Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
mBio ; 13(3): e0081922, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35491830

RESUMEN

The opportunistic bacterium Pseudomonas aeruginosa can infect mucosal tissues of the human body. To persist at the mucosal barrier, this highly adaptable pathogen has evolved many strategies, including invasion of host cells. Here, we show that the P. aeruginosa lectin LecB binds and cross-links fucosylated receptors at the apical plasma membrane of epithelial cells. This triggers a signaling cascade via Src kinases and phosphoinositide 3-kinase (PI3K), leading to the formation of patches enriched with the basolateral marker phosphatidylinositol (3,4,5)-trisphosphate (PIP3) at the apical plasma membrane. This identifies LecB as a causative bacterial factor for activating this well-known host cell response that is elicited upon apical binding of P. aeruginosa. Downstream from PI3K, Rac1 is activated to cause actin rearrangement and the outgrowth of protrusions at the apical plasma membrane. LecB-triggered PI3K activation also results in aberrant recruitment of caveolin-1 to the apical domain. In addition, we reveal a positive feedback loop between PI3K activation and apical caveolin-1 recruitment, which provides a mechanistic explanation for the previously observed implication of caveolin-1 in P. aeruginosa host cell invasion. Interestingly, LecB treatment also reversibly removes primary cilia. To directly prove the role of LecB for bacterial uptake, we coated bacterium-sized beads with LecB, which drastically enhanced their endocytosis. Furthermore, LecB deletion and LecB inhibition with l-fucose diminished the invasion efficiency of P. aeruginosa bacteria. Taken together, the results of our study identify LecB as a missing link that can explain how PI3K signaling and caveolin-1 recruitment are triggered to facilitate invasion of epithelial cells from the apical side by P. aeruginosa. IMPORTANCE An intriguing feature of the bacterium P. aeruginosa is its ability to colonize highly diverse niches. P. aeruginosa can, besides forming biofilms, also enter and proliferate within epithelial host cells. Moreover, research during recent years has shown that P. aeruginosa possesses many different mechanisms to invade host cells. In this study, we identify LecB as a novel invasion factor. In particular, we show that LecB activates PI3K signaling, which is connected via a positive feedback loop to apical caveolin-1 recruitment and leads to actin rearrangement at the apical plasma membrane. This provides a unifying explanation for the previously reported implication of PI3K and caveolin-1 in host cell invasion by P. aeruginosa. In addition, our study adds a further function to the remarkable repertoire of the lectin LecB, which is all brought about by the capability of LecB to recognize fucosylated glycans on many different niche-specific host cell receptors.


Asunto(s)
Lectinas , Pseudomonas aeruginosa , Actinas/metabolismo , Caveolina 1/metabolismo , Membrana Celular/metabolismo , Humanos , Lectinas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Pseudomonas aeruginosa/metabolismo
2.
Cell Mol Life Sci ; 78(24): 8165-8186, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34731252

RESUMEN

B cell superantigens crosslink conserved domains of B cell receptors (BCRs) and cause dysregulated, polyclonal B cell activation irrespective of normal BCR-antigen complementarity. The cells typically succumb to activation-induced cell death, which can impede the adaptive immune response and favor infection. In the present study, we demonstrate that the fucose-binding lectin of Burkholderia ambifaria, BambL, bears functional resemblance to B cell superantigens. By engaging surface glycans, the bacterial lectin activated human peripheral blood B cells, which manifested in the surface expression of CD69, CD54 and CD86 but became increasingly cytotoxic at higher concentrations. The effects were sensitive to BCR pathway inhibitors and excess fucose, which corroborates a glycan-driven mode of action. Interactome analyses in a model cell line suggest BambL binds directly to glycans of the BCR and regulatory coreceptors. In vitro, BambL triggered BCR signaling and induced CD19 internalization and degradation. Owing to the lectin's six binding sites, we propose a BCR activation model in which BambL functions as a clustering hub for receptor glycans, modulates normal BCR regulation, and induces cell death through exhaustive activation.


Asunto(s)
Linfocitos B/metabolismo , Proteínas Bacterianas/metabolismo , Burkholderia/metabolismo , Lectinas/metabolismo , Polisacáridos/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Superantígenos/metabolismo , Linfocitos B/inmunología , Proteínas Bacterianas/inmunología , Sitios de Unión , Humanos , Lectinas/inmunología , Polisacáridos/inmunología , Unión Proteica , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal , Superantígenos/inmunología
3.
mBio ; 11(2)2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32156827

RESUMEN

The opportunistic bacterium Pseudomonas aeruginosa produces the fucose-specific lectin LecB, which has been identified as a virulence factor. LecB has a tetrameric structure with four opposing binding sites and has been shown to act as a cross-linker. Here, we demonstrate that LecB strongly binds to the glycosylated moieties of ß1-integrins on the basolateral plasma membrane of epithelial cells and causes rapid integrin endocytosis. Whereas internalized integrins were degraded via a lysosomal pathway, washout of LecB restored integrin cell surface localization, thus indicating a specific and direct action of LecB on integrins to bring about their endocytosis. Interestingly, LecB was able to trigger uptake of active and inactive ß1-integrins and also of complete α3ß1-integrin-laminin complexes. We provide a mechanistic explanation for this unique endocytic process by showing that LecB has the additional ability to recognize fucose-bearing glycosphingolipids and causes the formation of membrane invaginations on giant unilamellar vesicles. In cells, LecB recruited integrins to these invaginations by cross-linking integrins and glycosphingolipids. In epithelial wound healing assays, LecB specifically cleared integrins from the surface of cells located at the wound edge and blocked cell migration and wound healing in a dose-dependent manner. Moreover, the wild-type P. aeruginosa strain PAO1 was able to loosen cell-substrate adhesion in order to crawl underneath exposed cells, whereas knockout of LecB significantly reduced crawling events. Based on these results, we suggest that LecB has a role in disseminating bacteria along the cell-basement membrane interface.IMPORTANCEPseudomonas aeruginosa is a ubiquitous environmental bacterium that is one of the leading causes of nosocomial infections. P. aeruginosa is able to switch between planktonic, intracellular, and biofilm-based lifestyles, which allows it to evade the immune system as well as antibiotic treatment. Hence, alternatives to antibiotic treatment are urgently required to combat P. aeruginosa infections. Lectins, like the fucose-specific LecB, are promising targets, because removal of LecB resulted in decreased virulence in mouse models. Currently, several research groups are developing LecB inhibitors. However, the role of LecB in host-pathogen interactions is not well understood. The significance of our research is in identifying cellular mechanisms of how LecB facilitates P. aeruginosa infection. We introduce LecB as a new member of the list of bacterial molecules that bind integrins and show that P. aeruginosa can move forward underneath attached epithelial cells by loosening cell-basement membrane attachment in a LecB-dependent manner.


Asunto(s)
Interacciones Huésped-Patógeno , Integrinas/metabolismo , Lectinas/metabolismo , Lectinas/farmacología , Pseudomonas aeruginosa/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Perros , Endocitosis , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células de Riñón Canino Madin Darby , Unión Proteica , Pseudomonas aeruginosa/patogenicidad , Factores de Virulencia/metabolismo
4.
Methods Mol Biol ; 1880: 341-357, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30610709

RESUMEN

Mass spectrometry (MS)-based identification and characterization of protein complexes is becoming a prerequisite for in-depth biochemical analyses of intracellular processes. Here, we describe two state-of-the-art MS-based approaches to characterize protein-protein interactions and multi-protein complexes involved in autophagy in mammalian cells. The combination of affinity purification (AP)-MS, which identifies binary protein-protein interactions, with size-exclusion chromatography (SEC)-protein correlation profiling (PCP), which helps monitor protein complex assemblies, is a powerful tool to acquire a full overview of the interlinkage and regulation of novel multi-protein complexes that might play a role in autophagy.


Asunto(s)
Autofagia/fisiología , Complejos Multiproteicos/metabolismo , Mapeo de Interacción de Proteínas/métodos , Espectrometría de Masas en Tándem/métodos , Autofagia/efectos de los fármacos , Cromatografía de Afinidad/instrumentación , Cromatografía de Afinidad/métodos , Cromatografía en Gel/instrumentación , Cromatografía en Gel/métodos , Humanos , Células MCF-7 , Macrólidos/farmacología , Mapeo de Interacción de Proteínas/instrumentación , Multimerización de Proteína/fisiología , Espectrometría de Masas en Tándem/instrumentación
5.
Cell Tissue Res ; 369(1): 41-51, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28432464

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a common monogenetic disorder that is caused by mutations in the genes PKD1 and PKD2 encoding polycystin-1 and polycystin-2, respectively. Polycystin-1 and -2 form a complex, interact with several proteins involved in signal transduction and localize to discrete subcellular positions, most importantly the primary cilium. Whereas the causative mutations leading to ADPKD are known, the underlying deregulated cellular pathways are not well understood. In the current review, we introduce state-of-the-art mass spectrometry (MS)-based proteomic techniques and summarize their use in kidney and ADPKD research. Proteomic profiling approaches, the elucidation of ADPKD-relevant protein-protein interactions and the regulation of posttranslational modifications are included. We also discuss the use of MS-based methods for ADPKD prognosis, diagnosis and disease monitoring by using protein- and peptide-based biomarkers.


Asunto(s)
Espectrometría de Masas/métodos , Riñón Poliquístico Autosómico Dominante/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Animales , Humanos , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Proteoma/genética
6.
EMBO J ; 36(5): 646-663, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28093501

RESUMEN

As a central element within the RAS/ERK pathway, the serine/threonine kinase BRAF plays a key role in development and homeostasis and represents the most frequently mutated kinase in tumors. Consequently, it has emerged as an important therapeutic target in various malignancies. Nevertheless, the BRAF activation cycle still raises many mechanistic questions as illustrated by the paradoxical action and side effects of RAF inhibitors. By applying SEC-PCP-SILAC, we analyzed protein-protein interactions of hyperactive BRAFV600E and wild-type BRAF (BRAFWT). We identified two macromolecular, cytosolic BRAF complexes of distinct molecular composition and phosphorylation status. Hyperactive BRAFV600E resides in large complexes of higher molecular mass and activity, while BRAFWT is confined to smaller, slightly less active complexes. However, expression of oncogenic K-RasG12V, either by itself or in combination with RAF dimer promoting inhibitors, induces the incorporation of BRAFWT into large, active complexes, whereas pharmacological inhibition of BRAFV600E has the opposite effect. Thus, the quaternary structure of BRAF complexes is shaped by its activation status, the conformation of its kinase domain, and clinically relevant inhibitors.


Asunto(s)
Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas B-raf/química , Proteínas Proto-Oncogénicas B-raf/metabolismo , Animales , Células Cultivadas , Cromatografía en Gel , Humanos , Espectrometría de Masas , Ratones , Fosforilación , Conformación Proteica
7.
J Cell Sci ; 130(2): 382-395, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27909246

RESUMEN

Retromer and the associated actin-polymerizing WASH complex are essential for the endocytic recycling of a wide range of integral membrane proteins. A hereditary Parkinson's-disease-causing point mutation (D620N) in the retromer subunit VPS35 perturbs retromer's association with the WASH complex and also with the uncharacterized protein ankyrin-repeat-domain-containing protein 50 (ANKRD50). Here, we firmly establish ANKRD50 as a new and essential component of the SNX27-retromer-WASH super complex. Depletion of ANKRD50 in HeLa or U2OS cells phenocopied the loss of endosome-to-cell-surface recycling of multiple transmembrane proteins seen upon suppression of SNX27, retromer or WASH-complex components. Mass-spectrometry-based quantification of the cell surface proteome of ANKRD50-depleted cells identified amino acid transporters of the SLC1A family, among them SLC1A4, as additional cargo molecules that depend on ANKRD50 and retromer for their endocytic recycling. Mechanistically, we show that ANKRD50 simultaneously engages multiple parts of the SNX27-retromer-WASH complex machinery in a direct and co-operative interaction network that is needed to efficiently recycle the nutrient transporters GLUT1 (also known as SLC2A1) and SLC1A4, and potentially many other surface proteins.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Mapas de Interacción de Proteínas , Proteínas de Transporte Vesicular/metabolismo , Secuencias de Aminoácidos , Transporte Biológico , Membrana Celular/metabolismo , Endocitosis , Endosomas/metabolismo , Receptores ErbB/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Integrinas/metabolismo , Espectrometría de Masas , Fosfoproteínas Fosfatasas/química , Unión Proteica , Proteolisis , Proteómica , Nexinas de Clasificación/metabolismo , Transferrina/metabolismo , Proteínas de Transporte Vesicular/química
8.
Oncotarget ; 7(18): 26628-52, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27034005

RESUMEN

B-Raf represents a critical physiological regulator of the Ras/RAF/MEK/ERK-pathway and a pharmacological target of growing clinical relevance, in particular in oncology. To understand how B-Raf itself is regulated, we combined mass spectrometry with genetic approaches to map its interactome in MCF-10A cells as well as in B-Raf deficient murine embryonic fibroblasts (MEFs) and B-Raf/Raf-1 double deficient DT40 lymphoma cells complemented with wildtype or mutant B-Raf expression vectors. Using a multi-protease digestion approach, we identified a novel ubiquitination site and provide a detailed B-Raf phospho-map. Importantly, we identify two evolutionary conserved phosphorylation clusters around T401 and S419 in the B-Raf hinge region. SILAC labelling and genetic/biochemical follow-up revealed that these clusters are phosphorylated in the contexts of oncogenic Ras, sorafenib induced Raf dimerization and in the background of the V600E mutation. We further show that the vemurafenib sensitive phosphorylation of the T401 cluster occurs in trans within a Raf dimer. Substitution of the Ser/Thr-residues of this cluster by alanine residues enhances the transforming potential of B-Raf, indicating that these phosphorylation sites suppress its signaling output. Moreover, several B-Raf phosphorylation sites, including T401 and S419, are somatically mutated in tumors, further illustrating the importance of phosphorylation for the regulation of this kinase.


Asunto(s)
Proteínas Proto-Oncogénicas B-raf/química , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Animales , Transformación Celular Neoplásica/química , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Ratones , Fosforilación , Proteómica , Transducción de Señal/fisiología
9.
Life Sci ; 141: 128-36, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26382596

RESUMEN

AIMS: The incidence of secondary systemic fungal infections has sharply increased in bacterial septic patients. Antimycotics exhibit immunomodulatory properties, yet these effects are incompletely understood in secondary systemic fungal infections following bacterial sepsis. We investigated a model of systemic inflammation to determine whether antimycotics (liposomal amphotericin B (L-AMB), itraconazol (ITC), and anidulafungin (ANI)) modulate the gene and protein expression as well as the phagocytic activity of lipopolysaccharide (LPS)-stimulated human monocytes. MAIN METHODS: THP-1 monocytes were incubated with L-AMB, ITC or ANI and LPS. Gene expression levels of cytokines (TNF-, IL-1, IL-6, and IL-10) were measured after 2h, 6h, and 24h. Cytokine protein levels were evaluated after 24h and phagocytic activity was determined following co-incubation with Escherichia coli. KEY FINDINGS: All antimycotics differentially modulated the gene and protein expression of cytokines in sepsis-like conditions. In the presence of LPS, we identified L-AMB as immunosuppressive, whereas ITC demonstrated pro-inflammatory properties. Both compounds induced remarkably less phagocytosis. SIGNIFICANCE: Our study suggests that antimycotics routinely used in septic patients alter the immune response in sepsis-like conditions by modulating cytokine gene and protein expression levels and phagocytic activity. Future treatment strategies should consider the immune status of the host and apply antimycotics accordingly in bacterial septic patients with secondary fungal infections.


Asunto(s)
Antifúngicos/farmacología , Citocinas/biosíntesis , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Fagocitosis/efectos de los fármacos , Sepsis/patología , Anfotericina B/farmacología , Anidulafungina , Células Cultivadas , Equinocandinas/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Inmunosupresores/farmacología , Inflamación/inducido químicamente , Inflamación/patología , Itraconazol/farmacología
10.
Autophagy ; 11(9): 1471-83, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26213203

RESUMEN

Autophagy describes an intracellular process responsible for the lysosome-dependent degradation of cytosolic components. The ULK1/2 complex comprising the kinase ULK1/2 and the accessory proteins ATG13, RB1CC1, and ATG101 has been identified as a central player in the autophagy network, and it represents the main entry point for autophagy-regulating kinases such as MTOR and AMPK. It is generally accepted that the ULK1 complex is constitutively assembled independent of nutrient supply. Here we report the characterization of the ATG13 region required for the binding of ULK1/2. This binding site is established by an extremely short peptide motif at the C terminus of ATG13. This motif is mandatory for the recruitment of ULK1 into the autophagy-initiating high-molecular mass complex. Expression of a ULK1/2 binding-deficient ATG13 variant in ATG13-deficient cells resulted in diminished but not completely abolished autophagic activity. Collectively, we propose that autophagy can be executed by mechanisms that are dependent or independent of the ULK1/2-ATG13 interaction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Autofagia , Mutación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencias de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Homólogo de la Proteína 1 Relacionada con la Autofagia , Estabilidad de Enzimas , Fibroblastos/metabolismo , Proteínas de Choque Térmico/metabolismo , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Péptidos/metabolismo , Fagosomas/metabolismo , Unión Proteica , Proteolisis , Proteína Sequestosoma-1
11.
J Antibiot (Tokyo) ; 68(8): 485-90, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25735844

RESUMEN

Conventional antibiotics exhibit immunomodulatory properties beneficial in the treatment of sepsis. Antibiotic-resistant Gram-positive bacteria have become a problem in sepsis therapy, giving rise to increased use of last-resort antibiotics; for example, linezolid (LIN), vancomycin (VAN) and daptomycin (DAP). As the immunomodulatory properties of these antibiotics in treating sepsis are unknown, this study examined the effect of VAN, LIN and DAP on the immune response under sepsis-like conditions in vitro. Lipopolysaccharide (LPS)-activated THP-1 monocytes were incubated with LIN, VAN or DAP. Gene expression of cytokines (TNFα, IL-1ß, IL-6, IL-10) and Toll-like receptors (TLR1, 2, 4, 6, 7 and 9) was monitored and phagocytosis was determined following coincubation with E. coli. The antibiotics differentially modulated the gene expression of the investigated cytokines. While LIN and VAN upregulated the expression of all TLRs, DAP downregulated mRNA levels of TLR1, TLR2 and TLR6, which recognize pathogen-associated molecular patterns from Gram-positive bacteria. In addition, LIN inhibited, whereas VAN promoted the phagocytic activity of monocytes. Our results suggest that LIN and VAN possess pro-inflammatory properties, whereas DAP might reduce the immune response to Gram-positive bacteria in sepsis. Furthermore, VAN might be beneficial in the prevention of Gram-negative infections by increasing the phagocytosis of E. coli.


Asunto(s)
Antibacterianos/farmacología , Citocinas/metabolismo , Factores Inmunológicos/farmacología , Fagocitosis/efectos de los fármacos , Sepsis/inmunología , Receptores Toll-Like/biosíntesis , Acetamidas/farmacología , Línea Celular , Daptomicina/farmacología , Escherichia coli/inmunología , Perfilación de la Expresión Génica , Bacterias Grampositivas/inmunología , Humanos , Linezolid , Modelos Teóricos , Monocitos/efectos de los fármacos , Monocitos/inmunología , Oxazolidinonas/farmacología , Vancomicina/farmacología
12.
Autophagy ; 10(2): 356-71, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24275748

RESUMEN

Under conditions of nutrient shortage autophagy is the primary cellular mechanism ensuring availability of substrates for continuous biosynthesis. Subjecting cells to starvation or rapamycin efficiently induces autophagy by inhibiting the MTOR signaling pathway triggering increased autophagic flux. To elucidate the regulation of early signaling events upon autophagy induction, we applied quantitative phosphoproteomics characterizing the temporal phosphorylation dynamics after starvation and rapamycin treatment. We obtained a comprehensive atlas of phosphorylation kinetics within the first 30 min upon induction of autophagy with both treatments affecting widely different cellular processes. The identification of dynamic phosphorylation already after 2 min demonstrates that the earliest events in autophagy signaling occur rapidly after induction. The data was subjected to extensive bioinformatics analysis revealing regulated phosphorylation sites on proteins involved in a wide range of cellular processes and an impact of the treatments on the kinome. To approach the potential function of the identified phosphorylation sites we performed a screen for MAP1LC3-interacting proteins and identified a group of binding partners exhibiting dynamic phosphorylation patterns. The data presented here provide a valuable resource on phosphorylation events underlying early autophagy induction.


Asunto(s)
Autofagia/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Línea Celular Tumoral , Humanos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteómica , Inanición/metabolismo , Factores de Tiempo
13.
Int Immunopharmacol ; 18(1): 27-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24239744

RESUMEN

The inflammatory response to pathogen-associated molecular patterns such as lipopolysaccharide (LPS) in sepsis is mediated via Toll-like receptors (TLRs). Since TLRs also trigger various immune functions, including phagocytosis, their modulation is a promising strategy in the treatment of sepsis. As antibiotics have immunomodulatory properties, this study examined the effect of commonly used classes of antibiotics on i) the expression of TLRs and cytokines and ii) the phagocytic activity under sepsis-like conditions in vitro. This was achieved by incubating THP-1 monocytes and peripheral blood mononuclear cells (PBMCs) obtained from patients after open-heart surgery with the addition of LPS and six key antibiotics (piperacillin, doxycycline, erythromycin, moxifloxacin or gentamicin). After 24h, mRNA levels of both cytokines (IL-1ß, IL-6) and TLRs (1, 2, 4, and 6) were monitored and phagocytosis was determined following coincubation with Escherichia coli. Each antibiotic differentially regulated the gene expression of the investigated TLRs and cytokines in monocytes. Erythromycin, moxifloxacin and doxycyclin displayed the strongest effects and changed mRNA-levels of the investigated genes up to 5.6-fold. Consistent with this, antibiotics and, in particular, moxifloxacin, regulated the TLR-and cytokine expression in activated PBMCs obtained from patients after open-heart surgery. Furthermore, piperacillin, doxycyclin and moxifloxacin inhibited the phagocytic activity of monocytes. Our results suggest that antibiotics regulate the immune response by modulating TLR- and cytokine expression as well as phagocytosis under septic conditions. Moxifloxacin, doxycycline and erythromycin were shown to possess the strongest immunomodulatory effects and these antibiotic classes should be considered for future immunomodulatory studies in sepsis.


Asunto(s)
Antibacterianos/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Escherichia coli/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Monocitos/efectos de los fármacos , Sepsis/tratamiento farmacológico , Receptores Toll-Like/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Infecciones por Escherichia coli/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunomodulación , Leucocitos Mononucleares/inmunología , Lipopolisacáridos/inmunología , Monocitos/inmunología , Fagocitosis/efectos de los fármacos , Sepsis/inmunología , Receptores Toll-Like/inmunología
14.
Cell Commun Signal ; 11(1): 30, 2013 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-23607741

RESUMEN

BACKGROUND: The Gab2 docking protein acts as an important signal amplifier downstream of various growth factor receptors and Bcr-Abl, the driver of chronic myeloid leukaemia (CML). Despite the success of Bcr-Abl tyrosine kinase inhibitors (TKI) in the therapy of CML, TKI-resistance remains an unsolved problem in the clinic. We have recently shown that Gab2 signalling counteracts the efficacy of four distinct Bcr-Abl inhibitors. In the course of that project, we noticed that two clinically relevant drugs, imatinib and dasatinib, provoke distinct alterations in the electrophoretic mobility of Gab2, its signalling output and protein interactions. As the signalling potential of the docking protein is highly modulated by its phosphorylation status, we set out to obtain more insights into the impact of TKIs on Gab2 phosphorylation. FINDINGS: Using stable isotope labelling by amino acids in cell culture (SILAC)-based quantitative mass spectrometry (MS), we show now that imatinib and dasatinib provoke distinct effects on the phosphorylation status and interactome of Gab2. This study identifies several new phosphorylation sites on Gab2 and confirms many sites previously known from other experimental systems. At equimolar concentrations, dasatinib is more effective in preventing Gab2 tyrosine and serine/threonine phosphorylation than imatinib. It also affects the phosphorylation status of more residues than imatinib. In addition, we also identify novel components of the Gab2 signalling complex, such as casein kinases, stathmins and PIP1 as well as known interaction partners whose association with Gab2 is disrupted by imatinib and/or dasatinib. CONCLUSIONS: By using MS-based proteomics, we have identified new and confirmed known phosphorylation sites and interaction partners of Gab2, which may play an important role in the regulation of this docking protein. Given the growing importance of Gab2 in several tumour entities we expect that our results will help to understand the complex regulation of Gab2 and how this docking protein can contribute to malignancy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...