Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
PLoS Pathog ; 13(8): e1006551, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28806745

RESUMEN

Pathogenic mycobacteria reside in macrophages where they avoid lysosomal targeting and degradation through poorly understood mechanisms proposed to involve arrest of phagosomal maturation at an early endosomal stage. A clear understanding of how this relates to host defenses elicited from various intracellular compartments is also missing and can only be studied using techniques allowing single cell and subcellular analyses. Using confocal imaging of human primary macrophages infected with Mycobacterium avium (Mav) we show evidence that Mav phagosomes are not arrested at an early endosomal stage, but mature to a (LAMP1+/LAMP2+/CD63+) late endosomal/phagolysosomal stage where inflammatory signaling and Mav growth restriction is initiated through a mechanism involving Toll-like receptors (TLR) 7 and 8, the adaptor MyD88 and transcription factors NF-κB and IRF-1. Furthermore, a fraction of the mycobacteria re-establish in a less hostile compartment (LAMP1-/LAMP2-/CD63-) where they not only evade destruction, but also recognition by TLRs, growth restriction and inflammatory host responses that could be detrimental for intracellular survival and establishment of chronic infections.


Asunto(s)
Macrófagos/microbiología , Infecciones por Mycobacterium/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Receptor Toll-Like 7/inmunología , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Lisosomas/inmunología , Macrófagos/inmunología , Microscopía Confocal , Mycobacterium avium , Fagosomas/inmunología , Reacción en Cadena de la Polimerasa
2.
PLoS Pathog ; 13(11): e1006712, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29112990

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1006551.].

3.
Cytokine ; 119: 188-196, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30954016

RESUMEN

INTRODUCTION: Human pregnancy is a state of elevated maternal systemic inflammation, and pregnancy complications are often associated with a dysfunctional immune response. The network of cytokines reflects this complex immune activity, and broad serum cytokine profiling provides a new tool to understand the changes in immune status during pregnancy. OBJECTIVE: This study aimed to determine how maternal serum cytokine patterns change during the first half of pregnancy. METHODS: Maternal peripheral serum samples collected at a mean gestation of 10, 13, 18 and 24 weeks were included from a prospective clinical study of healthy women (n = 110) in first half of normal pregnancy. The serum samples were analysed for 27 different cytokines using multiplex magnetic bead-based immunoassays, and high sensitivity C-reactive protein (CRP) was analysed by ELISA. Serum cytokine and CRP patterns were explored with linear mixed effects models (LMM) and multilevel partial least squares discriminant analysis (PLS-DA). RESULTS: Serum cytokine profiling provided partial overview of the maternal immune status and corresponding reference values for serum cytokine levels during the first half of pregnancy. Several cytokines decreased in concentration from first to second trimester. Cytokine pattern analysis revealed that chemokines provided the most sensitive measurement of variation with gestational age in normal pregnancies. The nine inflammatory cytokines showed the highest intra-group correlation during pregnancy, while CRP levels did not correlate with changes in the inflammatory cytokines. CONCLUSION: Chemokines showed the greatest gestational variation and inflammatory cytokines showed a strong intra-group correlation during the first half of pregnancy.


Asunto(s)
Citocinas/sangre , Adulto , Proteína C-Reactiva/metabolismo , Quimiocinas/sangre , Femenino , Edad Gestacional , Humanos , Inflamación/sangre , Embarazo , Complicaciones del Embarazo/sangre , Estudios Prospectivos , Valores de Referencia
4.
J Immunol ; 199(8): 2910-2920, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28855312

RESUMEN

Cholesterol crystals (CC) are abundant in atherosclerotic plaques and promote inflammatory responses via the complement system and inflammasome activation. Cyclic oligosaccharide 2-hydroxypropyl-ß-cyclodextrin (BCD) is a compound that solubilizes lipophilic substances. Recently we have shown that BCD has an anti-inflammatory effect on CC via suppression of the inflammasome and liver X receptor activation. The putative effects of BCD on CC-induced complement activation remain unknown. In this study, we found that BCD bound to CC and reduced deposition of Igs, pattern recognition molecules, and complement factors on CC in human plasma. Furthermore, BCD decreased complement activation as measured by terminal complement complex and lowered the expression of complement receptors on monocytes in whole blood in response to CC exposure. In line with this, BCD also reduced reactive oxygen species formation caused by CC in whole blood. Furthermore, BCD attenuated the CC-induced proinflammatory cytokine responses (e.g., IL-1α, MIP-1α, TNF, IL-6, and IL-8) as well as regulated a range of CC-induced genes in human PBMC. BCD also regulated complement-related genes in human carotid plaques treated ex vivo. Formation of terminal complement complex on other complement-activating structures such as monosodium urate crystals and zymosan was not affected by BCD. These data demonstrate that BCD inhibits CC-induced inflammatory responses, which may be explained by BCD-mediated attenuation of complement activation. Thus, these findings support the potential for using BCD in treatment of atherosclerosis.


Asunto(s)
Arterias Carótidas/fisiología , Colesterol/metabolismo , Ciclodextrinas/metabolismo , Inflamación/inmunología , Leucocitos Mononucleares/fisiología , Monocitos/fisiología , Placa Aterosclerótica/inmunología , Células Cultivadas , Colesterol/inmunología , Activación de Complemento , Proteínas del Sistema Complemento/biosíntesis , Ciclodextrinas/química , Citocinas/metabolismo , Humanos , Inmunomodulación , Inflamación/inducido químicamente , Mediadores de Inflamación/metabolismo , Placa Aterosclerótica/terapia , Especies Reactivas de Oxígeno/metabolismo
5.
J Immunol ; 195(3): 1100-11, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26085680

RESUMEN

Staphylococcus aureus may cause serious infections and is one of the most lethal and common causes of sepsis. TLR2 has been described as the main pattern recognition receptor that senses S. aureus and elicits production of proinflammatory cytokines via MyD88 -: NF-κB signaling. S. aureus can also induce the production of IFN-ß, a cytokine that requires IFN regulatory factors (IRFs) for its transcription, but the signaling mechanism for IFN-ß induction by S. aureus are unclear. Surprisingly, we demonstrate that activation of TLR2 by lipoproteins does not contribute to IFN-ß production but instead can suppress the induction of IFN-ß in human primary monocytes and monocyte-derived macrophages. The production of IFN-ß was induced by TLR8-mediated sensing of S. aureus RNA, which triggered IRF5 nuclear accumulation, and this could be antagonized by concomitant TLR2 signaling. The TLR8-mediated activation of IRF5 was dependent on TAK1 and IκB kinase (IKK)ß, which thus reveals a physiological role of the recently described IRF5-activating function of IKKß. TLR8 -: IRF5 signaling was necessary for induction of IFN-ß and IL-12 by S. aureus, and it also contributed to the induction of TNF. In conclusion, our study demonstrates a physiological role of TLR8 in the sensing of entire S. aureus in human primary phagocytes, including the induction of IFN-ß and IL-12 production via a TAK1 -: IKKß -: IRF5 pathway that can be inhibited by TLR2 signaling.


Asunto(s)
Factores Reguladores del Interferón/inmunología , Interferón beta/biosíntesis , Interleucina-12/biosíntesis , ARN Bacteriano/inmunología , Staphylococcus aureus/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 8/inmunología , Activación Enzimática/inmunología , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Factores Reguladores del Interferón/genética , Interferón beta/inmunología , Interleucina-12/inmunología , Quinasas Quinasa Quinasa PAM/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/genética , Monocitos/inmunología , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , ARN Bacteriano/genética , ARN Interferente Pequeño/genética , Transducción de Señal/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 8/genética , Factor de Necrosis Tumoral alfa/biosíntesis
6.
J Immunol ; 195(1): 257-64, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26026058

RESUMEN

Chronic inflammation of the arterial wall is a key element in the development of atherosclerosis, and cholesterol crystals (CC) that accumulate in plaques are associated with initiation and progression of the disease. We recently revealed a link between the complement system and CC-induced inflammasome caspase-1 activation, showing that the complement system is a key trigger in CC-induced inflammation. HDL exhibits cardioprotective and anti-inflammatory properties thought to explain its inverse correlation to cardiovascular risk. In this study, we sought to determine the effect of reconstituted HDL (rHDL) on CC-induced inflammation in a human whole blood model. rHDL bound to CC and inhibited the CC-induced complement activation as measured by soluble terminal C5b-9 formation and C3c deposition on the CC surface. rHDL attenuated the amount of CC-induced complement receptor 3 (CD11b/CD18) expression on monocytes and granulocytes, as well as reactive oxygen species generation. Moreover, addition of CC to whole blood resulted in release of proinflammatory cytokines that were inhibited by rHDL. Our results support and extend the notion that CC are potent triggers of inflammation, and that rHDL may have a beneficial role in controlling the CC-induced inflammatory responses by inhibiting complement deposition on the crystals.


Asunto(s)
Colesterol/efectos adversos , Activación de Complemento/efectos de los fármacos , Lipoproteínas HDL/farmacología , Células Sanguíneas/citología , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/inmunología , Antígeno CD11b/inmunología , Antígenos CD18/inmunología , Complemento C3c/antagonistas & inhibidores , Complemento C3c/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Cristalización , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Cultivo Primario de Células , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/inmunología
7.
J Infect Dis ; 214(1): 140-50, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26977050

RESUMEN

BACKGROUND: Single inhibition of the Toll-like receptor 4 (TLR4)-MD2 complex failed in treatment of sepsis. CD14 is a coreceptor for several TLRs, including TLR4 and TLR2. The aim of this study was to investigate the effect of single TLR4-MD2 inhibition by using eritoran, compared with the effect of CD14 inhibition alone and combined with the C3 complement inhibitor compstatin (Cp40), on the bacteria-induced inflammatory response in human whole blood. METHODS: Cytokines were measured by multiplex technology, and leukocyte activation markers CD11b and CD35 were measured by flow cytometry. RESULTS: Lipopolysaccharide (LPS)-induced inflammatory markers were efficiently abolished by both anti-CD14 and eritoran. Anti-CD14 was significantly more effective than eritoran in inhibiting LPS-binding to HEK-293E cells transfected with CD14 and Escherichia coli-induced upregulation of monocyte activation markers (P < .01). Combining Cp40 with anti-CD14 was significantly more effective than combining Cp40 with eritoran in reducing E. coli-induced interleukin 6 (P < .05) and monocyte activation markers induced by both E. coli (P < .001) and Staphylococcus aureus (P < .01). Combining CP40 with anti-CD14 was more efficient than eritoran alone for 18 of 20 bacteria-induced inflammatory responses (mean P < .0001). CONCLUSIONS: Whole bacteria-induced inflammation was inhibited more efficiently by anti-CD14 than by eritoran, particularly when combined with complement inhibition. Combined CD14 and complement inhibition may prove a promising treatment strategy for bacterial sepsis.


Asunto(s)
Antibacterianos/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/microbiología , Sepsis/tratamiento farmacológico , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/tratamiento farmacológico , Citocinas/sangre , Escherichia coli/efectos de los fármacos , Humanos , Inflamación/sangre , Receptores de Lipopolisacáridos/sangre , Receptores de Lipopolisacáridos/efectos de los fármacos , Sepsis/microbiología , Staphylococcus aureus/efectos de los fármacos , Receptor Toll-Like 4/efectos de los fármacos
8.
J Immunol ; 192(6): 2837-45, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24554772

RESUMEN

Inflammation is associated with development of atherosclerosis, and cholesterol crystals (CC) have long been recognized as a hallmark of atherosclerotic lesions. CC appear early in the atheroma development and trigger inflammation by NLRP3 inflammasome activation. In this study we hypothesized whether CC employ the complement system to activate inflammasome/caspase-1, leading to release of mature IL-1ß, and whether complement activation regulates CC-induced cytokine production. In this study we describe that CC activated both the classical and alternative complement pathways, and C1q was found to be crucial for the activation. CC employed C5a in the release of a number of cytokines in whole blood, including IL-1ß and TNF. CC induced minimal amounts of cytokines in C5-deficient whole blood, until reconstituted with C5. Furthermore, C5a and TNF in combination acted as a potent primer for CC-induced IL-1ß release by increasing IL-1ß transcripts. CC-induced complement activation resulted in upregulation of complement receptor 3 (CD11b/CD18), leading to phagocytosis of CC. Also, CC mounted a complement-dependent production of reactive oxygen species and active caspase-1. We conclude that CC employ the complement system to induce cytokines and activate the inflammasome/caspase-1 by regulating several cellular responses in human monocytes. In light of this, complement inhibition might be an interesting therapeutic approach for treatment of atherosclerosis.


Asunto(s)
Colesterol/farmacología , Proteínas del Sistema Complemento/inmunología , Citocinas/inmunología , Inflamasomas/efectos de los fármacos , Western Blotting , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células Cultivadas , Colesterol/metabolismo , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Complemento C1q/inmunología , Complemento C1q/metabolismo , Complemento C5/inmunología , Complemento C5/metabolismo , Complemento C5a/inmunología , Complemento C5a/metabolismo , Vía Alternativa del Complemento/efectos de los fármacos , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/efectos de los fármacos , Vía Clásica del Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Citocinas/metabolismo , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Antígeno de Macrófago-1/inmunología , Antígeno de Macrófago-1/metabolismo , Microscopía Confocal , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Fagocitosis/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
9.
J Sport Health Sci ; 13(2): 245-255, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37500010

RESUMEN

BACKGROUND: Exercise training promotes brain plasticity and is associated with protection against cognitive impairment and Alzheimer's disease (AD). These beneficial effects may be partly mediated by blood-borne factors. Here we used an in vitro model of AD to investigate effects of blood plasma from exercise-trained donors on neuronal viability, and an in vivo rat model of AD to test whether such plasma impacts cognitive function, amyloid pathology, and neurogenesis. METHODS: Mouse hippocampal neuronal cells were exposed to AD-like stress using amyloid-ß and treated with plasma collected from human male donors 3 h after a single bout of high-intensity exercise. For in vivo studies, blood was collected from exercise-trained young male Wistar rats (high-intensity intervals 5 days/week for 6 weeks). Transgenic AD rats (McGill-R-Thy1-APP) were injected 5 times/fortnight for 6 weeks at 2 months or 5 months of age with either (a) plasma from the exercise-trained rats, (b) plasma from sedentary rats, or (c) saline. Cognitive function, amyloid plaque pathology, and neurogenesis were assessed. The plasma used for the treatment was analyzed for 23 cytokines. RESULTS: Plasma from exercised donors enhanced cell viability by 44.1% (p = 0.032) and reduced atrophy by 50.0% (p < 0.001) in amyloid-ß-treated cells. In vivo exercised plasma treatment did not alter cognitive function or amyloid plaque pathology but did increase hippocampal neurogenesis by ∼3 fold, regardless of pathological stage, when compared to saline-treated rats. Concentrations of 7 cytokines were significantly reduced in exercised plasma compared to sedentary plasma. CONCLUSION: Our proof-of-concept study demonstrates that plasma from exercise-trained donors can protect neuronal cells in culture and promote adult hippocampal neurogenesis in the AD rat brain. This effect may be partly due to reduced pro-inflammatory signaling molecules in exercised plasma.


Asunto(s)
Enfermedad de Alzheimer , Ratas , Masculino , Ratones , Animales , Humanos , Placa Amiloide/patología , Placa Amiloide/prevención & control , Ratas Wistar , Hipocampo/patología , Péptidos beta-Amiloides/metabolismo , Neurogénesis/fisiología , Citocinas , Plasma/metabolismo
10.
Front Immunol ; 15: 1383505, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38686379

RESUMEN

Acute myocardial infarction (MI) results in tissue damage to affected areas of the myocardium. The initial inflammatory response is the most damaging for residual cardiac function, while at later stages inflammation is a prerequisite for proper healing and scar formation. Balancing the extent and duration of inflammation during various stages after MI is thus pivotal for preserving cardiac function. Recently, a signaling lymphocytic activation molecule 1 (SLAMF1)-derived peptide (P7) was shown to reduce the secretion of inflammatory cytokines and protected against acute lipopolysaccharide-induced death in mice. In the present study, we experimentally induced MI by permanent ligation of the left anterior descending artery (LAD) in mice and explored the beneficial effect of immediately administering P7, with the aim of dampening the initial inflammatory phase without compromising the healing and remodeling phase. Blood samples taken 9 h post-LAD surgery and P7 administration dampened the secretion of inflammatory cytokines, but this dampening effect of P7 was diminished after 3 days. Echocardiography revealed less deterioration of cardiac contraction in mice receiving P7. In line with this, less myocardial damage was observed histologically in P7-treated mice. In conclusion, the administration of a SLAMF1-derived peptide (P7) immediately after induction of MI reduces the initial myocardial inflammation, reduces infarct expansion, and leads to less deterioration of cardiac contraction.


Asunto(s)
Modelos Animales de Enfermedad , Infarto del Miocardio , Animales , Ratones , Masculino , Citocinas/metabolismo , Ratones Endogámicos C57BL , Antígenos CD/metabolismo , Ligadura , Miocardio/patología , Miocardio/metabolismo , Péptidos/farmacología , Receptores de Superficie Celular/metabolismo , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/patología
11.
Cell Rep Med ; 4(7): 101098, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37390829

RESUMEN

During differentiation, neutrophils undergo a spontaneous pro-inflammatory program that is hypothesized here to be under caspase-8 control. In mice, intraperitoneal administration of the caspase-8 inhibitor z-IETD-fmk is sufficient to unleash the production of pro-inflammatory cytokines and neutrophil influx in the absence of cell death. These effects are due to selective inhibition of caspase-8 and require tonic interferon-ß (IFN-ß) production and RIPK3 but not MLKL, the essential downstream executioner of necroptotic cell death. In vitro, stimulation with z-IETD-fmk is sufficient to induce significant cytokine production in murine neutrophils but not in macrophages. Therapeutic administration of z-IETD-fmk improves clinical outcome in models of lethal bacterial peritonitis and pneumonia by augmenting cytokine release, neutrophil influx, and bacterial clearance. Moreover, the inhibitor protects mice against high-dose endotoxin shock. Collectively, our data unveil a RIPK3- and IFN-ß-dependent pathway that is constitutively activated in neutrophils and can be harnessed therapeutically using caspase-8 inhibition.


Asunto(s)
Apoptosis , Infecciones Bacterianas , Animales , Ratones , Infecciones Bacterianas/tratamiento farmacológico , Caspasa 8/metabolismo , Caspasa 8/farmacología , Citocinas/metabolismo , Activación Neutrófila
12.
Life Sci Alliance ; 6(12)2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37788908

RESUMEN

Inflammation plays a crucial role in the development and progression of many diseases, and is often caused by dysregulation of signalling from pattern recognition receptors, such as TLRs. Inhibition of key protein-protein interactions is an attractive target for treating inflammation. Recently, we demonstrated that the signalling lymphocyte activation molecule family 1 (SLAMF1) positively regulates signalling downstream of TLR4 and identified the interaction interface between SLAMF1 and the TLR4 adaptor protein TRIF-related adapter molecule (TRAM). Based on these findings, we developed a SLAMF1-derived peptide, P7, which is linked to a cell-penetrating peptide for intracellular delivery. We found that P7 peptide inhibits the expression and secretion of IFNß and pro-inflammatory cytokines (TNF, IL-1ß, IL-6) induced by TLR4, and prevents death in mice subjected to LPS shock. The mechanism of action of P7 peptide is based on interference with several intracellular protein-protein interactions, including TRAM-SLAMF1, TRAM-Rab11FIP2, and TIRAP-MyD88 interactions. Overall, P7 peptide has a unique mode of action and demonstrates high efficacy in inhibiting TLR4-mediated signalling in vitro and in vivo.


Asunto(s)
Transducción de Señal , Receptor Toll-Like 4 , Animales , Ratones , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Péptidos/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inflamación
13.
Open Heart ; 10(2)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37591633

RESUMEN

BACKGROUND: Tocilizumab improves myocardial salvage index (MSI) in patients with ST-elevation myocardial infarction (STEMI), but its mechanisms of action are unclear. Here, we explored how cytokines were affected by tocilizumab and their correlations with neutrophils, C-reactive protein (CRP), troponin T, MSI and infarct size. METHODS: STEMI patients were randomised to receive a single dose of 280 mg tocilizumab (n=101) or placebo (n=98) before percutaneous coronary intervention. Blood samples were collected before infusion of tocilizumab or placebo at baseline, during follow-up at 24-36, 72-168 hours, 3 and 6 months. 27 cytokines were analysed using a multiplex cytokine assay. Cardiac MRI was performed during hospitalisation and 6 months. RESULTS: Repeated measures analysis of variance showed significant (p<0.001) between-group difference in changes for IL-6, IL-8 and IL-1ra due to an increase in the tocilizumab group during hospitalisation. IL-6 and IL-8 correlated to neutrophils in the placebo group (r=0.73, 0.68, respectively), which was attenuated in the tocilizumab group (r=0.28, 0.27, respectively). A similar pattern was seen for MSI and IL-6 and IL-8 in the placebo group (r=-0.29, -0.25, respectively) in patients presenting ≤3 hours from symptom onset, which was attenuated in the tocilizumab group (r=-0.09,-0.14, respectively). CONCLUSIONS: Tocilizumab increases IL-6, IL-8 and IL-1ra in STEMI. IL-6 and IL-8 show correlations to neutrophils/CRP and markers of cardiac injury in the placebo group that was attenuated in the tocilizumab group. This may suggest a beneficial effect of tocilizumab on the ischaemia-reperfusion injury in STEMI patients. TRIAL REGISTRATION NUMBER: NCT03004703.


Asunto(s)
Citocinas , Infarto del Miocardio con Elevación del ST , Humanos , Proteína Antagonista del Receptor de Interleucina 1 , Infarto del Miocardio con Elevación del ST/diagnóstico por imagen , Infarto del Miocardio con Elevación del ST/tratamiento farmacológico , Interleucina-6 , Interleucina-8 , Proteína C-Reactiva , Receptores de Interleucina-6
14.
Wound Repair Regen ; 19(3): 337-47, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21371163

RESUMEN

Acute wounds contain many biological active molecules, including several cytokines and growth factors. However, the cellular sources of each molecule, as well as the stimuli inducing them, are poorly characterized. We quantified the levels of 27 cytokines, chemokines, and growth factors in acute wound fluid in a luminex-based assay. The acute wound fluid contained particularly high levels of IL-6 and IL-8, as well as elevated levels of MCP-1, IL-1RA, PDGF, IP-10, IFN-γ, and TNF-α. Surprisingly, the amounts of IL-1ß and IL-10 were relatively low. To characterize the cellular sources of these molecules, we analyzed supernatants from monocytes, neutrophils, keratinocytes, fibroblasts, and endothelial cells stimulated with pro- and anti inflammatory cytokines, and different Toll-like receptor (TLR) ligands. The different cell types showed overlapping but distinct patterns of production of signal molecules, as well as sensitivity to ligands. Among pro-inflammatory cytokines, IL-1ß was the most potent inducer of signal molecule production. Furthermore, keratinocytes and endothelial cells were in particular responsive to the Toll-like receptor-3 ligand poly I:C. New interactions between cytokines and growth factors were revealed, which may have important roles in wound healing, including IL-1ß-induced IFN-γ and IL-10-induced VEGF.


Asunto(s)
Citocinas/análisis , Exudados y Transudados/química , Cicatrización de Heridas/fisiología , Proliferación Celular , Células Cultivadas , Quimiocinas/análisis , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/análisis , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/citología , Receptores Toll-Like/fisiología
15.
J Immunol ; 182(1): 588-95, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19109192

RESUMEN

Soluble proteins that bind LPS, like myeloid differentiation-2 (MD-2) and CD14, have essential roles in regulating LPS signaling through TLR4. During a gram-negative bacterial infection, the host may control the response by adjusting the levels of soluble MD-2 and CD14. To address the surface expression of MD-2 on human leukocytes, we developed a mAb, IIC1, that recognized MD-2 both free and when bound to TLR4. MD-2 was found on the surface of freshly isolated monocytes, on a subpopulation of CD19(+) B-cells and on CD15(+) neutrophils. LPS transiently reduced the MD-2 levels on monocytes, which is most likely due to endocytosis of the LPS receptor complex since MD-2 colocalized with TLR4 in early endosomes after LPS stimulation. In the absence of LPS, MD-2 partly colocalized with TLR4 in Golgi trans and medial compartments. Cultivating monocytes for 18-20 h resulted in loss of MD-2 expression on the surface, which was reversed either by LPS or IL-10. Furthermore, addition of IL-10, but not LPS, resulted in a considerable increase in mRNA for both MD-2 and CD14. Using ELISA, we demonstrated that IL-10 had a profound dose- and time-related effect on the release of soluble MD-2 and soluble CD14 from monocytes. In HIV-infected patients, the amounts of MD-2, CD14, and IL-10 increased significantly in the patient group with AIDS. Of interest, we found that IL-10, CD14, and MD-2 levels were positively correlated, suggesting that IL-10 may be a driving force for increased release of MD-2 and CD14 during systemic inflammation.


Asunto(s)
Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Interleucina-10/fisiología , Receptores de Lipopolisacáridos/biosíntesis , Antígeno 96 de los Linfocitos/biosíntesis , Monocitos/inmunología , Monocitos/metabolismo , Regulación hacia Arriba/inmunología , Adulto , Animales , Anticuerpos Monoclonales/metabolismo , Sitios de Unión de Anticuerpos , Células CHO , Línea Celular , Cricetinae , Cricetulus , Femenino , Humanos , Mediadores de Inflamación/fisiología , Interleucina-10/sangre , Receptores de Lipopolisacáridos/sangre , Receptores de Lipopolisacáridos/genética , Antígeno 96 de los Linfocitos/sangre , Antígeno 96 de los Linfocitos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/biosíntesis
16.
J Infect Dis ; 201(5): 783-92, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20121435

RESUMEN

Iron is an essential nutrient for microbes, and many pathogenic bacteria depend on siderophores to obtain iron. The mammalian innate immunity protein lipocalin 2 (Lcn2; also known as neutrophil gelatinase-associated lipocalin, 24p3, or siderocalin) binds the siderophore carboxymycobactin, an essential component of the iron acquisition apparatus of mycobacteria. Here we show that Lcn2 suppressed growth of Mycobacterium avium in culture, and M. avium induced Lcn2 production from mouse macrophages. Lcn2 also had elevated levels and initially limited the growth of M. avium in the blood of infected mice but did not impede growth in tissues and during long-term infections. M. avium is an intracellular pathogen. Subcellular imaging of infected macrophages revealed that Lcn2 trafficked to lysosomes separate from M. avium, whereas transferrin was efficiently transported to the mycobacteria. Thus, mycobacteria seem to reside in the Rab11(+) endocytic recycling pathway, thereby retaining access to nutrition and avoiding endocytosed immunoproteins like Lcn2.


Asunto(s)
Proteínas de Fase Aguda/inmunología , Proteínas de Fase Aguda/metabolismo , Lipocalinas/inmunología , Lipocalinas/metabolismo , Lisosomas/metabolismo , Lisosomas/microbiología , Mycobacterium avium/inmunología , Mycobacterium avium/patogenicidad , Proteínas Oncogénicas/inmunología , Proteínas Oncogénicas/metabolismo , Transferrina/metabolismo , Animales , Sangre/microbiología , Recuento de Colonia Microbiana , Lipocalina 2 , Lipocalinas/sangre , Hígado/microbiología , Lisosomas/química , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mycobacterium avium/crecimiento & desarrollo , Mycobacterium avium/metabolismo , Proteínas Oncogénicas/sangre , Bazo/microbiología , Tuberculosis/inmunología , Tuberculosis/microbiología , Proteínas de Unión al GTP rab/metabolismo
17.
Front Immunol ; 12: 679458, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34234780

RESUMEN

Macrophages are sentinels of the innate immune system, and the human monocytic cell line THP-1 is one of the widely used in vitro models to study inflammatory processes and immune responses. Several monocyte-to-macrophage differentiation protocols exist, with phorbol 12-myristate-13-acetate (PMA) being the most commonly used and accepted method. However, the concentrations and duration of PMA treatment vary widely in the published literature and could affect the probed phenotype, however their effect on protein expression is not fully deciphered. In this study, we employed a dimethyl labeling-based quantitative proteomics approach to determine the changes in the protein repertoire of macrophage-like cells differentiated from THP-1 monocytes by three commonly used PMA-based differentiation protocols. Employing an integrated network analysis, we show that variations in PMA concentration and duration of rest post-stimulation result in downstream differences in the protein expression and cellular signaling processes. We demonstrate that these differences result in altered inflammatory responses, including variation in the expression of cytokines upon stimulation with various Toll-like receptor (TLR) agonists. Together, these findings provide a valuable resource that significantly expands the knowledge of protein expression dynamics with one of the most common in vitro models for macrophages, which in turn has a profound impact on the immune as well as inflammatory responses being studied.


Asunto(s)
Inmunidad , Macrófagos/metabolismo , Monocitos/metabolismo , Proteoma , Proteómica , Biomarcadores , Diferenciación Celular/inmunología , Membrana Celular , Biología Computacional/métodos , Citocinas/metabolismo , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Monocitos/inmunología , Proteómica/métodos , Transducción de Señal , Células THP-1 , Acetato de Tetradecanoilforbol/inmunología , Transcriptoma
18.
J Leukoc Biol ; 109(1): 23-33, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32531827

RESUMEN

Mycobacterium avium (Mav) causes chronic infections in immunocompromised patients that require long-term antibiotic treatment. We have previously shown that Mav takes residence in host Mϕs and establishes a compartment (MavC) in which it is hidden from host defenses. Failure to establish the MavC traps Mav in Lamp1+ phagolysosomes where growth is prevented, and inflammatory signaling activated through TLRs 7/8. To elucidate how antibiotic treatment affects mycobacterial trafficking and host defenses, we infected human primary Mϕs with Mav for 4 days prior to treatment with a macrolide, aminoglycoside, and ethambutol. We show that Mav is killed and the MavC fuses with Lamp1+ lysosomes following antibiotic treatment. However, this does not result in nuclear translocation of NF-κB or production of inflammatory cytokines, suggesting different Lamp1+ lysosomal compartments can form that differ in their innate signaling capabilities. Thus, we show that upon antibiotic treatment of a chronic infection, Mav is quietly disposed of by Mϕs.


Asunto(s)
Antibióticos Antituberculosos/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Infección por Mycobacterium avium-intracellulare , Citocinas/biosíntesis , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos/inmunología , Complejo Mycobacterium avium/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/microbiología
19.
J Leukoc Biol ; 107(4): 673-683, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32083344

RESUMEN

We recently showed that TLR8 is critical for the detection of Gram-positive bacteria by human monocytes. Here, we hypothesized that TLR8 and complement together regulate antibacterial responses in human blood. Anticoagulated blood was treated with selective inhibitors of TLR8 and/or complement C5, and then challenged with live Streptococcus agalactiae (Group B streptococcus, GBS), Staphylococcus aureus, or Escherichia coli. Cytokine production, plasma membrane permeability, bacterial survival, phagocytosis, and activation of coagulation was examined. GBS and S. aureus, but not E. coli, triggered TLR8-dependent production of IL-12p70, IL-1ß, TNF, and IL-6 in fresh human whole blood. In purified polymorphonuclear neutrophils (PMN), GBS and S. aureus induced IL-8 release in part via TLR8, whereas PMN plasma membrane leakage and extracellular DNA levels increased independently of TLR8. TLR8 was more important than C5 for bacteria-induced production of IL-12p70, IL-1ß, and TNF in blood, whereas IL-8 release was more C5 dependent. Both TLR8 and C5 induced IL-6 release and activation of prothrombin cleavage, and here their combined effects were additive. Blocking of C5 or C5aR1 attenuated phagocytosis and increased the extracellular growth of GBS in blood, whereas TLR8 inhibition neither reduced phagocytosis nor intracellular killing of GBS and S. aureus. In conclusion, TLR8 is more important than C5 for production of IL-12p70, IL-1ß, and TNF upon GBS and S. aureus infection in blood, whereas C5 is central for IL-8 release and phagocytosis. Both TLR8 and C5 mediate IL-6 release and activation of coagulation during challenge with Gram-positive bacteria in blood.


Asunto(s)
Complemento C5/metabolismo , Citocinas/sangre , Bacterias Grampositivas/fisiología , Trombina/metabolismo , Receptor Toll-Like 8/sangre , Coagulación Sanguínea , Membrana Celular/metabolismo , Supervivencia Celular , ADN/metabolismo , Humanos , Interleucina-8/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Viabilidad Microbiana , Monocitos/metabolismo , Neutrófilos/metabolismo , Receptor Toll-Like 8/antagonistas & inhibidores , Receptor Toll-Like 8/metabolismo
20.
Nat Commun ; 11(1): 147, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31919342

RESUMEN

During HIV infection, cell-to-cell transmission results in endosomal uptake of the virus by target CD4+ T cells and potential exposure of the viral ssRNA genome to endosomal Toll-like receptors (TLRs). TLRs are instrumental in activating inflammatory responses in innate immune cells, but their function in adaptive immune cells is less well understood. Here we show that synthetic ligands of TLR8 boosted T cell receptor signaling, resulting in increased cytokine production and upregulation of surface activation markers. Adjuvant TLR8 stimulation, but not TLR7 or TLR9, further promoted T helper cell differentiation towards Th1 and Th17. In addition, we found that endosomal HIV induced cytokine secretion from CD4+ T cells in a TLR8-specific manner. TLR8 engagement also enhanced HIV-1 replication and potentiated the reversal of latency in patient-derived T cells. The adjuvant TLR8 activity in T cells can contribute to viral dissemination in the lymph node and low-grade inflammation in HIV patients. In addition, it can potentially be exploited for therapeutic targeting and vaccine development.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/inmunología , Células TH1/inmunología , Células Th17/inmunología , Receptor Toll-Like 8/metabolismo , Línea Celular , Infecciones por VIH/transmisión , Humanos , Inmunidad Innata/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 8/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA