Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Eur J Public Health ; 29(5): 943-947, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31219550

RESUMEN

BACKGROUND: The Italian National Institute of Health (Istituto Superiore di Sanità, ISS) considers health inequalities (HI) an important area of activity. As the scientific and technical body of the Ministry of Health and the National Health Service, ISS may play a key role to reduce HI. In order to enable ISS in addressing the new and crucial HI challenge, a Research Positioning Exercise was designed and implemented. METHODS: The Exercise included: (i) workshop to strengthen the institutional interest in the field of HI; (ii) review and analysis of ISS publications (years 2000-2017) to identify HI research topics; (iii) survey among ISS researchers regarding main research challenges to address HI in the coming years; and (iv) analysis of input on research challenges from HI international experts. RESULTS: The results of this Exercise suggest that the following points should be included in the future ISS agenda planning: (i) themes which ISS should continue working on (e.g. migrants/vulnerable groups); (ii) themes to be improved: (a) relationship between social determinants and mechanism of HI generation and (b) relationship between risk factors exposure and social determinants; and (iii) new themes to be addressed: (a) mechanisms underlying the resilience observed in Italy; (b) new socioeconomic indicators for HI monitoring; and (c) evidence-based policies aimed at reducing HI. CONCLUSION: Findings of this Exercise show that ISS researchers identified relevant areas, addressing inequalities in addressing the health. Because of ISS structural peculiarity that includes multidisciplinary expertise, the ISS could provide a significant contribution to HI research challenges and knowledge gaps.


Asunto(s)
Investigación Biomédica , Educación , Disparidades en el Estado de Salud , Proteínas de Arabidopsis , Investigación Biomédica/organización & administración , Agencias Gubernamentales/organización & administración , N-Metiltransferasa de Histona-Lisina , Humanos , Italia/epidemiología , Investigación , Factores de Riesgo , Determinantes Sociales de la Salud , Poblaciones Vulnerables
2.
PLoS One ; 12(10): e0185728, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28977040

RESUMEN

BACKGROUND: Few data are available on the virological and clinical outcomes of advanced liver disease patients retreated after first-line DAA failure. AIM: To evaluate DAA failure incidence and the retreatment clinical impact in patients treated in the advanced liver disease stage. METHODS: Data on HCV genotype, liver disease severity, and first and second line DAA regimens were prospectively collected in consecutive patients who reached the 12-week post-treatment and retreatment evaluations from January 2015 to December 2016 in 23 of the PITER network centers. RESULTS: Among 3,830 patients with advanced fibrosis (F3) or cirrhosis, 139 (3.6%) failed to achieve SVR. Genotype 3, bilirubin levels >1.5mg/dl, platelet count <120,000/mm3 and the sofosbuvir+ribavirin regimen were independent predictors of failure by logistic regression analysis. The failure rate was 7.6% for patients treated with regimens that are no longer recommended or considered suboptimal (sofosbuvir+ribavirin or simeprevir+sofosbuvir±ribavirin), whereas 1.4% for regimens containing sofosbuvir combined with daclatasvir or ledipasvir or other DAAs. Of the patients who failed to achieve SVR, 72 (51.8%) were retreated with a second DAA regimen, specifically 38 (52.7%) with sofosbuvir+daclatasvir, 27 (37.5%) with sofosbuvir+ledipasvir, and 7 (9.7%) with other DAAs ±ribavirin. Among these, 69 (96%) patients achieved SVR12 and 3 (4%) failed. During a median time of 6 months (range: 5-14 months) between failure and the second DAA therapy, the Child-Pugh class worsened in 12 (16.7%) patients: from A to B in 10 patients (19.6%) and from B to C in 2 patients (10.5%), whereas it did not change in the remaining 60 patients. Following the retreatment SVR12 (median time of 6 months; range: 3-12 months), the Child-Pugh class improved in 17 (23.6%) patients: from B to A in 14 (19.4%) patients, from C to A in 1 patient (1.4%) and from C to B in 2 (2.9%) patients; it remained unchanged in 53 patients (73.6%) and worsened in 2 (2.8%) patients. Of patients who were retreated, 3 (4%) had undergone OLT before retreatment (all reached SVR12 following retreatment) and 2 (2.8%) underwent OLT after having achieved retreatment SVR12. Two (70%) of the 3 patients who failed to achieve SVR12 after retreatment, and 2 (2.8%) of the 69 patients who achieved retreatment SVR12 died from liver failure (Child-Pugh class deteriorated from B to C) or HCC complications. CONCLUSIONS: Failure rate following the first DAA regimen in patients with advanced disease is similar to or lower than that reported in clinical trials, although the majority of patients were treated with suboptimal regimens. Interim findings showed that worsening of liver function after failure, in terms of Child Pugh class deterioration, was improved by successful retreatment in about one third of retreated patients within a short follow-up period; however, in some advanced liver disease patients, clinical outcomes (Child Pugh class, HCC development, liver failure and death) were independent of viral eradication.


Asunto(s)
Antivirales/uso terapéutico , Hepatitis C/tratamiento farmacológico , Hepatopatías/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antivirales/administración & dosificación , Quimioterapia Combinada , Femenino , Hepatitis C/genética , Hepatitis C/patología , Humanos , Incidencia , Hepatopatías/genética , Hepatopatías/patología , Masculino , Persona de Mediana Edad , Estudios Prospectivos
3.
PLoS One ; 7(4): e34115, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22523545

RESUMEN

Modulation of cerebral Rho GTPases activity in mice brain by intracerebral administration of Cytotoxic Necrotizing Factor 1 (CNF1) leads to enhanced neurotransmission and synaptic plasticity and improves learning and memory. To gain more insight into the interactions between CNF1 and neuronal cells, we used primary neuronal and astrocytic cultures from rat embryonic brain to study CNF1 effects on neuronal differentiation, focusing on dendritic tree growth and synapse formation, which are strictly modulated by Rho GTPases. CNF1 profoundly remodeled the cytoskeleton of hippocampal and cortical neurons, which showed philopodia-like, actin-positive projections, thickened and poorly branched dendrites, and a decrease in synapse number. CNF1 removal, however, restored dendritic tree development and synapse formation, suggesting that the toxin can reversibly block neuronal differentiation. On differentiated neurons, CNF1 had a similar effacing effect on synapses. Therefore, a direct interaction with CNF1 is apparently deleterious for neurons. Since astrocytes play a pivotal role in neuronal differentiation and synaptic regulation, we wondered if the beneficial in vivo effect could be mediated by astrocytes. Primary astrocytes from embryonic cortex were treated with CNF1 for 48 hours and used as a substrate for growing hippocampal neurons. Such neurons showed an increased development of neurites, in respect to age-matched controls, with a wider dendritic tree and a richer content in synapses. In CNF1-exposed astrocytes, the production of interleukin 1ß, known to reduce dendrite development and complexity in neuronal cultures, was decreased. These results demonstrate that astrocytes, under the influence of CNF1, increase their supporting activity on neuronal growth and differentiation, possibly related to the diminished levels of interleukin 1ß. These observations suggest that the enhanced synaptic plasticity and improved learning and memory described in CNF1-injected mice are probably mediated by astrocytes.


Asunto(s)
Astrocitos/fisiología , Toxinas Bacterianas/farmacología , Diferenciación Celular/efectos de los fármacos , Proteínas de Escherichia coli/farmacología , Neurogénesis/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Interleucina-1beta/biosíntesis , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Wistar , Sinapsis/efectos de los fármacos , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
4.
Viral Immunol ; 24(3): 245-50, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21668366

RESUMEN

HIV infections are mainly acquired by mucosal transmission, through oral, rectal, or genital mucosa. Epithelial cells (EC) are the first cells encountered by HIV during infection through sexual transmission and breastfeeding. EC express several receptors critical for both primary HIV infection and secondary transmission. The regulation of co-receptor expression correlates with changes in susceptibility to infection by HIV-1 strains with different tropism. Moreover, inflammatory responses at mucosal surfaces after HIV-1 transmission may influence disease outcome. In the present study, we analyzed the effect of the accessory HIV-1 Nef protein on mucosal EC, using unstimulated or IFN-γ-stimulated HEp-2, T24, and Caco2 cell lines as models for homeostatic or inflamed mucosal tracts. We found that Nef significantly upregulated the expression of CXCR4 on the Caco-2 cell surface and the expression of galactosylceramide on the T24 cell surface. In addition, Nef significantly upregulated IL-6 production by T24 and Caco-2 cells, and TNF-α release by all three cell lines analyzed. Notably, Nef abrogated the IFN-γ-induced modulation of co-receptor expression and cytokine secretion. Our findings suggest that Nef differently regulates co-receptor expression and cytokine secretion at the epithelial level, depending on the anatomical derivation of the cells and the inflammatory status.


Asunto(s)
Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/virología , VIH-1/inmunología , VIH-1/patogenicidad , Receptores del VIH/biosíntesis , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Línea Celular , Expresión Génica , Humanos , Mucosa Intestinal/virología , Laringe/virología , Vejiga Urinaria/virología
5.
Pain ; 145(1-2): 219-29, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19608345

RESUMEN

Cytotoxic Necrotizing Factor 1 (CNF1) is a protein toxin from Escherichia coli that constitutively activates the Rho, Rac and Cdc42 GTPases. These regulatory proteins oscillate between a cytosolic GDP-bound inactive form and a membrane-linked GTP-bound active form, orchestrating the actin cytoskeleton assembly and dynamics. We herein describe, for the first time, the ability of CNF1 to potently counteract the formalin-induced inflammatory pain in mice. The analgesic response due to CNF1 requires both the sustained activation of the Rac GTPase, with consequent cerebral actin cytoskeleton remodeling, and the up-regulation of the mu-opioid receptors (MORs), the most important receptors controlling pain perception. The crucial role of Rac is proved by the lack of analgesic activity in mice challenged with a recombinant CNF1, in which the enzymatic activity was abolished by substituting serine with cysteine at position 866. The importance of MORs is proved by the inability of CNF1 to induce any analgesic effect in MORs knockout mice and by the ability of naloxone to antagonize the analgesic effects. Furthermore, it is worth noting that the analgesic effect in mice occurs after both peripheral and central administration of CNF1. Hence, taken altogether, our findings provide new insights into the comprehension of intracellular mechanisms involved in pain modulation, and indicate this bacterial protein toxin as a novel tool in the field of pain control. Conceivably, this might pave the way for new therapeutic strategies.


Asunto(s)
Analgésicos/uso terapéutico , Toxinas Bacterianas/farmacología , Toxinas Bacterianas/uso terapéutico , Proteínas de Escherichia coli/farmacología , Proteínas de Escherichia coli/uso terapéutico , Receptores Opioides mu/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Línea Celular Tumoral , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Formaldehído/efectos adversos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Neuroblastoma/patología , Dolor/inducido químicamente , Dolor/patología , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides mu/deficiencia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Transfección/métodos , Regulación hacia Arriba/genética , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo
6.
Mol Biol Cell ; 18(7): 2735-44, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17507655

RESUMEN

Cytotoxic necrotizing factor 1 (CNF1) is a protein toxin produced by some pathogenic strains of Escherichia coli that specifically activates Rho, Rac, and Cdc42 GTPases. We previously reported that this toxin prevents the ultraviolet-B-induced apoptosis in epithelial cells, with a mechanism that remained to be defined. In this work, we show that the proteasomal degradation of the Rho GTPase is necessary to achieve cell death protection, because inhibition of Rho degradation abolishes the prosurvival activity of CNF1. We hypothesize that Rho inactivation allows the activity of Rac to become dominant. This in turn leads to stimulation of the phosphoinositide 3-kinase/Akt/IkappaB kinase/nuclear factor-kappaB prosurvival pathway and to a remarkable modification in the architecture of the mitochondrial network, mainly consisting in the appearance of elongated and interconnected mitochondria. Importantly, we found that Bcl-2 silencing reduces the ability of CNF1 to protect cells against apoptosis and that it also prevents the CNF1-induced mitochondrial changes. It is worth noting that the ability of a bacterial toxin to induce such a remodeling of the mitochondrial network is herein reported for the first time. The possible pathophysiological relevance of this finding is discussed.


Asunto(s)
Apoptosis/efectos de los fármacos , Toxinas Bacterianas/farmacología , Proteínas de Escherichia coli/farmacología , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Apoptosis/efectos de la radiación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/efectos de la radiación , Humanos , Cinética , Mitocondrias/efectos de los fármacos , Mitocondrias/efectos de la radiación , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/efectos de la radiación , Rayos Ultravioleta , Proteína bcl-X/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo
7.
Proc Natl Acad Sci U S A ; 104(2): 636-41, 2007 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-17202256

RESUMEN

The mechanism whereby the morphology and connectivity of the dendritic tree is regulated depends on an actin dynamics that, in turn, is controlled by Rho GTPases, a family of small GTP-binding proteins encompassing Rho, Rac, and Cdc42 subfamilies. Cytotoxic necrotizing factor 1 (CNF1), a protein toxin from Escherichia coli, constitutively activates Rho GTPases, thus leading to remodeling of the actin cytoskeleton in intact cells. Here, we show that the modulation of cerebral RhoA and Rac1 activity induced by CNF1 in mice leads to (i) rearrangement of cerebral actin cytoskeleton, (ii) enhanced neurotransmission and synaptic plasticity, and (iii) improved learning and memory in various behavioral tasks. The effects persist for weeks and are not observed in mice treated with a recombinant CNF1, in which the enzymatic activity was abolished by substituting serine to cysteine at position 866. The results suggest that learning ability can be improved through pharmacological manipulation of neural connectivity.


Asunto(s)
Encéfalo/enzimología , Aprendizaje/fisiología , Memoria/fisiología , Proteínas de Unión al GTP rho/metabolismo , Actinas/metabolismo , Animales , Toxinas Bacterianas/toxicidad , Encéfalo/efectos de los fármacos , Células Cultivadas , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Citotoxinas/toxicidad , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Dendritas/ultraestructura , Activación Enzimática , Proteínas de Escherichia coli/toxicidad , Miedo/efectos de los fármacos , Miedo/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/ultraestructura , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología , Transmisión Sináptica/efectos de los fármacos
8.
J Biol Chem ; 282(12): 9029-41, 2007 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-17220295

RESUMEN

Targeting to mitochondria is emerging as a common strategy that bacteria utilize to interact with these central executioners of apoptosis. Several lines of evidence have in fact indicated mitochondria as specific targets for bacterial protein toxins, regarded as the principal virulence factors of pathogenic bacteria. This work shows, for the first time, the ability of the Clostridium difficile toxin B (TcdB), a glucosyltransferase that inhibits the Rho GTPases, to impact mitochondria. In living cells, TcdB provokes an early hyperpolarization of mitochondria that follows a calcium-associated signaling pathway and precedes the final execution step of apoptosis (i.e. mitochondria depolarization). Importantly, in isolated mitochondria, the toxin can induce a calcium-dependent mitochondrial swelling, accompanied by the release of the proapoptogenic factor cytochrome c. This is consistent with a mitochondrial targeting that does not require the Rho-inhibiting activity of the toxin. Of interest, the mitochondrial ATP-sensitive potassium channels are also involved in the apoptotic response to TcdB and appear to be crucial for the cell death execution phase, as demonstrated by using specific modulators of these channels. To our knowledge, the involvement of these mitochondrial channels in the ability of a bacterial toxin to control cell fate is a hitherto unreported finding.


Asunto(s)
Adenosina Trifosfato/metabolismo , Apoptosis , Proteínas Bacterianas/fisiología , Células Epiteliales/metabolismo , Mitocondrias/metabolismo , Toxinas Bacterianas , Calcio/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Caspasas/metabolismo , Línea Celular , Citometría de Flujo , Humanos , Potenciales de la Membrana , Transducción de Señal , Factores de Tiempo
9.
Infect Immun ; 74(7): 3765-72, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16790748

RESUMEN

Evidence is accumulating that a growing number of bacterial toxins act by modulating the eukaryotic cell cycle machinery. In this context, we provide evidence that a protein toxin named cytotoxic necrotizing factor 1 (CNF1) from uropathogenic Escherichia coli is able to block cell cycle G(2)/M transition in the uroepithelial cell line T24. CNF1 permanently activates the small GTP-binding proteins of the Rho family that, beside controlling the actin cytoskeleton organization, also play a pivotal role in a large number of other cellular processes, including cell cycle regulation. The results reported here show that CNF1 is able to induce the accumulation of cells in the G(2)/M phase by sequestering cyclin B1 in the cytoplasm and down-regulating its expression. The possible role played by the Rho GTPases in the toxin-induced cell cycle deregulation has been investigated and discussed. The activity of CNF1 on cell cycle progression can offer a novel view of E. coli pathogenicity.


Asunto(s)
División Celular/fisiología , Citotoxinas/fisiología , Proteínas de Escherichia coli/fisiología , Escherichia coli/fisiología , Fase G2/fisiología , Inhibidores de Crecimiento/fisiología , Vejiga Urinaria/citología , Vejiga Urinaria/microbiología , Toxinas Bacterianas , Línea Celular Tumoral , Ciclina B/metabolismo , Ciclina B1 , Escherichia coli/patogenicidad , Humanos , Urotelio/microbiología , Proteínas de Unión al GTP rho/fisiología
10.
Environ Res ; 95(1): 82-91, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15068934

RESUMEN

Particulate matter (PM), a component of urban air pollution that derives primarily from the combustion of fossil fuels, is responsible for a number of health effects in humans. Recent studies have demonstrated that the fine particles (PM(2.5)) present in high numbers in PM samples can be more harmful than larger particles, since they are more efficiently retained in the peripheral lung. In the present study, we have investigated the biological effects of PM(2.5) on human lung epithelial cell line A549. Morphological analysis performed by immunofluorescence and electron microscopy showed that fine particles interact with the cell surface, where they induce evident alterations and, subsequently, are internalized in the cytoplasm. Cytoskeletal components, in particular microfilaments and microtubules, cause modifications upon challenge with PM(2.5). Of interest, an early cell response to the fine particulate is an increase of reactive oxygen species content, which can account for the observed cytoskeletal changes and the production of proinflammatory cytokines in A549 cells. In particular, exposure to PM(2.5) promoted a dose- and time-dependent release of TNF-alpha and IL-6 in the cell medium.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Apoptosis/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Células Epiteliales/ultraestructura , Pulmón/efectos de los fármacos , Anexina A5 , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Combustibles Fósiles , Humanos , Interleucina-6/metabolismo , Italia , Microscopía Electrónica , Microscopía Fluorescente , Tamaño de la Partícula , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Mol Biol Cell ; 15(3): 1124-33, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14668491

RESUMEN

Nuclear factor-kappaB (NF-kappaB) is a ubiquitously expressed transcription factor that plays a central role in directing a vast range of cellular functions. Its activation is controlled by the Rac GTPase and relies on the coordinated cooperation of the E3-ligase complex SCF(betaTrCP), composed by Skp-1/Cullin-1, Rbx/Roc1, and the beta-TrCP proteins. Recently, Cullin-1 has been reported to form a complex with the activated Rac GTPase. Here, we show that the specific activation of the Rac GTPase, besides directing its own positioning, induces the relocalization of the SCF component Cullin-1 to the ruffling membranes. This occurred only if the ruffles were stimulated by the Rac GTPase and was accompanied by the repositioning to the same intracellular compartment of the SCF protein Skp-1 and the ubiquitin-like molecule Nedd-8. The SCF substrate IkBalpha was also directed to the ruffling membranes in a Rac-dependent way. The novelty of these findings is in respect to the demonstration that the correct positioning at the ruffling membranes is crucial for the subsequent series of events that leads to IkBalpha proteasomal degradation and the resultant activation of NF-kappaB. Consequently, this points to the role of Rac as a docking molecule in NF-kappaB activation.


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , FN-kappa B/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteínas Cullin/metabolismo , Cisteína Endopeptidasas/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Complejos Multienzimáticos/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Factores de Transcripción/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo
12.
Toxicol In Vitro ; 17(5-6): 547-52, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14599443

RESUMEN

An epidemiological survey on mortality for malignant pleural neoplasm in Italy evidenced a number of patients in Biancavilla, a village located in a volcanic area of eastern Sicily, none of which had been significantly exposed to asbestos during their professional lives. Environmental studies suggested the involvement of the material derived from stone quarries in the disease onset. A detailed crystal-chemical analysis of amphiboles contained in this material allowed the discovery and the identification of a new fiber that was named fluoro-edenite. In order to define the mode of action of fluoro-edenite at a subcellular level, we have conducted a study by using A549 cells, a tumor-cell line from a human lung carcinoma with properties of alveolar epithelial cells. The results obtained showed a remarkable tropism of A549 cells toward fluoro-edenite fibers. In fact, these epithelial cells contacted the fibers via the extension of membrane ruffles and filopodia that allowed the capture and most probably the internalization of material into the cytoplasm. Moreover, fluoro-edenite interfered with epithelial cell physiology, by reducing the proliferation rate without perturbing the cell cycle and increasing the release of the proinflammatory cytokine IL-6, one of the main mediators of asbestos-induced pathophysiological response.


Asunto(s)
Asbestos Anfíboles/toxicidad , Células Epiteliales/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Adenocarcinoma , Asbestos Anfíboles/aislamiento & purificación , División Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Microanálisis por Sonda Electrónica , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Humanos , Interleucina-6/metabolismo , Neoplasias Pulmonares , Microscopía Electrónica de Rastreo , Fibras Minerales/toxicidad , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología
13.
Infect Immun ; 71(7): 4178-81, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12819113

RESUMEN

Uropathogenic Escherichia coli strains frequently produce a Rho-activating protein toxin named cytotoxic necrotizing factor type 1 (CNF1). We herein report that CNF1 promotes transcription and release of tumor necrosis factor alpha, gamma interferon, interleukin-6 (IL-6), and IL-8 proinflammatory cytokines and increases the production of reactive oxygen species (ROS) in uroepithelial T24 cells. The antioxidant N-acetyl-L-cysteine counteracts these phenomena, a fact which suggests a role for ROS-mediated signaling in CNF1-induced proinflammatory cytokine production.


Asunto(s)
Toxinas Bacterianas/toxicidad , Citocinas/biosíntesis , Citotoxinas/toxicidad , Proteínas de Escherichia coli , Especies Reactivas de Oxígeno , Transcripción Genética/efectos de los fármacos , Vejiga Urinaria/efectos de los fármacos , Línea Celular , Humanos , Vejiga Urinaria/metabolismo
14.
Comp Immunol Microbiol Infect Dis ; 26(3): 145-56, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12581745

RESUMEN

To investigate the mode of action of Clostridium perfringens epsilon-toxin, MDCK cells were treated with purified toxin and incubated at 37 degrees C for up to 24h. Exposure to epsilon-toxin caused a time-dependent decrease in cell-cell and cell-substrate interactions. After 30min of treatment retraction of the cell body and the emission of filopodia were detectable in a number of cells. Longer exposure resulted in cell rounding and cell blebbing which reached a maximum after 5h of toxin treatment. A parallel modification in the cytoskeleton was also detected. Actin marginalization and the entanglement of microtubules and intermediate filaments were observed by fluorescence microscopy after 30min of toxin exposure. Functional alterations of the plasma membrane of MDCK cells were assessed by flow cytometry. After 10 or 30min of intoxication an increase in cell volume was detected, indicating an alteration in plasma membrane permeability. These findings provide evidence for cytoskeletal changes and plasma membrane functional alterations in the in vitro cell response to C. perfringens epsilon-toxin.


Asunto(s)
Toxinas Bacterianas/toxicidad , Permeabilidad de la Membrana Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Clostridium perfringens/química , Citoesqueleto/efectos de los fármacos , Animales , Anticuerpos Monoclonales/inmunología , Toxinas Bacterianas/inmunología , Recuento de Células , Línea Celular , Membrana Celular/patología , Membrana Celular/fisiología , Citoesqueleto/patología , Citometría de Flujo/veterinaria , Cinética , Potenciales de la Membrana/efectos de los fármacos , Microscopía Fluorescente/veterinaria
15.
FEBS Lett ; 527(1-3): 269-73, 2002 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-12220672

RESUMEN

We report a study on the regulation by 2-chloro adenosine (2CA) of a glutamate (Glu) transporter in myogenic C2C12 cells. Long-term 2CA exposition significantly increased the V(max) of the Glu transporter. Moreover, 2CA-treated cells responded to Glu challenge by a rapid and transient increase in their intracellular calcium level. The above reported effects were totally abolished by treating C2C12 cells with the Na(+)-dependent Glu transporter inhibitors DL-threo-b-hydroxyaspartic acid and L-trans-pyrrolidine-2,4-dicarboxylic acid. We propose that the possible link between the Glu uptake increase and the Glu induction of calcium rise could be the depolarizing currents carried by Na(+) coupled with transporter activity.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Calcio/metabolismo , Ácido Glutámico/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Simportadores/metabolismo , 2-Cloroadenosina/farmacología , Sistema de Transporte de Aminoácidos X-AG/efectos de los fármacos , Animales , Ácido Aspártico/análogos & derivados , Ácido Aspártico/farmacología , Transporte Biológico/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cicloheximida/farmacología , Ácidos Dicarboxílicos/farmacología , Proteínas de Transporte de Glutamato en la Membrana Plasmática , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Purinas/metabolismo , Pirrolidinas/farmacología , Simportadores/efectos de los fármacos , Regulación hacia Arriba
16.
Int J Med Microbiol ; 291(6-7): 551-4, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11890557

RESUMEN

Some pathogenic Escherichia coli strains produce a protein toxin, named cytotoxic necrotizing factor 1 (CNF1), which permanently activates proteins belonging to the Rho family. In epithelial cells, the consequence of this activation is the rearrangement of the actin cytoskeleton and the promotion of an intense and generalized ruffling activity. This leads, in turn, to the induction of a phagocytic-like behavior called macropinocytosis that, in the case of CNF1, depends on the coordinate activation of Rho, Rac and Cdc42. Following internalization, the ingested material is discharged into Rab-7 and Lamp-1-positive acidic vesicles where it probably undergoes degradation. By exerting this activity, CNF1-activated epithelial cells might support the scavenging activity of macrophages during bacterial overgrowth.


Asunto(s)
Apoptosis , Toxinas Bacterianas/metabolismo , Citotoxinas/metabolismo , Endocitosis , Células Epiteliales/fisiología , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Antígenos CD , Toxinas Bacterianas/farmacología , Citoesqueleto/metabolismo , Citotoxinas/farmacología , Células Epiteliales/efectos de los fármacos , Humanos , Proteínas de Membrana de los Lisosomas , Glicoproteínas de Membrana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA