Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.278
Filter
1.
J Med Virol ; 96(8): e29804, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39092809

ABSTRACT

Although rhinoviruses play a major role in exacerbations of childhood asthma, the presence of rhinovirus (RV) RNA in plasma, referred to as viremia, has been investigated in a few studies. The aim of the study was to investigate the presence of rhinovirus viremia at the time of asthma exacerbation and to describe the molecular characteristics of rhinoviruses associated with viremia. We conducted an observational, prospective, multicenter study in eight pediatric hospitals (VIRASTHMA2). Preschool-aged recurrent wheezers (1-5 years) hospitalized for a severe exacerbation were included. Reverse-transcription polymerase chain reaction (RT-PCR) and molecular typing for RV/enteroviruses (EV) were performed on nasal swabs and plasma. Plasma specimens were available for 105 children with positive RT-PCR for RV/EV in respiratory specimens. Thirty-six (34.3%) had positive viremia. In plasma, 28 (82.4%) of the typable specimens were RV-C, five (14.7%) were EV-D68, and one was RV-A (2.9%). In all cases, the RV/EV type was identical in the plasma and respiratory specimens. In conclusion, RV/EV viremia is frequent in severe exacerbations of preschool recurrent wheezers, particularly in RV-C infections.


Subject(s)
Asthma , Picornaviridae Infections , Rhinovirus , Viremia , Humans , Viremia/virology , Child, Preschool , Rhinovirus/genetics , Rhinovirus/isolation & purification , Rhinovirus/classification , Asthma/virology , Male , Female , Prospective Studies , Picornaviridae Infections/virology , Infant , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Plasma/virology
2.
Virology ; 598: 110192, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39106585

ABSTRACT

In vitro studies have shown that deletion of nef and deleterious mutation in the Nef dimerization interface attenuates HIV replication and associated pathogenesis. Humanized rodents with human immune cells and lymphoid tissues are robust in vivo models for investigating the interactions between HIV and the human immune system. Here, we demonstrate that nef deletion impairs HIV replication and HIV-induced immune dysregulation in the blood and human secondary lymphoid tissue (human spleen) in bone marrow-liver-thymus-spleen (BLTS) humanized mice. Furthermore, we also show that nef defects (via deleterious mutations in the dimerization interface) impair HIV replication and HIV-induced immune dysregulation in the blood and human spleen in BLTS-humanized mice. We demonstrate that the reduced replication of nef-deleted and nef-defective HIV is associated with robust antiviral innate immune response, and T helper 1 response. Our results support the proposition that Nef may be a therapeutic target for adjuvants in HIV cure strategies.


Subject(s)
Disease Models, Animal , HIV Infections , HIV-1 , Liver , Spleen , Viremia , Virus Replication , nef Gene Products, Human Immunodeficiency Virus , Animals , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/immunology , HIV Infections/immunology , HIV Infections/virology , Mice , Humans , Viremia/immunology , Spleen/immunology , Spleen/virology , HIV-1/immunology , HIV-1/genetics , HIV-1/physiology , Liver/virology , Liver/immunology , Liver/pathology , Bone Marrow/virology , Bone Marrow/immunology , Thymus Gland/immunology , Thymus Gland/virology , Immunity, Innate
3.
Science ; 385(6709): eadn5866, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39116226

ABSTRACT

Antiviral therapies with reduced frequencies of administration and high barriers to resistance remain a major goal. For HIV, theories have proposed that viral-deletion variants, which conditionally replicate with a basic reproductive ratio [R0] > 1 (termed "therapeutic interfering particles" or "TIPs"), could parasitize wild-type virus to constitute single-administration, escape-resistant antiviral therapies. We report the engineering of a TIP that, in rhesus macaques, reduces viremia of a highly pathogenic model of HIV by >3log10 following a single intravenous injection. Animal lifespan was significantly extended, TIPs conditionally replicated and were continually detected for >6 months, and sequencing data showed no evidence of viral escape. A single TIP injection also suppressed virus replication in humanized mice and cells from persons living with HIV. These data provide proof of concept for a potential new class of single-administration antiviral therapies.


Subject(s)
Artificial Virus-Like Particles , Gene Deletion , HIV Infections , HIV-1 , Viral Interference , Virus Replication , Animals , Humans , Mice , Basic Reproduction Number , Disease Models, Animal , Genetic Engineering , HIV Infections/therapy , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Macaca mulatta , Proof of Concept Study , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Viremia/therapy , Viremia/virology
4.
PLoS Pathog ; 20(8): e1012328, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102417

ABSTRACT

Spring viremia of carp virus (SVCV) has a broad fish host spectrum and is responsible for a disease that generally affects juvenile fishes with a mortality rate of up to 90%. In the absence of treatments or vaccines against SVCV, the search for prophylactic or therapeutic solutions is thus relevant, particularly to identify solutions compatible with mass vaccination. In addition to being a threat to aquaculture and ecosystems, SVCV is a unique pathogen to study virus-host interactions in the zebrafish model. Establishing the first reverse genetics system for SVCV and the design of recombinant SVCV (rSVCV) expressing fluorescent or bioluminescent proteins adds a new dimension for the study of these interactions using innovative imaging techniques. The infection by bath immersion of zebrafish larvae with rSVCV expressing mCherry allows us to define the first SVCV replication sites and the host innate immune responses using different transgenic lines of zebrafish. The fins were found as the main initial sites of infection in both zebrafish and carp, its natural host. Hence, new insights into the physiopathology of SVCV infection have been described. We report that neutrophils are recruited at the sites of infection and persist up to the death of the animal leading to an uncontrolled inflammation correlated with the expression of the pro-inflammatory cytokine IL1ß. Tissue damage was observed at the site of initial replication, a likely consequence of virus-induced injury or the pro-inflammatory response. Interestingly, SVCV infection by bath immersion triggers a persistent pro-inflammatory response rather than activation of the antiviral IFN signaling pathway as observed following intravenous injection, highlighting the importance of the route of infection on the progression of pathogenicity. Thus, this model of zebrafish larvae infection by rSVCV offers new perspectives to study in detail virus-host interactions and to discover new prophylactic or therapeutic solutions.


Subject(s)
Carps , Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Zebrafish , Animals , Zebrafish/virology , Rhabdoviridae/physiology , Fish Diseases/virology , Rhabdoviridae Infections/virology , Rhabdoviridae Infections/immunology , Carps/virology , Animals, Genetically Modified , Disease Models, Animal , Immunity, Innate , Viremia
5.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125815

ABSTRACT

Neurological symptoms associated with COVID-19, acute and long term, suggest SARS-CoV-2 affects both the peripheral and central nervous systems (PNS/CNS). Although studies have shown olfactory and hematogenous invasion into the CNS, coinciding with neuroinflammation, little attention has been paid to susceptibility of the PNS to infection or to its contribution to CNS invasion. Here we show that sensory and autonomic neurons in the PNS are susceptible to productive infection with SARS-CoV-2 and outline physiological and molecular mechanisms mediating neuroinvasion. Our infection of K18-hACE2 mice, wild-type mice, and golden Syrian hamsters, as well as primary peripheral sensory and autonomic neuronal cultures, show viral RNA, proteins, and infectious virus in PNS neurons, satellite glial cells, and functionally connected CNS tissues. Additionally, we demonstrate, in vitro, that neuropilin-1 facilitates SARS-CoV-2 neuronal entry. SARS-CoV-2 rapidly invades the PNS prior to viremia, establishes a productive infection in peripheral neurons, and results in sensory symptoms often reported by COVID-19 patients.


Subject(s)
COVID-19 , Neuropilin-1 , SARS-CoV-2 , Animals , SARS-CoV-2/physiology , SARS-CoV-2/pathogenicity , COVID-19/virology , COVID-19/pathology , COVID-19/metabolism , Mice , Neuropilin-1/metabolism , Neuropilin-1/genetics , Viremia/virology , Central Nervous System/virology , Central Nervous System/pathology , Central Nervous System/metabolism , Sensory Receptor Cells/virology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Mesocricetus , Humans , Angiotensin-Converting Enzyme 2/metabolism , Mice, Inbred C57BL , Virus Internalization , Male
6.
Front Cell Infect Microbiol ; 14: 1413589, 2024.
Article in English | MEDLINE | ID: mdl-39170987

ABSTRACT

Background: About 20% of on-treatment patients with chronic hepatitis B (CHB) experienced low-level viraemia (LLV), which is associated with persistent low-grade inflammation, fibrosis progression, and increased risk of hepatocellular carcinoma. We aimed to investigate the high-risk factors related to LLV. Methods: In this retrospective study, patients receiving entecavir (ETV) treatment from January 2018 to January 2023 were enrolled, and were divided into a LLV (HBV DNA 20-2000 IU/mL) cohort and a complete virological response (CVR) (HBV DNA < 20 IU/mL) cohort according to the virological response at week 48 posttreatment. Treatment baseline characteristics were retrieved from electronic medical records. Multivariate logistic regression was performed. Results: Totally, 1653 patients were enrolled, male patients accounted for 73.0%; the median age was 44 years; the mean HBV DNA level was 5.9 Log10 IU/ml. Among them, 472 (28.6%) experienced LLV. Multivariate analysis showed that HBeAg positivity (OR = 2.650, 95% CI: 2.000-3.511, p < 0.001), HBV DNA ≥ 6.0 Log10 IU/mL (OR = 1.370, 95% CI: 1.054-1.780, p = 0.019), qHBsAg ≥ 9000 IU/mL (OR = 4.472, 95% CI: 3.410-5.866, p < 0.001), cirrhosis (OR = 1.650, 95% CI: 1.234-2.207, P = 0.001), LSM ≥ 13.0 kPa (OR = 1.644, 95% CI: 1.203-2.246, p = 0.002), and PLT < 100×109/L (OR = 1.450, 95% CI: 1.094-1.922, p = 0.010) at baseline were related to the development of LLV. Conclusions: High HBV DNA/HBsAg quantification/LSM, low PLT, HBeAg positivity, and liver cirrhosis were high-risk factors associated with LLV in patients receiving entecavir treatment.


Subject(s)
Antiviral Agents , DNA, Viral , Guanine , Hepatitis B virus , Hepatitis B, Chronic , Viremia , Humans , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/complications , Male , Guanine/analogs & derivatives , Guanine/therapeutic use , Female , Adult , Risk Factors , Antiviral Agents/therapeutic use , Retrospective Studies , Middle Aged , Hepatitis B virus/genetics , Hepatitis B virus/drug effects , DNA, Viral/blood , Hepatitis B e Antigens/blood , Liver Cirrhosis/virology , Viral Load/drug effects
7.
Virol J ; 21(1): 177, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107806

ABSTRACT

BACKGROUND: Reticuloendotheliosis virus (REV), a member of the family Retroviridae, is a hot area of research, and a previous study showed that exosomes purified from REV-positive semen were not blocked by REV-specific neutralizing antibodies and established productive infections. METHODS: To further verify the infectivity of exosomes from REV-infected cells, we isolated and purified exosomes from REV-infected DF-1 cells and identified them using Western blot and a transmission electron microscope. We then inoculated 7-day-old embryonated eggs, 1-day-old chicks and 23-week-old hens with and without antibody treatment. REV was administered simultaneously as a control. RESULTS: In the absence of antibodies, the results indicated that REV-exosomes and REV could infect chicks, resulting in viremia and viral shedding, compared with the infection caused by REV, REV-exosomes reduced the hatching rate and increased mortality after hatching, causing severe growth inhibition and immune organ damage in 1-day-old chicks; both REV and REV-exosomes also could infect hens, however, lead to transient infection. In the presence of antibodies, REV-exosomes were not blocked by REV-specific neutralizing antibodies and infected 7-day-old embryonated eggs. However, REV could not infect 1-day-old chicks and 23-week-old hens. CONCLUSION: In this study, we compared the infectious ability of REV-exosomes and REV, REV-exosomes could escape from REV-specific neutralizing antibodies in embryonated eggs, providing new insights into the immune escape mechanism of REV.


Subject(s)
Antibodies, Viral , Chickens , Exosomes , Poultry Diseases , Reticuloendotheliosis virus , Retroviridae Infections , Virus Shedding , Animals , Exosomes/virology , Exosomes/immunology , Antibodies, Viral/immunology , Chickens/virology , Reticuloendotheliosis virus/immunology , Poultry Diseases/virology , Poultry Diseases/transmission , Poultry Diseases/immunology , Retroviridae Infections/virology , Retroviridae Infections/transmission , Retroviridae Infections/immunology , Retroviridae Infections/veterinary , Antibodies, Neutralizing/immunology , Cell Line , Viremia/virology , Female
8.
J Biomed Sci ; 31(1): 80, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160510

ABSTRACT

BACKGROUND: Around 10% of people with HIV (PWH) exhibit a low-level viremia (LLV) under antiretroviral therapy (ART). However, its origin and clinical significance are largely unknown, particularly at viremias between 50 and 200 copies/mL and under modern ART based on integrase strand transfer inhibitors (INSTIs). Our aim was to characterize their poor immune response against HIV in comparison to individuals with suppressed viremia (SV) and non-HIV controls (NHC). METHODS: Transversal observational study in 81 matched participants: 27 PWH with LLV, 27 PWH with SV, and 27 NHC. Activation (CD25, HLA-DR, and CD38) and senescence [CD57, PD1, and HAVCR2 (TIM3)] were characterized in peripheral T-cell subsets by spectral flow cytometry. 45 soluble biomarkers of systemic inflammation were evaluated by immunoassays. Differences in cell frequencies and plasma biomarkers among groups were evaluated by a generalized additive model for location, scale, and shape (GAMLSS) and generalized linear model (GLM) respectively, adjusted by age, sex at birth, and ART regimen. RESULTS: The median age was 53 years and 77.8% were male. Compared to NHC, PWH showed a lower CD4+/CD8+ ratio and increased activation, senescence, and inflammation, highlighting IL-13 in LLV. In addition, LLV showed a downtrend in the frequency of CD8+ naive and effector memory (EM) type 1 compared to SV, along with higher activation and senescence in CD4+ and CD8+ EM and terminally differentiated effector memory RA+ (TEMRA) subpopulations. No significant differences in systemic inflammation were observed between PWH groups. CONCLUSION: LLV between 50 and 200 copies/mL leads to reduced cytotoxic activity and T-cell dysfunction that could affect cytokine production, being unable to control and eliminate infected cells. The increase in senescence markers suggests a progressive loss of immunological memory and a reduction in the proliferative capacity of immune cells. This accelerated immune aging could lead to an increased risk of developing future comorbidities. These findings strongly advocate for heightened surveillance of these PWH to promptly identify potential future complications.


Subject(s)
Cellular Senescence , HIV Infections , HIV-1 , Lymphocyte Activation , Viremia , Humans , Male , Middle Aged , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , Female , Lymphocyte Activation/drug effects , Viremia/immunology , Viremia/drug therapy , Cellular Senescence/drug effects , Aged , Adult , T-Lymphocytes/immunology
9.
J Med Virol ; 96(8): e29853, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39135474

ABSTRACT

This study aimed to retrospectively evaluate the baseline and follow-up viral loads and viral clearance times in cases followed for asymptomatic and symptomatic congenital cytomegalovirus (cCMV) infection between August 2010 and August 2022. Among 93 cases, they had asymptomatic (n: 55) and symptomatic (n: 38). The median baseline blood viral load detected in the symptomatic cCMV (ScCMV) infection (13 054 IU/mL) was significantly higher than that of asymptomatic cCMV (AcCMV) infection (4636 IU/mL) (p < 0.013). There was no difference in median viral clearance times (75 and 90 days, respectively) in baseline viremic cases in the ScCMV and AcCMV infection groups. There were no differences in median baseline blood viral load (6930 IU/mL and 14 268 IU/mL, respectively) and median viral clearance times (75 and 85 days, respectively) between the 6-week and 6-month antiviral treatment group. No correlation was found between baseline blood viral load, clinical severity, and the number of systems involved. However, in initial viremic cases, the viral load threshold for a symptomatic case was 8856 IU/mL, with 85.7% sensitivity and 54.5% specificity.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Viral Load , Humans , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/diagnosis , Retrospective Studies , Female , Male , Infant, Newborn , Cytomegalovirus/isolation & purification , Cytomegalovirus/genetics , Asymptomatic Infections , Infant , Antiviral Agents/therapeutic use , Viremia/virology
10.
J Infect Dev Ctries ; 18(7): 1108-1117, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39078797

ABSTRACT

INTRODUCTION: Human pegivirus-1 (HPgV-1) influences the pathogenesis and outcome of viral infections. We investigated the prevalence and impact of HPgV-1 due to the paucity of studies on Indian people living with HIV (PLHIV). METHODOLOGY: Samples were collected from 347 treatment-naïve PLHIV; and 100 blood donors negative for HIV, HBV, and HCV. CD4+ T-cell and HIV-1 viral load were measured using flow-cytometry and quantitative polymerase chain reaction (qPCR), respectively. HPgV-1 was quantified and genotyped by qPCR and Sanger sequencing, respectively. RESULTS: HPgV-1 viremia in PLHIV and controls was 11% (38/347) and 1% (1/100), respectively. We found HPgV-1 genotype-2a in PLHIV and genotype-2b in controls. Male preponderance was seen in HIV-1 mono-infection and co-infection groups (166 vs. 143 and 33 vs. 5; p < 0.0001). The peak prevalence of HPgV-1 was at 31-50 years (p = 0.02). CD4+ T-cell count (245.5 vs. 240; p = 0.59) and HIV-1 log viral load (4.7 vs. 4.9; p = 0.50) were not significantly different between the HIV-1 mono-infected and coinfected individuals. However, a direct correlation existed between HpgV-1 viral load and CD4+ T-cell count (r = 0.27, p = 0.05) and an inverse correlation with HIV-1 viral load (r = -0.21, p = 0.10). CONCLUSIONS: This is the first study in India to estimate the HPgV-1 prevalence in PLHIV with the predominance of genotype-2a. HPgV-1 viremia had a moderate impact on CD4+ T-cells and HIV-1 viral load, which requires a longitudinal study to identify the beneficial influence on HIV-1 disease progression and outcome.


Subject(s)
Disease Progression , Flaviviridae Infections , HIV Infections , HIV-1 , Viral Load , Humans , India/epidemiology , Male , HIV Infections/virology , HIV Infections/epidemiology , HIV Infections/complications , Adult , Female , Flaviviridae Infections/epidemiology , Flaviviridae Infections/virology , Prevalence , Middle Aged , HIV-1/genetics , HIV-1/isolation & purification , Young Adult , Coinfection/virology , Coinfection/epidemiology , Genotype , CD4 Lymphocyte Count , Pegivirus/genetics , Viremia/epidemiology
11.
Viruses ; 16(7)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39066265

ABSTRACT

Although a combination of immunoprophylaxis and antiviral therapy can effectively prevent mother-to-child transmission (MTCT) of hepatitis B virus (HBV), a considerable number of infants born to highly viremic mothers still develop occult HBV infection (OBI). To uncover the virological factor and risk predictor for OBI in infants, we found that the diversity and complexity of maternal HBV quasispecies in the case group were lower than those in the control group. Mutations with significant differences between the two groups were most enriched in the NTCPbd and PreC regions. Genetic distance at the amino-acid level of the PreC region, especially the combination of three amino-acid mutations in the PreC region, could strongly predict the risk of OBI in infants. HBV quasispecies in OBI infants were highly complex, and the non-synonymous substitutions were mainly found in the RT and HBsAg regions. The sK47E (rtQ55R) and sP49L mutations in OBI infants might contribute to OBI through inhibiting the production of HBV DNA and HBsAg, respectively. This study found the potential virological factors and risk predictors for OBI in infants born to highly viremic mothers, which might be helpful for controlling OBI in infants.


Subject(s)
DNA, Viral , Hepatitis B virus , Hepatitis B , Infectious Disease Transmission, Vertical , Mutation , Quasispecies , Viremia , Humans , Hepatitis B virus/genetics , Hepatitis B virus/classification , Hepatitis B virus/isolation & purification , Female , Quasispecies/genetics , Hepatitis B/virology , Hepatitis B/transmission , DNA, Viral/genetics , Infant , Pregnancy , Adult , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/blood , Infant, Newborn , Pregnancy Complications, Infectious/virology , Male , Mothers , Genotype
12.
Vet Res ; 55(1): 87, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982477

ABSTRACT

Hepatitis E virus (HEV) is a major cause of viral hepatitis worldwide. Pigs are the natural host of HEV genotype 3 and the main reservoir of HEV. As the host range of HEV genotype 3 expands, the possibility that HEV from various species can be transmitted to humans via pigs is increasing. We investigated the potential cross-species transmission of HEV by infecting minipigs with swine HEV (swHEV), rabbit HEV (rbHEV), and human HEV (huHEV) and examining their histopathological characteristics and distribution in various organs. Fifteen specific-pathogen-free Yucatan minipigs were infected with swHEV, rbHEV, huHEV, or a mock control. In the present study, we analysed faecal shedding, viremia, and serological parameters over a seven-week period. Our results indicated that swHEV exhibited more robust shedding and viremia than non-swHEVs. Only swHEV affected the serological parameters, suggesting strain-specific differences. Histopathological examination revealed distinct patterns in the liver, pancreas, intestine, and lymphoid tissues after infection with each HEV strain. Notably, all three HEVs induced histopathological changes in the pancreas, supporting the association of HEVs with acute pancreatitis. Our results also identified skeletal muscle as a site of HEV antigen presence, suggesting a potential link to myositis. In conclusion, this study provides valuable insights into the infection dynamics of different HEV strains in minipigs, emphasizing the strain-specific variations in virological, serological, and histological parameters. The observed differences in infection kinetics and tissue tropism will contribute to our understanding of HEV pathogenesis and the potential for cross-species transmission.


Subject(s)
Hepatitis E virus , Hepatitis E , Swine Diseases , Swine, Miniature , Animals , Swine , Hepatitis E/veterinary , Hepatitis E/virology , Hepatitis E/transmission , Hepatitis E virus/physiology , Swine Diseases/virology , Swine Diseases/transmission , Swine Diseases/pathology , Specific Pathogen-Free Organisms , Rabbits , Virus Shedding , Humans , Feces/virology , Female , Viremia/veterinary , Viremia/virology
13.
J Gen Virol ; 105(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38995674

ABSTRACT

Mayaro virus (MAYV), a mosquito-borne alphavirus, is considered an emerging threat to public health with epidemic potential. Phylogenetic studies show the existence of three MAYV genotypes. In this study, we provide a preliminary analysis of the pathogenesis of all three MAYV genotypes in cynomolgus macaques (Macaca facicularis, Mauritian origin). Significant MAYV-specific RNAemia and viremia were detected during acute infection in animals challenged intravenously with the three MAYV genotypes, and strong neutralizing antibody responses were observed. MAYV RNA was detected at high levels in lymphoid tissues, joint muscle and synovia over 1 month after infection, suggesting that this model could serve as a promising tool in studying MAYV-induced chronic arthralgia, which can persist for years. Significant leucopenia was observed across all MAYV genotypes, peaking with RNAemia. Notable differences in the severity of acute RNAemia and composition of cytokine responses were observed among the three MAYV genotypes. Our model showed no outward signs of clinical disease, but several major endpoints for future MAYV pathology and intervention studies are described. Disruptions to normal blood cell counts and cytokine responses were markedly distinct from those observed in macaque models of CHIKV infection, underlining the importance of developing non-human primate models specific to MAYV infection.


Subject(s)
Alphavirus Infections , Alphavirus , Genotype , Macaca fascicularis , RNA, Viral , Viremia , Animals , Macaca fascicularis/virology , Alphavirus/genetics , Alphavirus/pathogenicity , Alphavirus/classification , Alphavirus/isolation & purification , Alphavirus Infections/virology , Alphavirus Infections/veterinary , Viremia/virology , RNA, Viral/genetics , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Disease Models, Animal , Phylogeny , Cytokines/genetics , Cytokines/blood
14.
J Med Virol ; 96(7): e29806, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39007420

ABSTRACT

Optimization of individual immunosuppression, which reduces the risks of both graft loss and patients' death, is considered the best approach to improve long-term outcomes of renal transplantation. Torque Teno Virus (TTV) DNAemia has emerged as a potential biomarker reflecting the depth of therapeutic immunosuppression during the initial year post-transplantation. However, its efficacy in long-term monitoring remains uncertain. In a cohort study involving 34 stable kidney transplant recipients and 124 healthy volunteers, we established lower and upper TTV DNAemia thresholds (3.75-5.1 log10 cp/mL) correlating with T-cell activatability, antibody response against flu vaccine, and risk for subsequent serious infections or cancer over 50 months. Validation in an independent cohort of 92 recipients confirmed that maintaining TTV DNAemia within this range in >50% of follow-up time points was associated with reduced risks of complications due to inadequate immunosuppression, including de novo DSA, biopsy-proven antibody-mediated rejection, graft loss, infections, or cancer. Multivariate analysis highlighted "in-target" TTV DNAemia as the sole independent variable significantly linked to decreased risk for long-term complications due to inadequate immunosuppression (odds ratio [OR]: 0.27 [0.09-0.77]; p = 0.019). Our data suggest that the longitudinal monitoring of TTV DNAemia in kidney transplant recipients could help preventing the long-term complications due to inadequate immunosuppression.


Subject(s)
DNA Virus Infections , DNA, Viral , Immunosuppression Therapy , Kidney Transplantation , Torque teno virus , Transplant Recipients , Humans , Torque teno virus/genetics , Kidney Transplantation/adverse effects , Male , Female , Middle Aged , DNA, Viral/blood , Adult , DNA Virus Infections/virology , DNA Virus Infections/blood , DNA Virus Infections/immunology , Immunosuppression Therapy/adverse effects , Longitudinal Studies , Aged , Graft Rejection , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Cohort Studies , Viremia
15.
J Med Virol ; 96(7): e29814, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39015038

ABSTRACT

Novel biomarkers reflecting the degree of immunosuppression in transplant patients are required to ensure eventual personalized equilibrium between rejection and infection risks. With the above aim, Torque Teno Virus (TTV) viremia was precisely examined in a large cohort of transplanted immunocompromised patients (192 hematological and 60 solid organ transplant recipients) being monitored for Cytomegalovirus reactivation. TTV load was measured in 2612 plasma samples from 448 patients. The results revealed a significant increase in TTV viral load approximately 14 days following CMV reactivation/infection in solid organ transplant (SOT) patients. No recognizable difference in TTV load was noted among hematological patients during the entire timeframe analyzed. Furthermore, a temporal gap of approximately 30 days was noted between the viral load peaks reached by the two viruses, with Cytomegalovirus (CMV) preceding TTV. It was not possible to establish a correlation between CMV reactivation/infection and TTV viremia in hematological patients. On the other hand, the SOT patient cohort allowed us to analyze viral kinetics and draw intriguing conclusions. Taken together, the data suggest, to our knowledge for the first time, that CMV infection itself could potentially cause an increase in TTV load in the peripheral blood of patients undergoing immunosuppressive therapy.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , DNA Virus Infections , Immunocompromised Host , Torque teno virus , Viral Load , Viremia , Humans , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/blood , Male , DNA Virus Infections/virology , DNA Virus Infections/blood , DNA Virus Infections/immunology , Middle Aged , Female , Adult , Immunosuppression Therapy/adverse effects , Virus Activation , Transplant Recipients/statistics & numerical data , Aged , Cohort Studies
16.
BMC Pulm Med ; 24(1): 366, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39080682

ABSTRACT

BACKGROUND: Severe COVID-19 carries a high morbidity and mortality. Previous studies have shown an association between COVID-19 severity and SARS-CoV-2 viral load (VL). We sought to measure VL in multiple compartments (urine, plasma, lower respiratory tract) in patients admitted to the intensive care unit (ICU) with severe COVID-19 pneumonia and correlate with clinical outcomes. METHODS: Plasma, urine, and endotracheal aspirate (ETA) samples were obtained on days 1, 3, 7, 14, and 21 from subjects admitted to the ICU with severe COVID-19. VL was measured via reverse transcriptase polymerase chain reaction. Clinical data was collected from the electronic health record. Grouped comparisons were performed using Student's t-test or 1-way ANOVA. Linear regression was used to correlate VL from different compartments collected at the same time. Logistic regression was performed to model ventilator-freedom at 28 days as a function of peak plasma VL. RESULTS: We enrolled 57 subjects with severe COVID-19 and measured VL in plasma (n = 57), urine (n = 25), and ETA (n = 34). Ventilator-associated pneumonia developed in 63% of subjects. 49% of subjects were viremic on study day 1. VL in plasma and ETA both significantly decreased by day 14 (P < 0.05), and the two were weakly correlated on study day 1 (P = 0.0037, r2 = 0.2343) and on all study days (P < 0.001, r2 = 0.2211). VL were not detected in urine. While no associations were observed with peak ETA VL, subjects with higher peak plasma VL experienced a greater number of respiratory complications, including ventilator-associated pneumonia and fewer ventilator-free and hospital-free days. There was no association between VL in either plasma or ETA and mortality. In viremic patients, plasma VL was significantly lower in subjects that were ICU-free and ventilator-free (P < 0.05), with trends noted for hospital-freedom, ventilator-associated pneumonia, and survival to discharge (P < 0.1). By logistic regression, plasma VL was inversely associated with ventilator-freedom at 28 days (odds ratio 0.14, 95% confidence interval 0.02-0.50). CONCLUSIONS: Elevated SARS-CoV-2 VL in the plasma but not in the lower respiratory tract is a novel biomarker in severe COVID-19 for respiratory complications.


Subject(s)
COVID-19 , Intensive Care Units , SARS-CoV-2 , Viral Load , Viremia , Humans , COVID-19/complications , Male , Female , Middle Aged , Aged , Severity of Illness Index , Pneumonia, Ventilator-Associated/epidemiology , Pneumonia, Ventilator-Associated/virology , Adult
17.
Hum Vaccin Immunother ; 20(1): 2318814, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38961639

ABSTRACT

The present study aimed at investigating whether the hydroxychloroquine (HCQ) treatment would impact the neutralizing antibody production, viremia levels and the kinetics of serum soluble mediators upon planned 17DD-Yellow Fever (YF) primovaccination (Bio-Manguinhos-FIOCRUZ) of primary Sjögren's syndrome (pSS). A total of 34 pSS patients and 23 healthy controls (HC) were enrolled. The pSS group was further categorized according to the use of HCQ (HCQ and Non-HCQ). The YF-plaque reduction neutralization test (PRNT ≥1:50), YF viremia (RNAnemia) and serum biomarkers analyses were performed at baseline and subsequent time-points (Day0/Day3-4/Day5-6/Day7/Day14-D28). The pSS group showed PRNT titers and seropositivity rates similar to those observed for HC (GeoMean = 238 vs 440, p = .11; 82% vs 96%, p = .13). However, the HCQ subgroup exhibited lower seroconversion rates as compared to HC (GeoMean = 161 vs 440, p = .04; 69% vs 96%, p = .02) and Non-HQC (GeoMean = 161 vs 337, p = .582; 69% vs 94%, p = .049). No differences in YF viremia were observed amongst subgroups. Serum biomarkers analyses demonstrated that HCQ subgroup exhibited increased levels of CCL2, CXL10, IL-6, IFN-γ, IL1-Ra, IL-9, IL-10, and IL-2 at baseline and displayed a consistent increase of several biomarkers along the kinetics timeline up to D14-28. These results indicated that HCQ subgroup exhibited a deficiency in assembling YF-specific immune response elicited by 17DD-YF primovaccination as compared to Non-HCQ subgroup. Our findings suggested that hydroxychloroquine is associated with a decrease in the humoral immune response after 17DD-YF primovaccination.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Hydroxychloroquine , Seroconversion , Sjogren's Syndrome , Yellow Fever , Humans , Hydroxychloroquine/therapeutic use , Sjogren's Syndrome/drug therapy , Sjogren's Syndrome/immunology , Female , Middle Aged , Male , Adult , Yellow Fever/immunology , Yellow Fever/prevention & control , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Yellow Fever Vaccine/immunology , Aged , Viremia/drug therapy , Viremia/immunology , Yellow fever virus/immunology , Cytokines/blood , Biomarkers/blood
18.
Vet Res ; 55(1): 89, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010163

ABSTRACT

Since the reintroduction of African swine fever virus (ASFV) in Europe in 2007 and its subsequent spread to Asia, wild boar has played a crucial role in maintaining and disseminating the virus. There are significant gaps in the knowledge regarding infection dynamics and disease pathogenesis in domestic pigs and wild boar, particularly at the early infection stage. We aimed to compare domestic pigs and wild boar infected intranasally to mimic natural infection with one of the original highly virulent genotype II ASFV isolates (Armenia 2007). The study involved euthanising three domestic pigs and three wild boar on days 1, 2, 3, and 5 post-infection, while four domestic pigs and four wild boar were monitored until they reached a humane endpoint. The parameters assessed included clinical signs, macroscopic lesions, viremia levels, tissue viral load, and virus shedding in nasal and rectal swabs from day 1 post-infection. Compared with domestic pigs, wild boar were more susceptible to ASFV, with a shorter incubation period and earlier onset of clinical signs. While wild boar reached a humane endpoint earlier than domestic pigs did, the macroscopic lesions were comparatively less severe. In addition, wild boar had earlier viremia, and the virus was also detected earlier in tissues. The medial retropharyngeal lymph nodes were identified as key portals for ASFV infection in both subspecies. No viral genome was detected in nasal or rectal swabs until shortly before reaching the humane endpoint in both domestic pigs and wild boar, suggesting limited virus shedding in acute infections.


Subject(s)
African Swine Fever Virus , African Swine Fever , Genotype , Sus scrofa , Animals , African Swine Fever Virus/genetics , African Swine Fever Virus/physiology , African Swine Fever/virology , Swine , Virus Shedding , Viremia/veterinary , Viremia/virology , Viral Load/veterinary , Virulence
19.
Int J Med Sci ; 21(9): 1661-1671, 2024.
Article in English | MEDLINE | ID: mdl-39006848

ABSTRACT

Background and aim: Patients with chronic hepatitis B patients (CHB) with low-level viremia (LLV) are not necessarily at low risk of developing hepatocellular carcinoma (HCC). The question of whether CHB patients with LLV require immediate antiviral agent (AVT) or long-term AVT remains controversial. The study aims to investigate the risk of HCC development and the risk factors in CHB patients with LLV and construct a nomogram model predicting the risk of HCC. Methods: We conducted a retrospective cohort study that enrolled 16,895 CHB patients from January 2008 to December 2020. The patients were divided into three groups for comparison: the LLV group, maintained virological response (MVR) group and HBV-DNA>2000 group. The cumulative incidence of progression to HCC was assessed. Cox regression analysis was performed to determine the final risk factors, and a nomogram model was constructed. The 10-fold Cross-Validation method was utilized for internal validation. Results: A total of 408 new cases of HCC occurred during the average follow-up period of 5.78 years. The 3, 5, and 10-year cumulative HCC risks in the LLV group were 3.56%, 4.96%, and 9.51%, respectively. There was a significant difference in the cumulative risk of HCC between the HBV-DNA level > 2000 IU/mL and LLV groups (p = 0.049). Independent risk factors for HCC development in LLV group included male gender, age, presence of cirrhosis, and platelets count. The Area Under the Curve (AUC) values for the 3-year and 5-year prediction from our HCC risk prediction model were 0.75 and 0.76, respectively. Conclusion: Patients with LLV and MVR are still at risk for developing HCC. The nomogram established for CHB patient with LLV, incorporating identified significant risk factors, serves as an effective tool for predicting HCC-free outcomes. This nomogram model provides valuable information for determining appropriate surveillance strategies and prescribing AVT.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B virus , Hepatitis B, Chronic , Liver Neoplasms , Nomograms , Viremia , Humans , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/virology , Liver Neoplasms/epidemiology , Liver Neoplasms/etiology , Male , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/virology , Female , Middle Aged , Retrospective Studies , Risk Factors , Viremia/complications , Adult , Hepatitis B virus/isolation & purification , Antiviral Agents/therapeutic use , Incidence , DNA, Viral/blood
20.
Lancet Glob Health ; 12(8): e1312-e1322, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39030062

ABSTRACT

BACKGROUND: Children and adolescents with HIV taking antiretroviral therapy (ART) have high rates of viraemia. We assessed if genotypic resistance testing (GRT) to inform onward treatment improved treatment outcomes in Lesotho and Tanzania, two countries with little access to GRT. METHODS: The Genotype-Informed Versus Empirical Management of Viremia (GIVE MOVE) open-label, parallel-group randomised controlled trial enrolled children and adolescents with HIV between the ages of 6 months and 19 years, taking ART, and with a viral load at least 400 copies per mL. Participants were recruited from ten clinical centres and hospitals in Lesotho and Tanzania. Participants were electronically randomly allocated 1:1 to receive either GRT with expert recommendation (GRT group) or repeat viral-load testing and empirical onward treatment (usual care group). Participants and study staff were not masked, but the endpoint committee and laboratory staff conducting viral-load testing were. Participants in both groups received at least three sessions of enhanced adherence counselling, and in the GRT group, blood for GRT assessed via Sanger sequencing was drawn at enrolment. The composite primary endpoint was death, hospitalisation, a new WHO HIV clinical stage 4 event, or not having documented viral suppression of less than 50 copies per mL at 36 weeks in the modified intention-to-treat population, which excluded participants who were retrospectively found to be ineligible after randomisation. Serious adverse events were analysed in the modified intention-to-treat population. The trial was registered with ClinicalTrials.gov (NCT04233242) and the trial status is completed. FINDINGS: Between March 3, 2020, and July 5, 2022, 286 participants were enrolled and 284 were included in the modified intention-to-treat analysis (144 in the GRT group and 140 in the usual care group). Of these participants, 158 (56%) were female and 126 (44%) were male. Five (3%) in the GRT group and four (3%) in the usual care group did not complete follow-up but were included in the primary analysis. The median age across both groups was 14 years (IQR 9-16). The composite primary endpoint occurred in 67 (47%) participants in the GRT group and 73 (52%) in the usual care group (adjusted odds ratio 0·79 [95% CI 0·49 to 1·27]; adjusted risk difference -0·06 [95% CI -0·17 to 0·06]; p=0·34); all participants reaching the composite primary endpoint had no documented viral suppression at 36 weeks. No deaths were recorded, and only one clinical stage 4 event requiring hospitalisation occurred (in the usual care group); this was the only serious adverse event recorded in the study. INTERPRETATION: GRT-informed management did not significantly improve treatment outcomes for children and adolescents with viraemia while taking ART. FUNDING: Fondation Botnar, Swiss National Science Foundation, and Gottfried and Julia Bangerter-Rhyner Foundation. TRANSLATIONS: For the Sesotho and Swahili translations of the abstract see Supplementary Materials section.


Subject(s)
HIV Infections , Viremia , Humans , HIV Infections/drug therapy , Male , Female , Tanzania/epidemiology , Adolescent , Lesotho , Child , Viremia/drug therapy , Child, Preschool , Viral Load , Infant , Drug Resistance, Viral , Anti-HIV Agents/therapeutic use , Young Adult , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL