Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 17924, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31784575

ABSTRACT

In the present study, patients with acute OROV fever were classified as early seroconverters (IgM/IgG positive at baseline) or late seroconverters (IgM/IgG negative at baseline) and the timeline kinetics of the production of chemokines and cytokines were assessed at 1-3, 4-7, 8-10 and ≥11 days after patients have reported the first symptoms. Regardless immunoglobulin profile, all OROV fever patients presented higher levels of CXCL8, and IFN-α and lower levels of TNF and IL-10 at baseline as compared to healthy donors (HD). Lower levels of CCL2, CXCL10, and IFN-γ and higher levels of CCL2, CXCL10, IL-6, and IL-17A were detected in early and late seroconverters, respectively, as compared to HD. While early seroconverters presented the increasing levels of CCL2 along the timeline, late seroconverters displayed decreasing levels of CCL2, CXCL10, and IL-6 following days of disease onset. Noteworthy was that IFN-α was revealed as universal biomarker of human OROV fever, while CXCL8 & IL-5 and CXCL10 & IL-17 were consistently observed in early and late seroconverters, respectively. Thus, our results suggest that the production of IFN-α, CXCL10, and IL-17 precede the seroconversion bringing novel insights on the immunological events triggered by the OROV disease.


Subject(s)
Bunyaviridae Infections/blood , Interferon-alpha/blood , Seroconversion , Biomarkers/blood , Bunyaviridae Infections/immunology , Bunyaviridae Infections/pathology , Chemokines/blood , Humans , Interferon-gamma/blood , Interleukin-27/blood , Interleukin-6/blood , Serologic Tests/methods , Serologic Tests/standards , Time
2.
Front Immunol ; 10: 2192, 2019.
Article in English | MEDLINE | ID: mdl-31616412

ABSTRACT

The Yellow Fever (YF) vaccination is recommended for people living in endemic areas and represents the most effective strategy to reduce the risk of infection. Previous studies have warned that booster regimens should be considered to guarantee the long-term persistence of 17DD-YF-specific memory components in adults living in areas with YF-virus circulation. Considering the lower seroconversion rates observed in children (9-12 months of age) as compared to adults, this study was designed in order to access the duration of immunity in single-dose vaccinated children in a 10-years cross-sectional time-span. The levels of neutralizing antibodies (PRNT) and the phenotypic/functional memory status of T and B-cells were measured at a baseline, 30-45 days, 1, 2, 4, 7, and 10 years following primary vaccination. The results revealed that a single dose induced 85% of seropositivity at 30-45 days and a progressive time-dependent decrease was observed as early as 2 years and declines toward critical values (below 60%) at time-spans of ≥4-years. Moreover, short-lived YF-specific cellular immunity, mediated by memory T and B-cells was also observed after 4-years. Predicted probability and resultant memory analysis emphasize that correlates of protection (PRNT; effector memory CD8+ T-cells; non-classical memory B-cells) wane to critical values within ≥4-years after primary vaccination. Together, these results clearly demonstrate the decline of 17DD-YF-specific memory response along time in children primarily vaccinated at 9-12 months of age and support the need of booster regimen to guarantee the long-term persistence of memory components for children living in areas with high risk of YF transmission.


Subject(s)
Immunity/immunology , Yellow Fever Vaccine/immunology , Yellow Fever/immunology , Yellow Fever/prevention & control , Yellow fever virus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Immunization, Secondary/methods , Infant , Male , Vaccination/methods
3.
Arch Virol ; 164(4): 1187-1192, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30725182

ABSTRACT

Yellow fever virus (YFV) penetrates the skin through the bite of a vector mosquito and spreads to various organs, mainly the liver, where it causes lesions and induces necrosis and apoptosis. We evaluated the mRNA expression of various cytokines and the activation of caspases in HepG2 cells infected with YFV. We observed that interferon-α (IFN-α) expression decreased and IFN-ß, transforming growth factor (TGF)-ß IIIR, interleukin (IL)-6, and IL-8 expression increased in cells infected with genotype 1. In contrast, TNF-α expression increased in cells infected with genotype 2 but not with genotype 1. This provides insights into the role of cytokine regulation in yellow fever.


Subject(s)
Caspase 3/metabolism , Caspase 7/metabolism , Cytokines/genetics , Liver Neoplasms/genetics , Yellow fever virus/physiology , Caspase 3/genetics , Caspase 7/genetics , Cell Line , Cytokines/metabolism , Hep G2 Cells , Host-Pathogen Interactions , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Yellow fever virus/genetics
4.
Infect Genet Evol ; 68: 16-22, 2019 03.
Article in English | MEDLINE | ID: mdl-30504003

ABSTRACT

Oropouche orthobunyavirus (OROV) has significant impact in public health in Amazon region. This arbovirus is one of the most common causes of febrile illness in Brazil, and is responsible for several epidemics since 1960's. In this study, we sequenced and characterized the complete coding sequences (S-, M- and L-RNA) of 35 OROV isolates from Brazil. Here, we classified 20 strains in genotype I from Pará and Maranhão states, nine as genotype II from Pará and Rondônia states confirmed, four classified into genotype III from Acre, Maranhão, Minas Gerais and Rondônia states and two genotype IV from Amazonas State. Also, we did not observe reassortment events involving the OROV isolates. In addition, we developed novel RT-PCR tools to identify reassortment events among OROV strains. These data will be useful to better understand the molecular epidemiology and diagnostic of OROV infections.


Subject(s)
Bunyaviridae Infections/virology , Genome, Viral , Genomics , Orthobunyavirus/genetics , Reassortant Viruses/genetics , Animals , Brazil/epidemiology , Chlorocebus aethiops , Computational Biology/methods , Genomics/methods , Genotype , Geography, Medical , Humans , Molecular Epidemiology , Molecular Sequence Annotation , Molecular Typing , Orthobunyavirus/classification , Phylogeny , Vero Cells
5.
PLoS Negl Trop Dis ; 12(6): e0006462, 2018 06.
Article in English | MEDLINE | ID: mdl-29879134

ABSTRACT

In this investigation, machine-enhanced techniques were applied to bring about scientific insights to identify a minimum set of phenotypic/functional memory-related biomarkers for post-vaccination follow-up upon yellow fever (YF) vaccination. For this purpose, memory status of circulating T-cells (Naïve/early-effector/Central-Memory/Effector-Memory) and B-cells (Naïve/non-Classical-Memory/Classical-Memory) along with the cytokine profile (IFN/TNF/IL-5/IL-10) were monitored before-NV(day0) and at distinct time-points after 17DD-YF primary vaccination-PV(day30-45); PV(year1-9) and PV(year10-11). A set of biomarkers (eEfCD4; EMCD4; CMCD19; EMCD8; IFNCD4; IL-5CD8; TNFCD4; IFNCD8; TNFCD8; IL-5CD19; IL-5CD4) were observed in PV(day30-45), but not in NV(day0), with most of them still observed in PV(year1-9). Deficiencies of phenotypic/functional biomarkers were observed in NV(day0), while total lack of memory-related attributes was observed in PV(year10-11), regardless of the age at primary vaccination. Venn-diagram analysis pre-selected 10 attributes (eEfCD4, EMCD4, CMCD19, EMCD8, IFNCD4, IL-5CD8, TNFCD4, IFNCD8, TNFCD8 and IL-5CD4), of which the overall mean presented moderate accuracy to discriminate PV(day30-45)&PV(year1-9) from NV(day0)&PV(year10-11). Multi-parameter approaches and decision-tree algorithms defined the EMCD8 and IL-5CD4 attributes as the top-two predictors with moderated performance. Together with the PRNT titers, the top-two biomarkers led to a resultant memory status observed in 80% and 51% of volunteers in PV(day30-45) and PV(year1-9), contrasting with 0% and 29% found in NV(day0) and PV(year10-11), respectively. The deficiency of memory-related attributes observed at PV(year10-11) underscores the conspicuous time-dependent decrease of resultant memory following17DD-YF primary vaccination that could be useful to monitor potential correlates of protection in areas under risk of YF transmission.


Subject(s)
Antibodies, Viral/blood , Biomarkers/blood , Vaccination , Yellow Fever Vaccine/immunology , Yellow Fever/prevention & control , Yellow fever virus/immunology , Adolescent , Adult , Aged , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/blood , Humans , Male , Middle Aged , Time Factors , Yellow Fever/immunology , Yellow Fever/virology , Young Adult
6.
Viral Immunol ; 31(8): 583-588, 2018 10.
Article in English | MEDLINE | ID: mdl-29878881

ABSTRACT

Zika virus (ZIKV) is an arbovirus belonging to the genus Flavivirus (Flaviviridae). ZIKV infection is associated with alterations in various organs, including the liver, lungs, and kidneys. Studies on the influence of posttranscriptional control on viral infections have demonstrated that microRNAs (miRNAs) interfere with different stages of the replicative cycle of several viruses and may influence the disease outcome. To shed light on ZIKV-induced regulation of host miRNA-processing machinery in the above organs, we analyzed the expression of genes encoding key proteins of the miRNA pathway in different ZIKV-infected continuous primate cell lineages (HepG2, A549, and MA104) by reverse-transcription quantitative polymerase chain reaction (RT-qPCR). Expression of the genes encoding the miRNA-related proteins DGCR8, Ago1, and Ago3 in HepG2 cells and Drosha, Dicer, Ago2, and Ago3 in A549 and MA104 cells was significantly altered in the presence of ZIKV. Our results suggest that ZIKV modulates miRNA levels during infection in liver, lung, and kidney cells, which may be an additional mechanism of host cell subversion in these organs.


Subject(s)
Kidney/cytology , Liver/cytology , Lung/cytology , MicroRNAs/genetics , Zika Virus/immunology , Animals , Cell Lineage , Chlorocebus aethiops , Gene Expression Regulation , Hep G2 Cells , Host-Pathogen Interactions/genetics , Humans , Kidney/virology , Liver/virology , Lung/virology , Virus Replication
7.
Viral Immunol ; 30(5): 336-341, 2017 06.
Article in English | MEDLINE | ID: mdl-28388330

ABSTRACT

Yellow fever is a zoonotic disease caused by the yellow fever virus (YFV) and transmitted by mosquitoes of the family Culicidae. It is well known that cellular and viral microRNAs (miRNAs) are involved in modulation of viral and cellular gene expression, as well as immune response, and are considered by the scientific community as possible targets for an effective therapy against viral infections. This regulation may be involved in different levels of infection and clinical symptomatology. We used viral titration techniques, viral kinetics from 24 to 96 hours postinfection (hpi), and analyzed the expression of key proteins related to the miRNA pathway by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). The expression of Dicer was different when compared over the course of infection by the distinct YFV genotypes. Drosha expression was similar during infection by YFV genotype 1 or 2, with a decrease in their expression over time and a slight increase in 96 hpi. Ago1, Ago2, and Ago4 showed different levels of expression between the viral genotypes: for YFV genotype 1 infection, Ago1 presented a positive expression, while for YFV genotype 2, it showed a negative expression, when compared with negative controls. We conclude that YFV infection modulates the proteins involved in miRNA biogenesis, which can regulate both viral replication and cellular immune response.


Subject(s)
Gene Expression Profiling , Host-Pathogen Interactions , MicroRNAs/biosynthesis , Yellow fever virus/physiology , Gene Expression Regulation , Hep G2 Cells , Humans , Real-Time Polymerase Chain Reaction , Time Factors , Viral Load
8.
Rev. Inst. Med. Trop. Säo Paulo ; 35(2): 141-148, Mar.-Apr. 1993.
Article in Portuguese | LILACS | ID: lil-320576

ABSTRACT

We report the first outbreak of dengue fever caused by dengue 2 (DEN 2) in Araguaina, Tocantins State. Four hundred people of 74 families, living at S. João, Araguaina Sul and Neblina districts were questioned and then bled, in order to obtain sera to test for anti-dengue antibodies. If a person was sick, a small quantity of blood was collected for virus isolation. The main clinical picture of disease was characterized by fever, headache, myalgias, arthralgias and skin rash. Were obtained 1,105 (56 females and 45 males of Culex quinquefasciatus and 567 females and 437 males of Aedes aegypti) mosquitoes from larvae collected in Araguaina. The females of Aedes aegypti obtained from larvae were allowed to feed on 8 febrile patients. The diagnosis of infection was made by both virus isolation into Aedes albopictus (C6/36) cells, and serology, by Hemagglutination-inhibition (HI) and IgM capture ELISA (MAC ELISA). No virus was isolated from mosquitoes. Although five strains of DEN 2 were obtained from humans, and another 111 infections were diagnosed serologically (IgM positive). The positivity rate of the samples was 27.75 (111 of 400), while that of the families was 66.2 (45 of 72), where at least one member of the each family was infected. It was also detected 26.1 of asymptomatic infections. All age groups were affected. Therefore, the infection was more frequent in females (33.5) than males (23.8).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Humans , Male , Female , Infant, Newborn , Infant , Child, Preschool , Child , Adolescent , Adult , Middle Aged , Dengue , Age Distribution , Brazil , Dengue Virus , Sex Distribution
9.
In. Brasil. Ministerio da Saude; Organizacao Pan-Americana da Saude; Minas Gerais. Secretaria da Saúde. 1§ Seminario Nacional de Zoonoses. s.l, Brasil. Ministerio da Saude, out. 1985. p.57-62.
Monography in Portuguese | LILACS | ID: lil-76090
SELECTION OF CITATIONS
SEARCH DETAIL
...