Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Clin Rheumatol ; 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39264828

RESUMEN

BACKGROUND: Hospital-acquired bacterial infections are associated with high morbidity and mortality rates in patients with systemic lupus erythematosus (SLE). This study aimed to develop and validate predictive models for the risk of hospital-acquired bacterial infections in patients with SLE. METHODS: A historical cohort study was designed for development, and another bidirectional cohort study was used for external validation. The risk of bacterial infection was assessed upon admission and after 5 days of hospitalization. Predictor selection employed the least absolute shrinkage and selection operator (LASSO) techniques. Multiple imputations were used to handle missing data. Logistic regression models were applied, and the properties of discrimination, calibration, and decision curve analysis were evaluated. RESULTS: The development cohort comprised 1686 patients and 237 events (14.1%) from 3 tertiary hospitals. The external validation cohort included 531 patients and 84 infection outcomes (15.8%) from 10 hospital centers in Colombia (secondary and tertiary level). The models applied at admission and after 120 hours of stay exhibited good discrimination (AUC > 0.74). External validation demonstrated good performance among patients from the same tertiary institutions where the models were developed. However, geographic validation at other institutions has been suboptimal. CONCLUSIONS: Two predictive models for nosocomial bacterial infections in patients with SLE are presented. All infection prevention recommendations should be maximized in patients at moderate/high risk. Further validation studies in diverse contexts, as well as clinical impact trials, are necessary before potential applications in research and clinical care.

2.
PLoS Pathog ; 17(9): e1009488, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34492091

RESUMEN

Arenavirus entry into host cells occurs through a low pH-dependent fusion with late endosomes that is mediated by the viral glycoprotein complex (GPC). The mechanisms of GPC-mediated membrane fusion and of virus targeting to late endosomes are not well understood. To gain insights into arenavirus fusion, we examined cell-cell fusion induced by the Old World Lassa virus (LASV) GPC complex. LASV GPC-mediated cell fusion is more efficient and occurs at higher pH with target cells expressing human LAMP1 compared to cells lacking this cognate receptor. However, human LAMP1 is not absolutely required for cell-cell fusion or LASV entry. We found that GPC-induced fusion progresses through the same lipid intermediates as fusion mediated by other viral glycoproteins-a lipid curvature-sensitive intermediate upstream of hemifusion and a hemifusion intermediate downstream of acid-dependent steps that can be arrested in the cold. Importantly, GPC-mediated fusion and LASV pseudovirus entry are specifically augmented by an anionic lipid, bis(monoacylglycero)phosphate (BMP), which is highly enriched in late endosomes. This lipid also specifically promotes cell fusion mediated by Junin virus GPC, an unrelated New World arenavirus. We show that BMP promotes late steps of LASV fusion downstream of hemifusion-the formation and enlargement of fusion pores. The BMP-dependence of post-hemifusion stages of arenavirus fusion suggests that these viruses evolved to use this lipid as a cofactor to selectively fuse with late endosomes.


Asunto(s)
Endosomas/metabolismo , Fiebre de Lassa/metabolismo , Virus Lassa/fisiología , Lisofosfolípidos/metabolismo , Monoglicéridos/metabolismo , Internalización del Virus , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Proteínas del Envoltorio Viral/metabolismo
3.
Sensors (Basel) ; 22(21)2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36365951

RESUMEN

The aim of this study is the preparation of nanostructured copper(II) oxide-based materials (CuONPs) through a facile additive-free polyol procedure that consists of the hydrolysis of copper(II) acetate in 1,4-butane diol and its application in hydrogen peroxide sensing. The nonenzymatic electrochemical sensor for hydrogen peroxide determination was constructed by drop casting the CuONP sensing material on top of a glassy carbon electrode (GCE) modified by a layer of poly(3,4-ethylenedioxythiophene) conducting polymer (PEDOT). The PEDOT layer was prepared on GCE using the sinusoidal voltage method. The XRD pattern of the CuONPs reveals the formation of the monoclinic tenorite phase, CuO, with average crystallite sizes of 8.7 nm, while the estimated band gap from UV-vis spectroscopy is of 1.2 eV. The SEM, STEM, and BET analyses show the formation of quasi-prismatic microaggregates of nanoparticles, with dimensions ranging from 1 µm up to ca. 200 µm, with a mesoporous structure. The developed electrochemical sensor exhibited a linear response toward H2O2 in the concentration range from 0.04 to 10 mM, with a low detection limit of 8.5 µM of H2O2. Furthermore, the obtained sensor possessed an excellent anti-interference capability in H2O2 determination in the presence of interfering compounds such as KNO3 and KNO2.


Asunto(s)
Peróxido de Hidrógeno , Nanopartículas , Peróxido de Hidrógeno/química , Óxidos/química , Electrodos , Nanopartículas/química , Carbono/química , Técnicas Electroquímicas/métodos
4.
PLoS Pathog ; 15(1): e1007532, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30640957

RESUMEN

Late endosome-resident interferon-induced transmembrane protein 3 (IFITM3) inhibits fusion of diverse viruses, including Influenza A virus (IAV), by a poorly understood mechanism. Despite the broad antiviral activity of IFITM3, viruses like Lassa virus (LASV), are fully resistant to its inhibitory effects. It is currently unclear whether resistance arises from a highly efficient fusion machinery that is capable of overcoming IFITM3 restriction or the ability to enter from cellular sites devoid of this factor. Here, we constructed and validated a functional IFITM3 tagged with EGFP or other fluorescent proteins. This breakthrough allowed live cell imaging of virus co-trafficking and fusion with endosomal compartments in cells expressing fluorescent IFITM3. Three-color single virus and endosome tracking revealed that sensitive (IAV), but not resistant (LASV), viruses become trapped within IFITM3-positive endosomes where they underwent hemifusion but failed to release their content into the cytoplasm. IAV fusion with IFITM3-containing compartments could be rescued by amphotericin B treatment, which has been previously shown to antagonize the antiviral activity of this protein. By comparison, virtually all LASV particles trafficked and fused with endosomes lacking detectable levels of fluorescent IFITM3, implying that this virus escapes restriction by utilizing endocytic pathways that are distinct from the IAV entry pathways. The importance of virus uptake and transport pathways is further reinforced by the observation that LASV glycoprotein-mediated cell-cell fusion is inhibited by IFITM3 and other members of the IFITM family expressed in target cells. Together, our results strongly support a model according to which IFITM3 accumulation at the sites of virus fusion is a prerequisite for its antiviral activity and that this protein traps viral fusion at a hemifusion stage by preventing the formation of fusion pores. We conclude that the ability to utilize alternative endocytic pathways for entry confers IFITM3-resistance to otherwise sensitive viruses.


Asunto(s)
Endosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/fisiología , Células A549/metabolismo , Animales , Antivirales/metabolismo , Células COS/metabolismo , Chlorocebus aethiops , Endosomas/virología , Células HEK293/metabolismo , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/patogenicidad , Interferones/metabolismo , Virus Lassa/patogenicidad , Imagen Óptica/métodos , Transporte de Proteínas , Internalización del Virus
5.
Sensors (Basel) ; 22(1)2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-35009659

RESUMEN

In this work, the development of an electrochemical sensor for melatonin determination is presented. The sensor was based on Sonogel-Carbon electrode material (SNGCE) and Au nanoparticles (AuNPs). The low-cost and environmentally friendly SNGCE material was prepared by the ultrasound-assisted sonogel method. AuNPs were prepared by a chemical route and narrow size distribution was obtained. The electrochemical characterization of the SNGCE/AuNP sensor was carried out by cyclic voltammetry in the presence of a redox probe. The analytical performance of the SNGCE/AuNP sensor in terms of linear response range, repeatability, selectivity, and limit of detection was investigated. The optimized SNGCE/AuNP sensor displayed a low detection limit of 8.4 nM melatonin in synthetic samples assessed by means of the amperometry technique. The potential use of the proposed sensor in real sample analysis and the anti-matrix capability were assessed by a recovery study of melatonin detection in human peripheral blood serum with good accuracy.


Asunto(s)
Melatonina , Nanopartículas del Metal , Carbono , Técnicas Electroquímicas , Electrodos , Oro , Humanos , Límite de Detección
6.
J Biol Chem ; 292(19): 7817-7827, 2017 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-28341742

RESUMEN

Enveloped viruses infect host cells by fusing their membranes with those of the host cell, a process mediated by viral glycoproteins upon binding to cognate host receptors or entering into acidic intracellular compartments. Whereas the effect of receptor density on viral infection has been well studied, the role of cell type-specific factors/processes, such as pH regulation, has not been characterized in sufficient detail. Here, we examined the effects of cell-extrinsic factors (buffer environment) and cell-intrinsic factors (interferon-inducible transmembrane proteins, IFITMs), on the pH regulation in early endosomes and on the efficiency of acid-dependent fusion of the avian sarcoma and leukosis virus (ASLV), with endosomes. First, we found that a modest elevation of external pH can raise the pH in early endosomes in a cell type-dependent manner and thereby delay the acid-induced fusion of endocytosed ASLV. Second, we observed a cell type-dependent delay between the low pH-dependent and temperature-dependent steps of viral fusion, consistent with the delayed enlargement of the fusion pore. Third, ectopic expression of IFITMs, known to potently block influenza virus fusion with late compartments, was found to only partially inhibit ASLV fusion with early endosomes. Interestingly, IFITM expression promoted virus uptake and the acidification of endosomal compartments, resulting in an accelerated fusion rate when driven by the glycosylphosphatidylinositol-anchored, but not by the transmembrane isoform of the ASLV receptor. Collectively, these results highlight the role of cell-extrinsic and cell-intrinsic factors in regulating the efficiency and kinetics of virus entry and fusion with target cells.


Asunto(s)
Virus del Sarcoma Aviar/fisiología , Fusión de Membrana , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Internalización del Virus , Células A549 , Ácidos/química , Animales , Línea Celular , Endocitosis , Endosomas/metabolismo , Regulación Viral de la Expresión Génica , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Isoformas de Proteínas/metabolismo , Receptores Virales/metabolismo , Temperatura , Vesículas Transportadoras/metabolismo
7.
J Biol Chem ; 292(14): 6014-6026, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28179429

RESUMEN

The host proteins, SERINC3 and SERINC5, have been recently shown to incorporate into HIV-1 particles and compromise their ability to fuse with target cells, an effect that is antagonized by the viral Nef protein. Envelope (Env) glycoproteins from different HIV-1 isolates exhibit a broad range of sensitivity to SERINC-mediated restriction, and the mechanism by which SERINCs interfere with HIV-1 fusion remains unclear. Here, we show that incorporation of SERINC5 into virions in the absence of Nef inhibits the formation of small fusion pores between viruses and cells. Strikingly, we found that SERINC5 promotes spontaneous functional inactivation of sensitive but not resistant Env glycoproteins. Although SERINC5-Env interaction was not detected by co-immunoprecipitation, incorporation of this protein enhanced the exposure of the conserved gp41 domains and sensitized the virus to neutralizing antibodies and gp41-derived inhibitory peptides. These results imply that SERINC5 restricts HIV-1 fusion at a step prior to small pore formation by selectively inactivating sensitive Env glycoproteins, likely through altering their conformation. The increased HIV-1 sensitivity to anti-gp41 antibodies and peptides suggests that SER5 also delays refolding of the remaining fusion-competent Env trimers.


Asunto(s)
Proteína gp41 de Envoltorio del VIH/metabolismo , VIH-1/metabolismo , Proteínas de la Membrana/metabolismo , Replegamiento Proteico , Células HEK293 , Anticuerpos Anti-VIH/farmacología , Proteína gp41 de Envoltorio del VIH/genética , VIH-1/genética , Humanos , Proteínas de la Membrana/genética , Dominios Proteicos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
8.
PLoS Pathog ; 12(6): e1005709, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27322072

RESUMEN

Disassembly of the cone-shaped HIV-1 capsid in target cells is a prerequisite for establishing a life-long infection. This step in HIV-1 entry, referred to as uncoating, is critical yet poorly understood. Here we report a novel strategy to visualize HIV-1 uncoating using a fluorescently tagged oligomeric form of a capsid-binding host protein cyclophilin A (CypA-DsRed), which is specifically packaged into virions through the high-avidity binding to capsid (CA). Single virus imaging reveals that CypA-DsRed remains associated with cores after permeabilization/removal of the viral membrane and that CypA-DsRed and CA are lost concomitantly from the cores in vitro and in living cells. The rate of loss is modulated by the core stability and is accelerated upon the initiation of reverse transcription. We show that the majority of single cores lose CypA-DsRed shortly after viral fusion, while a small fraction remains intact for several hours. Single particle tracking at late times post-infection reveals a gradual loss of CypA-DsRed which is dependent on reverse transcription. Uncoating occurs both in the cytoplasm and at the nuclear membrane. Our novel imaging assay thus enables time-resolved visualization of single HIV-1 uncoating in living cells, and reveals the previously unappreciated spatio-temporal features of this incompletely understood process.


Asunto(s)
Infecciones por VIH , VIH-1 , Imagen de Lapso de Tiempo/métodos , Desencapsidación Viral/fisiología , Western Blotting , Ciclofilina A , Colorantes Fluorescentes , Células HEK293 , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía Confocal , Imagen Óptica/métodos
9.
J Biol Chem ; 290(10): 6558-73, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25589785

RESUMEN

Whether HIV-1 enters cells by fusing with the plasma membrane or with endosomes is a subject of active debate. The ability of HIV-1 to mediate fusion between adjacent cells, a process referred to as "fusion-from-without" (FFWO), shows that this virus can fuse with the plasma membrane. To compare FFWO occurring at the cell surface with HIV-cell fusion through a conventional entry route, we designed an experimental approach that enabled the measurements of both processes in the same sample. The following key differences were observed. First, a very small fraction of viruses fusing with target cells participated in FFWO. Second, whereas HIV-1 fusion with adherent cells was insensitive to actin inhibitors, post-CD4/coreceptor binding steps during FFWO were abrogated. A partial dependence of HIV-cell fusion on actin remodeling was observed in CD4(+) T cells, but this effect appeared to be due to the actin dependence of virus uptake. Third, deletion of the cytoplasmic tail of HIV-1 gp41 dramatically enhanced the ability of the virus to promote FFWO, while having a modest effect on virus-cell fusion. Distinct efficiencies and actin dependences of FFWO versus HIV-cell fusion are consistent with the notion that, except for a minor fraction of particles that mediate fusion between the plasma membranes of adjacent cells, HIV-1 enters through an endocytic pathway. We surmise, however, that cell-cell contacts enabling HIV-1 fusion with the plasma membrane could be favored at the sites of high density of target cells, such as lymph nodes.


Asunto(s)
Endocitosis/genética , Infecciones por VIH/genética , VIH-1/genética , Fusión de Membrana/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/virología , Citoesqueleto/genética , Citoesqueleto/metabolismo , Endosomas/metabolismo , Endosomas/virología , Células HEK293 , Proteína gp41 de Envoltorio del VIH/metabolismo , Infecciones por VIH/virología , VIH-1/patogenicidad , Humanos , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo
10.
J Virol ; 89(18): 9368-82, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26136569

RESUMEN

UNLABELLED: HIV-1 Env glycoprotein-mediated fusion is initiated upon sequential binding of Env to CD4 and the coreceptor CXCR4 or CCR5. Whereas these interactions are thought to be necessary and sufficient to promote HIV-1 fusion, other host factors can modulate this process. Previous studies reported potent inhibition of HIV-1 fusion by selective P2X1 receptor antagonists, including NF279, and suggested that these receptors play a role in HIV-1 entry. Here we investigated the mechanism of antiviral activity of NF279 and found that this compound does not inhibit HIV-1 fusion by preventing the activation of P2X1 channels but effectively blocks the binding of the virus to CXCR4 or CCR5. The notion of an off-target effect of NF279 on HIV-1 fusion is supported by the lack of detectable expression of P2X1 receptors in cells used in fusion experiments and by the fact that the addition of ATP or the enzymatic depletion of ATP in culture medium does not modulate viral fusion. Importantly, NF279 fails to inhibit HIV-1 fusion with cell lines and primary macrophages when added at an intermediate stage downstream of Env-CD4-coreceptor engagement. Conversely, in the presence of NF279, HIV-1 fusion is arrested downstream of CD4 binding but prior to coreceptor engagement. NF279 also antagonizes the signaling function of CCR5, CXCR4, and another chemokine receptor, as evidenced by the suppression of calcium responses elicited by specific ligands and by recombinant gp120. Collectively, our results demonstrate that NF279 is a dual HIV-1 coreceptor inhibitor that interferes with the functional engagement of CCR5 and CXCR4 by Env. IMPORTANCE: Inhibition of P2X receptor activity suppresses HIV-1 fusion and replication, suggesting that P2X signaling is involved in HIV-1 entry. However, mechanistic experiments conducted in this study imply that P2X1 receptor is not expressed in target cells or involved in viral fusion. Instead, we found that inhibition of HIV-1 fusion by a specific P2X1 receptor antagonist, NF279, is due to the blocking of virus interactions with both the CXCR4 and CCR5 coreceptors. The ability of NF279 to abrogate cellular calcium signaling induced by the respective chemokines showed that this compound acts as a dual-coreceptor antagonist. P2X1 receptor antagonists could thus represent a new class of dual-coreceptor inhibitors with a structure and a mechanism of action that are distinct from those of known HIV-1 coreceptor antagonists.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , VIH-1/metabolismo , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Suramina/análogos & derivados , Internalización del Virus/efectos de los fármacos , Linfocitos T CD4-Positivos/virología , Femenino , Células HEK293 , Humanos , Masculino , Receptores Purinérgicos P2X1/metabolismo , Suramina/farmacología
11.
PLoS Pathog ; 10(4): e1004048, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24699674

RESUMEN

Interferon-induced transmembrane proteins (IFITMs) inhibit infection of diverse enveloped viruses, including the influenza A virus (IAV) which is thought to enter from late endosomes. Recent evidence suggests that IFITMs block virus hemifusion (lipid mixing in the absence of viral content release) by altering the properties of cell membranes. Consistent with this mechanism, excess cholesterol in late endosomes of IFITM-expressing cells has been reported to inhibit IAV entry. Here, we examined IAV restriction by IFITM3 protein using direct virus-cell fusion assay and single virus imaging in live cells. IFITM3 over-expression did not inhibit lipid mixing, but abrogated the release of viral content into the cytoplasm. Although late endosomes of IFITM3-expressing cells accumulated cholesterol, other interventions leading to aberrantly high levels of this lipid did not inhibit virus fusion. These results imply that excess cholesterol in late endosomes is not the mechanism by which IFITM3 inhibits the transition from hemifusion to full fusion. The IFITM3's ability to block fusion pore formation at a post-hemifusion stage shows that this protein stabilizes the cytoplasmic leaflet of endosomal membranes without adversely affecting the lumenal leaflet. We propose that IFITM3 interferes with pore formation either directly, through partitioning into the cytoplasmic leaflet of a hemifusion intermediate, or indirectly, by modulating the lipid/protein composition of this leaflet. Alternatively, IFITM3 may redirect IAV fusion to a non-productive pathway, perhaps by promoting fusion with intralumenal vesicles within multivesicular bodies/late endosomes.


Asunto(s)
Endosomas/metabolismo , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Fusión de Membrana , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Internalización del Virus , Animales , Células CHO , Colesterol/genética , Colesterol/inmunología , Colesterol/metabolismo , Cricetinae , Cricetulus , Endosomas/genética , Endosomas/inmunología , Células HEK293 , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Gripe Humana/genética , Gripe Humana/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología
12.
Retrovirology ; 12: 88, 2015 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-26511606

RESUMEN

BACKGROUND: HIV-1 Vpr is recruited into virions during assembly and appears to remain associated with the viral core after the reverse transcription and uncoating steps of entry. This feature has prompted the use of fluorescently labeled Vpr to visualize viral particles and to follow trafficking of post-fusion HIV-1 cores in the cytoplasm. RESULTS: Here, we tracked single pseudovirus entry and fusion and observed that fluorescently tagged Vpr gradually dissociates from post-fusion viral cores over the course of several minutes and accumulates in the nucleus. Kinetics measurements showed that fluorescent Vpr released from the cores very rapidly entered the cell nucleus. More than 10,000 Vpr molecules can be delivered into the cell nucleus within 45 min of infection by HIV-1 particles pseudotyped with the avian sarcoma and leukosis virus envelope glycoprotein. The fraction of Vpr from cell-bound viruses that accumulated in the nucleus was proportional to the extent of virus-cell fusion and was fully blocked by viral fusion inhibitors. Entry of virus-derived Vpr into the nucleus occurred independently of envelope glycoproteins or target cells. Fluorescence correlation spectroscopy revealed two forms of nuclear Vpr-monomers and very large complexes, likely involving host factors. The kinetics of viral Vpr entering the nucleus after fusion was not affected by point mutations in the capsid protein that alter the stability of the viral core. CONCLUSIONS: The independence of Vpr shedding of capsid stability and its relatively rapid dissociation from post-fusion cores suggest that this process may precede capsid uncoating, which appears to occur on a slower time scale. Our results thus demonstrate that a bulk of fluorescently labeled Vpr incorporated into HIV-1 particles is released shortly after fusion. Future studies will address the question whether the quick and efficient nuclear delivery of Vpr derived from incoming viruses can regulate subsequent steps of HIV-1 infection.


Asunto(s)
Núcleo Celular/metabolismo , VIH-1/fisiología , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Transporte Activo de Núcleo Celular , Proteínas de la Cápside/metabolismo , Núcleo Celular/ultraestructura , Núcleo Celular/virología , Citoplasma/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Células HeLa , Humanos , Espectrometría de Fluorescencia/métodos , Virión/fisiología , Internalización del Virus , Replicación Viral
13.
PLoS Pathog ; 9(6): e1003431, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785290

RESUMEN

Human defensins are at the forefront of the host responses to HIV and other pathogens in mucosal tissues. However, their ability to inactivate HIV in the bloodstream has been questioned due to the antagonistic effect of serum. In this study, we have examined the effect of sub-inhibitory concentrations of human α-defensin HNP-1 on the kinetics of early steps of fusion between HIV-1 and target cells in the presence of serum. Direct measurements of HIV-cell fusion using an enzymatic assay revealed that, in spite of the modest effect on the extent of fusion, HNP-1 prolonged the exposure of functionally important transitional epitopes of HIV-1 gp41 on the cell surface. The increased lifetime of gp41 intermediates in the presence of defensin was caused by a delay in the post-coreceptor binding steps of HIV-1 entry that correlated with the marked enhancement of the virus' sensitivity to neutralizing anti-gp41 antibodies. By contrast, the activity of antibodies to gp120 was not affected. HNP-1 appeared to specifically potentiate antibodies and peptides targeting the first heptad repeat domain of gp41, while its effect on inhibitors and antibodies to other gp41 domains was less prominent. Sub-inhibitory concentrations of HNP-1 also promoted inhibition of HIV-1 entry into peripheral blood mononuclear cells by antibodies and, more importantly, by HIV-1 immune serum. Our findings demonstrate that: (i) sub-inhibitory doses of HNP-1 potently enhance the activity of a number of anti-gp41 antibodies and peptide inhibitors, apparently by prolonging the lifetime of gp41 intermediates; and (ii) the efficiency of HIV-1 fusion inhibitors and neutralizing antibodies is kinetically restricted. This study thus reveals an important role of α-defensin in enhancing adaptive immune responses to HIV-1 infection and suggests future strategies to augment these responses.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Anticuerpos Anti-VIH/farmacología , Proteína gp41 de Envoltorio del VIH/metabolismo , VIH-1/metabolismo , alfa-Defensinas/metabolismo , Anticuerpos Neutralizantes/inmunología , Anticuerpos Anti-VIH/inmunología , Proteína gp41 de Envoltorio del VIH/antagonistas & inhibidores , Proteína gp41 de Envoltorio del VIH/genética , Proteína gp41 de Envoltorio del VIH/inmunología , VIH-1/genética , VIH-1/inmunología , Células HeLa , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/virología , Estructura Terciaria de Proteína , Internalización del Virus/efectos de los fármacos , alfa-Defensinas/síntesis química , alfa-Defensinas/química , alfa-Defensinas/genética , alfa-Defensinas/inmunología
14.
Proc Natl Acad Sci U S A ; 109(43): 17627-32, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23047692

RESUMEN

Diverse enveloped viruses enter host cells through endocytosis and fuse with endosomal membranes upon encountering acidic pH. Currently, the pH dynamics in virus-carrying endosomes and the relationship between acidification and viral fusion are poorly characterized. Here, we examined the entry of avian retrovirus that requires two sequential stimuli--binding to a cognate receptor and low pH--to undergo fusion. A genetically encoded sensor incorporated into the viral membrane was used to measure the pH in virus-carrying endosomes. Acid-induced virus fusion was visualized as the release of a fluorescent viral content marker into the cytosol. The pH values in early acidic endosomes transporting the virus ranged from 5.6 to 6.5 but were relatively stable over time for a given vesicle. Analysis of viral motility and luminal pH showed that cells expressing the transmembrane isoform of the receptor (TVA950) preferentially sorted the virus into slowly trafficking, less acidic endosomes. In contrast, viruses internalized by cells expressing the GPI-anchored isoform (TVA800) were uniformly distributed between stationary and mobile compartments. We found that the lag times between acidification and fusion were significantly shorter and fusion pores were larger in dynamic endosomes than in more stationary compartments. Despite the same average pH within mobile compartments of cells expressing alternative receptor isoforms, TVA950 supported faster fusion than TVA800 receptor. Collectively, our results suggest that fusion steps downstream of the low-pH trigger are modulated by properties of intracellular compartments harboring the virus.


Asunto(s)
Ácidos/metabolismo , Endosomas/metabolismo , Fusión de Membrana , Retroviridae/fisiología , Animales , Línea Celular , Humanos
15.
J Biol Chem ; 288(18): 12416-25, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23493401

RESUMEN

Viral glycoproteins mediate fusion between viral and cellular membranes upon binding to cognate receptors and/or experiencing low pH. Although activation of viral glycoproteins is thought to be necessary and sufficient for fusion, accumulating evidence suggests that additional cellular factors, including lipids, can modulate the fusion process. Understanding the role of lipids in virus entry via endocytosis is impeded by poor accessibility and the highly diverse nature of endosomes. Here we imaged fusion of single retroviral particles pseudotyped with the vesicular stomatitis virus (VSV) G protein with dextran-supported lipid bilayers. Incorporation of diffusible fluorescent labels into the viral membrane and the viral interior enabled detection of the lipid mixing (hemifusion) and content transfer (full fusion) steps of VSV G-mediated fusion at low pH. Although single virus fusion with supported bilayers made of zwitterionic lipids could not be detected, inclusion of anionic lipids, phosphatidylserine, and bis(monoacylglycero)phosphate (BMP), greatly enhanced the efficiency of hemifusion and permitted full fusion. Importantly, lipid mixing always preceded the opening of a fusion pore, demonstrating that VSV G-mediated fusion proceeds through a long-lived hemifusion intermediate. Kinetic analysis of lipid and content transfer showed that the lags between lipid and content mixing defining the lifetime of a hemifusion intermediate were significantly shorter for BMP-containing compared with PS-containing bilayers. The strong fusion-enhancing effect of BMP, a late endosome-resident lipid, is consistent with the model that VSV initiates fusion in early endosomes but releases its core into the cytosol after reaching late endosomal compartments.


Asunto(s)
Membrana Dobles de Lípidos/metabolismo , Glicoproteínas de Membrana/metabolismo , Fosfolípidos/metabolismo , Vesiculovirus/fisiología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Endocitosis , Endosomas/genética , Endosomas/metabolismo , Endosomas/virología , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Cinética , Glicoproteínas de Membrana/genética , Proteínas del Envoltorio Viral/genética
16.
Retrovirology ; 11: 47, 2014 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-24935247

RESUMEN

BACKGROUND: The majority of viruses enter host cells via endocytosis. Current knowledge of viral entry pathways is largely based upon infectivity measurements following genetic and/or pharmacological interventions that disrupt vesicular trafficking and maturation. Imaging of single virus entry in living cells provides a powerful means to delineate viral trafficking pathways and entry sites under physiological conditions. RESULTS: Here, we visualized single avian retrovirus co-trafficking with markers for early (Rab5) and late (Rab7) endosomes, acidification of endosomal lumen and the resulting viral fusion measured by the viral content release into the cytoplasm. Virus-carrying vesicles either merged with the existing Rab5-positive early endosomes or slowly accumulated Rab5. The Rab5 recruitment to virus-carrying endosomes correlated with acidification of their lumen. Viral fusion occurred either in early (Rab5-positive) or intermediate (Rab5- and Rab7-positive) compartments. Interestingly, different isoforms of the cognate receptor directed virus entry from distinct endosomes. In cells expressing the transmembrane receptor, viruses preferentially entered and fused with slowly maturing early endosomes prior to accumulation of Rab7. By comparison, in cells expressing the GPI-anchored receptor, viruses entered both slowly and quickly maturing endosomes and fused with early (Rab5-positive) and intermediate (Rab5- and Rab7-positive) compartments. CONCLUSIONS: Since the rate of low pH-triggered fusion was independent of the receptor isoform, we concluded that the sites of virus entry are determined by the kinetic competition between endosome maturation and viral fusion. Our findings demonstrate the ability of this retrovirus to enter cells via alternative endocytic pathways and establish infection by releasing its content from distinct endosomal compartments.


Asunto(s)
Receptores de Superficie Celular/metabolismo , Retroviridae/fisiología , Internalización del Virus , Transporte Biológico , Línea Celular , Citoplasma/metabolismo , Citoplasma/fisiología , Citoplasma/virología , Endosomas/metabolismo , Endosomas/fisiología , Endosomas/virología , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Isoformas de Proteínas , Retroviridae/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Proteínas de Unión a GTP rab7
17.
PLoS Pathog ; 8(5): e1002694, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22589725

RESUMEN

Disparate enveloped viruses initiate infection by fusing with endosomes. However, the highly diverse and dynamic nature of endosomes impairs mechanistic studies of fusion and identification of sub-cellular sites supporting the nucleocapsid release. We took advantage of the extreme stability of avian retrovirus-receptor complexes at neutral pH and of acid-dependence of virus-endosome fusion to isolate the latter step from preceding asynchronous internalization/trafficking steps. Viruses were trapped within endosomes in the presence of NH4Cl. Removal of NH4Cl resulted in a quick and uniform acidification of all subcellular compartments, thereby initiating synchronous viral fusion. Single virus imaging demonstrated that fusion was initiated within seconds after acidification and often culminated in the release of the viral core from an endosome. Comparative studies of cells expressing either the transmembrane or GPI-anchored receptor isoform revealed that the transmembrane receptor delivered the virus to more fusion-permissive compartments. Thus the identity of endosomal compartments, in addition to their acidity, appears to modulate viral fusion. A more striking manifestation of the virus delivery to distinct compartments in the presence of NH4Cl was the viral core release into the cytosol of cells expressing the transmembrane receptor and into endosomes of cells expressing the GPI-anchored isoform. In the latter cells, the newly released cores exhibited restricted mobility and were exposed to a more acidic environment than the cytoplasm. These cores appear to enter into the cytosol after an additional slow temperature-dependent step. We conclude that the NH4Cl block traps the virus within intralumenal vesicles of late endosomes in cells expressing the GPI-anchored receptor. Viruses surrounded by more than one endosomal membrane release their core into the cytoplasm in two steps--fusion with an intralumenal vesicle followed by a yet unknown temperature-dependent step that liberates the core from late endosomes.


Asunto(s)
Virus del Sarcoma Aviar/genética , Virus del Sarcoma Aviar/metabolismo , Endosomas/virología , Proteínas del Núcleo Viral/metabolismo , Proteínas Virales de Fusión/metabolismo , Cloruro de Amonio/química , Animales , Compartimento Celular , Línea Celular , Chlorocebus aethiops , Endosomas/metabolismo , Células HEK293 , Humanos , Isoformas de Proteínas/biosíntesis , Transporte de Proteínas , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/metabolismo , Proteínas del Núcleo Viral/genética , Proteínas Virales de Fusión/genética , Internalización del Virus
18.
ACS Cent Sci ; 10(7): 1332-1347, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39071064

RESUMEN

Assays that detect viral infections play a significant role in limiting the spread of diseases such as SARS-CoV-2. Here, we present Rolosense, a virus sensing platform that leverages the motion of 5 µm DNA-based motors on RNA fuel chips to transduce the presence of viruses. Motors and chips are modified with aptamers, which are designed for multivalent binding to viral targets and lead to stalling of motion. Therefore, the motors perform a "mechanical test" of the viral target and stall in the presence of whole virions, which represents a unique mechanism of transduction distinct from conventional assays. Rolosense can detect SARS-CoV-2 spiked in artificial saliva and exhaled breath condensate with a sensitivity of 103 copies/mL and discriminates among other respiratory viruses. The assay is modular and amenable to multiplexing, as demonstrated by our one-pot detection of influenza A and SARS-CoV-2. As a proof of concept, we show that readout can be achieved using a smartphone camera with a microscopic attachment in as little as 15 min without amplification reactions. Taken together, these results show that mechanical detection using Rolosense can be broadly applied to any viral target and has the potential to enable rapid, low-cost point-of-care screening of circulating viruses.

19.
Biosensors (Basel) ; 14(7)2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-39056596

RESUMEN

The development of low-cost, sensitive, and simple analytical tools for biomolecule detection in health status monitoring is nowadays a growing research topic. Sensing platforms integrating nanocomposite materials as recognition elements in the monitoring of various biomolecules and biomarkers are addressing this challenging objective. Herein, we have developed electrochemical sensing platforms by means of a novel fabrication procedure for biomolecule detection. The platforms are based on commercially available low-cost conductive substrates like glassy carbon and/or screen-printed carbon electrodes selectively functionalized with nanocomposite materials composed of Ag and Au metallic nanoparticles and an organic polymer, poly(3,4-ethylenedioxythiophene). The novel fabrication method made use of alternating currents with controlled amplitude and frequency. The frequency of the applied alternating current was 100 mHz for the polymer deposition, while a frequency value of 50 mHz was used for the in situ electrodeposition of Ag and Au nanoparticles. The selected frequency values ensured the successful preparation of the composite materials. The use of readily available composite materials is intended to produce cost-effective analytical tools. The judicious modification of the organic conductive matrix by various metallic nanoparticles, such as Ag and Au, extends the potential applications of the sensing platform toward a range of biomolecules like quercetin and epinephrine, chosen as benchmark analytes for proof-of-concept antioxidant and neurotransmitter detection. The sensing platforms were tested successfully for quercetin and epinephrine determination on synthetic and real samples. Wide linear response ranges and low limit-of-detection values were obtained for epinephrine and quercetin detection.


Asunto(s)
Técnicas Biosensibles , Compuestos Bicíclicos Heterocíclicos con Puentes , Técnicas Electroquímicas , Epinefrina , Oro , Nanopartículas del Metal , Nanocompuestos , Polímeros , Quercetina , Quercetina/análisis , Epinefrina/análisis , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Nanopartículas del Metal/química , Polímeros/química , Oro/química , Nanocompuestos/química , Humanos , Plata/química , Electrodos
20.
Biomolecules ; 14(5)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38785977

RESUMEN

Host restriction factor SERINC5 (SER5) incorporates into the HIV-1 membrane and inhibits infectivity by a poorly understood mechanism. Recently, SER5 was found to exhibit scramblase-like activity leading to the externalization of phosphatidylserine (PS) on the viral surface, which has been proposed to be responsible for SER5's antiviral activity. This and other reports that document modulation of HIV-1 infectivity by viral lipid composition prompted us to investigate the role of PS in regulating SER5-mediated HIV-1 restriction. First, we show that the level of SER5 incorporation into virions correlates with an increase in PS levels in the outer leaflet of the viral membrane. We developed an assay to estimate the PS distribution across the viral membrane and found that SER5, but not SER2, which lacks antiviral activity, abrogates PS asymmetry by externalizing this lipid. Second, SER5 incorporation diminished the infectivity of pseudoviruses produced from cells lacking a flippase subunit CDC50a and, therefore, exhibited a higher baseline level of surface-accessible PS. Finally, exogenous manipulation of the viral PS levels utilizing methyl-alpha-cyclodextrin revealed a lack of correlation between external PS and virion infectivity. Taken together, our study implies that the increased PS exposure to SER5-containing virions itself is not directly linked to HIV-1 restriction.


Asunto(s)
VIH-1 , Proteínas de la Membrana , Fosfatidilserinas , VIH-1/metabolismo , Fosfatidilserinas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Virión/metabolismo , Células HEK293 , Membrana Celular/metabolismo , Infecciones por VIH/virología , Infecciones por VIH/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA