Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Sci Rep ; 13(1): 11096, 2023 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-37423933

RESUMEN

Acute kidney injury (AKI) is a common complication after complex aortic procedures and it is associated with relevant mortality and morbidity. Biomarkers for early and specific AKI detection are lacking. The aim of this work is to investigate the reliability of the NephroCheck bedside system for diagnosing stage 3 AKI following open aortic surgery. In this prospective, multicenter, observational study,- https://clinicaltrials.gov/ct2/show/NCT04087161 -we included 45 patients undergoing open thoracoabdominal aortic repair. AKI risk (AKIRisk-Index) was calculated from urine samples at 5 timepoints: baseline, immediately postoperatively and at 12, 24, 48, and 72 h post-surgery. AKIs were classified according to the KDIGO criteria. Contributing factors were identified in univariable and multivariable logistic regression. Predictive ability was assessed with the area under the receiver operator curve (ROCAUC). Among 31 patients (68.8%) that developed AKIs, 21 (44.9%) developed stage-3 AKIs, which required dialysis. AKIs were correlated with increased in-hospital mortality (p = .006), respiratory complications (p < .001), sepsis (p < .001), and multi-organ dysfunction syndrome (p < .001). The AKIRisk-Index showed reliable diagnostic accuracy starting at 24 h post-surgery (ROCAUC: .8056, p = .001). In conclusion, starting at 24 h after open aortic repair, the NephroCheck system showed adequate diagnostic accuracy for detecting the patients at risk for stage 3 AKIs.


Asunto(s)
Lesión Renal Aguda , Diálisis Renal , Humanos , Estudios Prospectivos , Reproducibilidad de los Resultados , Diálisis Renal/efectos adversos , Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/etiología , Biomarcadores/orina
3.
J Clin Invest ; 133(11)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37014703

RESUMEN

Current therapies for Fabry disease are based on reversing intracellular accumulation of globotriaosylceramide (Gb3) by enzyme replacement therapy (ERT) or chaperone-mediated stabilization of the defective enzyme, thereby alleviating lysosomal dysfunction. However, their effect in the reversal of end-organ damage, like kidney injury and chronic kidney disease, remains unclear. In this study, ultrastructural analysis of serial human kidney biopsies showed that long-term use of ERT reduced Gb3 accumulation in podocytes but did not reverse podocyte injury. Then, a CRISPR/Cas9-mediated α-galactosidase knockout podocyte cell line confirmed ERT-mediated reversal of Gb3 accumulation without resolution of lysosomal dysfunction. Transcriptome-based connectivity mapping and SILAC-based quantitative proteomics identified α-synuclein (SNCA) accumulation as a key event mediating podocyte injury. Genetic and pharmacological inhibition of SNCA improved lysosomal structure and function in Fabry podocytes, exceeding the benefits of ERT. Together, this work reconceptualizes Fabry-associated cell injury beyond Gb3 accumulation, and introduces SNCA modulation as a potential intervention, especially for patients with Fabry nephropathy.


Asunto(s)
Enfermedad de Fabry , Podocitos , Humanos , Podocitos/patología , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Enfermedad de Fabry/genética , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/patología , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo , alfa-Galactosidasa/uso terapéutico , Riñón/metabolismo , Trihexosilceramidas/metabolismo , Trihexosilceramidas/farmacología , Trihexosilceramidas/uso terapéutico
4.
Front Cell Dev Biol ; 11: 1076250, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36846596

RESUMEN

Short-chain fatty acids (SCFAs) like butyrate (BUT) largely influence vascular integrity and are closely associated with the onset and progression of cardiovascular diseases. However, their impact on vascular endothelial cadherin (VEC), a major vascular adhesion and signaling molecule, is largely unknown. Here, we explored the effect of the SCFA BUT on the phosphorylation of specific tyrosine residues of VEC (Y731, Y685, and Y658), which are reported to be critical for VEC regulation and vascular integrity. Moreover, we shed light on the signaling pathway engaged by BUT to affect the phosphorylation of VEC. Thereby, we used phospho-specific antibodies to evaluate the phosphorylation of VEC in response to the SCFA sodium butyrate in human aortic endothelial cells (HAOECs) and performed dextran assays to analyze the permeability of the EC monolayer. The role of c-Src and SCFA receptors FFAR2 and FFAR3 in the induction of VEC phosphorylation was analyzed using inhibitors and antagonists for c-Src family kinases and FFAR2/3, respectively, as well as by RNAi-mediated knockdown. Localization of VEC in response to BUT was assessed by fluorescence microscopy. BUT treatment of HAOEC resulted in the specific phosphorylation of Y731 at VEC with minor effects on Y685 and Y658. Thereby, BUT engages FFAR3, FFAR2, and c-Src kinase to induce phosphorylation of VEC. VEC phosphorylation correlated with enhanced endothelial permeability and c-Src-dependent remodeling of junctional VEC. Our data suggest that BUT, an SCFA and gut microbiota-derived metabolite, impacts vascular integrity by targeting VEC phosphorylation with potential impact on the pathophysiology and therapy of vascular diseases.

5.
Diagnostics (Basel) ; 12(10)2022 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-36292213

RESUMEN

The aim of this study was to compare the mortality rates, re-intervention rates, and volumetric changes in aortas following surgery, in terms of the true lumen and false lumen changes, using conventional hemi-arch repair (CET) and frozen elephant trunk (FET) techniques. During the period from 2015 to 2018, 66 patients underwent surgical treatment for acute aortic dissection (Debakey type 1). Demographic and procedure-related data were evaluated. We measured volumetric change before surgical treatment, at discharge, and at 12- and 24-month time points based on computed tomography angiography. The study cohort was divided into two groups (FET vs. CET). The mean age of the patients was 56.9 ± 9.4 years in the FET group versus 63.6 ± 11 years in the CET group (p = 0.063). The mean follow-up time was 24 ± 6 and 25 ± 5 months for the FET and CET groups, respectively. There were no significant differences between the two groups in terms of the medical histories of the cohorts. The results showed a significant increase in true lumen volume after the FET procedure (within 24 months postoperatively; p = 0.005), and no significant changes in total (p = 0.392) or false lumen (p = 0.659) volumes were noted. After the CET procedure, there were significant increases in total and false lumen volumes (p = 0.013, p = 0.042), while no significant change in true lumen was observed (p = 0.219). The volume increase in true lumen after the FET procedure was higher compared to the CET group at all postoperative time points (at discharge, 12 months, and 24 months) without significant evidence (p = 0.416, p = 0.422, p = 0.268). At two years, the volume increase in false lumen was significantly higher among the CET group compared to the FET group (p = 0.02). The Kaplan-Meier curve analysis showed that patients who underwent the CET procedure underwent significantly more re-interventions due to false lumen expansion of the descending aorta (p = 0.047). Present study results indicate that the true and false lumen changes in the aorta following the FET and CET procedures were different. FET led to a significant increase in true lumen volume, while false lumen volume remained stable; however, after the CET procedure, significant false lumen enlargement was noted at mid-term follow-up time points. The re-intervention rate after CET was higher due to false lumen expansion.

6.
Cell Rep ; 41(3): 111510, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36261008

RESUMEN

Septin GTPases polymerize into higher-ordered structures as a part of the cytoskeleton and are involved in interactions of the host with a wide spectrum of pathogens. Many pathogens foster an ambiguous relationship with septins. They exploit septins for uptake, but septins also prevent their intracellular replication and target them for autophagy. We demonstrate that septins are involved in a defense mechanism against the pathogen Pseudomonas aeruginosa, which enters cells via a lipid zippering mechanism relying on interaction of the lectin LecA with the glycosphingolipid Gb3 on the host membrane. LecA-dependent invagination of the plasma membrane triggers septin recruitment to the site of bacterial attachment. We also find a septin-dependent reinforcement of cortical actin at attachment sites. Atomic force microscopy reveals formation of a septin-dependent rigid barrier below the membrane, preventing bacterial penetration. Our data suggest that septin barriers represent a cellular defense against bacteria inducing membrane curvature for invasion.


Asunto(s)
Pseudomonas aeruginosa , Septinas , Animales , Septinas/metabolismo , Pseudomonas aeruginosa/metabolismo , Actinas/metabolismo , Glicoesfingolípidos/metabolismo , Lectinas/metabolismo , Mamíferos/metabolismo
7.
J Thorac Dis ; 13(7): 4311-4321, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34422358

RESUMEN

BACKGROUND: Post-implantation syndrome (PIS) is defined as non-infectious continuous fever and a concomitant rise in inflammatory markers shortly after endovascular aortic repair. PIS occurrence after hybrid procedures, such as the frozen elephant trunk (FET) technique, has not been adequately investigated. The current study aims to define the incidence of PIS after the FET and to identify possible risk factors associated with its occurrence. METHODS: The clinical charts of 59 patients undergoing the FET between February 2015 and April 2020 were reviewed retrospectively. The occurrence of PIS was defined as the presence of fever (>38 °C lasting longer than one day during the hospitalisation) and leucocytosis (white blood cell count >12,000/µL). Patients with concomitant conditions possibly leading to fever and/or leucocytosis were excluded. Beside demographic and procedure-related data, serum/plasma inflammatory markers were evaluated before surgery and daily up to seven days postoperatively. Computed tomography scans (CT) were examined to calculate the volume of pre-existent and new-onset mural thrombus after the FET. RESULTS: Thirty-eight patients met the inclusion criteria. The study cohort was divided into two groups based on the occurrence of PIS (17 cases; 44.7%). Patients with PIS were significantly younger than those without PIS (53.5±8.9 vs. 62.5±9.6 years; P=0.005). Female patients were less likely to develop PIS (5.2% vs. 26.3%, P=0.018). Patients with PIS had a higher volume of new-onset thrombus in the postoperative CT (P<0.001). Patients treated for post-dissection aneurysm had, postoperatively, significantly more thrombus material developed in a false lumen (P=0.02). Among the PIS markers, CRP (C-reactive protein) levels on the third postoperative day were independently associated with the volume of new-onset thrombus (P=0.011). After multivariate analysis, the volume of new-onset thrombus (P=0.028) and age (P=0.036) remained the variable associated with a statistically significant increased incidence of PIS. CONCLUSIONS: PIS can occur after the frozen elephant trunk procedure. The volume of new-onset thrombus seems to be associated with an increased incidence of PIS. These findings need to be confirmed in larger patient cohorts.

8.
Cell Mol Life Sci ; 78(7): 3637-3656, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33555391

RESUMEN

The opportunistic pathogen Pseudomonas aeruginosa has gained precedence over the years due to its ability to develop resistance to existing antibiotics, thereby necessitating alternative strategies to understand and combat the bacterium. Our previous work identified the interaction between the bacterial lectin LecA and its host cell glycosphingolipid receptor globotriaosylceramide (Gb3) as a crucial step for the engulfment of P. aeruginosa via the lipid zipper mechanism. In this study, we define the LecA-associated host cell membrane domain by pull-down and mass spectrometry analysis. We unraveled a predilection of LecA for binding to saturated, long fatty acyl chain-containing Gb3 species in the extracellular membrane leaflet and an induction of dynamic phosphatidylinositol (3,4,5)-trisphosphate (PIP3) clusters at the intracellular leaflet co-localizing with sites of LecA binding. We found flotillins and the GPI-anchored protein CD59 not only to be an integral part of the LecA-interacting membrane domain, but also majorly influencing bacterial invasion as depletion of either of these host cell proteins resulted in about 50% reduced invasiveness of the P. aeruginosa strain PAO1. In summary, we report that the LecA-Gb3 interaction at the extracellular leaflet induces the formation of a plasma membrane domain enriched in saturated Gb3 species, CD59, PIP3 and flotillin thereby facilitating efficient uptake of PAO1.


Asunto(s)
Antígenos CD59/metabolismo , Membrana Celular/metabolismo , Interacciones Huésped-Patógeno , Pulmón/microbiología , Proteínas de la Membrana/metabolismo , Pseudomonas aeruginosa/aislamiento & purificación , Trihexosilceramidas/metabolismo , Transporte Biológico , Antígenos CD59/genética , Endocitosis , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Pulmón/metabolismo , Pulmón/patología , Proteínas de la Membrana/genética , Pseudomonas aeruginosa/fisiología , Transducción de Señal
9.
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
10.
Nat Commun ; 10(1): 4644, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604911

RESUMEN

In mammalian cells, the internal and external leaflets of the plasma membrane (PM) possess different phospholipids. Phosphatidylserine (PS) is normally confined to the inner (cytoplasmic) membrane leaflet. Here we report that the adhesin CPn0473 of the human pathogenic bacterium Chlamydia pneumoniae (Cpn) binds to the PM of human cells and induces PS externalization but unexpectedly not apoptosis. PS externalization is increased in human cells exposed to infectious Cpn cells expressing increased CPn0473 and reduced in exposure to Cpn expressing decreased CPn0473. CPn0473 binds specifically to synthetic membranes carrying PS and stimulates pore formation. Asymmetric giant unilamellar vesicles (GUVs) in which PS is restricted to the inner leaflet reveal that CPn0473 induces PS externalization in the absence of other proteins. Thus our identification of CPn0473 as a bacterial PS translocator capable of specific and apoptosis-independent PS externalization during infection extends the spectrum of mechanisms intracellular pathogens use to enter host cells.


Asunto(s)
Adhesinas Bacterianas/fisiología , Chlamydophila pneumoniae/fisiología , Fosfatidilserinas/metabolismo , Adhesinas Bacterianas/metabolismo , Interacciones Microbiota-Huesped , Humanos , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo
11.
Angew Chem Int Ed Engl ; 56(24): 6762-6766, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28504473

RESUMEN

Pathogens frequently rely on lectins for adhesion and cellular entry into the host. Since these interactions typically result from multimeric binding of lectins to cell-surface glycans, novel therapeutic strategies are being developed with the use of glycomimetics as competitors of such interactions. Herein we study the benefit of nucleic acid based oligomeric assemblies with PNA-fucose conjugates. We demonstrate that the interactions of a lectin with epithelial cells can be inhibited with conjugates that do not form stable assemblies in solution but benefit from cooperativity between ligand-protein interactions and PNA hybridization to achieve high affinity. A dynamic dimeric assembly fully blocked the binding of the fucose-binding lectin BambL of Burkholderia ambifaria, a pathogenic bacterium, to epithelial cells with an efficiency of more than 700-fold compared to l-fucose.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Lectinas/metabolismo , Polisacáridos/química , Biomimética , Burkholderia/metabolismo , Línea Celular Tumoral , Humanos , Ácidos Nucleicos de Péptidos/metabolismo , Unión Proteica , Ralstonia solanacearum/metabolismo
12.
Biochim Biophys Acta Mol Cell Res ; 1864(7): 1236-1245, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28428058

RESUMEN

The human pathogen Pseudomonas aeruginosa induces phosphorylation of the adaptor protein CrkII by activating the non-receptor tyrosine kinase Abl to promote its uptake into host cells. So far, specific factors of P. aeruginosa, which induce Abl/CrkII signalling, are entirely unknown. In this research, we employed human lung epithelial cells H1299, Chinese hamster ovary cells and P. aeruginosa wild type strain PAO1 to study the invasion process of P. aeruginosa into host cells by using microbiological, biochemical and cell biological approaches such as Western Blot, immunofluorescence microscopy and flow cytometry. Here, we demonstrate that the host glycosphingolipid globotriaosylceramide, also termed Gb3, represents a signalling receptor for the P. aeruginosa lectin LecA to induce CrkII phosphorylation at tyrosine 221. Alterations in Gb3 expression and LecA function correlate with CrkII phosphorylation. Interestingly, phosphorylation of CrkIIY221 occurs independently of Abl kinase. We further show that Src family kinases transduce the signal induced by LecA binding to Gb3, leading to CrkY221 phosphorylation. In summary, we identified LecA as a bacterial factor, which utilizes a so far unrecognized mechanism for phospho-CrkIIY221 induction by binding to the host glycosphingolipid receptor Gb3. The LecA/Gb3 interaction highlights the potential of glycolipids to mediate signalling processes across the plasma membrane and should be further elucidated to gain deeper insights into this non-canonical mechanism of activating host cell processes.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Globósidos/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Pseudomonas aeruginosa/patogenicidad , Transducción de Señal , Trihexosilceramidas/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Interacciones Huésped-Patógeno , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Pseudomonas aeruginosa/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Familia-src Quinasas/metabolismo
13.
Expert Opin Drug Deliv ; 14(2): 141-153, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27935765

RESUMEN

OBJECTIVES: Epithelial cell layers as well as endothelia forming the blood-brain barrier can drastically reduce the efficiency of drug targeting. Our goal was to investigate lectins recognizing the glycosphingolipid globotriaosylceramide (Gb3) for their potential as carriers for transcytotic drug delivery. METHODS: We utilized an in vitro model based on Madin-Darby canine kidney cells transfected with Gb3 synthase to characterize transcytosis of the Gb3-binding lectins LecA from Pseudomonas aeruginosa and the B-subunit of Shiga toxin (StxB). RESULTS: Both lectins were rapidly transcytosed from the apical to the basolateral plasma membrane and vice versa. Whereas StxB proceeded on retrograde and transcytotic routes, LecA avoided retrograde transport. This differential trafficking could be explained by our observation that LecA and StxB segregated into different domains during endocytosis. Furthermore, inhibiting the small GTPase Rab11a, which organizes trafficking through apical recycling endosomes, blocked basolateral to apical transcytosis of both lectins. CONCLUSIONS: Gb3-binding lectins are promising candidates for transcytotic drug delivery. Our findings highlight that LecA and StxB, which both bind Gb3 but exhibit dissimilar valence and molecular structures of their carbohydrate binding sites and can take divergent intracellular trafficking routes. This opens up the possibility of developing tailor-made glycosphingolipid-binding carrier lectins, which take optimized trafficking pathways.


Asunto(s)
Sistemas de Liberación de Medicamentos , Células Epiteliales/metabolismo , Lectinas/metabolismo , Trihexosilceramidas/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Línea Celular , Membrana Celular/metabolismo , Perros , Endocitosis , Células de Riñón Canino Madin Darby , Transporte de Proteínas
14.
Biochim Biophys Acta ; 1863(6 Pt A): 1106-18, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26862060

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen that induces severe lung infections such as ventilator-associated pneumonia and acute lung injury. Under these conditions, the bacterium diminishes epithelial integrity and inhibits tissue repair mechanisms, leading to persistent infections. Understanding the involved bacterial virulence factors and their mode of action is essential for the development of new therapeutic approaches. In our study we discovered a so far unknown effect of the P. aeruginosa lectin LecB on host cell physiology. LecB alone was sufficient to attenuate migration and proliferation of human lung epithelial cells and to induce transcriptional activity of NF-κB. These effects are characteristic of impaired tissue repair. Moreover, we found a strong degradation of ß-catenin, which was partially recovered by the proteasome inhibitor lactacystin. In addition, LecB induced loss of cell-cell contacts and reduced expression of the ß-catenin targets c-myc and cyclin D1. Blocking of LecB binding to host cell plasma membrane receptors by soluble l-fucose prevented these changes in host cell behavior and signaling, and thereby provides a powerful strategy to suppress LecB function. Our findings suggest that P. aeruginosa employs LecB as a virulence factor to induce ß-catenin degradation, which then represses processes that are directly linked to tissue recovery.


Asunto(s)
Proteínas Bacterianas/farmacología , Células Epiteliales/efectos de los fármacos , Lectinas/farmacología , beta Catenina/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Proteínas Bacterianas/genética , Western Blotting , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Células Epiteliales/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Integrina beta1/metabolismo , Lectinas/genética , Microscopía Confocal , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Factor de Transcripción ReIA/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
15.
Proc Natl Acad Sci U S A ; 111(35): 12895-900, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25136128

RESUMEN

Glycosphingolipids are important structural constituents of cellular membranes. They are involved in the formation of nanodomains ("lipid rafts"), which serve as important signaling platforms. Invasive bacterial pathogens exploit these signaling domains to trigger actin polymerization for the bending of the plasma membrane and the engulfment of the bacterium--a key process in bacterial uptake. However, it is unknown whether glycosphingolipids directly take part in the membrane invagination process. Here, we demonstrate that a "lipid zipper," which is formed by the interaction between the bacterial surface lectin LecA and its cellular receptor, the glycosphingolipid Gb3, triggers plasma membrane bending during host cell invasion of the bacterium Pseudomonas aeruginosa. In vitro experiments with Gb3-containing giant unilamellar vesicles revealed that LecA/Gb3-mediated lipid zippering was sufficient to achieve complete membrane engulfment of the bacterium. In addition, theoretical modeling elucidated that the adhesion energy of the LecA-Gb3 interaction is adequate to drive the engulfment process. In cellulo experiments demonstrated that inhibition of the LecA/Gb3 lipid zipper by either lecA knockout, Gb3 depletion, or application of soluble sugars that interfere with LecA binding to Gb3 significantly lowered P. aeruginosa uptake by host cells. Of note, membrane engulfment of P. aeruginosa occurred independently of actin polymerization, thus corroborating that lipid zippering alone is sufficient for this crucial first step of bacterial host-cell entry. Our study sheds new light on the impact of glycosphingolipids in the cellular invasion of bacterial pathogens and provides a mechanistic explication of the initial uptake processes.


Asunto(s)
Actinas/metabolismo , Glicoesfingolípidos/metabolismo , Microdominios de Membrana/microbiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/metabolismo , Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana/fisiología , Membrana Celular/metabolismo , Membrana Celular/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Glucolípidos/metabolismo , Membrana Dobles de Lípidos/metabolismo , Microdominios de Membrana/metabolismo , Modelos Biológicos , Transducción de Señal/fisiología , Esfingolípidos/metabolismo
16.
Angew Chem Int Ed Engl ; 53(34): 8885-9, 2014 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-25044671

RESUMEN

Lectin LecA is a virulence factor of Pseudomonas aeruginosa involved in lung injury, mortality, and cellular invasion. Ligands competing with human glycoconjugates for LecA binding are thus promising candidates to counteract P. aeruginosa infections. We have identified a novel divalent ligand from a focused galactoside(Gal)-conjugate array which binds to LecA with very high affinity (Kd = 82 nM). Crystal structures of LecA complexed with the ligand together with modeling studies confirmed its ability to chelate two binding sites of LecA. The ligand lowers cellular invasiveness of P. aeruginosa up to 90 % when applied in the range of 0.05-5 µM. Hence, this ligand might lead to the development of drugs against P. aeruginosa infection.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Galactósidos/química , Pseudomonas aeruginosa/patogenicidad , Interacciones Huésped-Patógeno , Ligandos , Pseudomonas aeruginosa/metabolismo
17.
J Am Chem Soc ; 134(49): 20013-6, 2012 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-23186060

RESUMEN

The photoreduction of azide-based immolative linker by Ru(II) conjugates to uncage rhodamine was achieved using different oligomeric protein templates. The generality of the approach was validated with three sets of ligand having varying affinity to their target (biotin, desthiobiotin and raloxifene). The reaction rates of the templated reaction was found to be at least 30-fold faster than the untemplated reaction providing a clear fluorescent signal in response to the protein oligomer within 30 min. The templated reaction was found to also proceed in cellulo and could be used to identify acetyl coenzyme A carboxylase (ACC) in Pseudomonas aeruginosa and human cell lines as well the and estrogen receptor (ER).


Asunto(s)
Azidas/química , Compuestos Organometálicos/química , Proteínas/química , Rodaminas/química , Rutenio/química , Acetil-CoA Carboxilasa/metabolismo , Biotina/análogos & derivados , Biotina/química , Línea Celular , Humanos , Células MCF-7 , Estructura Molecular , Oxidación-Reducción , Procesos Fotoquímicos , Pseudomonas aeruginosa/enzimología , Clorhidrato de Raloxifeno/química , Receptores de Estrógenos/química , Receptores de Estrógenos/metabolismo
18.
PLoS Pathog ; 6(9): e1001099, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20844577

RESUMEN

Influenza A viruses (IAV) bind to sialic-acids at cellular surfaces and enter cells by using endocytotic routes. There is evidence that this process does not occur constitutively but requires induction of specific cellular signals, including activation of PI3K that promotes virus internalization. This implies engagement of cellular signaling receptors during viral entry. Here, we present first indications for an interplay of IAV with receptor tyrosine kinases (RTKs). As representative RTK family-members the epidermal growth factor receptor (EGFR) and the c-Met receptor were studied. Modulation of expression or activity of both RTKs resulted in altered uptake of IAV, showing that these receptors transmit entry relevant signals upon virus binding. More detailed studies on EGFR function revealed that virus binding lead to clustering of lipid-rafts, suggesting that multivalent binding of IAV to cells induces a signaling platform leading to activation of EGFR and other RTKs that in turn facilitates IAV uptake.


Asunto(s)
Membrana Celular/virología , Receptores ErbB/metabolismo , Virus de la Influenza A/fisiología , Gripe Humana/virología , Infecciones por Orthomyxoviridae/virología , Animales , Transporte Biológico , Western Blotting , Células Cultivadas , Perros , Endocitosis , Receptores ErbB/genética , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Gripe Humana/metabolismo , Gripe Humana/patología , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Transducción de Señal
19.
Microbes Infect ; 12(1): 81-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19782761

RESUMEN

Pathogens such as influenza A viruses (IAV) have to overcome a number of barriers defined and maintained by the host, to successfully establish an infection. One of the initial barriers is collectively characterized as the innate immune system. This is a broad anti-pathogen defense program that ranges from the action of natural killer cells to the induction of an antiviral cytokine response. In this article we will focus on new developments and discoveries concerning the interaction of IAV with the cellular innate immune signaling. We discuss new mechanisms of interference of IAV with the pathogen recognition receptor RIG-I and the type I IFN antagonist NS1 in the background of already known and established concepts. Further we summarize progress related to recently identified IFN induced proteins and the role of RNA interference in the context of IAV infection.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Virus de la Influenza A/inmunología , Transducción de Señal , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/antagonistas & inhibidores , Humanos , Interferón Tipo I/antagonistas & inhibidores , Receptores Inmunológicos , Proteínas no Estructurales Virales/inmunología , Factores de Virulencia/inmunología
20.
Biol Chem ; 390(5-6): 509-15, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19335204

RESUMEN

The uptake of influenza A viruses (IAV) into cells represents an attractive antiviral drug target, e.g., by interfering with essential cellular or viral entry factors. So far, this process could only be studied by time-consuming microscopical methods. Thus, there is a lack of rapid and easy assay systems to monitor viral entry. Here, we describe a rapid procedure to analyse internalisation of IAV via Western blot detection of virion-associated matrix protein (M1), the most abundant protein within the viral particle. The assay is broadly applicable and detects different virus strains of various subtypes. As a proof of principle, treatment of cells with various known or presumed entry inhibitors resulted in reduced M1 levels. Removal of sialic acids, the receptors for IAV, led to a complete loss of the M1 signal, indicating that virus internalisation can be monitored already at the stage of attachment. Prevention of endosomal acidification resulted in a delayed degradation of M1 indicative of IAV particles trapped in endosomes. Thus, early detection of the virus-associated M1 protein is a rapid method to monitor different steps of influenza virus internalisation and has potential for application as a screening method for drugs that interfere with the uptake of IAV.


Asunto(s)
Western Blotting/métodos , Virus de la Influenza A/fisiología , Gripe Humana/diagnóstico , Proteínas de la Matriz Viral/análisis , Internalización del Virus , Animales , Línea Celular , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Subtipo H7N7 del Virus de la Influenza A/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...