Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 24(9): e56901, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37497756

RESUMEN

Interferons control viral infection by inducing the expression of antiviral effector proteins encoded by interferon-stimulated genes (ISGs). The field has mostly focused on identifying individual antiviral ISG effectors and defining their mechanisms of action. However, fundamental gaps in knowledge about the interferon response remain. For example, it is not known how many ISGs are required to protect cells from a particular virus, though it is theorized that numerous ISGs act in concert to achieve viral inhibition. Here, we used CRISPR-based loss-of-function screens to identify a markedly limited set of ISGs that confer interferon-mediated suppression of a model alphavirus, Venezuelan equine encephalitis virus (VEEV). We show via combinatorial gene targeting that three antiviral effectors-ZAP, IFIT3, and IFIT1-together constitute the majority of interferon-mediated restriction of VEEV, while accounting for < 0.5% of the interferon-induced transcriptome. Together, our data suggest a refined model of the antiviral interferon response in which a small subset of "dominant" ISGs may confer the bulk of the inhibition of a given virus.


Asunto(s)
Virus de la Encefalitis Equina Venezolana , Virus , Animales , Caballos , Interferones , Línea Celular , Replicación Viral , Antivirales/farmacología , Virus de la Encefalitis Equina Venezolana/fisiología
2.
Biomed Pharmacother ; 162: 114614, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37068330

RESUMEN

The continuing heavy toll of the COVID-19 pandemic necessitates development of therapeutic options. We adopted structure-based drug repurposing to screen FDA-approved drugs for inhibitory effects against main protease enzyme (Mpro) substrate-binding pocket of SARS-CoV-2 for non-covalent and covalent binding. Top candidates were screened against infectious SARS-CoV-2 in a cell-based viral replication assay. Promising candidates included atovaquone, mebendazole, ouabain, dronedarone, and entacapone, although atovaquone and mebendazole were the only two candidates with IC50s that fall within their therapeutic plasma concentration. Additionally, we performed Mpro assays on the top hits, which demonstrated inhibition of Mpro by dronedarone (IC50 18 µM), mebendazole (IC50 19 µM) and entacapone (IC50 9 µM). Atovaquone showed only modest Mpro inhibition, and thus we explored other potential mechanisms. Although atovaquone is Dihydroorotate dehydrogenase (DHODH) inhibitor, we did not observe inhibition of DHODH at the respective SARS-CoV-2 IC50. Metabolomic profiling of atovaquone treated cells showed dysregulation of purine metabolism pathway metabolite, where ecto-5'-nucleotidase (NT5E) was downregulated by atovaquone at concentrations equivalent to its antiviral IC50. Atovaquone and mebendazole are promising candidates with SARS-CoV-2 antiviral activity. While mebendazole does appear to target Mpro, atovaquone may inhibit SARS-CoV-2 viral replication by targeting host purine metabolism.


Asunto(s)
Antivirales , COVID-19 , Humanos , Antivirales/farmacología , SARS-CoV-2 , Dihidroorotato Deshidrogenasa , Reposicionamiento de Medicamentos , Dronedarona/farmacología , Pandemias , Atovacuona/farmacología , Mebendazol/farmacología , Purinas/farmacología , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/farmacología , Simulación de Dinámica Molecular
3.
bioRxiv ; 2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36865157

RESUMEN

Interferons control viral infection by inducing the expression of antiviral effector proteins encoded by interferon-stimulated genes (ISGs). The field has mostly focused on identifying individual antiviral ISG effectors and defining their mechanisms of action. However, fundamental gaps in knowledge about the interferon response remain. For example, it is not known how many ISGs are required to protect cells from a particular virus, though it is theorized that numerous ISGs act in concert to achieve viral inhibition. Here, we used CRISPR-based loss-of-function screens to identify a markedly limited set of ISGs that confer interferon-mediated suppression of a model alphavirus, Venezuelan equine encephalitis virus (VEEV). We show via combinatorial gene targeting that three antiviral effectors - ZAP, IFIT3, and IFIT1 - together constitute the majority of interferon-mediated restriction of VEEV, while accounting for less than 0.5% of the interferon-induced transcriptome. Together, our data suggests a refined model of the antiviral interferon response in which a small subset of "dominant" ISGs may confer the bulk of the inhibition of a given virus.

4.
Cell Rep ; 42(2): 112076, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36753415

RESUMEN

During translation of the genomic RNA of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative virus in the COVID-19 pandemic, host ribosomes undergo programmed ribosomal frameshifting (PRF) at a conserved structural element. Although PRF is essential for coronavirus replication, host factors that regulate this process have not yet been identified. Here we perform genome-wide CRISPR-Cas9 knockout screens to identify regulators of SARS-CoV-2 PRF. These screens reveal that loss of ribosome recycling factors markedly decreases frameshifting efficiency and impairs SARS-CoV-2 viral replication. Mutational studies support a model wherein efficient removal of ribosomal subunits at the ORF1a stop codon is required for frameshifting of trailing ribosomes. This dependency upon ribosome recycling is not observed with other non-pathogenic human betacoronaviruses and is likely due to the unique position of the ORF1a stop codon in the SARS clade of coronaviruses. These findings therefore uncover host factors that support efficient SARS-CoV-2 translation and replication.


Asunto(s)
COVID-19 , Sistema de Lectura Ribosómico , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , COVID-19/metabolismo , Codón de Terminación/genética , Codón de Terminación/metabolismo , Pandemias , Replicación Viral/genética , Ribosomas/metabolismo , ARN Viral/metabolismo
5.
mBio ; 14(2): e0012723, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36809113

RESUMEN

Viruses are known to co-opt host machinery for translation initiation, but less is known about which host factors are required for the formation of ribosomes used to synthesize viral proteins. Using a loss-of-function CRISPR screen, we show that synthesis of a flavivirus-encoded fluorescent reporter depends on multiple host factors, including several 60S ribosome biogenesis proteins. Viral phenotyping revealed that two of these factors, SBDS, a known ribosome biogenesis factor, and the relatively uncharacterized protein SPATA5, were broadly required for replication of flaviviruses, coronaviruses, alphaviruses, paramyxoviruses, an enterovirus, and a poxvirus. Mechanistic studies revealed that loss of SPATA5 caused defects in rRNA processing and ribosome assembly, suggesting that this human protein may be a functional ortholog of yeast Drg1. These studies implicate specific ribosome biogenesis proteins as viral host dependency factors that are required for synthesis of virally encoded protein and accordingly, optimal viral replication. IMPORTANCE Viruses are well known for their ability to co-opt host ribosomes to synthesize viral proteins. The specific factors involved in translation of viral RNAs are not fully described. In this study, we implemented a unique genome-scale CRISPR screen to identify previously uncharacterized host factors that are important for the synthesis of virally encoded protein. We found that multiple genes involved in 60S ribosome biogenesis were required for viral RNA translation. Loss of these factors severely impaired viral replication. Mechanistic studies on the AAA ATPase SPATA5 indicate that this host factor is required for a late step in ribosome formation. These findings reveal insight into the identity and function of specific ribosome biogenesis proteins that are critical for viral infections.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Flavivirus , Humanos , Ribosomas/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , ARN Viral/genética , ARN Viral/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo
6.
Elife ; 122023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36695568

RESUMEN

Most of the cholesterol in the plasma membranes (PMs) of animal cells is sequestered through interactions with phospholipids and transmembrane domains of proteins. However, as cholesterol concentration rises above the PM's sequestration capacity, a new pool of cholesterol, called accessible cholesterol, emerges. The transport of accessible cholesterol between the PM and the endoplasmic reticulum (ER) is critical to maintain cholesterol homeostasis. This pathway has also been implicated in the suppression of both bacterial and viral pathogens by immunomodulatory oxysterols. Here, we describe a mechanism of depletion of accessible cholesterol from PMs by the oxysterol 25-hydroxycholesterol (25HC). We show that 25HC-mediated activation of acyl coenzyme A: cholesterol acyltransferase (ACAT) in the ER creates an imbalance in the equilibrium distribution of accessible cholesterol between the ER and PM. This imbalance triggers the rapid internalization of accessible cholesterol from the PM, and this depletion is sustained for long periods of time through 25HC-mediated suppression of SREBPs and continued activation of ACAT. In support of a physiological role for this mechanism, 25HC failed to suppress Zika virus and human coronavirus infection in ACAT-deficient cells, and Listeria monocytogenes infection in ACAT-deficient cells and mice. We propose that selective depletion of accessible PM cholesterol triggered by ACAT activation and sustained through SREBP suppression underpins the immunological activities of 25HC and a functionally related class of oxysterols.


Asunto(s)
Oxiesteroles , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Ratones , Oxiesteroles/metabolismo , Aciltransferasas/metabolismo , Colesterol/metabolismo , Membrana Celular/metabolismo , Bacterias/metabolismo
7.
Pediatr Pulmonol ; 57(2): 519-528, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34842360

RESUMEN

BACKGROUND: Biomarkers that can risk-stratify children with influenza virus lower respiratory infection may identify patients for targeted intervention. Early elevation of alveolar-related proteins in the bloodstream in these patients could indicate more severe lung damage portending worse outcomes. METHODS: We used a mouse model of human influenza infection and evaluated relationships between lung pathophysiology and surfactant protein D (SP-D), SP-A, and Club cell protein 16 (CC16). We then measured SP-A, SP-D, and CC16 levels in plasma samples from 94 children with influenza-associated acute respiratory failure (PICFLU cohort), excluding children with underlying conditions explaining disease severity. We tested for associations between levels of circulating proteins and disease severity including the diagnosis of acute respiratory distress syndrome (ARDS), mechanical ventilator, intensive care unit and hospital days, and hospital mortality. RESULTS: Circulating SP-D showed a greater increase than SP-A and CC16 in mice with increased alveolar-vascular permeability following influenza infection. In the PICFLU cohort, SP-D was associated with moderate-severe ARDS diagnosis (p = 0.01) and with mechanical ventilator (r = 0.45, p = 0.002), ICU (r = 0.44, p = 0.002), and hospital days (r = 0.37, p = 0.001) in influenza-infected children without bacterial coinfection. Levels of SP-D were lower in children with secondary bacterial pneumonia (p = 0.01) and not associated with outcomes. CC16 and SP-A levels did not differ with bacterial coinfection and were not consistently associated with severe outcomes. CONCLUSIONS: SP-D has potential as an early circulating biomarker reflecting a degree of lung damage caused directly by influenza virus infection in children. Secondary bacterial pneumonia alters SP-D biomarker performance.


Asunto(s)
Gripe Humana , Lesión Pulmonar , Síndrome de Dificultad Respiratoria , Animales , Biomarcadores , Niño , Humanos , Gripe Humana/complicaciones , Lesión Pulmonar/complicaciones , Ratones , Proteína D Asociada a Surfactante Pulmonar
8.
Nat Microbiol ; 3(11): 1214-1223, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30224801

RESUMEN

The endoplasmic reticulum (ER) is an architecturally diverse organelle that serves as a membrane source for the replication of multiple viruses. Flaviviruses, including yellow fever virus, West Nile virus, dengue virus and Zika virus, induce unique single-membrane ER invaginations that house the viral replication machinery1. Whether this virus-induced ER remodelling is vulnerable to antiviral pathways is unknown. Here, we show that flavivirus replication at the ER is targeted by the interferon (IFN) response. Through genome-scale CRISPR screening, we uncovered an antiviral mechanism mediated by a functional gene pairing between IFI6 (encoding IFN-α-inducible protein 6), an IFN-stimulated gene cloned over 30 years ago2, and HSPA5, which encodes the ER-resident heat shock protein 70 chaperone BiP. We reveal that IFI6 is an ER-localized integral membrane effector that is stabilized through interactions with BiP. Mechanistically, IFI6 prophylactically protects uninfected cells by preventing the formation of virus-induced ER membrane invaginations. Notably, IFI6 has little effect on other mammalian RNA viruses, including the related Flaviviridae family member hepatitis C virus, which replicates in double-membrane vesicles that protrude outwards from the ER. These findings support a model in which the IFN response is armed with a membrane-targeted effector that discriminately blocks the establishment of virus-specific ER microenvironments that are required for replication.


Asunto(s)
Antivirales/farmacología , Retículo Endoplásmico/metabolismo , Interferón-alfa/farmacología , Proteínas Mitocondriales/metabolismo , Replicación Viral , Fiebre Amarilla/metabolismo , Virus de la Fiebre Amarilla/efectos de los fármacos , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Chaperón BiP del Retículo Endoplásmico , Técnicas de Inactivación de Genes , Estudio de Asociación del Genoma Completo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas Mitocondriales/genética , Unión Proteica , Especificidad de la Especie , Fiebre Amarilla/virología , Virus de la Fiebre Amarilla/fisiología
10.
Artículo en Inglés | MEDLINE | ID: mdl-28807912

RESUMEN

MHAA4549A, a human monoclonal antibody targeting the hemagglutinin stalk region of influenza A virus (IAV), is being developed as a therapeutic for patients hospitalized with severe IAV infection. The safety and efficacy of MHAA4549A were assessed in a randomized, double-blind, placebo-controlled, dose-ranging study in a human IAV challenge model. One hundred healthy volunteers were inoculated with A/Wisconsin/67/2005 (H3N2) IAV and, 24 to 36 h later, administered a single intravenous dose of either placebo, MHAA4549A (400, 1,200, or 3,600 mg), or a standard oral dose of oseltamivir. Subjects were assessed for safety, pharmacokinetics (PK), and immunogenicity. The intent-to-treat-infected (ITTI) population was assessed for changes in viral load, influenza symptoms, and inflammatory biomarkers. MHAA4549A was well tolerated in all IAV challenge subjects. The 3,600-mg dose of MHAA4549A significantly reduced the viral burden relative to that of the placebo as determined by the area under the curve (AUC) of nasopharyngeal virus infection, quantified using quantitative PCR (98%) and 50% tissue culture infective dose (TCID50) (100%) assays. Peak viral load, duration of viral shedding, influenza symptom scores, mucus weight, and inflammatory biomarkers were also reduced. Serum PK was linear with a half-life of ∼23 days. No MHAA4549A-treated subjects developed anti-drug antibodies. In conclusion, MHAA4549A was well tolerated and demonstrated statistically significant and substantial antiviral activity in an IAV challenge model. (This study has been registered at ClinicalTrials.gov under identifier NCT01980966.).


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antivirales/efectos adversos , Antivirales/farmacología , Gripe Humana/tratamiento farmacológico , Adulto , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Neutralizantes/farmacología , Antivirales/farmacocinética , Farmacorresistencia Viral/efectos de los fármacos , Voluntarios Sanos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Masculino , Enfermedades Nasofaríngeas/virología , Resultado del Tratamiento , Carga Viral , Esparcimiento de Virus , Adulto Joven
11.
Elife ; 42015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25821985

RESUMEN

Most kinesins transport cargoes bound to their C-termini and use N-terminal motor domains to move along microtubules. We report here a novel function for KIF1C: it transports Rab6A-vesicles and can influence Golgi complex organization. These activities correlate with KIF1C's capacity to bind the Golgi protein Rab6A directly, both via its motor domain and C-terminus. Rab6A binding to the motor domain inhibits microtubule interaction in vitro and in cells, decreasing the amount of motile KIF1C. KIF1C depletion slows protein delivery to the cell surface, interferes with vesicle motility, and triggers Golgi fragmentation. KIF1C can protect Golgi membranes from fragmentation in cells lacking an intact microtubule network. Rescue of fragmentation requires sequences that enable KIF1C to bind Rab6A at both ends, but not KIF1C motor function. Rab6A binding to KIF1C's motor domain represents an entirely new mode of regulation for a kinesin motor, and likely has important consequences for KIF1C's cellular functions.


Asunto(s)
Aparato de Golgi/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Sitios de Unión , Chlorocebus aethiops , Aparato de Golgi/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Nocodazol/farmacología , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Imagen de Lapso de Tiempo , Transfección , Células Vero
12.
J Immunol ; 194(7): 3236-45, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25710914

RESUMEN

Type I IFN production is an important host immune response against viral and bacterial infections. However, little is known about the ligands and corresponding host receptors that trigger type I IFN production during bacterial infections. We used a model intracellular pathogen, Francisella novicida, to begin characterizing the type I IFN response to bacterial pathogens. F. novicida replicates in the cytosol of host cells and elicits a robust type I IFN response that is largely TLR independent, but is dependent on the adapter molecule STING, suggesting that the type I IFN stimulus during F. novicida infection is cytosolic. In this study, we report that the cytosolic DNA sensors, cyclic GMP-AMP synthase (cGAS) and Ifi204, are both required for the STING-dependent type I IFN response to F. novicida infection in both primary and immortalized murine macrophages. We created cGAS, Ifi204, and Sting functional knockouts in RAW264.7 macrophages and demonstrated that cGAS and Ifi204 cooperate to sense dsDNA and activate the STING-dependent type I IFN pathway. In addition, we show that dsDNA from F. novicida is an important type I IFN stimulating ligand. One outcome of cGAS-STING signaling is the activation of the absent in melanoma 2 inflammasome in response to F. novicida infection. Whereas the absent in melanoma 2 inflammasome is beneficial to the host during F. novicida infection, type I IFN signaling by STING and IFN regulatory factor 3 is detrimental to the host during F. novicida infection. Collectively, our studies indicate that cGAS and Ifi204 cooperate to sense cytosolic dsDNA and F. novicida infection to produce a strong type I IFN response.


Asunto(s)
Francisella/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/metabolismo , Interferón Tipo I/metabolismo , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/metabolismo , Fosfoproteínas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Línea Celular , Citosol/inmunología , Citosol/metabolismo , ADN/inmunología , Modelos Animales de Enfermedad , Expresión Génica , Regulación de la Expresión Génica , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/mortalidad , Inflamasomas/metabolismo , Factor 3 Regulador del Interferón/deficiencia , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Nucleotidiltransferasas/genética , Fosfoproteínas/genética , Unión Proteica , Interferencia de ARN , Transducción de Señal
13.
Gut Microbes ; 3(2): 62-70, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22198618

RESUMEN

The innate immune system provides the first line of defense against invading microorganisms by inducing a variety of inflammatory and antimicrobial responses. These responses are particularly important in the gastrointestinal tract, where the needs for efficient nutrient uptake and host defense collide. Many pathogens have evolved to specifically colonize the intestine, causing millions of cases of enteric infections a year. A paradigm of an enteric pathogen is Salmonella enterica, a gram-negative bacterium that causes a wide range of gastrointestinal and systemic diseases. Infections with Salmonella enterica serovar Typhimurium (S. typhimurium) lead to an acute intestinal inflammation in human and animal hosts, as a result of the bacterium invading the mucosa. A distinctive feature of Salmonella is that it has not only adapted to survive in a strong inflammatory environment, but it also uses this adaptation as a strategy to gain a growth advantage over the intestinal microbiota. We will use the model organism S. typhimurium to discuss the innate immune mechanisms employed by the mammalian gastrointestinal system and how the pathogen responds and subverts these mechanisms. In particular, we focus on the recognition of extra- and intra-cellular Salmonellae by germline-encoded pattern recognition receptors of the TLR and NLR families, and how Salmonella might profit from the activation of these receptors.


Asunto(s)
Inmunidad Innata , Salmonelosis Animal/inmunología , Salmonelosis Animal/patología , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/patología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Animales , Humanos , Evasión Inmune , Infecciones por Salmonella/microbiología , Salmonelosis Animal/microbiología
14.
Cell Host Microbe ; 4(5): 434-46, 2008 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-18996344

RESUMEN

The Salmonella typhimurium type III secretion effector protein SifA is essential for inducing tubulation of the Salmonella phagosome and binds the mammalian kinesin-binding protein SKIP. Coexpression of SifA with the effector SseJ induced tubulation of mammalian cell endosomes, similar to that induced by Salmonella infection. Interestingly, GTP-bound RhoA, RhoB, and RhoC also induced endosomal tubulation when coexpressed with SseJ, indicating that SifA likely mimics or activates a RhoA family GTPase. The structure of SifA in complex with the PH domain of SKIP revealed that SifA has two distinct domains; the amino terminus binds SKIP, and the carboxyl terminus has a fold similar to SopE, a Salmonella effector with Rho GTPase guanine nucleotide exchange factor activity (GEF). Similar to GEFs, SifA interacted with GDP-bound RhoA, and purified SseJ and RhoA formed a protein complex, suggesting that SifA, SKIP, SseJ, and RhoA family GTPases cooperatively promote host membrane tubulation.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Endosomas/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteínas Bacterianas/genética , Endosomas/química , Endosomas/genética , Glicoproteínas/genética , Células HeLa , Humanos , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Familia de Multigenes , Monoéster Fosfórico Hidrolasas/genética , Unión Proteica , Estructura Terciaria de Proteína , Infecciones por Salmonella/microbiología , Salmonella typhimurium/química , Salmonella typhimurium/genética , Proteínas de Unión al GTP rho/genética
15.
Infect Immun ; 76(6): 2722-35, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18411289

RESUMEN

Salmonella enterica serovar Typhimurium grows within host cells in a permissive compartment termed the Salmonella-containing vacuole (SCV). These bacteria use two distinct type III secretion systems (T3SS) to deliver virulence proteins (effectors) into cells. Effectors secreted by the Salmonella pathogenicity island 1 (SPI-1)-encoded T3SS mediate invasion and early SCV maturation steps, while those secreted by the SPI-2 T3SS affect the SCV at later stages postinfection. Some SPI-2 effectors modulate microtubule motor activity on the SCV. Here, we show that the actin-based motor myosin II also affects SCV dynamics during infection. Following invasion, myosin II is required for SCV positioning near the nucleus of host cells. Later, myosin II counteracts the activities of the SPI-2 effectors PipB2 and SseJ to maintain SCV positioning and stability, respectively. Myosin II activity was required for maximal bacterial growth in macrophages. Rho kinase activity was required for SCV positioning. The effector SopB, a known activator of Rho GTPases, was found to be required for SCV positioning, and transfection of cells with SopB was sufficient to induce myosin II phosphorylation. These studies reveal a novel role for myosin II in controlling SCV dynamics during infection and suggest that SopB activates myosin II.


Asunto(s)
Miosina Tipo II/metabolismo , Salmonella typhimurium/fisiología , Vacuolas/microbiología , Animales , Proteínas Bacterianas/metabolismo , División Celular/fisiología , Núcleo Celular , Regulación de la Expresión Génica , Células HeLa , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Macrófagos/microbiología , Ratones , Miosina Tipo II/antagonistas & inhibidores , Miosina Tipo II/genética , Salmonella typhimurium/citología , Salmonella typhimurium/patogenicidad , Vacuolas/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
16.
Nat Rev Microbiol ; 6(1): 53-66, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18026123

RESUMEN

Salmonellae are important causes of enteric diseases in all vertebrates. Characterization of the molecular mechanisms that underpin the interactions of salmonellae with their animal hosts has advanced greatly over the past decade, mainly through the study of Salmonella enterica serovar Typhimurium in tissue culture and animal models of infection. Knowledge of these bacterial processes and host responses has painted a dynamic and complex picture of the interaction between salmonellae and animal cells. This Review focuses on the molecular mechanisms of these host-pathogen interactions, in terms of their context, significance and future perspectives.


Asunto(s)
Interacciones Huésped-Patógeno , Enfermedades Intestinales/microbiología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/fisiología , Animales , Humanos
17.
Infect Immun ; 73(10): 6249-59, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16177296

RESUMEN

Salmonella enterica serovar Typhimurium utilizes a type III secretion system (TTSS) encoded on Salmonella pathogenicity island-2 (SPI2) to promote intracellular replication during infection, but little is known about the molecular function of SPI2-translocated effectors and how they contribute to this process. SseJ is a SPI2 TTSS effector protein that is homologous to enzymes called glycerophospholipid-cholesterol acyltransferases and, following translocation, localizes to the Salmonella-containing vacuole and Salmonella-induced filaments. Full virulence requires SseJ, as sseJ null mutants exhibit decreased replication in cultured cells and host tissues. This work demonstrates that SseJ is an enzyme with deacylase activity in vitro and identifies three active-site residues. Catalytic SseJ mutants display wild-type translocation and subcellular localization but fail to complement the virulence defect of an sseJ null mutant. In contrast to the wild type, SseJ catalytic mutants fail to down regulate Salmonella-induced filament formation and fail to restore the sifA null mutant phenotype of loss of phagosomal membrane to sifA sseJ null double mutants, suggesting that wild-type SseJ modifies the vacuolar membrane. This is the first demonstration of an enzymatic activity for a SPI2 effector protein and provides support for the hypothesis that the deacylation of lipids on the Salmonella-containing vacuole membrane is important to bacterial pathogenesis.


Asunto(s)
Aciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Salmonella typhimurium/enzimología , Salmonella typhimurium/patogenicidad , Aciltransferasas/análisis , Aciltransferasas/genética , Animales , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Dominio Catalítico , Femenino , Prueba de Complementación Genética , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Transporte de Proteínas , Vacuolas/enzimología , Virulencia
18.
Infect Immun ; 73(2): 1204-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15664965

RESUMEN

Salmonella enterica serovar Typhimurium has the fascinating ability to form tubular structures known as Salmonella-induced filaments (Sifs) in host cells. Here, we show that the prevalence of the Sif phenotype in HeLa cells is affected by host cell density, growth, and the multiplicity of infection. Sif formation was observed in cells that displayed rapid intracellular bacterial replication and was found to be dynamic, being maximal 8 to 10 h postinfection and declining thereafter. The virulence factors SpvB and SseJ were found to negatively modulate Sif formation. Our findings demonstrate the complex and dynamic nature of the Sif phenotype.


Asunto(s)
División Celular/fisiología , Membrana Celular/fisiología , Células Epiteliales/microbiología , Salmonella typhimurium/fisiología , Humanos , Infecciones por Salmonella , Salmonella typhimurium/citología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...