Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Infect Chemother ; 30(8): 793-795, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38242284

RESUMEN

The management of persistent symptomatic coronavirus disease 2019 (COVID-19) infections in immunocompromised patients remains unclear. Here, we present the first case of successful antiviral therapy (nirmatrelvir/ritonavir and remdesivir) in combination with intravenous immunoglobulin (IVIg) in a patient who had received CD20 depleting therapy for follicular lymphoma and experienced recurrent COVID-19 relapses. After the patient received IVIg treatment, the viral load decreased without recurrence. Subsequently, it was found that the anti-spike antibody titer in the administered immunoglobulin was high at 9528.0 binding antibody units/mL. Our case highlights the potential of combination therapy with selective IVIg and antiviral drugs for relapsed immunocompromised COVID-19 patients who have received CD20 depleting therapy.


Asunto(s)
Adenosina Monofosfato , Alanina , Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Huésped Inmunocomprometido , Inmunoglobulinas Intravenosas , Linfoma Folicular , Ritonavir , SARS-CoV-2 , Humanos , Linfoma Folicular/tratamiento farmacológico , Linfoma Folicular/inmunología , Alanina/análogos & derivados , Alanina/uso terapéutico , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Adenosina Monofosfato/administración & dosificación , Ritonavir/uso terapéutico , Antivirales/uso terapéutico , Inmunoglobulinas Intravenosas/uso terapéutico , COVID-19/inmunología , SARS-CoV-2/inmunología , Masculino , Persona de Mediana Edad , Antígenos CD20/inmunología , Resultado del Tratamiento , Quimioterapia Combinada/métodos , Rituximab/uso terapéutico , Rituximab/administración & dosificación , Carga Viral/efectos de los fármacos , Anticuerpos Monoclonales Humanizados
2.
Intern Med ; 63(16): 2283-2287, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38171874

RESUMEN

Objective Prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been reported in immunocompromised patients, as they poorly develop antibodies against SARS-CoV-2. We conducted a clinical trial to determine the efficacy of Imdevimab/Casirivimab (Imde/Casiri), an anti-viral monoclonal antibody (mAb), for prolonged infection at our institution. Methods Nine patients with hematological malignancies (six with malignant lymphoma and three with multiple myeloma) in our institution presented with coronavirus disease 2019 caused by SARS-CoV-2 omicron variants (one, five, and one with BA.2, BA.5, and BF.7, respectively; two undetermined). Although not all nine patients developed severe disease, viral mRNA was detected in all patients after treatment with remdesivir or molnupiravir. Imde/casiri was infused 11-49 days after the disease onset. Results Within seven days of infusion, viral RNA was undetectable in five of the nine cases. Because all seven viruses isolated from patients whose viral RNA became undetectable showed low or no sensitivity to this monoclonal antibody cocktail, the disappearance of viral RNA in these cases may not be attributable to the antibody cocktail. Conclusion It may be worth considering the use of monoclonal antibodies that show some activity against these virus variants to treat persistent SARS-CoV-2 infection in immunocompromised patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Neoplasias Hematológicas , SARS-CoV-2 , Humanos , Masculino , Persona de Mediana Edad , Femenino , Anciano , Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/tratamiento farmacológico , Tratamiento Farmacológico de COVID-19 , Antivirales/uso terapéutico , Resultado del Tratamiento , COVID-19/inmunología , COVID-19/complicaciones , Adulto , ARN Viral/sangre , Huésped Inmunocomprometido , Anciano de 80 o más Años , Combinación de Medicamentos , Anticuerpos Neutralizantes
3.
BMC Microbiol ; 24(1): 6, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172680

RESUMEN

BACKGROUND: People living with HIV (PLWH) with chronic inflammation may have an increasing risk for coronavirus disease 2019 (COVID-19) severity; however, the impact of their gut microbiota on COVID-19 is not fully elucidated. Here, we analyzed the temporal changes in the gut microbiota composition of hospitalized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected PLWH (PLWH-CoV) and their correlation with COVID-19 severity. RESULT: The 16S rRNA analysis results using stool samples (along the timeline from disease onset) from 12 hospitalized PLWH-CoV, whose median CD4 + T cell count was 671 cells/µl, were compared to those of 19 healthy people and 25 PLWH. Bacterial diversity in PLWH-CoV is not significantly different from that of healthy people and SARS-CoV-2 non-infected PLWH, but a significant difference in the microbiota diversity was observed in the classification according to the disease severity. Immediately after the disease onset, remarkable changes were observed in the gut microbiota of PLWH-CoV, and the changing with a decrease in some short-chain fatty acid-producing bacteria and an increase in colitis-related pathobiont. In the second week after disease onset, relative amounts of specific bacteria distinguished between disease severity. One month after the disease onset, dysbiosis of the gut microbiota persisted, and the number of Enterobacteriaceae, mainly Escherichia-Shigella, which is potentially pathogenic, increased and were enriched in patients who developed post-acute sequelae of COVID-19 (PASC). CONCLUSION: The changes in the gut microbiota associated with SARS-CoV-2 infection observed in PLWH in this study indicated a persistent decrease in SCFA-producing bacteria and an intestinal environment with an increase in opportunistic pathogens associated with enteritis. This report demonstrates that the intestinal environment in PLWH tends to show delayed improvement even after COVID-19 recovery, and highlights the importance of the dysbiosis associated with SARS-CoV-2 infection as a potential factor in the COVID-19 severity and the PASC in PLWH.


Asunto(s)
COVID-19 , Microbioma Gastrointestinal , Infecciones por VIH , Humanos , VIH , COVID-19/complicaciones , Disbiosis , ARN Ribosómico 16S/genética , SARS-CoV-2 , Infecciones por VIH/complicaciones
4.
Jpn J Infect Dis ; 75(6): 608-611, 2022 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-35768273

RESUMEN

Immunocompromised patients are more likely to develop severe COVID-19, and exhibit high mortality. It is also hypothesized that chronic infection in these patients can be a risk factor for developing new variants. We describe a patient with prolonged active infection of COVID-19 who became infected during treatment with an anti-CD20 antibody (obinutuzumab) for follicular lymphoma. This patient had persistent RT-PCR positivity and live virus isolation for nine months despite treatment with remdesivir and other potential antiviral therapies. The computed tomography image of the chest showed that the viral pneumonia repeatedly appeared and disappeared in different lobes, as if a new infection had occurred continuously. The patient's SARS-CoV-2 antibody titer was negative throughout the illness, even after two doses of the BNT162b2 mRNA vaccine were administered in the seventh month of infection. A combination of monoclonal antibody therapy against COVID-19 (casirivimab and imdevimab) and antivirals resulted in negative RT-PCR results, and the virus was no longer isolated. The patient was clinically cured. During the 9-month active infection period, no fixed mutations in the spike (S) protein were detected, and the in vitro susceptibility to remdesivir was retained. Therapeutic administration of anti-SARS-CoV-2 monoclonal antibodies is essential in immunocompromised patients. Therefore, measures to prevent resistance against these key drugs are urgently needed.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Linfoma Folicular , Humanos , Linfoma Folicular/tratamiento farmacológico , Linfoma Folicular/patología , Vacuna BNT162 , SARS-CoV-2 , Anticuerpos Antivirales , Vacunas de ARNm
5.
Sci Transl Med ; 14(657): eabm4908, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35579540

RESUMEN

The SARS-CoV-2 B.1.621 (Mu) variant emerged in January 2021 and was categorized as a variant of interest by the World Health Organization in August 2021. This designation prompted us to study the sensitivity of this variant to antibody neutralization. In a live virus neutralization assay with serum samples from individuals vaccinated with the Pfizer/BioNTech or Moderna mRNA vaccines, we measured neutralization antibody titers against B.1.621, an early isolate (spike 614D), and a variant of concern (B.1.351, Beta variant). We observed reduced neutralizing antibody titers against the B.1.621 variant (3.4- to 7-fold reduction, depending on the serum sample and time after the second vaccination) compared to the early isolate and a similar reduction when compared to B.1.351. Likewise, convalescent serum from hamsters previously infected with an early isolate neutralized B.1.621 to a lower degree. Despite this antibody titer reduction, hamsters could not be efficiently rechallenged with the B.1.621 variant, suggesting that the immune response to the first infection is adequate to provide protection against a subsequent infection with the B.1.621 variant.


Asunto(s)
COVID-19 , Proteínas del Envoltorio Viral , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/terapia , Humanos , Inmunización Pasiva , Glicoproteínas de Membrana/genética , Pruebas de Neutralización , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética , Vacunación , Proteínas del Envoltorio Viral/genética , Sueroterapia para COVID-19
6.
mBio ; 12(5): e0198721, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34579572

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has raised concerns about the detrimental effects of antibodies. Antibody-dependent enhancement (ADE) of infection is one of the biggest concerns in terms of not only the antibody reaction to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) upon reinfection with the virus but also the reaction to COVID-19 vaccines. In this study, we evaluated ADE of infection by using COVID-19 convalescent-phase plasma and BHK cells expressing human Fcγ receptors (FcγRs). We found that FcγRIIA and FcγRIIIA mediated modest ADE of infection against SARS-CoV-2. Although ADE of infection was observed in monocyte-derived macrophages infected with SARS-CoV-2, including its variants, proinflammatory cytokine/chemokine expression was not upregulated in macrophages. SARS-CoV-2 infection thus produces antibodies that elicit ADE of infection, but these antibodies do not contribute to excess cytokine production by macrophages. IMPORTANCE Viruses infect cells mainly via specific receptors at the cell surface. Antibody-dependent enhancement (ADE) of infection is an alternative mechanism of infection for viruses to infect immune cells that is mediated by antibodies and IgG receptors (FcγRs). Because ADE of infection contributes to the pathogenesis of some viruses, such as dengue virus and feline coronavirus, it is important to evaluate the precise mechanism of ADE and its contribution to the pathogenesis of SARS-CoV-2. Here, using convalescent-phase plasma from COVID-19 patients, we found that two types of FcγRs, FcγRIIA and FcγRIIIA, mediate ADE of SARS-CoV-2 infection. Although ADE of infection was observed for SARS-CoV-2 and its recent variants, proinflammatory cytokine production in monocyte-derived macrophages was not upregulated. These observations suggest that SARS-CoV-2 infection produces antibodies that elicit ADE of infection, but these antibodies may not be involved in aberrant cytokine release by macrophages during SARS-CoV-2 infection.


Asunto(s)
Citocinas/metabolismo , Macrófagos/metabolismo , Receptores de IgG/metabolismo , SARS-CoV-2/patogenicidad , Animales , Acrecentamiento Dependiente de Anticuerpo/fisiología , Línea Celular , Cricetinae , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de IgG/genética
7.
J Vet Med Sci ; 80(6): 1020-1024, 2018 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-29669959

RESUMEN

Influenza B virus has been known to infect humans and other animals, including seals. Vaccination efficacy varies across seasons. Human monoclonal antibodies (mAbs) can be useful for developing novel vaccines, guided by epitope analysis, and can be used therapeutically. Hybridoma technology has been used to make mAbs. Here we evaluated SPYMEG as a fusion partner cell line for human mAb generation specific to influenza B hemagglutinin (HA). SPYMEG is a human/murine myeloma partner cell line that has previously been used to generate human mAbs that recognize the HA of influenza A and B viruses. Peripheral blood mononuclear cells were obtained from 16 volunteers, previously vaccinated with the 2014-2015 trivalent seasonal influenza vaccine, and were fused with SPYMEG to yield hybridomas. The resulting hybridomas were screened for antigen-specific antibody secretion and cloned by limiting dilution. We obtained 32 stable clones secreting anti-influenza B HA human IgG, although most of these clones were obtained from one volunteer (SeaV-29) who had a robust immune response. We conclude that SPYMEG is a good fusion partner cell line, although cloning by limiting dilution may lead to significant loss of hybridomas.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Hibridomas/inmunología , Virus de la Influenza B , Animales , Línea Celular , Glicoproteínas Hemaglutininas del Virus de la Influenza , Humanos , Leucocitos Mononucleares , Ratones
8.
J Biol Chem ; 293(19): 7126-7138, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29555684

RESUMEN

Histone acetylation plays crucial roles in transcriptional regulation and chromatin organization. Viral RNA of the influenza virus interacts with its nucleoprotein (NP), whose function corresponds to that of eukaryotic histones. NP regulates viral replication and has been shown to undergo acetylation by the cAMP-response element (CRE)-binding protein (CBP) from the host. However, whether NP is the target of other host acetyltransferases is unknown. Here, we show that influenza virus NP undergoes acetylation by the two host acetyltransferases GCN5 and P300/CBP-associated factor (PCAF) and that this modification affects viral polymerase activities. Western blot analysis with anti-acetyl-lysine antibody on cultured A549 human lung adenocarcinoma epithelial cells infected with different influenza virus strains indicated acetylation of the viral NP. A series of biochemical analyses disclosed that the host lysine acetyltransferases GCN5 and PCAF acetylate NP in vitro MS experiments identified three lysine residues as acetylation targets in the host cells and suggested that Lys-31 and Lys-90 are acetylated by PCAF and GCN5, respectively. RNAi-mediated silencing of GCN5 and PCAF did not change acetylation levels of NP. However, interestingly, viral polymerase activities were increased by the PCAF silencing and were decreased by the GCN5 silencing, suggesting that acetylation of the Lys-31 and Lys-90 residues has opposing effects on viral replication. Our findings suggest that epigenetic control of NP via acetylation by host acetyltransferases contributes to regulation of polymerase activity in the influenza A virus.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Virus de la Influenza A/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas del Núcleo Viral/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Células A549 , Acetilación , Secuencia de Aminoácidos , Western Blotting , Cromatografía Liquida , Epigénesis Genética , Células Epiteliales/virología , Histona Acetiltransferasas/genética , Humanos , Virus de la Influenza A/enzimología , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Lisina/metabolismo , Proteínas de la Nucleocápside , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Viral/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Espectrometría de Masas en Tándem , Transcripción Genética , Proteínas del Núcleo Viral/química , Proteínas del Núcleo Viral/genética , Replicación Viral , Factores de Transcripción p300-CBP/genética
9.
J Virol ; 90(1): 444-56, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491155

RESUMEN

UNLABELLED: Over the past 2 decades, several novel influenza virus proteins have been identified that modulate viral infections in vitro and/or in vivo. The PB2 segment, which is one of the longest influenza A virus segments, is known to encode only one viral protein, PB2. In the present study, we used reverse transcription-PCR (RT-PCR) targeting viral mRNAs transcribed from the PB2 segment to look for novel viral proteins encoded by spliced mRNAs. We identified a new viral protein, PB2-S1, encoded by a novel spliced mRNA in which the region corresponding to nucleotides 1513 to 1894 of the PB2 mRNA is deleted. PB2-S1 was detected in virus-infected cells and in cells transfected with a protein expression plasmid encoding PB2. PB2-S1 localized to mitochondria, inhibited the RIG-I-dependent interferon signaling pathway, and interfered with viral polymerase activity (dependent on its PB1-binding capability). The nucleotide sequences around the splicing donor and acceptor sites for PB2-S1 were highly conserved among pre-2009 human H1N1 viruses but not among human H1N1pdm and H3N2 viruses. PB2-S1-deficient viruses, however, showed growth kinetics in MDCK cells and virulence in mice similar to those of wild-type virus. The biological significance of PB2-S1 to the replication and pathogenicity of seasonal H1N1 influenza A viruses warrants further investigation. IMPORTANCE: Transcriptome analysis of cells infected with influenza A virus has improved our understanding of the host response to viral infection, because such analysis yields considerable information about both in vitro and in vivo viral infections. However, little attention has been paid to transcriptomes derived from the viral genome. Here we focused on the splicing of mRNA expressed from the PB2 segment and identified a spliced viral mRNA encoding a novel viral protein. This result suggests that other, as yet unidentified viral proteins encoded by spliced mRNAs could be expressed in virus-infected cells. A viral transcriptome including the viral spliceosome should be evaluated to gain new insights into influenza virus infection.


Asunto(s)
Perfilación de la Expresión Génica , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Isoformas de Proteínas/genética , Empalme del ARN , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/antagonistas & inhibidores , Perros , Células Epiteliales/virología , Femenino , Expresión Génica , Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Mitocondrias/química , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Isoformas de Proteínas/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Receptores Inmunológicos , Proteínas Virales/metabolismo , Virulencia
10.
J Biol Chem ; 289(36): 24980-94, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25063805

RESUMEN

The PA, PB1, and PB2 subunits, components of the RNA-dependent RNA polymerase of influenza A virus, are essential for viral transcription and replication. The PB2 subunit binds to the host RNA cap (7-methylguanosine triphosphate (m(7)GTP)) and supports the endonuclease activity of PA to "snatch" the cap from host pre-mRNAs. However, the structure of PB2 is not fully understood, and the functional sites remain unknown. In this study, we describe a novel Val/Arg/Gly (VRG) site in the PB2 cap-binding domain, which is involved in interaction with acetyl-CoA found in eukaryotic histone acetyltransferases (HATs). In vitro experiments revealed that the recombinant PB2 cap-binding domain that includes the VRG site interacts with acetyl-CoA; moreover, it was found that this interaction could be blocked by CoA and various HAT inhibitors. Interestingly, m(7)GTP also inhibited this interaction, suggesting that the same active pocket is capable of interacting with acetyl-CoA and m(7)GTP. To elucidate the importance of the VRG site on PB2 function and viral replication, we constructed a PB2 recombinant protein and recombinant viruses including several patterns of amino acid mutations in the VRG site. Substitutions of the valine and arginine residues or of all 3 residues of the VRG site to alanine significantly reduced the binding ability of PB2 to acetyl-CoA and its RNA polymerase activity. Recombinant viruses containing the same mutations could not be replicated in cultured cells. These results indicate that the PB2 VRG sequence is a functional site that is essential for acetyl-CoA interaction, RNA polymerase activity, and viral replication.


Asunto(s)
Acetilcoenzima A/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Acetiltransferasas/química , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Línea Celular Tumoral , Perros , Células Eucariotas/enzimología , Humanos , Subtipo H3N2 del Virus de la Influenza A/enzimología , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Células de Riñón Canino Madin Darby , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Mutación , Células Procariotas/enzimología , Unión Proteica , Estructura Terciaria de Proteína , Caperuzas de ARN/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Proteínas Virales/química , Proteínas Virales/genética
11.
Proc Natl Acad Sci U S A ; 110(36): 14753-8, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959904

RESUMEN

Enterovirus 71 (EV71) typically causes mild hand-foot-and-mouth disease in children, but it can also cause severe neurological disease. Recently, epidemic outbreaks of EV71 with significant mortality have been reported in the Asia-Pacific region, and EV71 infection has become a serious public health concern worldwide. However, there is little information available concerning EV71 neuropathogenesis, and no vaccines or anti-EV71 drugs have been developed. Previous studies of this disease have used monkeys and neonatal mice that are susceptible to some EV71 strains as models. The monkey model is problematic for ethical and economical reasons, and mice that are more than a few weeks old lose their susceptibility to EV71. Thus, the development of an appropriate small animal model would greatly contribute to the study of this disease. Mice lack EV71 susceptibility due to the absence of a receptor for this virus. Previously, we identified the human scavenger receptor class B, member 2 (hSCARB2) as a cellular receptor for EV71. In the current study, we generated a transgenic (Tg) mouse expressing hSCARB2 with an expression profile similar to that in humans. Tg mice infected with EV71 exhibited ataxia, paralysis, and death. The most severely affected cells were neurons in the spinal cord, brainstem, cerebellum, hypothalamus, thalamus, and cerebrum. The pathological features in these Tg mice were generally similar to those of EV71 encephalomyelitis in humans and experimentally infected monkeys. These results suggest that this Tg mouse could represent a useful animal model for the study of EV71 infection.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Modelos Animales de Enfermedad , Infecciones por Enterovirus/genética , Proteínas de Membrana de los Lisosomas/genética , Receptores Depuradores/genética , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Línea Celular Tumoral , Enfermedades del Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/virología , Chlorocebus aethiops , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/virología , Interacciones Huésped-Patógeno , Humanos , Inmunohistoquímica , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones , Ratones Transgénicos , Receptores Depuradores/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/virología , Factores de Tiempo , Células Vero
12.
Nat Med ; 15(7): 798-801, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19543282

RESUMEN

Enterovirus 71 (EV71) belongs to human enterovirus species A of the genus Enterovirus within the family Picornaviridae. EV71, together with coxsackievirus A16 (CVA16), are most frequently associated with hand, foot and mouth disease (HFMD). Although HFMD is considered a mild exanthematous infection, infections involving EV71, but not CVA16, can progress to severe neurological disease, including fatal encephalitis, aseptic meningitis and acute flaccid paralysis. In recent years, epidemic and sporadic outbreaks of neurovirulent EV71 infections have been reported in Taiwan, Malaysia, Singapore, Japan and China. Here, we show that human scavenger receptor class B, member 2 (SCARB2, also known as lysosomal integral membrane protein II or CD36b like-2) is a receptor for EV71. EV71 binds soluble SCARB2 or cells expressing SCARB2, and the binding is inhibited by an antibody to SCARB2. Expression of human SCARB2 enables normally unsusceptible cell lines to support EV71 propagation and develop cytopathic effects. EV71 infection is hampered by the antibody to SCARB2 and soluble SCARB2. SCARB2 also supports the infection of the milder pathogen CVA16. The identification of SCARB2 as an EV71 and CVA16 receptor contributes to a better understanding of the pathogenicity of these viruses.


Asunto(s)
Enterovirus Humano A/patogenicidad , Proteínas de Membrana de los Lisosomas/fisiología , Receptores Depuradores/fisiología , Receptores Virales/fisiología , Animales , Perfilación de la Expresión Génica , Humanos , Proteínas de Membrana de los Lisosomas/análisis , Proteínas de Membrana de los Lisosomas/genética , Ratones , Receptores Depuradores/análisis , Receptores Depuradores/genética , Receptores Virales/análisis , Rabdomiosarcoma/virología
13.
Cell Host Microbe ; 3(3): 168-77, 2008 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-18329616

RESUMEN

The Ebola virus matrix protein VP40 plays an important role in virion formation and viral egress from cells. However, the host cell proteins and mechanisms responsible for intracellular transport of VP40 prior to its contribution to virion formation remain to be elucidated. Therefore we used coimmunoprecipitation and mass spectrometric analyses to identify host proteins interacting with VP40. We found that Sec24C, a component of the host COPII vesicular transport system, interacts specifically with VP40 via VP40 amino acids 303 to 307. Coimmunoprecipitation and dominant-negative mutant studies indicated that the COPII transport system plays a critical role in VP40 intracellular transport to the plasma membrane. Marburg virus VP40 was also shown to use the COPII transport system for intracellular transport. These findings identify a conserved intersection between a host pathway and filovirus replication, an intersection that can be targeted in the development of new antiviral drugs.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Ebolavirus/fisiología , Nucleoproteínas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas del Núcleo Viral/metabolismo , Línea Celular , Membrana Celular/química , Citosol/química , Humanos , Inmunoprecipitación , Marburgvirus/fisiología , Espectrometría de Masas , Unión Proteica , Mapeo de Interacción de Proteínas , Transporte de Proteínas
14.
J Virol ; 81(1): 30-41, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17050598

RESUMEN

The RNA genome of influenza A virus, which forms viral ribonucleoprotein complexes (vRNPs) with viral polymerase subunit proteins (PA, PB1, and PB2) and nucleoprotein (NP), is transcribed and replicated in the nucleus. NP, the major component of vRNPs, has at least two amino acid sequences that serve as nuclear localization signals (NLSs): an unconventional NLS (residues 3 to 13; NLS1) and a bipartite NLS (residues 198 to 216; NLS2). Although both NLSs are known to play a role in nuclear transport, their relative contributions to viral replication are poorly understood. We therefore investigated their contributions to NP subcellular/subnuclear localization, viral RNA (vRNA) transcription, and viral replication. Abolishing the unconventional NLS caused NP to localize predominantly to the cytoplasm and affected its activity in vRNA transcription. However, we were able to create a virus whose NP contained amino acid substitutions in NLS1 known to abolish its nuclear localization function, although this virus was highly attenuated. These results indicate that while the unconventional NLS is not essential for viral replication, it is necessary for efficient viral mRNA synthesis. On the other hand, the bipartite NLS, whose contribution to the nuclear transport of NP is limited, was essential for vRNA transcription and NP's nucleolar accumulation. A virus with nonfunctional NLS2 could not be generated. Thus, the bipartite NLS, but not the unconventional NLS, of NP is essential for influenza A virus replication.


Asunto(s)
Virus de la Influenza A/fisiología , Señales de Localización Nuclear , Nucleoproteínas/química , Proteínas de Unión al ARN/química , Proteínas del Núcleo Viral/química , Replicación Viral , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Humanos , Virus de la Influenza A/metabolismo , Mutación , Proteínas de la Nucleocápside , Nucleoproteínas/metabolismo , Transporte de Proteínas , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Proteínas del Núcleo Viral/metabolismo , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA