, we have sought to target CD14 to achieve TLR modulation. Here we discuss the design, activity and therapeutic development options of TLR-derived peptides that interact with CD14 and enhance its co-receptor activity, thus amplifying TLR-mediated responses. This strategy represents a promising alternative to current TLR-based therapies, as it has the potential to amplify responses to different pathogens mediated by different TLRs by targeting the common TLR co-receptor, CD14.


Assuntos
Antígenos de Histocompatibilidade Classe II/imunologia , Receptores de Lipopolissacarídeos/imunologia , Animais , Humanos , Peptídeos/imunologia , Transdução de Sinais , Receptores Toll-Like/imunologia , Vacinas/imunologia
8.
Sci Transl Med ; 5(185): 185ra64, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23677593

RESUMO

Dysregulation of Toll-like receptor (TLR) responses to pathogens can lead to pathological inflammation or to immune hyporesponsiveness and susceptibility to infections, and may affect adaptive immune responses. TLRs are therefore attractive therapeutic targets. We assessed the potential of the TLR co-receptor CD14 as a target for therapeutics by investigating the magnitude of its influence on TLR responses. We studied the interaction of CD14 with TLR2 by conducting peptide screening and site-directed mutagenesis analysis and found TLR2 leucine-rich repeats 5, 9, 15, and 20 involved in interaction with CD14. Peptides representing these regions interacted with CD14 and enhanced TLR2- and TLR4-mediated proinflammatory responses to bacterial pathogens in vitro. Notably, the peptides' immune boosting capacity helped to rescue proinflammatory responses of immunosuppressed sepsis patients ex vivo. In vivo, peptide treatment increased phagocyte recruitment and accelerated bacterial clearance in murine models of Gram-negative and Gram-positive bacterial peritonitis. Up-modulating CD14's co-receptor activity with TLR2-derived peptides also enhanced antigen-induced dendritic cell (DC) maturation and interleukin-2 production and, most notably, differentially affected DC cytokine profile upon antigen stimulation, promoting a T helper 1-skewed adaptive immune response. Biochemical, cell imaging, and molecular docking studies showed that peptide binding to CD14 accelerates microbial ligand transfer from CD14 to TLR2, resulting in increased and sustained ligand occupancy of TLR2 and receptor clustering for signaling. These findings reveal the influence that CD14 exerts on TLR activities and describe a potential therapeutic strategy to amplify responses to different pathogens mediated by different TLRs by targeting the common TLR co-receptor, CD14.


Assuntos
Bactérias/imunologia , Imunidade/imunologia , Receptores de Lipopolissacarídeos/imunologia , Peptídeos/imunologia , Receptor 2 Toll-Like/química , Sequência de Aminoácidos , Animais , Bactérias/efeitos dos fármacos , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Células HEK293 , Humanos , Imunidade/efeitos dos fármacos , Terapia de Imunossupressão , Inflamação/complicações , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Proteínas de Repetições Ricas em Leucina , Ligantes , Lipopolissacarídeos/farmacologia , Lipoproteínas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Viabilidade Microbiana/efeitos dos fármacos , Dados de Sequência Molecular , Peptídeos/química , Peritonite/imunologia , Peritonite/microbiologia , Peritonite/patologia , Fagócitos/citologia , Fagócitos/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas/imunologia , Sepse/complicações , Sepse/imunologia , Sepse/microbiologia , Sepse/patologia
9.
Nephrol Dial Transplant ; 26(12): 4079-90, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21633096

RESUMO

BACKGROUND: Bacterial infection remains a major cause of morbidity and mortality in peritoneal dialysis (PD) patients worldwide. Previous studies have identified a key role for mesothelial cells, lining the peritoneal cavity, in coordinating inflammation and host defense. Toll-like receptor (TLR) involvement in early activation events within the mesothelium, however, remains poorly defined. To investigate the initiation of bacterial peritonitis, we characterized TLR activation by bacterial ligands in human peritoneal mesothelial cells (HPMC). METHODS: Primary HPMC were isolated from omental biopsies and TLR expression detected by real-time polymerase chain reaction (PCR), reverse transcription (RT)-PCR and flow cytometry. The responsiveness of HPMC to specific bacterial TLR agonists was determined using chemokine production as a biological readout. The requirement for CD14 in HPMC responses to a clinically relevant Staphylococcus epidermidis cell-free supernatant (SES) was investigated using soluble CD14 or anti-CD14-blocking antibodies. RESULTS: Real-time PCR detected TLR1-6 messenger RNA expression in HPMC and responses to TLR2/1 and TLR2/6 ligands and SES. No cell surface TLR4 expression or responses to lipopolysaccharide were detectable in HPMC, but they did respond to flagellin, a TLR5 ligand. SES-mediated responses were dependent on TLR2 but did not require CD14 in HPMC for optimal efficiency, unlike peripheral blood mononuclear cells. HPMC expression of TLR2 was also modulated by TLR2 ligands and inflammatory cytokines. CONCLUSIONS: These data suggest that mesothelial cell activation by TLR2/1, TLR2/6 and TLR5 contributes to bacterial recognition influencing the course of the infective process and has implications for improving treatment of infection in PD patients.


Assuntos
Proteínas de Bactérias/fisiologia , Células Epiteliais/fisiologia , Peritônio/citologia , Receptores Toll-Like/fisiologia , Células Cultivadas , Humanos , Ligantes
10.
Eur J Immunol ; 41(9): 2741-52, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21630250

RESUMO

TLR and complement activation ensures efficient clearance of infection. Previous studies documented synergism between TLRs and the receptor for the pro-inflammatory complement peptide C5a (C5aR/CD88), and regulation of TLR-induced pro-inflammatory responses by C5aR, suggesting crosstalk between TLRs and C5aR. However, it is unclear whether and how TLRs modulate C5a-induced pro-inflammatory responses. We demonstrate a marked positive modulatory effect of TLR activation on cell sensitivity to C5a in vitro and ex vivo and identify an underlying mechanistic target. Pre-exposure of PBMCs and whole blood to diverse TLR ligands or bacteria enhanced C5a-induced pro-inflammatory responses. This effect was not observed in TLR4 signalling-deficient mice. TLR-induced hypersensitivity to C5a did not result from C5aR upregulation or modulation of C5a-induced Ca(2+) mobilization. Rather, TLRs targeted another C5a receptor, C5L2 (acting as a negative modulator of C5aR), by reducing C5L2 activity. TLR-induced hypersensitivity to C5a was mimicked by blocking C5L2 and was not observed in C5L2KO mice. Furthermore, TLR activation inhibited C5L2 expression upon C5a stimulation. These findings identify a novel pathway of crosstalk within the innate immune system that amplifies innate host defense at the TLR-complement interface. Unravelling the mutually regulated activities of TLRs and complement may reveal new therapeutic avenues to control inflammation.


Assuntos
Complemento C5a/metabolismo , Leucócitos Mononucleares/metabolismo , Receptores de Quimiocinas/metabolismo , Receptores de Complemento/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Sinalização do Cálcio/imunologia , Células Cultivadas , Complemento C5a/imunologia , Retroalimentação Fisiológica , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Interleucina-8/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Knockout , Receptor Cross-Talk/imunologia , Receptor da Anafilatoxina C5a , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/imunologia , Receptores de Complemento/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
11.
J Immunol ; 183(1): 506-17, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19542461

RESUMO

TLR overactivation may lead to end organ damage and serious acute and chronic inflammatory conditions. TLR responses must therefore be tightly regulated to control disease outcomes. We show in this study the ability of the soluble form of TLR2 (sTLR2) to regulate proinflammatory responses, and demonstrate the mechanisms underlying sTLR2 regulatory capacity. Cells overexpressing sTLR2, or stimulated in the presence of the sTLR2 protein, are hyporesponsive to TLR2 ligands. Regulation was TLR2 specific, and affected NF-kappaB activation, phagocytosis, and superoxide production. Natural sTLR2-depleted serum rendered leukocytes hypersensitive to TLR2-mediated stimulation. Mice administered sTLR2 together with Gram-positive bacteria-derived components showed lower peritoneal levels of the neutrophil (PMN) chemoattractant, keratinocyte-derived chemokine; lower PMN numbers; and a reduction in late apoptotic PMN. Mononuclear cell recruitment remained unaffected, and endogenous peritoneal sTLR2 levels increased. Notably, the capacity of sTLR2 to modulate acute inflammatory parameters did not compromise the ability of mice to clear live Gram-positive bacteria-induced infection. Mechanistically, sTLR2 interfered with TLR2 mobilization to lipid rafts for signaling, acted as a decoy microbial receptor, and disrupted the interaction of TLR2 with its coreceptor, CD14, by associating with CD14. These findings establish sTLR2 as a regulator of TLR2-mediated inflammatory responses, capable of blunting immune responses without abrogating microbial recognition and may inform the design of novel therapeutics against acute and chronic inflammatory conditions.


Assuntos
Mediadores da Inflamação/fisiologia , Peritonite/imunologia , Peritonite/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Receptor 2 Toll-Like/fisiologia , Doença Aguda , Sequência de Aminoácidos , Animais , Aderência Bacteriana/genética , Aderência Bacteriana/imunologia , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Humanos , Imunidade Inata/genética , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/sangue , Ligantes , Receptores de Lipopolissacarídeos/metabolismo , Receptores de Lipopolissacarídeos/fisiologia , Microdomínios da Membrana/genética , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/microbiologia , Microdomínios da Membrana/patologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Peritonite/patologia , Peritonite/prevenção & controle , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Infecções Estafilocócicas/patologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus epidermidis/imunologia , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/sangue , Receptor 2 Toll-Like/genética
12.
J Immunol ; 176(6): 3742-52, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16517743

RESUMO

The mechanisms controlling innate microbial recognition in the neonatal gut are still to be fully understood. We have sought specific regulatory mechanisms operating in human breast milk relating to TLR-mediated microbial recognition. In this study, we report a specific and differential modulatory effect of early samples (days 1-5) of breast milk on ligand-induced cell stimulation via TLRs. Although a negative modulation was exerted on TLR2 and TLR3-mediated responses, those via TLR4 and TLR5 were enhanced. This effect was observed in human adult and fetal intestinal epithelial cell lines, monocytes, dendritic cells, and PBMC as well as neonatal blood. In the latter case, milk compensated for the low capacity of neonatal plasma to support responses to LPS. Cell stimulation via the IL-1R or TNFR was not modulated by milk. This, together with the differential effect on TLR activation, suggested that the primary effect of milk is exerted upstream of signaling proximal to TLR ligand recognition. The analysis of TLR4-mediated gene expression, used as a model system, showed that milk modulated TLR-related genes differently, including those coding for signal intermediates and regulators. A proteinaceous milk component of > or =80 kDa was found to be responsible for the effect on TLR4. Notably, infant milk formulations did not reproduce the modulatory activity of breast milk. Together, these findings reveal an unrecognized function of human milk, namely, its capacity to influence neonatal microbial recognition by modulating TLR-mediated responses specifically and differentially. This in turn suggests the existence of novel mechanisms regulating TLR activation.


Assuntos
Imunidade Inata/imunologia , Leite Humano/imunologia , Receptores Toll-Like/imunologia , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Hemócitos/efeitos dos fármacos , Hemócitos/imunologia , Hemócitos/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Fórmulas Infantis/farmacologia , Recém-Nascido , Lipopolissacarídeos/farmacologia , Receptores de Interleucina-1/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Toll-Like/metabolismo
13.
J Immunol ; 171(12): 6680-9, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14662871

RESUMO

Dysregulation of the initial, innate immune response to bacterial infection may lead to septic shock and death. Toll-like receptors (TLRs) play a crucial role in this innate immune response, and yet the regulatory mechanisms controlling microbial-induced TLR triggering are still to be fully understood. We have therefore sought specific regulatory mechanisms that may modulate TLR signaling. In this study, we tested for the possible existence of a functionally active soluble form of TLR2. We demonstrated the existence of natural soluble forms of TLR2 (sTLR2), which we show to be capable of modulating cell activation. We found that blood monocytes released sTLR2 constitutively and that the kinetics of sTLR2 release increased upon cell activation. Analysis of cells expressing the human TLR2 cDNA or its c-myc-tagged version indicated that sTLR2 resulted from the posttranslational modification of the TLR2 protein in an intracellular compartment. Moreover, an intracellular pool of sTLR2 is maintained. sTLR2 was found naturally expressed in breast milk and plasma. Milk sTLR2 levels mirrored those of the TLR coreceptor soluble CD14. Depletion of sTLR2 from serum resulted in an increased cellular response to bacterial lipopeptide. Notably, serum sTLR2 was lower in tuberculosis patients. Coimmunoprecipitation experiments and computational molecular docking studies showed an interaction between sTLR2 and soluble CD14 in plasma and milk. These findings suggest the existence of a novel and specific innate immune mechanism regulating microbial-induced TLR triggering, and may lead to new therapeutics for the prevention and/or treatment of severe infectious diseases.


Assuntos
Cisteína/análogos & derivados , Glicoproteínas de Membrana/sangue , Glicoproteínas de Membrana/fisiologia , Leite Humano/imunologia , Leite Humano/metabolismo , Receptores de Superfície Celular/sangue , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/imunologia , Sequência de Aminoácidos , Linhagem Celular , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Cisteína/antagonistas & inibidores , Cisteína/farmacologia , DNA Complementar/biossíntese , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Lactação , Receptores de Lipopolissacarídeos/sangue , Receptores de Lipopolissacarídeos/metabolismo , Lipoproteínas/antagonistas & inibidores , Lipoproteínas/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Leite Humano/química , Dados de Sequência Molecular , Monensin/farmacologia , Monócitos/imunologia , Monócitos/metabolismo , Peptídeos/sangue , Peptídeos/isolamento & purificação , Peptídeos/metabolismo , Peptídeos/fisiologia , Testes de Precipitina , Isoformas de Proteínas/sangue , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/imunologia , Transporte Proteico/imunologia , Receptores de Superfície Celular/genética , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Receptor 2 Toll-Like , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...