Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Lancet Microbe ; 5(7): 633-644, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38705163

RESUMO

BACKGROUND: Artemether-lumefantrine is widely used for uncomplicated Plasmodium falciparum malaria; sulfadoxine-pyrimethamine plus amodiaquine is used for seasonal malaria chemoprevention. We aimed to determine the efficacy of artemether-lumefantrine with and without primaquine and sulfadoxine-pyrimethamine plus amodiaquine with and without tafenoquine for reducing gametocyte carriage and transmission to mosquitoes. METHODS: In this phase 2, single-blind, randomised clinical trial conducted in Ouelessebougou, Mali, asymptomatic individuals aged 10-50 years with P falciparum gametocytaemia were recruited from the community and randomly assigned (1:1:1:1) to receive either artemether-lumefantrine, artemether-lumefantrine with a single dose of 0·25 mg/kg primaquine, sulfadoxine-pyrimethamine plus amodiaquine, or sulfadoxine-pyrimethamine plus amodiaquine with a single dose of 1·66 mg/kg tafenoquine. All trial staff other than the pharmacist were masked to group allocation. Participants were not masked to group allocation. Randomisation was done with a computer-generated randomisation list and concealed with sealed, opaque envelopes. The primary outcome was the median within-person percent change in mosquito infection rate in infectious individuals from baseline to day 2 (artemether-lumefantrine groups) or day 7 (sulfadoxine-pyrimethamine plus amodiaquine groups) after treatment, assessed by direct membrane feeding assay. All participants who received any trial drug were included in the safety analysis. This study is registered with ClinicalTrials.gov, NCT05081089. FINDINGS: Between Oct 13 and Dec 16, 2021, 1290 individuals were screened and 80 were enrolled and randomly assigned to one of the four treatment groups (20 per group). The median age of participants was 13 (IQR 11-20); 37 (46%) of 80 participants were female and 43 (54%) were male. In individuals who were infectious before treatment, the median percentage reduction in mosquito infection rate 2 days after treatment was 100·0% (IQR 100·0-100·0; n=19; p=0·0011) with artemether-lumefantrine and 100·0% (100·0-100·0; n=19; p=0·0001) with artemether-lumefantrine with primaquine. Only two individuals who were infectious at baseline infected mosquitoes on day 2 after artemether-lumefantrine and none at day 5. By contrast, the median percentage reduction in mosquito infection rate 7 days after treatment was 63·6% (IQR 0·0-100·0; n=20; p=0·013) with sulfadoxine-pyrimethamine plus amodiaquine and 100% (100·0-100·0; n=19; p<0·0001) with sulfadoxine-pyrimethamine plus amodiaquine with tafenoquine. No grade 3-4 or serious adverse events occurred. INTERPRETATION: These data support the effectiveness of artemether-lumefantrine alone for preventing nearly all mosquito infections. By contrast, there was considerable post-treatment transmission after sulfadoxine-pyrimethamine plus amodiaquine; therefore, the addition of a transmission-blocking drug might be beneficial in maximising its community impact. FUNDING: Bill & Melinda Gates Foundation.


Assuntos
Amodiaquina , Antimaláricos , Combinação Arteméter e Lumefantrina , Combinação de Medicamentos , Fluorenos , Malária Falciparum , Plasmodium falciparum , Primaquina , Pirimetamina , Sulfadoxina , Humanos , Antimaláricos/uso terapêutico , Antimaláricos/administração & dosagem , Pirimetamina/uso terapêutico , Pirimetamina/administração & dosagem , Amodiaquina/uso terapêutico , Amodiaquina/administração & dosagem , Sulfadoxina/uso terapêutico , Sulfadoxina/administração & dosagem , Masculino , Adulto , Feminino , Adolescente , Criança , Malária Falciparum/transmissão , Malária Falciparum/prevenção & controle , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Método Simples-Cego , Pessoa de Meia-Idade , Primaquina/uso terapêutico , Primaquina/administração & dosagem , Combinação Arteméter e Lumefantrina/uso terapêutico , Combinação Arteméter e Lumefantrina/administração & dosagem , Adulto Jovem , Fluorenos/administração & dosagem , Fluorenos/uso terapêutico , Mali/epidemiologia , Plasmodium falciparum/efeitos dos fármacos , Artemisininas/administração & dosagem , Artemisininas/uso terapêutico , Aminoquinolinas/administração & dosagem , Aminoquinolinas/uso terapêutico , Aminoquinolinas/efeitos adversos , Etanolaminas/administração & dosagem , Etanolaminas/uso terapêutico , Animais , Quimioterapia Combinada
2.
J Infect Dis ; 226(4): 708-713, 2022 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-35578987

RESUMO

Achieving malaria elimination requires a better understanding of the transmissibility of human infections in different transmission settings. This study aimed to characterize the human infectious reservoir in a high endemicity setting in eastern Uganda, using gametocyte quantification and mosquito feeding assays. In asymptomatic infections, gametocyte densities were positively associated with the proportion of infected mosquitoes (ß = 1.60; 95% CI, 1.32-1.92; P < .0001). Combining transmissibility and abundance in the population, symptomatic and asymptomatic infections were estimated to contribute to 5.3% and 94.7% of the infectious reservoir, respectively. School-aged children (5-15 years old) contributed to 50.4% of transmission events and were important drivers of malaria transmission.


Assuntos
Anopheles , Linfoma de Burkitt , Malária Falciparum , Malária , Adolescente , Animais , Infecções Assintomáticas/epidemiologia , Criança , Pré-Escolar , Humanos , Malária/epidemiologia , Malária Falciparum/epidemiologia , Plasmodium falciparum , Uganda/epidemiologia
3.
JCI Insight ; 7(6)2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35167490

RESUMO

Sporozoite-based approaches currently represent the most effective vaccine strategies for induction of sterile protection against Plasmodium falciparum (Pf) malaria. Clinical development of subunit vaccines is almost exclusively centered on the circum-sporozoite protein (CSP), an abundantly expressed protein on the sporozoite membrane. Anti-CSP antibodies are able to block sporozoite invasion and development in human hepatocytes and subsequently prevent clinical malaria. Here, we have investigated whether sporozoite-induced human antibodies with specificities different from CSP can reduce Pf-liver stage development. IgG preparations were obtained from 12 volunteers inoculated with a protective immunization regime of whole sporozoites under chloroquine prophylaxis. These IgGs were depleted for CSP specificity by affinity chromatography. Recovered non-CSP antibodies were tested for sporozoite membrane binding and for functional inhibition of sporozoite invasion of a human hepatoma cell line and hepatocytes both in vitro and in vivo. Postimmunization IgGs depleted for CS specificity of 9 of 12 donors recognized sporozoite surface antigens. Samples from 5 of 12 donors functionally reduced parasite-liver cell invasion or development using the hepatoma cell line HC-04 and FRG-huHep mice containing human liver cells. The combined data provide clear evidence that non-CSP proteins, as yet undefined, do represent antibody targets for functional immunity against Pf parasites responsible for malaria.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Vacinas Antimaláricas , Malária Falciparum , Malária , Parasitos , Animais , Anticorpos Antiprotozoários , Carcinoma Hepatocelular/tratamento farmacológico , Hepatócitos , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Malária/tratamento farmacológico , Camundongos , Plasmodium falciparum , Proteínas de Protozoários , Esporozoítos
4.
J Infect Dis ; 223(10): 1822-1830, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31875909

RESUMO

BACKGROUND: Plasmodium falciparum transmission depends on mature gametocytes that can be ingested by mosquitoes taking a blood meal on human skin. Although gametocyte skin sequestration has long been hypothesized as important contributor to efficient malaria transmission, this has never been formally tested. METHODS: In naturally infected gametocyte carriers from Burkina Faso, we assessed infectivity to mosquitoes by direct skin feeding and membrane feeding. We directly quantified male and female gametocytes and asexual parasites in finger-prick and venous blood samples, skin biopsy samples, and in of mosquitoes that fed on venous blood or directly on skin. Gametocytes were visualized in skin tissue with confocal microscopy. RESULTS: Although more mosquitoes became infected when feeding directly on skin then when feeding on venous blood (odds ratio, 2.01; 95% confidence interval, 1.21-3.33; P = .007), concentrations of gametocytes were not higher in the subdermal skin vasculature than in other blood compartments; only sparse gametocytes were observed in skin tissue. DISCUSSION: Our data strongly suggest that there is no significant skin sequestration of P. falciparum gametocytes. Gametocyte densities in peripheral blood are thus informative for predicting onward transmission potential to mosquitoes and can be used to target and monitor malaria elimination initiatives.


Assuntos
Anopheles , Malária Falciparum , Animais , Anopheles/parasitologia , Burkina Faso , Humanos , Malária Falciparum/epidemiologia , Plasmodium falciparum
5.
Malar J ; 18(1): 430, 2019 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-31852480

RESUMO

BACKGROUND: Sulfadoxine-pyrimethamine (SP) is a cornerstone of malaria chemoprophylaxis and is considered for programmes in the Democratic Republic of Congo (DRC). However, SP efficacy is threatened by drug resistance, that is conferred by mutations in the dhfr and dhps genes. The World Health Organization has specified that intermittent preventive treatment for infants (IPTi) with SP should be implemented only if the prevalence of the dhps K540E mutation is under 50%. There are limited current data on the prevalence of resistance-conferring mutations available from Eastern DRC. The current study aimed to address this knowledge gap. METHODS: Dried blood-spot samples were collected from clinically suspected malaria patients [outpatient department (OPD)] and pregnant women attending antenatal care (ANC) in four sites in North and South Kivu, DRC. Quantitative PCR (qPCR) was performed on samples from individuals with positive and with negative rapid diagnostic test (RDT) results. Dhps K450E and A581G and dhfr I164L were assessed by nested PCR followed by allele-specific primer extension and detection by multiplex bead-based assays. RESULTS: Across populations, Plasmodium falciparum parasite prevalence was 47.9% (1160/2421) by RDT and 71.7 (1763/2421) by qPCR. Median parasite density measured by qPCR in RDT-negative qPCR-positive samples was very low with a median of 2.3 parasites/µL (IQR 0.5-25.2). Resistance genotyping was successfully performed in RDT-positive samples and RDT-negative/qPCR-positive samples with success rates of 86.2% (937/1086) and 55.5% (361/651), respectively. The presence of dhps K540E was high across sites (50.3-87.9%), with strong evidence for differences between sites (p < 0.001). Dhps A581G mutants were less prevalent (12.7-47.2%). The dhfr I164L mutation was found in one sample. CONCLUSIONS: The prevalence of the SP resistance marker dhps K540E exceeds 50% in all four study sites in North and South Kivu, DRC. K540E mutations regularly co-occurred with mutations in dhps A581G but not with the dhfr I164L mutation. The current results do not support implementation of IPTi with SP in the study area.


Assuntos
Antimaláricos/farmacologia , Resistência a Medicamentos , Malária/prevenção & controle , Plasmodium/efeitos dos fármacos , Pirimetamina/farmacologia , Sulfadoxina/farmacologia , Adolescente , Biomarcadores/sangue , Quimioprevenção/estatística & dados numéricos , Criança , Pré-Escolar , República Democrática do Congo , Combinação de Medicamentos , Feminino , Humanos , Lactente , Recém-Nascido , Masculino
6.
Cell Rep ; 10(4): 600-15, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25640182

RESUMO

Itraconazole (ITZ) is a well-known antifungal agent that also has anticancer activity. In this study, we identify ITZ as a broad-spectrum inhibitor of enteroviruses (e.g., poliovirus, coxsackievirus, enterovirus-71, rhinovirus). We demonstrate that ITZ inhibits viral RNA replication by targeting oxysterol-binding protein (OSBP) and OSBP-related protein 4 (ORP4). Consistently, OSW-1, a specific OSBP/ORP4 antagonist, also inhibits enterovirus replication. Knockdown of OSBP inhibits virus replication, whereas overexpression of OSBP or ORP4 counteracts the antiviral effects of ITZ and OSW-1. ITZ binds OSBP and inhibits its function, i.e., shuttling of cholesterol and phosphatidylinositol-4-phosphate between membranes, thereby likely perturbing the virus-induced membrane alterations essential for viral replication organelle formation. ITZ also inhibits hepatitis C virus replication, which also relies on OSBP. Together, these data implicate OSBP/ORP4 as molecular targets of ITZ and point to an essential role of OSBP/ORP4-mediated lipid exchange in virus replication that can be targeted by antiviral drugs.


Assuntos
Enterovirus/efeitos dos fármacos , Enterovirus/metabolismo , Itraconazol/farmacologia , Receptores de Esteroides/metabolismo , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Linhagem Celular Tumoral , Humanos
7.
J Virol ; 89(3): 1913-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25410869

RESUMO

PI4KIIIß recruitment to Golgi membranes relies on GBF1/Arf and ACBD3. Enteroviruses such as poliovirus and coxsackievirus recruit PI4KIIIß to their replication sites via their 3A proteins. Here, we show that human rhinovirus (HRV) 3A also recruited PI4KIIIß to replication sites. Unlike other enterovirus 3A proteins, HRV 3A failed to bind GBF1. Although HRV 3A was previously shown to interact with ACBD3, our data suggest that PI4KIIIß recruitment occurred independently of both GBF1 and ACBD3.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Interações Hospedeiro-Patógeno , Proteínas de Membrana/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Rhinovirus/fisiologia , Proteínas do Core Viral/metabolismo , Humanos , Ligação Proteica
8.
Antiviral Res ; 103: 17-24, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24393668

RESUMO

Proteolytical cleavage of the picornaviral polyprotein is essential for viral replication. Therefore, viral proteases are attractive targets for anti-viral therapy. Most assays available for testing proteolytical activity of proteases are performed in vitro, using heterologously expressed proteases and peptide substrates. To deal with the disadvantages associated with in vitro assays, we modified a cell-based protease assay for picornavirus proteases. The assay is based on the induction of expression of a firefly luciferase reporter by a chimeric transcription factor in which the viral protease and cleavage sites are inserted between the GAL4 binding domain and the VP16 activation domain. Firefly luciferase expression is dependent on cleavage of the transcription factor by the viral protease. This biosafe assay enables testing the effect of compounds on protease activity in cells while circumventing the need for infection. We designed the assay for 3C proteases (3C(pro)) of various enteroviruses as well as of viruses of several other picornavirus genera, and show that the assay is amenable for use in a high-throughput setting. Furthermore, we show that the spectrum of activity of 3C(pro) inhibitor AG7088 (rupintrivir) not only encompasses enterovirus 3C(pro) but also 3C(pro) of foot-and-mouth disease virus (FMDV), an aphthovirus. In contrary, AG7404 (compound 1), an analogue of AG7088, had no effect on FMDV 3C(pro) activity, for which we provide a structural explanation.


Assuntos
Antivirais/isolamento & purificação , Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Picornaviridae/efeitos dos fármacos , Picornaviridae/enzimologia , Proteínas Virais/antagonistas & inibidores , Proteases Virais 3C , Animais , Linhagem Celular , Cisteína Endopeptidases , Genes Reporter , Humanos , Luciferases de Vaga-Lume/análise , Luciferases de Vaga-Lume/genética , Inibidores de Proteases/isolamento & purificação , Inibidores de Proteases/farmacologia
9.
J Virol ; 88(6): 3369-78, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24390337

RESUMO

UNLABELLED: RIG-I-like receptors (RLRs) MDA5 and RIG-I are key players in the innate antiviral response. Upon recognition of viral RNA, they interact with MAVS, eventually inducing type I interferon production. The interferon induction pathway is commonly targeted by viruses. How enteroviruses suppress interferon production is incompletely understood. MDA5 has been suggested to undergo caspase- and proteasome-mediated degradation during poliovirus infection. Additionally, MAVS is reported to be cleaved during infection with coxsackievirus B3 (CVB3) by the CVB3 proteinase 3C(pro), whereas MAVS cleavage by enterovirus 71 has been attributed to 2A(pro). As yet, a detailed examination of the RLR pathway as a whole during any enterovirus infection is lacking. We performed a comprehensive analysis of crucial factors of the RLR pathway, including MDA5, RIG-I, LGP2, MAVS, TBK1, and IRF3, during infection of CVB3, a human enterovirus B (HEV-B) species member. We show that CVB3 inhibits the RLR pathway upstream of TBK1 activation, as demonstrated by limited phosphorylation of TBK1 and a lack of IRF3 phosphorylation. Furthermore, we show that MDA5, MAVS, and RIG-I all undergo proteolytic degradation in CVB3-infected cells through a caspase- and proteasome-independent manner. We convincingly show that MDA5 and MAVS cleavages are both mediated by CVB3 2A(pro), while RIG-I is cleaved by 3C(pro). Moreover, we show that proteinases 2A(pro) and 3C(pro) of poliovirus (HEV-C) and enterovirus 71 (HEV-A) exert the same functions. This study identifies a critical role of 2A(pro) by cleaving MDA5 and MAVS and shows that enteroviruses use a common strategy to counteract the interferon response in infected cells. IMPORTANCE: Human enteroviruses (HEVs) are important pathogens that cause a variety of diseases in humans, including poliomyelitis, hand, foot, and mouth disease, viral meningitis, cardiomyopathy, and more. Like many other viruses, enteroviruses target the host immune pathways to gain replication advantage. The MDA5/MAVS pathway is responsible for recognizing enterovirus infections in the host cell and leads to expression of type I interferons (IFN-I), crucial antiviral signaling molecules. Here we show that three species of HEVs all employ the viral proteinase 2A (2A(pro)) to proteolytically target MDA5 and MAVS, leading to an efficient blockade upstream of IFN-I transcription. These observations suggest that MDA5/MAVS antagonization is an evolutionarily conserved and beneficial mechanism of enteroviruses. Understanding the molecular mechanisms of enterovirus immune evasion strategies will help to develop countermeasures to control infections with these viruses in the future.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cisteína Endopeptidases/metabolismo , RNA Helicases DEAD-box/metabolismo , Enterovirus Humano B/enzimologia , Infecções por Enterovirus/metabolismo , Proteínas Virais/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Cisteína Endopeptidases/genética , RNA Helicases DEAD-box/genética , Enterovirus Humano B/genética , Enterovirus Humano B/fisiologia , Infecções por Enterovirus/enzimologia , Infecções por Enterovirus/genética , Infecções por Enterovirus/virologia , Interações Hospedeiro-Patógeno , Humanos , Helicase IFIH1 Induzida por Interferon , Fosforilação , Proteólise , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais , Proteínas Virais/genética
10.
Cell Res ; 22(11): 1576-92, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22945356

RESUMO

RNA viruses can rapidly mutate and acquire resistance to drugs that directly target viral enzymes, which poses serious problems in a clinical context. Therefore, there is a growing interest in the development of antiviral drugs that target host factors critical for viral replication, since they are unlikely to mutate in response to therapy. We recently demonstrated that phosphatidylinositol-4-kinase IIIß (PI4KIIIß) and its product phosphatidylinositol-4-phosphate (PI4P) are essential for replication of enteroviruses, a group of medically important RNA viruses including poliovirus (PV), coxsackievirus, rhinovirus, and enterovirus 71. Here, we show that enviroxime and GW5074 decreased PI4P levels at the Golgi complex by directly inhibiting PI4KIIIß. Coxsackievirus mutants resistant to these inhibitors harbor single point mutations in the non-structural protein 3A. These 3A mutations did not confer compound-resistance by restoring the activity of PI4KIIIß in the presence of the compounds. Instead, replication of the mutant viruses no longer depended on PI4KIIIß, since their replication was insensitive to siRNA-mediated depletion of PI4KIIIß. The mutant viruses also did not rely on other isoforms of PI4K. Consistently, no high level of PI4P could be detected at the replication sites induced by the mutant viruses in the presence of the compounds. Collectively, these findings indicate that through specific single point mutations in 3A, CVB3 can bypass an essential host factor and lipid for its propagation, which is a new example of RNA viruses acquiring resistance against antiviral compounds, even when they directly target host factors.


Assuntos
1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , Antivirais/farmacologia , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/fisiologia , Fosfatos de Fosfatidilinositol/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Proteínas não Estruturais Virais/genética , 1-Fosfatidilinositol 4-Quinase/metabolismo , Animais , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Farmacorresistência Viral/genética , Enterovirus Humano B/genética , Células HeLa , Humanos , Indóis/farmacologia , Oximas , Fenóis/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Mutação Puntual , Interferência de RNA , RNA Interferente Pequeno , Sulfonamidas , Replicação Viral/efeitos dos fármacos
11.
Diabetes ; 61(8): 2030-6, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22596052

RESUMO

Enteroviruses of the human enterovirus B species (HEV-Bs) (e.g., coxsackie B viruses [CVBs] and echoviruses) have been implicated as environmental factors that trigger/accelerate type 1 diabetes, but the underlying mechanism remains elusive. The aim of this study was to gain insight into the cytokines and chemokines that are produced by human pancreatic islets upon infection with CVBs. To this end, we studied the response of human islets of Langerhans upon mock or CVB3 infection. Using quantitative PCR, we showed that upon CVB3 infection, transcription of interferon (IFN), IFN-stimulated genes, and inflammatory genes was induced. Analysis of secreted cytokines and chemokines by Luminex technology confirmed production and secretion of proinflammatory cytokines (e.g., interleukin [IL]-6 and tumor necrosis factor-α) as well as various chemotactic proteins, such as IFN-γ-induced protein 10, macrophage inflammatory protein (MIP)-1α, MIP-1ß, and IL-8. Infection with other HEV-Bs induced similar responses, yet their extent depended on replication efficiency. Ultra violet-inactivated CVB3 did not induce any response, suggesting that virus replication is a prerequisite for antiviral responses. Our data represent the first comprehensive overview of inflammatory mediators that are secreted by human islets of Langerhans upon CVB infection and may shed light on the role of enteroviruses in type 1 diabetes pathogenesis.


Assuntos
Quimiocinas/biossíntese , Infecções por Coxsackievirus/metabolismo , Citocinas/biossíntese , Enterovirus Humano B , Ilhotas Pancreáticas/metabolismo , Diabetes Mellitus Tipo 1/virologia , Humanos
12.
Viral Immunol ; 23(1): 99-104, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20121407

RESUMO

Previous studies have shown that enteroviral RNA can be detected in blood at the onset of type 1 diabetes (T1D). The infection may play a role in triggering T1D and genetic host factors may contribute to this process. We investigated (1) whether enterovirus is present at the onset of T1D in peripheral blood mononuclear cells (PBMC), plasma, throat, or stool, and (2) whether enteroviral presence is linked with HLA-DR type and/or polymorphisms in melanoma differentiation-associated gene 5 (MDA5) and 2'-5' oligoadenylate synthetase 1 (OAS1), factors of antiviral immunity. To this end, PBMC, plasma, throat, and stool samples from 10 T1D patients and 20 unrelated controls were tested for the presence of enteroviruses (RT-PCR), for HLA-DR type, and polymorphisms in MDA5 and OAS1. Enterovirus RNA was detected in PBMC of 4/10 T1D patients, but none of 20 controls. Plasma was positive in 2/10 T1D patients and none of 20 controls, suggesting that enteroviruses found at the onset of T1D are mainly present in PBMC. All throat samples from positive T1D patients were virus-negative and only 1 fecal sample was positive. The negative results for all throat and most stool samples argues against acute infection. Enterovirus presence was linked with HLA-DR4, but not with polymorphisms in MDA5 or OAS1.


Assuntos
Sangue/virologia , Diabetes Mellitus Tipo 1/virologia , Infecções por Enterovirus/complicações , Infecções por Enterovirus/virologia , Enterovirus/isolamento & purificação , Leucócitos Mononucleares/virologia , RNA Viral/isolamento & purificação , 2',5'-Oligoadenilato Sintetase/genética , Adolescente , Criança , Pré-Escolar , RNA Helicases DEAD-box/genética , Enterovirus/genética , Fezes/virologia , Feminino , Antígenos HLA-DR/genética , Humanos , Helicase IFIH1 Induzida por Interferon , Masculino , Faringe/virologia , Plasma/virologia , Polimorfismo Genético , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
BMJ ; 340: c1018, 2010 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-20185493

RESUMO

OBJECTIVE: The presence of the retrovirus xenotropic murine leukaemia virus-related virus (XMRV) has been reported in peripheral blood mononuclear cells of patients with chronic fatigue syndrome. Considering the potentially great medical and social relevance of such a discovery, we investigated whether this finding could be confirmed in an independent European cohort of patients with chronic fatigue syndrome. DESIGN: Analysis of a well defined cohort of patients and matched neighbourhood controls by polymerase chain reaction. SETTING: Certified (ISO 15189) laboratory of clinical virology in a university hospital in the Netherlands. Population Between December 1991 and April 1992, peripheral blood mononuclear cells were isolated from 76 patients and 69 matched neighbourhood controls. In this study we tested cells from 32 patients and 43 controls from whom original cryopreserved phials were still available. MAIN OUTCOME MEASURES: Detection of XMRV in peripheral blood mononuclear cells by real time polymerase chain reaction assay targeting the XMRV integrase gene and/or a nested polymerase chain reaction assay targeting the XMRV gag gene. RESULTS: We detected no XMRV sequences in any of the patients or controls in either of the assays, in which relevant positive and negative isolation controls and polymerase chain reaction controls were included. Spiking experiments showed that we were able to detect at least 10 copies of XMRV sequences per 10(5) peripheral blood mononuclear cells by real time as well as by nested polymerase chain reaction, demonstrating high sensitivity of both assays. CONCLUSIONS: This study failed to show the presence of XMRV in peripheral blood mononuclear cells of patients with chronic fatigue syndrome from a Dutch cohort. These data cast doubt on the claim that XMRV is associated with chronic fatigue syndrome in the majority of patients.


Assuntos
Síndrome de Fadiga Crônica/virologia , Vírus da Leucemia Murina/isolamento & purificação , Infecções por Retroviridae/complicações , Adulto , Estudos de Casos e Controles , DNA Viral , Feminino , Humanos , Leucócitos Mononucleares/virologia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Estudos Retrospectivos
14.
Diabetes ; 59(5): 1182-91, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20071599

RESUMO

OBJECTIVE: Type 1 diabetes is a chronic endocrine disorder in which enteroviruses, such as coxsackie B viruses and echoviruses, are possible environmental factors that can trigger or accelerate disease. The development or acceleration of type 1 diabetes depends on the balance between autoreactive effector T-cells and regulatory T-cells. This balance is particularly influenced by dendritic cells (DCs). The goal of this study was to investigate the interaction between enterovirus-infected human pancreatic islets and human DCs. RESEARCH DESIGN AND METHODS: In vitro phagocytosis of human or porcine primary islets or Min6 mouse insuloma cells by DCs was investigated by flow cytometry and confocal analysis. Subsequent innate DC responses were monitored by quantitative PCR and Western blotting of interferon-stimulated genes (ISGs). RESULTS: In this study, we show that both mock- and coxsackievirus B3 (CVB3)-infected human and porcine pancreatic islets were efficiently phagocytosed by human monocyte-derived DCs. Phagocytosis of CVB3-infected, but not mock-infected, human and porcine islets resulted in induction of ISGs in DCs, including the retinoic acid-inducible gene (RIG)-I-like helicases (RLHs), RIG-I, and melanoma differentiation-associated gene 5 (Mda5). Studies with murine Min6 insuloma cells, which were also efficiently phagocytosed, revealed that increased ISG expression in DCs upon encountering CVB-infected cells resulted in an antiviral state that protected DCs from subsequent enterovirus infection. The observed innate antiviral responses depended on RNA within the phagocytosed cells, required endosomal acidification, and were type I interferon dependent. CONCLUSIONS: Human DCs can phagocytose enterovirus-infected pancreatic cells and subsequently induce innate antiviral responses, such as induction of RLHs. These responses may have important consequences for immune homeostasis in vivo and may play a role in the etiology of type 1 diabetes.


Assuntos
Células Dendríticas/imunologia , Enterovirus/fisiologia , Imunidade Inata/imunologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/virologia , Fagocitose/imunologia , Animais , Células Cultivadas , Enterovirus/efeitos dos fármacos , Humanos , Células Secretoras de Insulina/imunologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/virologia , Camundongos , Suínos , Células Tumorais Cultivadas
15.
Antimicrob Agents Chemother ; 53(5): 1850-7, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19237651

RESUMO

A novel compound, TTP-8307, was identified as a potent inhibitor of the replication of several rhino- and enteroviruses. TTP-8307 inhibits viral RNA synthesis in a dose-dependent manner, without affecting polyprotein synthesis and/or processing. Drug-resistant variants of coxsackievirus B3 were all shown to carry at least one amino acid mutation in the nonstructural protein 3A. In particular, three mutations located in a nonstructured region preceding the hydrophobic domain (V45A, I54F, and H57Y) appeared to contribute to the drug-resistant phenotype. This region has previously been identified as a hot sport for mutations that resulted in resistance to enviroxime, the sole 3A-targeting enterovirus inhibitor reported thus far. This was corroborated by the fact that TTP-8307 and enviroxime proved cross-resistant. It is hypothesized that TTP-8307 and enviroxime disrupt proper interactions of 3A(B) with other viral or cellular proteins that are required for efficient replication.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/genética , Enterovirus/efeitos dos fármacos , Mutação , Proteínas não Estruturais Virais/genética , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/química , Benzimidazóis/química , Benzimidazóis/farmacologia , Chlorocebus aethiops , Enterovirus/genética , Enterovirus/metabolismo , Enterovirus/fisiologia , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Enterovirus Humano B/fisiologia , Células HeLa/virologia , Humanos , Testes de Sensibilidade Microbiana/métodos , Oximas , Poliovirus/efeitos dos fármacos , Poliovirus/genética , Rhinovirus/efeitos dos fármacos , Rhinovirus/genética , Rhinovirus/metabolismo , Rhinovirus/fisiologia , Sulfonamidas , Células Vero/virologia , Proteínas não Estruturais Virais/química
16.
J Virol ; 82(10): 4720-30, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18337578

RESUMO

TBZE-029 {1-(2,6-difluorophenyl)-6-trifluoromethyl-1H,3H-thiazolo[3,4-a]benzimidazole} is a novel selective inhibitor of the replication of several enteroviruses. We show that TBZE-029 exerts its antiviral activity through inhibition of viral RNA replication, without affecting polyprotein processing. To identify the viral target of TBZE-029, drug-resistant coxsackievirus B3 (CVB3) was selected. Genotyping of resistant clones led to the identification of three amino acid mutations in nonstructural protein 2C, clustered at amino acid positions 224, 227, and 229, immediately downstream of NTPase/helicase motif C. The mutations were reintroduced, either alone or combined, into an infectious full-length CVB3 clone. In particular the mutations at positions 227 and 229 proved essential for the altered sensitivity of CVB3 to TBZE-029. Resistant virus exhibited cross-resistance to the earlier-reported antienterovirus agents targeting 2C, namely, guanidine hydrochloride, HBB [2-(alpha-hydroxybenzyl)-benzimidazole], and MRL-1237 {1-(4-fluorophenyl)-2-[(4-imino-1,4-dihydropyridin-1-yl)methyl]benzimidazole hydrochloride}. The ATPase activity of 2C, however, remained unaltered in the presence of TBZE-029.


Assuntos
Antivirais/farmacologia , Benzimidazóis/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Enterovirus Humano B/efeitos dos fármacos , Tiazóis/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Adenosina Trifosfatases/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Chlorocebus aethiops , Farmacorresistência Viral/genética , Enterovirus Humano B/genética , Guanidina/farmacologia , Estrutura Molecular , Mutação de Sentido Incorreto , Piridinas/farmacologia , RNA Viral/biossíntese , Células Vero , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
17.
J Virol ; 82(6): 2930-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18184700

RESUMO

Dendritic cells (DCs) play a central role in instructing antiviral immune responses. DCs, however, can become targeted by different viruses themselves. We recently demonstrated that human DCs can be productively infected with echoviruses (EVs), but not coxsackie B viruses (CVBs), both of which are RNA viruses belonging to the Enterovirus genus of the Picornaviridae family. We now show that phagocytosis of CVB-infected, type I interferon-deficient cells induces an antiviral state in human DCs. Uptake of infected cells increased the expression of the cytoplasmic RNA helicases retinoic acid-inducible gene I and melanoma differentiation-associated gene 5 as well as other interferon-stimulated genes and protected DCs against subsequent infection with EV9. These effects depended on recognition of viral RNA and could be mimicked by exposure to the synthetic double-stranded RNA analogue poly(I:C) but not other Toll-like receptor (TLR) ligands. Blocking endosomal acidification abrogated protection, suggesting a role for TLRs in the acquisition of an antiviral state in DCs. In conclusion, recognition of viral RNA rapidly induces an antiviral state in human DCs. This might provide a mechanism by which DCs protect themselves against viruses when attracted to an environment with ongoing infection.


Assuntos
Células Dendríticas/imunologia , Fagocitose , Picornaviridae/imunologia , RNA Viral/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Humanos , Microscopia Confocal , Reação em Cadeia da Polimerase , Regulação para Cima , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA