Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Immunol ; 193(4): 1954-65, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25024378

RESUMEN

Canonical neutrophil antimicrobial effector mechanisms, such as degranulation, production of reactive oxygen species, and release of neutrophil extracellular traps (NETs), can result in severe pathology. Activation of neutrophils through immune complexes (ICs) plays a central role in the pathogenesis of many autoimmune inflammatory diseases. In this study, we report that immobilized ICs (iICs), which are hallmarks of several autoimmune diseases, induce the release of NETs from primary human neutrophils. The iIC-induced NET formation was found to require production of reactive oxygen species by NADPH oxidase and myeloperoxidase and to be mediated by FcγRIIIb. Blocking of the ß2 integrin macrophage-1 Ag but not lymphocyte function-associated Ag-1 abolished iIC-induced NET formation. This suggests that FcγRIIIb signals in association with macrophage-1 Ag. As intracellular signaling pathways involved in iIC-induced NET formation we identified the tyrosine kinase Src/Syk pathway, which downstream regulates the PI3K/Akt, p38 MAPK, and ERK1/2 pathways. To our knowledge, the present study shows for the first time that iICs induce NET formation. Thus, we conclude that NETs contribute to pathology in autoimmune inflammatory disorders associated with surface-bound ICs.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Antígeno de Macrófago-1/inmunología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Receptores de IgG/inmunología , Aminopirina/farmacología , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Enfermedades Autoinmunes/inmunología , Butadienos/farmacología , Antígeno CD11a/metabolismo , Antígenos CD18/metabolismo , Degranulación de la Célula , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Proteínas Ligadas a GPI/inmunología , Humanos , Imidazoles/farmacología , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Antígeno de Macrófago-1/metabolismo , Mesalamina/farmacología , Nitrilos/farmacología , Compuestos Onio/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Piridinas/farmacología , Pirimidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptores de IgG/antagonistas & inhibidores , Quinasa Syk , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/inmunología
2.
Int J Med Microbiol ; 305(6): 493-500, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26005182

RESUMEN

The obligatory intracellular bacterium Chlamydia pneumoniae (C. pneumoniae) can survive and multiply in neutrophil granulocytes. Since neutrophils are short living cells, inhibition of neutrophil apoptosis appears to play a major role in the productive infection of neutrophils by C. pneumoniae. In the present study, we have investigated which survival pathways and which events of the apoptotic process are modulated in C. pneumoniae-infected neutrophils. All infection experiments were carried out using primary human neutrophils in vitro. We show that infection with C. pneumoniae activates PI3K/Akt as well as the ERK1/2 and p38 MAP kinases and present evidence that activation of the PI3K/Akt and ERK1/2 pathways are essential to initiate the apoptosis delay in C. pneumoniae-infected neutrophils. Both the PI3K/Akt and ERK1/2 pathways are involved in the maintained expression of the anti-apoptotic protein Mcl-1. In addition, we also showed that the PI3K/Akt pathway leads to the activation of NF-κB-dependent release of IL-8 by infected neutrophils. Infection with C. pneumoniae activates the PI3K/Akt and ERK1/2 MAPK survival pathways in neutrophils, induces the NF-κB dependent release of IL-8 and leads to the maintenance of Mcl-1 expression in neutrophils.


Asunto(s)
Apoptosis , Chlamydophila pneumoniae/crecimiento & desarrollo , Neutrófilos/microbiología , Regulación de la Expresión Génica , Humanos , Interleucina-8/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Med Microbiol Immunol ; 202(1): 25-35, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22661217

RESUMEN

Neutrophil granulocytes provide the first line of defense against bacterial, fungal, and parasitic infections. They phagocytose and kill many invading pathogens. Certain pathogenic microorganisms such as the intracellular protozoan parasite Leishmania major (L. major) can survive inside neutrophils. Mature neutrophils have a very short life span due to spontaneous apoptosis. Previously, we have reported that infections with L. major are able to delay spontaneous apoptosis. In the present study, we addressed the underlying mechanisms of regulation of both extrinsic and intrinsic apoptosis. We show that interaction with L. major transiently activates ERK1/2 phosphorylation. Pharmacological inhibition of ERK1/2 phosphorylation reversed the apoptosis delay. Moreover, infection leads to the enhanced and sustainable expression of the anti-apoptotic proteins Bcl-2 and Bfl-1, respectively. As downstream events, the release of cytochrome c from mitochondria and processing of caspase-6 were inhibited. We also confirm that infection with L. major results in reduced FAS expression on the surface of neutrophils. The presented data indicate that infection with L. major affects both intrinsic as well as extrinsic pathways of neutrophil apoptosis. Enhanced life span of host neutrophils enables the parasite to survive within neutrophils.


Asunto(s)
Apoptosis , Leishmania major/patogenicidad , Sistema de Señalización de MAP Quinasas/inmunología , Neutrófilos/inmunología , Neutrófilos/parasitología , Caspasa 6/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Humanos , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Receptor fas/biosíntesis
4.
Mediators Inflamm ; 2013: 710239, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24381411

RESUMEN

Neutrophil extracellular traps (NETs) have been suggested to play a pathophysiological role in several autoimmune diseases. Since NET-formation in response to several biological and chemical stimuli is mostly ROS dependent, in theory any substance that inhibits or scavenges ROS could prevent ROS-dependent NET release. Therefore, in the present comprehensive study, several antioxidative substances were assessed for their capacity to inhibit NET formation of primary human neutrophils in vitro. We could show that the flavonoids (-)-epicatechin, (+)-catechin hydrate, and rutin trihydrate as well as vitamin C and the pharmacological substances N-acetyl-L-cysteine and 5-aminosalicylic acid inhibited PMA induced ROS production and NET formation. Therefore, a broad spectrum of antioxidative substances that reduce ROS production of primary human neutrophils also inhibits ROS-dependent NET formation. It is tempting to speculate that such antioxidants can have beneficial therapeutic effects in diseases associated with ROS-dependent NET formation.


Asunto(s)
Antioxidantes/farmacología , Flavonoides/farmacología , Mesalamina/farmacología , Neutrófilos/efectos de los fármacos , Acetilcisteína/farmacología , Apoptosis/efectos de los fármacos , Ácido Ascórbico/farmacología , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Humanos , NADPH Oxidasas/antagonistas & inhibidores , Neutrófilos/inmunología , Peroxidasa/metabolismo , Fagocitosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Acetato de Tetradecanoilforbol/farmacología
5.
Infect Immun ; 80(4): 1615-23, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22252875

RESUMEN

Anaplasma phagocytophilum, a Gram-negative, obligate intracellular bacterium infects primarily neutrophil granulocytes. Infection with A. phagocytophilum leads to inhibition of neutrophil apoptosis and consequently contributes to the longevity of the host cells. Previous studies demonstrated that the infection inhibits the executionary apoptotic machinery in neutrophils. However, little attempt has been made to explore which survival signals are modulated by the pathogen. The aim of the present study was to clarify whether the phosphatidylinositol 3-kinase (PI3K)/Akt and NF-κB signaling pathways, which are considered as important survival pathways in neutrophils, are involved in A. phagocytophilum-induced apoptosis delay. Our data show that infection of neutrophils with A. phagocytophilum activates the PI3K/Akt pathway and suggest that this pathway, which in turn maintains the expression of the antiapoptotic protein Mcl-1, contributes to the infection-induced apoptosis delay. In addition, the PI3K/Akt pathway is involved in the activation of NF-κB in A. phagocytophilum-infected neutrophils. Activation of NF-κB leads to the release of interleukin-8 (IL-8) from infected neutrophils, which, in an autocrine manner, delays neutrophil apoptosis. In addition, enhanced expression of the antiapoptotic protein cIAP2 was observed in A. phagocytophilum-infected neutrophils. Taken together, the data indicate that upstream of the apoptotic cascade, signaling via the PI3K/Akt pathway plays a major role for apoptosis delay in A. phagocytophilum-infected neutrophils.


Asunto(s)
Anaplasma phagocytophilum/patogenicidad , Apoptosis , FN-kappa B/metabolismo , Neutrófilos/microbiología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Anaplasma phagocytophilum/metabolismo , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Células Cultivadas , Humanos , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Interleucina-8/biosíntesis , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , FN-kappa B/biosíntesis , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Fagocitosis , Fosfatidilinositol 3-Quinasa/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Transducción de Señal , Ubiquitina-Proteína Ligasas
6.
J Immunol ; 184(1): 391-400, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19949068

RESUMEN

Neutrophil granulocytes are rapidly recruited from the bloodstream to the site of acute inflammation where they die in large numbers. Because release of toxic substances from dead neutrophils can propagate the inflammatory response leading to tissue destruction, clearance of dying inflammatory neutrophils has a critical function in the resolution of the inflammatory response. Apoptotic neutrophils are phagocytosed primarily by macrophages, provided these cells are present in adequate numbers. However, macrophages are rare at sites of acute inflammation, whereas the number of neutrophils can be extremely high. In the current study, in vitro experiments with human neutrophils were carried out to investigate whether neutrophils can ingest apoptotic neutrophils. We show that naïve granulocytes isolated from venous blood have a limited capacity to phagocytose apoptotic cells. However, exposure to activating stimuli such as LPS, GM-CSF and/or IFN-gamma results in enhanced phagocytosis of apoptotic cells. The efficient uptake of apoptotic cells by neutrophils was found to depend on the presence of heat labile serum factors. Importantly, the contact to or uptake of apoptotic cells inhibited neutrophil functions such as respiratory burst and the release of the proinflammatory cytokines TNF-alpha and interferon-inducible protein-10. Contact to apoptotic cells, however, induced the secretion of IL-8 and growth-related oncogene-alpha, which was independent of NF-kappaB and p38 MAPK but involved C5a and the ERK1/2 pathway. The data suggest that activated neutrophils participate in the clearance of apoptotic cells. In addition, because apoptotic cells inhibit proinflammatory functions of neutrophils, uptake of apoptotic cells by neutrophils contributes to the resolution of inflammation.


Asunto(s)
Apoptosis/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Células Cultivadas , Citocinas/biosíntesis , Citocinas/inmunología , Humanos , Estallido Respiratorio/inmunología , Transducción de Señal/inmunología
7.
Mediators Inflamm ; 2012: 849136, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22481865

RESUMEN

The formation of neutrophil extracellular traps (NETs) depends on the generation of reactive oxygen species (ROS). Previous studies revealed that both NADPH oxidase and myeloperoxidase (MPO) are required for NET release. However, the contribution of various ROS as well as the role of mitochondria-derived ROS has not been addressed so far. In the present study we aimed to investigate in a systematic and comprehensive manner the contribution of various ROS and ROS-generating pathways to the PMA-induced NET release. By using specific inhibitors, the role of both NADPH oxidase- and mitochondria-derived ROS as well as the contribution of superoxide dismutase (SOD) and MPO on the NET release was assessed. We could demonstrate that NADPH oxidase function is crucial for the formation of NETs. In addition, we could clearly show the involvement of MPO-derived ROS in NET release. Our results, however, did not provide evidence for the role of SOD- or mitochondria-derived ROS in NET formation.


Asunto(s)
Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adulto , Apoptosis/genética , Supervivencia Celular/genética , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Neutrófilos/efectos de los fármacos , Peroxidasa/antagonistas & inhibidores , Peroxidasa/metabolismo , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/metabolismo
8.
Front Immunol ; 12: 747049, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34733282

RESUMEN

Although macrophages are considered for host cells for the multiplication of Leishmania, recent studies indicate the important role of neutrophil granulocytes as host cells for these intracellular parasites. Neutrophils have been shown to be massively and rapidly recruited to the site of Leishmania infection where they represent the first cells to encounter the parasites. Exposure to ATP and UTP have been shown to enhance anti-Leishmania activity of macrophages and intralesional injection of UTP led to strongly reduced parasite load in vivo. Since the in vivo anti-leishmanial effect of extracellular UTP correlated with enhanced neutrophil recruitment and enhanced ROS production at the site of Leishmania infection we hypothesized that exposure to extracellular nucleotides can directly enhance the killing of Leishmania by neutrophils. Since purinergic signaling is an essential mechanism of neutrophil activation the aim of the present study was to assess whether purinergic exposure results in the activation of anti-leishmanial neutrophil functions and, therefore, represent an essential component of enhanced anti-leishmanial defense in leishmaniasis. We could show that exposure to ATP and UTP led to activation and enhanced CD11b expression of primary human neutrophils in vitro. Leishmania-induced ROS production was strongly enhanced by extracellular ATP and UTP. Importantly, exposure to ATP and UTP resulted in enhanced killing of Leishmania donovani by neutrophils. In addition, ATP strongly enhanced the secretion of IL-8 and IL-1ß by Leishmania-exposed neutrophils. Our results suggest that signaling via the P2 receptor and phosphorylation of Erk1/2, Akt and p38 are involved in the purinergic enhancement of anti-leishmanial functions of neutrophils.


Asunto(s)
Adenosina Trifosfato/inmunología , Leishmania donovani/inmunología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Uridina Trifosfato/inmunología , Células Cultivadas , Humanos , Leishmaniasis Visceral/inmunología
9.
Front Immunol ; 12: 636954, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34168641

RESUMEN

Neutrophil extracellular traps (NETs) consist of decondensed nuclear chromatin that is associated with proteins and are released by neutrophils during an inflammatory response. Released NETs are able to capture pathogens, prevent their dissemination and potentially kill them via antimicrobial peptides and proteins that are associated with the decondensed chromatin. In addition to their antimicrobial functions, NETs have also been shown to exert immunomodulatory effects by activation and differentiation of macrophages, dendritic cells and T cells. However, the effect of NETs on neutrophil functions is poorly understood. Here we report the first comprehensive study regarding the effects of NETs on human primary neutrophils in vitro. NETs were isolated from cultures of PMA-exposed neutrophils. Exposure of neutrophils to isolated NETs resulted in the activation of several neutrophil functions in a concentration-dependent manner. NETs induced exocytosis of granules, the production of reactive oxygen species (ROS) by the NADPH oxidase NOX2, NOX2-dependent NET formation, increased the phagocytosis and killing of microbial pathogens. Furthermore, NETs induced the secretion of the proinflammatory chemokine IL-8 and the B-cell-activating cytokine BAFF. We could show that the NET-induced activation of neutrophils occurs by pathways that involve the phosphorylation of Akt, ERK1/2 and p38. Taken together our results provide further insights into the proinflammatory role of NETs by activating neutrophil effector function and further supports the view that NETs can amplify inflammatory events. On the one hand the amplified functions enhance the antimicrobial defense. On the other hand, NET-amplified neutrophil functions can be involved in the pathophysiology of NET-associated diseases. In addition, NETs can connect the innate and adaptive immune system by inducing the secretion of the B-cell-activating cytokine BAFF.


Asunto(s)
Trampas Extracelulares/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Factor Activador de Células B/metabolismo , Células Cultivadas , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Interleucina-8/metabolismo , Sistema de Señalización de MAP Quinasas , NADPH Oxidasa 2/metabolismo , Activación Neutrófila , Fagocitosis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo
10.
Front Immunol ; 12: 564720, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679729

RESUMEN

Neutrophil granulocytes represent the first line of defense against invading pathogens. In addition to the production of Reactive Oxygen Species, degranulation, and phagocytosis, these specialized cells are able to extrude Neutrophil Extracellular Traps. Extensive work was done to elucidate the mechanism of this special form of cell death. However, the exact mechanisms are still not fully uncovered. Here we demonstrate that the small GTPase Cdc42 is a negative regulator of NET formation in primary human and murine neutrophils. We present a functional role for Cdc42 activity in NET formation that differs from the already described NETosis pathways. We show that Cdc42 deficiency induces NETs independent of the NADPH-oxidase but dependent on protein kinase C. Furthermore, we demonstrate that Cdc42 deficiency induces NETosis through activation of SK-channels and that mitochondria play a crucial role in this process. Our data therefore suggests a mechanistic role for Cdc42 activity in primary human neutrophils, and identify Cdc42 activity as a target to modulate the formation of Neutrophil Extracellular Traps.


Asunto(s)
Trampas Extracelulares/metabolismo , Mitocondrias/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Neutrófilos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Animales , Células Cultivadas , Trampas Extracelulares/genética , Humanos , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Neutrófilos/citología , Canales de Potasio Calcio-Activados/metabolismo , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/genética
11.
Infect Immun ; 78(1): 358-63, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19858302

RESUMEN

Anaplasma phagocytophilum, the causative agent of tick-borne human granulocytic anaplasmosis (HGA), is an intracellular bacterium which survives and multiplies inside polymorphonuclear neutrophil granulocytes (PMN). Increased bacterial burden in gamma interferon (IFN-gamma)-deficient mice suggested a major role of IFN-gamma in the control of A. phagocytophilum. Here we investigated whether infection of human PMN with A. phagocytophilum impairs IFN-gamma signaling thus facilitating intracellular survival of the bacterium. The secretion of the IFN-gamma-inducible chemokines IP-10/CXCL10 and MIG/CXCL9 was markedly inhibited in infected neutrophils. Molecular analyses revealed that, compared to uninfected PMN, A. phagocytophilum decreased the expression of the IFN-gamma receptor alpha-chain CD119, diminished the IFN-gamma-induced phosphorylation of STAT1, and enhanced the expression of SOCS1 and SOCS3 in PMN. Since IFN-gamma activates various antibacterial effector mechanisms of PMN, the impaired IFN-gamma signaling in infected cells likely contributes to the survival of A. phagocytophilum inside PMN and to HGA disease development.


Asunto(s)
Anaplasma phagocytophilum/fisiología , Interferón gamma/metabolismo , Neutrófilos/microbiología , Transducción de Señal/fisiología , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo
12.
Front Immunol ; 11: 532, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32411122

RESUMEN

Neutrophils act as the first line of defense against invading pathogens. Although traditionally considered in context of their antimicrobial effector functions, the importance of tumor-associated neutrophils (TANs) in the development of cancer has become increasingly clear during the last decade. With regard to their high plasticity, neutrophils were shown to acquire an anti-tumorigenic N1 or a pro-tumorigenic N2 phenotype. Despite the urgent need to get a comprehensive understanding of the interaction of TANs with their tumor microenvironment, most studies still rely on murine tumor models. Here we present for the first time a polarization attempt to generate N1 and N2 neutrophils from primary human neutrophils in vitro. Our results underscore that N1-polarized neutrophils have a pro-inflammatory phenotype characterized among others by a higher level of intercellular adhesion molecule (ICAM)-1 and high secretion of interferon (IFN)γ-induced protein 10 (IP-10)/C-X-C motif chemokine 10 (CXCL10) and tumor necrosis factor (TNF). Further, we demonstrate that neutrophils incubated under a tumor-mimicking in vitro environment show a high cell surface expression of C-X-C motif chemokine receptor 2 (CXCR2) and secrete high levels of interleukin (IL)-8. These findings suggest that it is feasible to polarize blood-derived primary human neutrophils toward N1- and N2-like phenotypes in vitro. Further, we hypothesized that the presence of anti-inflammatory neutrophil phenotype is not a phenomenon limited to cancer but also occurs when neutrophils are infected with intracellular pathogens. Indeed, our findings indicate that N2-polarized neutrophils exert a markedly decreased capacity to kill the protozoan parasite Leishmania donovani and therefore permit parasite persistence.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular/inmunología , Neutrófilos/inmunología , Humanos , Leishmania donovani/inmunología , Fenotipo
13.
Front Immunol ; 11: 1197, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32595647

RESUMEN

Neutrophil granulocytes are key components of the innate immune system. As the first responders to inflammatory cues, they rapidly migrate toward the site of infection or inflammation and fulfill diverse effector functions. Since these effector functions can be both beneficial and harmful to the host and surrounding tissue, they require a strict control. The small GTPase Cdc42 is known to regulate neutrophil locomotion by controlling cytoskeleton rearrangement in murine neutrophils. However, the role of Cdc42 in other neutrophil functions in human neutrophils is still poorly understood. Here we demonstrate that in primary human neutrophils, Cdc42 controls directed and random migration, activation, and degranulation as well as the formation of reactive oxygen species, in a stimulus dependent manner. In addition, we show that Cdc42 regulates pathogen killing efficiency, both in murine and human neutrophils. Cdc42 regulation of neutrophil functions is linked to differential regulation of Akt, p38, and p42/44. Our data, therefore, suggests a mechanistic role for Cdc42 activity in primary human neutrophil biology, and identify Cdc42 activity as a target to modulate neutrophil effector mechanisms and killing efficacy.


Asunto(s)
Neutrófilos/inmunología , Proteína de Unión al GTP cdc42/inmunología , Animales , Degranulación de la Célula/inmunología , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Humanos , Ratones
14.
Front Immunol ; 8: 184, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28293240

RESUMEN

The inflammatory microenvironment is commonly characterized by extracellular acidosis (pH < 7.35). Sensitivity to pH, CO2 or bicarbonate concentrations allows neutrophils to react to changes in their environment and to detect inflamed areas in the tissue. One important antimicrobial effector mechanism is the production of neutrophil extracellular traps (NETs), which are released during a programmed reactive oxygen species (ROS)-dependent cell death, the so-called NETosis. Although several functions of neutrophils have been analyzed under acidic conditions, the effect of extracellular acidosis on NETosis remains mainly unexplored and the available experimental results are contradictory. We performed a comprehensive study with the aim to elucidate the effect of extracellular acidosis on ROS-dependent NETosis of primary human neutrophils and to identify the underlying mechanisms. The study was performed in parallel in a CO2-bicabonate-buffered culture medium, which mimics in vivo conditions, and under HEPES-buffered conditions to verify the effect of pH independent of CO2 or bicarbonate. We could clearly show that extracellular acidosis (pH 6.5, 6.0, and 5.5) and intracellular acidification inhibit the release of ROS-dependent NETs upon stimulation of neutrophils with phorbol myristate acetate and immobilized immune complexes. Moreover, our findings suggest that the diminished NET release is a consequence of reduced ROS production and diminished glycolysis of neutrophils under acidic conditions. It was suggested previously that neutrophils can sense the border of inflamed tissue by the pH gradient and that a drop in pH serves as an indicator for the progress of inflammation. Following this hypothesis, our data indicate that an acidic inflammatory environment results in inhibition of extracellular operating effector mechanisms of neutrophils such as release of ROS and NETs. This way the release of toxic components and tissue damage can be avoided. However, we observed that major antimicrobial effector mechanisms such as phagocytosis and the killing of pathogens by neutrophils remain functional under acidic conditions.

15.
J Invest Dermatol ; 136(1): 117-26, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26763431

RESUMEN

Host defense against pathogens relies on neutrophil activation. Inadequate neutrophil activation is often associated with chronic inflammatory diseases. Neutrophils also constitute a significant portion of infiltrating cells in chronic inflammatory diseases, for example, psoriasis and multiple sclerosis. Fumarates improve the latter diseases, which so far has been attributed to the effects on lymphocytes and dendritic cells. Here, we focused on the effects of dimethylfumarate (DMF) on neutrophils. In vitro, DMF inhibited neutrophil activation, including changes in surface marker expression, reactive oxygen species production, formation of neutrophil extracellular traps, and migration. Phagocytic ability and autoantibody-induced, neutrophil-dependent tissue injury ex vivo was also impaired by DMF. Regarding the mode of action, DMF modulates-in a stimulus-dependent manner-neutrophil activation using the phosphoinositide 3-kinase/Akt-p38 mitogen-activated protein kinase and extracellular signal-regulated kinase 1/2 pathways. For in vivo validation, mouse models of epidermolysis bullosa acquisita, an organ-specific autoimmune disease caused by autoantibodies to type VII collagen, were employed. In the presence of DMF, blistering induced by injection of anti-type VII collagen antibodies into mice was significantly impaired. DMF treatment of mice with clinically already-manifested epidermolysis bullosa acquisita led to disease improvement. Collectively, we demonstrate a profound inhibitory activity of DMF on neutrophil functions. These findings encourage wider use of DMF in patients with neutrophil-mediated diseases.


Asunto(s)
Dimetilfumarato/farmacología , Epidermólisis Ampollosa Adquirida/inmunología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Animales , Western Blotting , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Intervalos de Confianza , Modelos Animales de Enfermedad , Epidermólisis Ampollosa Adquirida/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA