Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 34(1): 1665-1678, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914676

RESUMEN

Bacterial infectious diseases can lead to death or to serious illnesses. These outcomes are partly the consequence of pore-forming toxins, which are secreted by the pathogenic bacteria (eg, pneumolysin of Streptococcus pneumoniae). Pneumolysin binds to cholesterol within the plasma membrane of host cells and assembles to form trans-membrane pores, which can lead to Ca2+ influx and cell death. Membrane repair mechanisms exist that limit the extent of damage. Immune cells which are essential to fight bacterial infections critically rely on survival mechanisms after detrimental pneumolysin attacks. This study investigated the susceptibility of different immune cell types to pneumolysin. As a model system, we used the lymphoid T-cell line Jurkat, and myeloid cell lines U937 and THP-1. We show that Jurkat T cells are highly susceptible to pneumolysin attack. In contrast, myeloid THP-1 and U937 cells are less susceptible to pneumolysin. In line with these findings, human primary T cells are shown to be more susceptible to pneumolysin attack than monocytes. Differences in susceptibility to pneumolysin are due to (I) preferential binding of pneumolysin to Jurkat T cells and (II) cell type specific plasma membrane repair capacity. Myeloid cell survival is mostly dependent on Ca2+ induced expelling of damaged plasma membrane areas as microvesicles. Thus, in myeloid cells, first-line defense cells in bacterial infections, a potent cellular repair machinery ensures cell survival after pneumolysin attack. In lymphoid cells, which are important at later stages of infections, less efficient repair mechanisms and enhanced toxin binding renders the cells more sensitive to pneumolysin.


Asunto(s)
Toxinas Bacterianas/metabolismo , Estructuras de la Membrana Celular/metabolismo , Estructuras de la Membrana Celular/patología , Membrana Celular/metabolismo , Membrana Celular/patología , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/patología , Calcio/metabolismo , Muerte Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Humanos , Células Jurkat , Monocitos/metabolismo , Monocitos/patología , Células Mieloides/metabolismo , Células Mieloides/patología , Streptococcus pneumoniae/patogenicidad , Células THP-1 , Células U937
2.
FASEB J ; 33(1): 275-285, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29979630

RESUMEN

Bacterial pore-forming toxins compromise plasmalemmal integrity, leading to Ca2+ influx, leakage of the cytoplasm, and cell death. Such lesions can be repaired by microvesicular shedding or by the endocytic uptake of the injured membrane sites. Cells have at their disposal an entire toolbox of repair proteins for the identification and elimination of membrane lesions. Sphingomyelinases catalyze the breakdown of sphingomyelin into ceramide and phosphocholine. Sphingomyelin is predominantly localized in the outer leaflet, where it is hydrolyzed by acid sphingomyelinase (ASM) after lysosomal fusion with the plasma membrane. The magnesium-dependent neutral sphingomyelinase (NSM)-2 is found at the inner leaflet of the plasmalemma. Because either sphingomyelinase has been ascribed a role in the cellular stress response, we investigated their role in plasma membrane repair and cellular survival after treatment with the pore-forming toxins listeriolysin O (LLO) or pneumolysin (PLY). Jurkat T cells, in which ASM or NSM-2 was down-regulated [ASM knockdown (KD) or NSM-2 KD cells], showed inverse reactions to toxin-induced membrane damage: ASM KD cells displayed reduced toxin resistance, decreased viability, and defects in membrane repair. In contrast, the down-regulation of NSM-2 led to an increase in viability and enhanced plasmalemmal repair. Yet, in addition to the increased plasmalemmal repair, the enhanced toxin resistance of NSM-2 KD cells also appeared to be dependent on the activation of p38/MAPK, which was constitutively activated, whereas in ASM KD cells, the p38/MAPK activation was constitutively blunted.-Schoenauer, R., Larpin, Y., Babiychuk, E. B., Drücker, P., Babiychuk, V. S., Avota, E., Schneider-Schaulies, S., Schumacher, F., Kleuser, B., Köffel, R., Draeger, A. Down-regulation of acid sphingomyelinase and neutral sphingomyelinase-2 inversely determines the cellular resistance to plasmalemmal injury by pore-forming toxins.


Asunto(s)
Toxinas Bacterianas/farmacología , Membrana Celular/metabolismo , Proteínas de Choque Térmico/farmacología , Proteínas Hemolisinas/farmacología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Estreptolisinas/farmacología , Proteínas Bacterianas/farmacología , Transporte Biológico , Sistemas CRISPR-Cas , Calcio/metabolismo , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Supervivencia Celular , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/efectos de los fármacos , Micropartículas Derivadas de Células/metabolismo , Humanos , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(8): 795-805, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29679741

RESUMEN

Nucleated cells eliminate lesions induced by bacterial pore-forming toxins, such as pneumolysin via shedding patches of damaged plasmalemma into the extracellular milieu. Recently, we have shown that the majority of shed pneumolysin is present in the form of inactive pre-pores. This finding is surprising considering that shedding is triggered by Ca2+-influx following membrane perforation and therefore is expected to positively discriminate for active pores versus inactive pre-pores. Here we provide evidence for the existence of plasmalemmal domains that are able to attract pneumolysin at high local concentrations. Within such a domain an immediate plasmalemmal perforation induced by a small number of pneumolysin pores would be capable of triggering the elimination of a large number of not yet active pre-pores/monomers and thus pre-empt more frequent and perilous perforation events. Our findings provide further insights into the functioning of the cellular repair machinery which benefits from an inhomogeneous plasmalemmal distribution of pneumolysin.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Membrana Dobles de Lípidos/metabolismo , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/fisiología , Proteínas Bacterianas/metabolismo , Derrame de Bacterias/inmunología , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/microbiología , Colesterol/metabolismo , Células HEK293 , Humanos , Microscopía Intravital , Membrana Dobles de Lípidos/inmunología , Microfluídica , Infecciones Neumocócicas/microbiología , Estreptolisinas/metabolismo
4.
Semin Cell Dev Biol ; 45: 39-47, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26481974

RESUMEN

The perforation of the plasmalemma by pore-forming toxins causes an influx of Ca(2+) and an efflux of cytoplasmic constituents. In order to ensure survival, the cell needs to identify, plug and remove lesions from its membrane. Quarantined by membrane folds and isolated by membrane fusion, the pores are removed from the plasmalemma and expelled into the extracellular space. Outward vesiculation and microparticle shedding seem to be the strategies of choice to eliminate toxin-perforated membrane regions from the plasmalemma of host cells. Depending on the cell type and the nature of injury, the membrane lesion can also be taken up by endocytosis and degraded internally. Host cells make excellent use of an initial, moderate rise in intracellular [Ca(2+)], which triggers containment of the toxin-inflicted damage and resealing of the damaged plasmalemma. Additional Ca(2+)-dependent defensive cellular actions range from the release of effector molecules in order to warn neighbouring cells, to the activation of caspases for the initiation of apoptosis in order to eliminate heavily damaged, dysregulated cells. Injury to the plasmalemma by bacterial toxins can be prevented by the early sequestration of bacterial toxins. Artificial liposomes can act as a decoy system preferentially binding and neutralizing bacterial toxins.


Asunto(s)
Toxinas Bacterianas/farmacología , Membrana Celular/fisiología , Animales , Anexinas/fisiología , Señalización del Calcio , Supervivencia Celular/efectos de los fármacos , Micropartículas Derivadas de Células/fisiología , Endocitosis , Humanos
5.
Biochim Biophys Acta ; 1860(11 Pt A): 2498-2509, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27481675

RESUMEN

BACKGROUND: Streptococcus pneumoniae is a potent human pathogen. Its pore-forming exotoxin pneumolysin is instrumental for breaching the host's epithelial barrier and for the incapacitation of the immune system. METHODS AND RESULTS: Using a combination of life imaging and cryo-electron microscopy we show that pneumolysin, released by cultured bacteria, is capable of permeabilizing the plasmalemma of host cells. However, such permeabilization does not lead to cell lysis since pneumolysin is actively removed by the host cells. The process of pore elimination starts with the formation of pore-bearing plasmalemmal nanotubes and proceeds by the shedding of pores that are embedded in the membrane of released microvesicles. Pneumolysin prepores are likewise removed. The protein composition of the toxin-induced microvesicles, assessed by mass spectrometry, is suggestive of a Ca(2+)-triggered mechanism encompassing the proteins of the annexin family and members of the endosomal sorting complex required for transport (ESCRT) complex. CONCLUSIONS: S. pneumoniae releases sufficient amounts of pneumolysin to perforate the plasmalemma of host cells, however, the immediate cell lysis, which is frequently reported as a result of treatment with purified and artificially concentrated toxin, appears to be an unlikely event in vivo since the toxin pores are efficiently eliminated by microvesicle shedding. Therefore the dysregulation of cellular homeostasis occurring as a result of transient pore formation/elimination should be held responsible for the damaging toxin action. GENERAL SIGNIFICANCE: We have achieved a comprehensive view of a general plasma membrane repair mechanism after injury by a major bacterial toxin.


Asunto(s)
Membrana Celular/ultraestructura , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/farmacología , Proteínas Bacterianas/farmacología , Proteínas Bacterianas/toxicidad , Membrana Celular/efectos de los fármacos , Membrana Celular/microbiología , Permeabilidad de la Membrana Celular , Células HEK293 , Células HeLa , Humanos , Estreptolisinas/toxicidad
6.
Biochim Biophys Acta ; 1853(9): 2045-54, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25219550

RESUMEN

Pneumolysin (PLY), a key virulence factor of Streptococcus pneumoniae, permeabilizes eukaryotic cells by forming large trans-membrane pores. PLY imposes a puzzling multitude of diverse, often mutually excluding actions on eukaryotic cells. Whereas cytotoxicity of PLY can be directly attributed to the pore-mediated effects, mechanisms that are responsible for the PLY-induced activation of host cells are poorly understood. We show that PLY pores can be repaired and thereby PLY-induced cell death can be prevented. Pore-induced Ca²âº entry from the extracellular milieu is of paramount importance for the initiation of plasmalemmal repair. Nevertheless, active Ca²âº sequestration that prevents excessive Ca²âº elevation during the execution phase of plasmalemmal repair is of no less importance. The efficacy of plasmalemmal repair does not only define the fate of targeted cells but also intensity, duration and repetitiveness of PLY-induced Ca²âº signals in cells that were able to survive after PLY attack. Intracellular Ca²âº dynamics evoked by the combined action of pore formation and their elimination mimic the pattern of receptor-mediated Ca²âº signaling, which is responsible for the activation of host immune responses. Therefore, we postulate that plasmalemmal repair of PLY pores might provoke cellular responses that are similar to those currently ascribed to the receptor-mediated PLY effects. Our data provide new insights into the understanding of the complexity of cellular non-immune defense responses to a major pneumococcal toxin that plays a critical role in the establishment and the progression of life-threatening diseases. Therapies boosting plasmalemmal repair of host cells and their metabolic fitness might prove beneficial for the treatment of pneumococcal infections. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.


Asunto(s)
Calcio/metabolismo , Streptococcus pneumoniae/química , Estreptolisinas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Membrana Celular , Células HEK293 , Humanos , Estreptolisinas/química
7.
Biochim Biophys Acta ; 1843(5): 915-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24487066

RESUMEN

In the majority of cells, the integrity of the plasmalemma is recurrently compromised by mechanical or chemical stress. Serum complement or bacterial pore-forming toxins can perforate the plasma membrane provoking uncontrolled Ca(2+) influx, loss of cytoplasmic constituents and cell lysis. Plasmalemmal blebbing has previously been shown to protect cells against bacterial pore-forming toxins. The activation of the P2X7 receptor (P2X7R), an ATP-gated trimeric membrane cation channel, triggers Ca(2+) influx and induces blebbing. We have investigated the role of the P2X7R as a regulator of plasmalemmal protection after toxin-induced membrane perforation caused by bacterial streptolysin O (SLO). Our results show that the expression and activation of the P2X7R furnishes cells with an increased chance of surviving attacks by SLO. This protective effect can be demonstrated not only in human embryonic kidney 293 (HEK) cells transfected with the P2X7R, but also in human mast cells (HMC-1), which express the receptor endogenously. In addition, this effect is abolished by treatment with blebbistatin or A-438079, a selective P2X7R antagonist. Thus blebbing, which is elicited by the ATP-mediated, paracrine activation of the P2X7R, is part of a cellular non-immune defense mechanism. It pre-empts plasmalemmal damage and promotes cellular survival. This mechanism is of considerable importance for cells of the immune system which carry the P2X7R and which are specifically exposed to toxin attacks.


Asunto(s)
Receptores Purinérgicos P2X7/fisiología , Estreptolisinas/toxicidad , Proteínas Bacterianas/toxicidad , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa
8.
Am J Pathol ; 182(2): 431-48, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23201090

RESUMEN

Defects in urothelial integrity resulting in leakage and activation of underlying sensory nerves are potential causative factors of bladder pain syndrome, a clinical syndrome of pelvic pain and urinary urgency/frequency in the absence of a specific cause. Herein, we identified the microRNA miR-199a-5p as an important regulator of intercellular junctions. On overexpression in urothelial cells, it impairs correct tight junction formation and leads to increased permeability. miR-199a-5p directly targets mRNAs encoding LIN7C, ARHGAP12, PALS1, RND1, and PVRL1 and attenuates their expression levels to a similar extent. Using laser microdissection, we showed that miR-199a-5p is predominantly expressed in bladder smooth muscle but that it is also detected in mature bladder urothelium and primary urothelial cultures. In the urothelium, its expression can be up-regulated after activation of cAMP signaling pathways. While validating miR-199a-5p targets, we delineated novel functions of LIN7C and ARHGAP12 in urothelial integrity and confirmed the essential role of PALS1 in establishing and maintaining urothelial polarity and junction assembly. The present results point to a possible link between miR-199a-5p expression and the control of urothelial permeability in bladder pain syndrome. Up-regulation of miR-199a-5p and concomitant down-regulation of its multiple targets might be detrimental to the establishment of a tight urothelial barrier, leading to chronic pain.


Asunto(s)
Cistitis Intersticial/genética , Cistitis Intersticial/patología , Urotelio/metabolismo , Urotelio/patología , Regiones no Traducidas 3'/genética , Secuencia de Bases , Sitios de Unión/genética , Diferenciación Celular/genética , Línea Celular , AMP Cíclico/metabolismo , Regulación hacia Abajo/genética , Impedancia Eléctrica , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Luciferasas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , Músculo Liso/metabolismo , Músculo Liso/patología , Permeabilidad , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Regulación hacia Arriba/genética , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Vejiga Urinaria/ultraestructura , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
9.
FASEB J ; 27(6): 2156-64, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23413360

RESUMEN

Skeletal muscle complaints are a common consequence of cholesterol-lowering therapy. Transverse tubular (T-tubular) vacuolations occur in patients having statin-associated myopathy and, to a lesser extent, in statin-treated patients without myopathy. We have investigated quantitative changes in T-tubular morphology and looked for early indicators of T-tubular membrane repair in skeletal muscle biopsy samples from patients receiving cholesterol-lowering therapy who do not have myopathic side effects. Gene expression and protein levels of incipient membrane repair proteins were monitored in patients who tolerated statin treatment without myopathy and in statin-naive subjects. In addition, morphometry of the T-tubular system was performed. Only the gene expression for annexin A1 was up-regulated, whereas the expression of other repair genes remained unchanged. However, annexin A1 and dysferlin protein levels were significantly increased. In statin-treated patients, the volume fraction of the T-tubular system was significantly increased, but the volume fraction of the sarcoplasmic reticulum remained unchanged. A complex surface structure in combination with high mechanical loads makes skeletal muscle plasma membranes susceptible to injury. Ca(2+)-dependent membrane repair proteins such as dysferlin and annexin A1 are deployed at T-tubular sites. The up-regulation of annexin A1 gene expression and protein points to this protein as a biomarker for T-tubular repair.


Asunto(s)
Anexina A1/biosíntesis , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Anciano , Anciano de 80 o más Años , Anexina A1/genética , Biomarcadores/metabolismo , Estudios de Casos y Controles , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Masculino , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/lesiones , Regeneración/efectos de los fármacos , Regeneración/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
10.
J Urol ; 190(1): 325-33, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23376147

RESUMEN

PURPOSE: We examined the role of annexins in bladder urothelium. We characterized expression and distribution in normal bladders, biopsies from patients with bladder pain syndrome, cultured human urothelium and urothelial TEU-2 cells. MATERIALS AND METHODS: Annexin expression in bladder layers was analyzed by quantitative reverse transcriptase-polymerase chain reaction and immunofluorescence. We assessed cell survival after exposure to the pore forming bacterial toxin streptolysin O by microscopy and alamarBlue® assay. Bladder dome biopsies were obtained from 8 asymptomatic controls and 28 patients with symptoms of bladder pain syndrome. RESULTS: Annexin A1, A2, A5 and A6 were differentially distributed in bladder layers. Annexin A6 was abundant in detrusor smooth muscle and low in urothelium, while annexin A1 was the highest in urothelium. Annexin A2 was localized to the lateral membrane of umbrella cells but excluded from tight junctions. TEU-2 cell differentiation caused up-regulation of annexin A1 and A2 and down-regulation of annexin A6 mRNA. Mature urothelium dedifferentiation during culture caused the opposite effect, decreasing annexin A1 and increasing annexin A6. Annexin A2 influenced TEU-2 cell epithelial permeability. siRNA mediated knockdown of annexin A1 in TEU-2 cells caused significantly decreased cell survival after streptolysin O exposure. Annexin A1 was significantly reduced in biopsies from patients with bladder pain syndrome. CONCLUSIONS: Several annexins are expressed in human bladder and TEU-2 cells, in which levels are regulated during urothelial differentiation. Annexin A1 down-regulation in patients with bladder pain syndrome might decrease cell survival and contribute to compromised urothelial function.


Asunto(s)
Anexina A1/genética , Supervivencia Celular/genética , Cistitis Intersticial/genética , Estreptolisinas/farmacología , Urotelio/patología , Análisis de Varianza , Anexina A1/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Cistitis Intersticial/patología , Regulación hacia Abajo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , ARN Mensajero/análisis , ARN Interferente Pequeño/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Valores de Referencia , Estadísticas no Paramétricas , Transfección , Vejiga Urinaria/citología , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/patología , Urotelio/efectos de los fármacos
11.
Handb Exp Pharmacol ; (216): 341-53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23563665

RESUMEN

The perforation of the plasmalemma by pore-forming toxins causes an influx of Ca(2+) and an efflux of cytoplasmic proteins. In order to ensure cellular survival, lesions have to be identified, plugged and removed from the membrane. The Ca(2+)-driven fusion of lysosomes with the plasma membrane leads to hydrolysis of sphingomyelin by acid sphingomyelinase and a formation of ceramide platforms in the outer leaflet of the lipid bilayer. We propose that the negative curvature, promoted by tighter packing of lipids in the outer layer, leads to an inward vesiculation of the damaged area for its endocytotic uptake and internal degradation. In contrast, the activation of neutral sphingomyelinase triggers the production of ceramide within the inner leaflet of the lipid bilayer, thereby promoting an outward curvature, which enables the cell to shed the membrane-containing toxin pore into the extracellular space. In this process, ceramide is supported by members of the annexin protein family which act as Ca(2+) sensors and as membrane fusion agents.


Asunto(s)
Membrana Celular/metabolismo , Ceramidas/metabolismo , Transducción de Señal , Animales , Membrana Celular/patología , Supervivencia Celular , Micropartículas Derivadas de Células/metabolismo , Endocitosis , Exocitosis , Humanos , Fusión de Membrana , Mitocondrias/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo
12.
J Biol Chem ; 286(20): 17982-91, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21454475

RESUMEN

The annexins, a family of Ca(2+)- and lipid-binding proteins, are involved in a range of intracellular processes. Recent findings have implicated annexin A1 in the resealing of plasmalemmal injuries. Here, we demonstrate that another member of the annexin protein family, annexin A6, is also involved in the repair of plasmalemmal lesions induced by a bacterial pore-forming toxin, streptolysin O. An injury-induced elevation in the intracellular concentration of Ca(2+) ([Ca(2+)](i)) triggers plasmalemmal repair. The highly Ca(2+)-sensitive annexin A6 responds faster than annexin A1 to [Ca(2+)](i) elevation. Correspondingly, a limited plasmalemmal injury can be promptly countered by annexin A6 even without the participation of annexin A1. However, its high Ca(2+) sensitivity makes annexin A6 highly amenable to an unproductive binding to the uninjured plasmalemma; during an extensive injury accompanied by a massive elevation in [Ca(2+)](i), its active pool is severely depleted. In contrast, annexin A1 with a much lower Ca(2+) sensitivity is ineffective at the early stages of injury; however, it remains available for the repair even at high [Ca(2+)](i). Our findings highlight the role of the annexins in the process of plasmalemmal repair; a number of annexins with different Ca(2+)-sensitivities provide a cell with the means to react promptly to a limited injury in its early stages and, at the same time, to withstand a sustained injury accompanied by the continuous formation of plasmalemmal lesions.


Asunto(s)
Anexina A1/metabolismo , Anexina A6/metabolismo , Calcio/metabolismo , Membrana Celular/metabolismo , Proteínas Bacterianas/farmacología , Línea Celular Tumoral , Membrana Celular/patología , Células HEK293 , Humanos , Estreptolisinas/farmacología
13.
Cells ; 11(1)2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-35011729

RESUMEN

The increasing antibiotic resistance of bacterial pathogens fosters the development of alternative, non-antibiotic treatments. Antivirulence therapy, which is neither bacteriostatic nor bactericidal, acts by depriving bacterial pathogens of their virulence factors. To establish a successful infection, many bacterial pathogens secrete exotoxins/cytolysins that perforate the host cell plasma membrane. Recently developed liposomal nanotraps, mimicking the outer layer of the targeted cell membranes, serve as decoys for exotoxins, thus diverting them from attacking host cells. In this study, we develop a liposomal nanotrap formulation that is capable of protecting immortalized immune cells from the whole palette of cytolysins secreted by Streptococcus pyogenes and Streptococcus dysgalactiae subsp. equisimilis-important human pathogens that can cause life-threatening bacteremia. We show that the mixture of cholesterol-containing liposomes with liposomes composed exclusively of phospholipids is protective against the combined action of all streptococcal exotoxins. Our findings pave the way for further development of liposomal antivirulence therapy in order to provide more efficient treatment of bacterial infections, including those caused by antibiotic resistant pathogens.


Asunto(s)
Citotoxinas/toxicidad , Leucocitos/metabolismo , Liposomas/química , Streptococcus pyogenes/metabolismo , Streptococcus/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Línea Celular Transformada , Colesterol/metabolismo , Humanos , Leucocitos/efectos de los fármacos , Pruebas de Neutralización
14.
Front Microbiol ; 13: 867449, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35369481

RESUMEN

Engineered liposomes composed of the naturally occurring lipids sphingomyelin (Sm) and cholesterol (Ch) have been demonstrated to efficiently neutralize toxins secreted by Gram-positive bacteria such as Streptococcus pneumoniae and Staphylococcus aureus. Here, we hypothesized that liposomes are capable of neutralizing cytolytic virulence factors secreted by the Gram-negative pathogen Pseudomonas aeruginosa. We used the highly virulent cystic fibrosis P. aeruginosa Liverpool Epidemic Strain LESB58 and showed that sphingomyelin (Sm) and a combination of sphingomyelin with cholesterol (Ch:Sm; 66 mol/% Ch and 34 mol/% Sm) liposomes reduced lysis of human bronchial and red blood cells upon challenge with the Pseudomonas secretome. Mass spectrometry of liposome-sequestered Pseudomonas proteins identified the virulence-promoting hemolytic phospholipase C (PlcH) as having been neutralized. Pseudomonas aeruginosa supernatants incubated with liposomes demonstrated reduced PlcH activity as assessed by the p-nitrophenylphosphorylcholine (NPPC) assay. Testing the in vivo efficacy of the liposomes in a murine cutaneous abscess model revealed that Sm and Ch:Sm, as single dose treatments, attenuated abscesses by >30%, demonstrating a similar effect to that of a mutant lacking plcH in this infection model. Thus, sphingomyelin-containing liposome therapy offers an interesting approach to treat and reduce virulence of complex infections caused by P. aeruginosa and potentially other Gram-negative pathogens expressing PlcH.

15.
Am J Pathol ; 177(1): 291-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20489141

RESUMEN

In skeletal muscle of patients with clinically diagnosed statin-associated myopathy, discrete signs of structural damage predominantly localize to the T-tubular region and are suggestive of a calcium leak. The impact of statins on skeletal muscle of non-myopathic patients is not known. We analyzed the expression of selected genes implicated in the molecular regulation of calcium and membrane repair, in lipid homeostasis, myocyte remodeling and mitochondrial function. Microscopic and gene expression analyses were performed using validated TaqMan custom arrays on skeletal muscle biopsies of 72 age-matched subjects who were receiving statin therapy (n = 38), who had discontinued therapy due to statin-associated myopathy (n = 14), and who had never undergone statin treatment (n = 20). In skeletal muscle, obtained from statin-treated, non-myopathic patients, statins caused extensive changes in the expression of genes of the calcium regulatory and the membrane repair machinery, whereas the expression of genes responsible for mitochondrial function or myocyte remodeling was unaffected. Discontinuation of treatment due to myopathic symptoms led to a normalization of gene expression levels, the genes encoding the ryanodine receptor 3, calpain 3, and dystrophin being the most notable exceptions. Hence, even in clinically asymptomatic (non-myopathic) patients, statin therapy leads to an upregulation in the expression of genes that are concerned with skeletal muscle regulation and membrane repair.


Asunto(s)
Calcio/metabolismo , Membrana Celular , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Músculo Esquelético , Enfermedades Musculares , Anciano , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/patología , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipercolesterolemia/tratamiento farmacológico , Persona de Mediana Edad , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/genética , Enfermedades Musculares/patología
16.
Toxins (Basel) ; 13(2)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572185

RESUMEN

Pore-forming toxins (PFTs) form multimeric trans-membrane pores in cell membranes that differ in pore channel diameter (PCD). Cellular resistance to large PFTs (>20 nm PCD) was shown to rely on Ca2+ influx activated membrane repair mechanisms. Small PFTs (<2 nm PCD) were shown to exhibit a high cytotoxic activity, but host cell response and membrane repair mechanisms are less well studied. We used monocytic immune cell lines to investigate the cellular resistance and host membrane repair mechanisms to small PFTs lysenin (Eisenia fetida) and aerolysin (Aeromonas hydrophila). Lysenin, but not aerolysin, is shown to induce Ca2+ influx from the extracellular space and to activate Ca2+ dependent membrane repair mechanisms. Moreover, lysenin binds to U937 cells with higher efficiency as compared to THP-1 cells, which is in line with a high sensitivity of U937 cells to lysenin. In contrast, aerolysin equally binds to U937 or THP-1 cells, but in different plasma membrane areas. Increased aerolysin induced cell death of U937 cells, as compared to THP-1 cells, is suggested to be a consequence of cap-like aerolysin binding. We conclude that host cell resistance to small PFTs attack comprises binding efficiency, pore localization, and capability to induce Ca2+ dependent membrane repair mechanisms.


Asunto(s)
Toxinas Bacterianas/toxicidad , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Monocitos/efectos de los fármacos , Proteínas Citotóxicas Formadoras de Poros/toxicidad , Toxinas Biológicas/toxicidad , Muerte Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/patología , Resistencia a Medicamentos , Genes Reporteros , Humanos , Monocitos/metabolismo , Monocitos/patología , Células THP-1 , Células U937
17.
Traffic ; 9(10): 1757-75, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18694456

RESUMEN

Upon its genesis during apoptosis, ceramide promotes gross reorganization of the plasma membrane structure involving clustering of signalling molecules and an amplification of vesicle formation, fusion and trafficking. The annexins are a family of proteins, which in the presence of Ca(2+), bind to membranes containing negatively charged phospholipids. Here, we show that ceramide increases affinity of annexin A1-membrane interaction. In the physiologically relevant range of Ca(2+) concentrations, this leads to an increase in the Ca(2+)sensitivity of annexin A1-membrane interaction. In fixed cells, using a ceramide-specific antibody, we establish a direct interaction of annexin A1 with areas of the plasma membrane enriched in ceramide (ceramide platforms). In living cells, the intracellular dynamics of annexin A1 match those of plasmalemmal ceramide. Among proteins of the annexin family, the interaction with ceramide platforms is restricted to annexin A1 and is conveyed by its unique N-terminal domain. We demonstrate that intracellular Ca(2+)overload occurring at the conditions of cellular stress induces ceramide production. Using fluorescently tagged annexin A1 as a reporter for ceramide platforms and annexin A6 as a non-selective membrane marker, we visualize ceramide platforms for the first time in living cells and provide evidence for a ceramide-driven segregation and internalization of membrane-associated proteins.


Asunto(s)
Anexina A1/metabolismo , Apoptosis/fisiología , Calcio/metabolismo , Membrana Celular/metabolismo , Ceramidas/fisiología , Animales , Apoptosis/efectos de los fármacos , Cadmio/farmacología , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Membrana Celular/ultraestructura , Ceramidas/metabolismo , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Humanos , Ionomicina/farmacología , Microscopía Confocal , Microsomas/efectos de los fármacos , Microsomas/metabolismo , Unión Proteica , Transporte de Proteínas , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/metabolismo , Porcinos , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo
18.
J Biol Chem ; 284(25): 17227-17242, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19386597

RESUMEN

The annexins are a family of Ca(2+)- and phospholipid-binding proteins, which interact with membranes upon increase of [Ca(2+)](i) or during cytoplasmic acidification. The transient nature of the membrane binding of annexins complicates the study of their influence on intracellular processes. To address the function of annexins at the plasma membrane (PM), we fused fluorescent protein-tagged annexins A6, A1, and A2 with H- and K-Ras membrane anchors. Stable PM localization of membrane-anchored annexin A6 significantly decreased the store-operated Ca(2+) entry (SOCE), but did not influence the rates of Ca(2+) extrusion. This attenuation was specific for annexin A6 because PM-anchored annexins A1 and A2 did not alter SOCE. Membrane association of annexin A6 was necessary for a measurable decrease of SOCE, because cytoplasmic annexin A6 had no effect on Ca(2+) entry as long as [Ca(2+)](i) was below the threshold of annexin A6-membrane translocation. However, when [Ca(2+)](i) reached the levels necessary for the Ca(2+)-dependent PM association of ectopically expressed wild-type annexin A6, SOCE was also inhibited. Conversely, knockdown of the endogenous annexin A6 in HEK293 cells resulted in an elevated Ca(2+) entry. Constitutive PM localization of annexin A6 caused a rearrangement and accumulation of F-actin at the PM, indicating a stabilized cortical cytoskeleton. Consistent with these findings, disruption of the actin cytoskeleton using latrunculin A abolished the inhibitory effect of PM-anchored annexin A6 on SOCE. In agreement with the inhibitory effect of annexin A6 on SOCE, constitutive PM localization of annexin A6 inhibited cell proliferation. Taken together, our results implicate annexin A6 in the actin-dependent regulation of Ca(2+) entry, with consequences for the rates of cell proliferation.


Asunto(s)
Actinas/metabolismo , Anexina A6/metabolismo , Señalización del Calcio/fisiología , Anexina A1/metabolismo , Anexina A2/metabolismo , Anexina A6/antagonistas & inhibidores , Anexina A6/genética , Secuencia de Bases , Canales de Calcio/metabolismo , Línea Celular , Membrana Celular/metabolismo , Proliferación Celular , Citoesqueleto/metabolismo , Cartilla de ADN/genética , Retículo Endoplásmico/metabolismo , Humanos , Interferencia de ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
19.
Cell Mol Life Sci ; 66(16): 2623-42, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19381436

RESUMEN

Annexins are a family of structurally related, Ca2+-sensitive proteins that bind to negatively charged phospholipids and establish specific interactions with other lipids and lipid microdomains. They are present in all eukaryotic cells and share a common folding motif, the "annexin core", which incorporates Ca2+- and membrane-binding sites. Annexins participate in a variety of intracellular processes, ranging from the regulation of membrane dynamics to cell migration, proliferation, and apoptosis. Here we focus on the role of annexins in cellular signaling during stress. A chronic stress response triggers the activation of different intracellular pathways, resulting in profound changes in Ca2+ and pH homeostasis and the production of lipid second messengers. We review the latest data on how these changes are sensed by the annexins, which have the ability to simultaneously interact with specific lipid and protein moieties at the plasma membrane, contributing to stress adaptation via regulation of various signaling pathways.


Asunto(s)
Anexinas/fisiología , Transducción de Señal , Estrés Fisiológico , Anexinas/química , Calcio/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Células Cultivadas , Ceramidas/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Humanos , Concentración de Iones de Hidrógeno , Metabolismo de los Lípidos , Proteína Quinasa C/metabolismo , Proteínas ras/metabolismo
20.
Nat Commun ; 11(1): 1338, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32165633

RESUMEN

Macrophages have important protective functions during infection with herpes simplex virus type 1 (HSV-1). However, molecular mechanisms that restrict viral propagation and protect from severe disease are unclear. Here we show that macrophages take up HSV-1 via endocytosis and transport the virions into multivesicular bodies (MVBs). In MVBs, acid ceramidase (aCDase) converts ceramide into sphingosine and increases the formation of sphingosine-rich intraluminal vesicles (ILVs). Once HSV-1 particles reach MVBs, sphingosine-rich ILVs bind to HSV-1 particles, which restricts fusion with the limiting endosomal membrane and prevents cellular infection. Lack of aCDase in macrophage cultures or in Asah1-/- mice results in replication of HSV-1 and Asah1-/- mice die soon after systemic or intravaginal inoculation. The treatment of macrophages with sphingosine enhancing compounds blocks HSV-1 propagation, suggesting a therapeutic potential of this pathway. In conclusion, aCDase loads ILVs with sphingosine, which prevents HSV-1 capsids from penetrating into the cytosol.


Asunto(s)
Ceramidasa Ácida/metabolismo , Herpes Simple/enzimología , Herpes Simple/prevención & control , Herpesvirus Humano 1/fisiología , Macrófagos/enzimología , Cuerpos Multivesiculares/virología , Ceramidasa Ácida/genética , Animales , Femenino , Herpes Simple/virología , Humanos , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA