Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 252
Filter
1.
Vet Res Commun ; 46(4): 1319-1324, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35854050

ABSTRACT

Generalized and fatal felid alphaherpesvirus-1 (FeHV-1) natural infection with liver involvement is rarely reported in cats, and the occurrence of herpesvirus viraemia with internal organ histologic lesions in adult cats is unknown. A 1.5-year-old cat, female, mixed breed, positive for feline leukaemia virus (FeLV) presented in a veterinary teaching hospital with sneezing, nasal discharge, anorexia, and diarrhoea after two weeks, evolving to inspiratory dyspnoea. Complete blood count and serum biochemistry analysis showed marked leukopenia and thrombocytopenia. After clinical worsening and lack of treatment response, the cat was euthanized. Pathological findings included hepatic necrosis, fibrinonecrotic tracheitis, and bronchointerstitial pneumonia. Marked amounts of coccobacillary bacteria were observed covering the necrotic tracheal and bronchial mucosa, at the cytoplasm of alveolar macrophages, and free in alveoli lumen, mimicking a primary bacterial tracheitis and pneumonia. Both lung and tracheal bacteria exhibited marked immunolabeling in anti-Escherichia coli immunohistochemistry. In addition, rare epithelial cells of bronchi contained round, eosinophilic, intranuclear viral inclusion bodies (4-7 µm) that marginate the chromatin, characteristic of FeHV-1 infection. Strong multifocal anti-FeHV-1 immunolabeling was observed in necrotic epithelial cells of the liver, trachea, and lungs. Generalized herpesvirus infection with the occurrence of acute hepatic necrosis and severe respiratory illness is a potential differential diagnosis in FeLV-positive cats with respiratory signs. The immunodepression in these cats probably favours a FeHV-1 viraemia in addition to the development of opportunistic bacterial infections, such as Escherichia coli, and it is associated with a poor outcome.


Subject(s)
Cat Diseases , Tracheitis , Cats , Female , Animals , Leukemia Virus, Feline , Tracheitis/pathology , Tracheitis/veterinary , Viremia/veterinary , Viremia/pathology , Hospitals, Animal , Hospitals, Teaching , Necrosis/pathology , Necrosis/veterinary , Liver/pathology
3.
Front Immunol ; 12: 769901, 2021.
Article in English | MEDLINE | ID: mdl-34880866

ABSTRACT

The zebrafish is extensively used as an animal model for human and fish diseases. However, our understanding of the structural organization of its immune system remains incomplete, especially the mucosa-associated lymphoid tissues (MALTs). Teleost MALTs are commonly perceived as diffuse and scattered populations of immune cells throughout the mucosa. Yet, structured MALTs have been recently discovered in Atlantic salmon (Salmo salar L.), including the interbranchial lymphoid tissue (ILT) in the gills. The existence of the ILT was only recently identified in zebrafish and other fish species, highlighting the need for in-depth characterizations of the gill-associated lymphoid tissue (GIALT) in teleosts. Here, using 3-D high-resolution microscopy, we analyze the GIALT of adult zebrafish with an immuno-histology approach that reveals the organization of lymphoid tissues via the labeling of T/NK cells with an antibody directed to a highly conserved epitope on the kinase ZAP70. We show that the GIALT in zebrafish is distributed over at least five distinct sub-regions, an organization found in all pairs of gill arches. The GIALT is diffuse in the pharyngeal part of the gill arch, the interbranchial septum and the filaments/lamellae, and structured in two sub-regions: the ILT, and a newly discovered lymphoid structure located along each side of the gill arch, which we named the Amphibranchial Lymphoid Tissue (ALT). Based on RAG2 expression, neither the ILT nor the ALT constitute additional thymi. The ALT shares several features with the ILT such as presence of abundant lymphoid cells and myeloid cells embedded in a network of reticulated epithelial cells. Further, the ILT and the ALT are also a site for T/NK cell proliferation. Both ILT and ALT show structural changes after infection with Spring Viraemia of Carp Virus (SVCV). Together, these data suggest that ALT and ILT play an active role in immune responses. Comparative studies show that whereas the ILT seems absent in most neoteleosts ("Percomorphs"), the ALT is widely present in cyprinids, salmonids and neoteleosts, suggesting that it constitutes a conserved tissue involved in the protection of teleosts via the gills.


Subject(s)
Fish Diseases/pathology , Gills/immunology , Imaging, Three-Dimensional/methods , Lymphoid Tissue/diagnostic imaging , Zebrafish/immunology , Animals , Gills/anatomy & histology , Gills/diagnostic imaging , Lymphoid Tissue/cytology , Viremia/pathology , Zebrafish/anatomy & histology
4.
PLoS Pathog ; 17(11): e1010034, 2021 11.
Article in English | MEDLINE | ID: mdl-34762717

ABSTRACT

Siglec-9 is an MHC-independent inhibitory receptor expressed on a subset of natural killer (NK) cells. Siglec-9 restrains NK cytotoxicity by binding to sialoglycans (sialic acid-containing glycans) on target cells. Despite the importance of Siglec-9 interactions in tumor immune evasion, their role as an immune evasion mechanism during HIV infection has not been investigated. Using in vivo phenotypic analyses, we found that Siglec-9+ CD56dim NK cells, during HIV infection, exhibit an activated phenotype with higher expression of activating receptors and markers (NKp30, CD38, CD16, DNAM-1, perforin) and lower expression of the inhibitory receptor NKG2A, compared to Siglec-9- CD56dim NK cells. We also found that levels of Siglec-9+ CD56dim NK cells inversely correlate with viral load during viremic infection and CD4+ T cell-associated HIV DNA during suppressed infection. Using in vitro cytotoxicity assays, we confirmed that Siglec-9+ NK cells exhibit higher cytotoxicity towards HIV-infected cells compared to Siglec-9- NK cells. These data are consistent with the notion that Siglec-9+ NK cells are highly cytotoxic against HIV-infected cells. However, blocking Siglec-9 enhanced NK cells' ability to lyse HIV-infected cells, consistent with the known inhibitory function of the Siglec-9 molecule. Together, these data support a model in which the Siglec-9+ CD56dim NK subpopulation is highly cytotoxic against HIV-infected cells even whilst being restrained by the inhibitory effects of Siglec-9. To harness the cytotoxic capacity of the Siglec-9+ NK subpopulation, which is dampened by Siglec-9, we developed a proof-of-concept approach to selectively disrupt Siglec/sialoglycan interactions between NK and HIV-infected cells. We achieved this goal by conjugating Sialidase to several HIV broadly neutralizing antibodies. These conjugates selectively desialylated HIV-infected cells and enhanced NK cells' capacity to kill them. In summary, we identified a novel, glycan-based interaction that may contribute to HIV-infected cells' ability to evade NK immunosurveillance and developed an approach to break this interaction.


Subject(s)
Antigens, CD/metabolism , CD56 Antigen/immunology , HIV Infections/pathology , HIV/physiology , Killer Cells, Natural/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Viral Load , Viremia/pathology , Antigens, CD/genetics , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/virology , Humans , Killer Cells, Natural/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Viremia/immunology , Viremia/metabolism , Viremia/virology
5.
EMBO J ; 40(22): e108966, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34618370

ABSTRACT

Viremia in the vertebrate host is a major determinant of arboviral reservoir competency, transmission efficiency, and disease severity. However, immune mechanisms that control arboviral viremia are poorly defined. Here, we identify critical roles for the scavenger receptor MARCO in controlling viremia during arthritogenic alphavirus infections in mice. Following subcutaneous inoculation, arthritogenic alphavirus particles drain via the lymph and are rapidly captured by MARCO+ lymphatic endothelial cells (LECs) in the draining lymph node (dLN), limiting viral spread to the bloodstream. Upon reaching the bloodstream, alphavirus particles are cleared from the circulation by MARCO-expressing Kupffer cells in the liver, limiting viremia and further viral dissemination. MARCO-mediated accumulation of alphavirus particles in the draining lymph node and liver is an important host defense mechanism as viremia and viral tissue burdens are elevated in MARCO-/- mice and disease is more severe. In contrast to prior studies implicating a key role for lymph node macrophages in limiting viral dissemination, these findings exemplify a previously unrecognized arbovirus-scavenging role for lymphatic endothelial cells and improve our mechanistic understanding of viremia control during arthritogenic alphavirus infection.


Subject(s)
Alphavirus Infections/virology , Lymph Nodes/cytology , Receptors, Immunologic/metabolism , Viremia/pathology , Alphavirus/pathogenicity , Animals , Chikungunya Fever/genetics , Chikungunya Fever/virology , Endothelial Cells/virology , Host-Pathogen Interactions , Kupffer Cells/virology , Lymph Nodes/virology , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , RNA, Viral/metabolism , Receptors, Immunologic/genetics , Single-Cell Analysis , Viremia/virology
6.
JCI Insight ; 6(23)2021 12 08.
Article in English | MEDLINE | ID: mdl-34699383

ABSTRACT

Understanding viral rebound in pediatric HIV-1 infection may inform the development of alternatives to lifelong antiretroviral therapy (ART) to achieve viral remission. We thus investigated viral rebound after analytical treatment interruption (ATI) in 10 infant macaques orally infected with SHIV.C.CH505 and treated with long-term ART. Rebound viremia was detected within 7 to 35 days of ATI in 9 of 10 animals, with posttreatment control of viremia seen in 5 of 5 Mamu-A*01+ macaques. Single-genome sequencing revealed that initial rebound virus was similar to viral DNA present in CD4+ T cells from blood, rectum, and lymph nodes before ATI. We assessed the earliest sites of viral reactivation immediately following ATI using ImmunoPET imaging. The largest increase in signal that preceded detectable viral RNA in plasma was found in the gastrointestinal (GI) tract, a site with relatively high SHIV RNA/DNA ratios in CD4+ T cells before ATI. Thus, the GI tract may be an initial source of rebound virus, but as ATI progresses, viral reactivation in other tissues likely contributes to the composition of plasma virus. Our study provides potentially novel insight into the features of viral rebound in pediatric infection and highlights the application of a noninvasive technique to monitor areas of HIV-1 expression in children.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Simian Acquired Immunodeficiency Syndrome/virology , Viremia/etiology , Animals , Female , Macaca , Male , Viremia/pathology
7.
Bull Exp Biol Med ; 171(4): 513-516, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34542764

ABSTRACT

Various biological models are used to isolate West Nile virus, but their role as a selection factor that facilitates selection of isolates with certain properties is usually not evaluated. We compared pathogenic properties of three strains of the West Nile virus obtained from one sample of virus-containing material using different models: WNV Volgograd 900m/18 (on the model of suckling mice), WNV Volgograd 900a/18 (on C6/36 cells) and WNV Volgograd 900v/18 (on Vero cells). WNV Volgograd 900m/18 strain demonstrated virulent (LD50 5×103±0.005×104 PFU, p≤0.05) and neuroinvasive properties, induced viremia and pathomorphological changes in the liver, lymph nodes, and brain of nonlinear white mice. WNV Volgograd 900v/18 strain had similar characteristics except for neuroinvasiveness. WNV Volgograd 900a/18 variant demonstrated minimum virulence (LD50 5×104±0.005×104 PFU, p≤0.05), did not cause neurological symptoms, and was not isolated from the blood of infected animals.


Subject(s)
Host-Pathogen Interactions/physiology , Models, Biological , West Nile virus/pathogenicity , Animals , Animals, Suckling , Cells, Cultured , Chlorocebus aethiops , Culicidae , Disease Models, Animal , Male , Mice , Vero Cells , Viremia/pathology , West Nile Fever/pathology , West Nile Fever/virology , West Nile virus/isolation & purification
8.
Front Immunol ; 12: 691879, 2021.
Article in English | MEDLINE | ID: mdl-34163488

ABSTRACT

Increasing human Adenovirus (HAdV) infections complicated with acute respiratory distress syndrome (ARDS) even fatal outcome were reported in immunocompetent adolescent and adult patients. Here, we characterized the cytokine/chemokine expression profiles of immunocompetent patients complicated with ARDS during HAdV infection and identified biomarkers for disease severity/progression. Forty-eight cytokines/chemokines in the plasma samples from 19 HAdV-infected immunocompetent adolescent and adult patients (ten complicated with ARDS) were measured and analyzed in combination with clinical indices. Immunocompetent patients with ARDS caused by severe acute respiratory disease coronavirus (SARS-CoV)-2, 2009 pandemic H1N1 (panH1N1) or bacteria were included for comparative analyses. Similar indices of disease course/progression were found in immunocompetent patients with ARDS caused by HAdV, SARS-CoV-2 or panH1N infections, whereas the HAdV-infected group showed a higher prevalence of viremia, as well as increased levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT) and creatine kinase (CK). Expression levels of 33 cytokines/chemokines were increased significantly in HAdV-infected patients with ARDS compared with that in healthy controls, and many of them were also significantly higher than those in SARS-CoV-2-infected and panH1N1-infected patients. Expression of interferon (IFN)-γ, interleukin (IL)-1ß, hepatocyte growth factor (HGF), monokine induced by IFN-γ (MIG), IL-6, macrophage-colony stimulating factor (M-CSF), IL-10, IL-1α and IL-2Ra was significantly higher in HAdV-infected patients with ARDS than that in those without ARDS, and negatively associated with the ratio of the partial pressure of oxygen in arterial blood/fraction of inspired oxygen (PaO2/FiO2). Analyses of the receiver operating characteristic curve (ROC) showed that expression of IL-10, M-CSF, MIG, HGF, IL-1ß, IFN-γ and IL-2Ra could predict the progression of HAdV infection, with the highest area under the curve (AUC) of 0.944 obtained for IL-10. Of note, the AUC value for the combination of IL-10, IFN-γ, and M-CSF reached 1. In conclusion, the "cytokine storm" occurred during HAdV infection in immunocompetent patients, and expression of IL-10, M-CSF, MIG, HGF, IL-1ß, IFN-γ and IL-2Ra was closely associated with disease severity and could predict disease progression.


Subject(s)
Adenovirus Infections, Human/blood , Cytokines/blood , Respiratory Distress Syndrome/blood , Adenovirus Infections, Human/complications , Adenovirus Infections, Human/pathology , Adenoviruses, Human , Adolescent , Adult , Bacteria , Bacterial Infections/blood , Bacterial Infections/complications , Bacterial Infections/pathology , Biomarkers/blood , COVID-19/blood , COVID-19/complications , COVID-19/pathology , Disease Progression , Female , Humans , Influenza A Virus, H1N1 Subtype , Influenza, Human/blood , Influenza, Human/complications , Influenza, Human/pathology , Male , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/pathology , SARS-CoV-2 , Severity of Illness Index , Viremia/blood , Viremia/complications , Viremia/pathology , Young Adult
9.
Front Immunol ; 12: 617042, 2021.
Article in English | MEDLINE | ID: mdl-33968019

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic, caused by the SARS-CoV-2 virus, is wreaking havoc around the world. Considering that extracellular vesicles (EVs) released from SARS-CoV-2 infected cells might play a role in a viremic phase contributing to disease progression and that standard methods for EV isolation have been reported to co-isolate viral particles, we would like to recommend the use of heightened laboratory safety measures during the isolation of EVs derived from SARS-CoV-2 infected tissue and blood from COVID-19 patients. Research needs to be conducted to better understand the role of EVs in SARS-CoV-2 infectivity, disease progression, and transmission. EV isolation procedures should include approaches for protection from SARS-CoV-2 contamination. We recommend the EV and virology scientific communities develop collaborative projects where relationships between endogenous EVs and potentially lethal enveloped viruses are addressed to better understand the risks and pathobiology involved.


Subject(s)
COVID-19/pathology , COVID-19/transmission , Containment of Biohazards/methods , Extracellular Vesicles/virology , Endocytosis/physiology , Humans , RNA, Viral/blood , RNA, Viral/genetics , SARS-CoV-2 , Viral Genome Packaging , Viremia/pathology
10.
J Med Virol ; 93(9): 5452-5457, 2021 09.
Article in English | MEDLINE | ID: mdl-33969515

ABSTRACT

Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA is generally detected in nasopharyngeal swabs, viral RNA can be found in other samples including blood. Recently, associations between SARS-CoV-2 RNAaemia and disease severity and mortality have been reported in adults, while no reports are available in pediatric patients with coronavirus disease 2019 (COVID-19). The aim of this study was to evaluate the mortality, severity, clinical, and laboratory findings of SARS-CoV-2 RNA detection in blood in 96 pediatric patients with confirmed COVID-19. Among all patients, 6 (6%) had SARS-CoV-2 RNAaemia. Out of the six patients with SARS-CoV-2 RNAaemia, four (67%) had a severe form of the disease, and two out of the 6 patients with SARS-CoV-2 RNAaemia passed away (33%). Our results show that the symptoms more commonly found in the cases of COVID-19 in the study (fever, cough, tachypnea, and vomiting), were found at a higher percentage in the patients with SARS-CoV-2 RNAaemia. Creatine phosphokinase and magnesium tests showed significant differences between the positive and negative SARS-CoV-2 RNAaemia groups. Among all laboratory tests, magnesium and creatine phosphokinase could better predict SARS-CoV-2 RNAemia with area under the curve  levels of 0.808 and 0.748, respectively. In conclusion, 67% of individuals with SARS-CoV-2 RNAaemia showed a severe COVID-19 and one-third of the patients with SARS-CoV-2 RNAaemia passed away. Our findings suggest that magnesium and creatine phosphokinase might be considered as markers to estimate the SARS-CoV-2 RNAaemia.


Subject(s)
COVID-19/pathology , Creatine Kinase/blood , Magnesium/blood , RNA, Viral/blood , SARS-CoV-2/pathogenicity , Viremia/pathology , Adolescent , Biomarkers/blood , COVID-19/diagnosis , COVID-19/mortality , COVID-19/virology , COVID-19 Nucleic Acid Testing , Child , Child, Preschool , Cough/diagnosis , Cough/mortality , Cough/pathology , Cough/virology , Female , Fever/diagnosis , Fever/mortality , Fever/pathology , Fever/virology , Hospitals , Humans , Infant , Infant, Newborn , Iran , Male , RNA, Viral/genetics , SARS-CoV-2/genetics , Severity of Illness Index , Survival Analysis , Tachypnea/diagnosis , Tachypnea/mortality , Tachypnea/pathology , Tachypnea/virology , Viremia/diagnosis , Viremia/mortality , Viremia/virology
11.
PLoS Negl Trop Dis ; 15(3): e0009300, 2021 03.
Article in English | MEDLINE | ID: mdl-33780452

ABSTRACT

Ebola virus has been responsible for two major epidemics over the last several years and there has been a strong effort to find potential treatments that can improve the disease outcome. Antiviral favipiravir was thus tested on non-human primates infected with Ebola virus. Half of the treated animals survived the Ebola virus challenge, whereas the infection was fully lethal for the untreated ones. Moreover, the treated animals that did not survive died later than the controls. We evaluated the hematological, virological, biochemical, and immunological parameters of the animals and performed proteomic analysis at various timepoints of the disease. The viral load strongly correlated with dysregulation of the biological functions involved in pathogenesis, notably the inflammatory response, hemostatic functions, and response to stress. Thus, the management of viral replication in Ebola virus disease is of crucial importance in preventing the immunopathogenic disorders and septic-like shock syndrome generally observed in Ebola virus-infected patients.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Cytokine Release Syndrome/prevention & control , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Pyrazines/pharmacology , Viral Load/drug effects , Animals , Cytokines/blood , Disease Models, Animal , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/veterinary , Macaca fascicularis , T-Lymphocytes/immunology , Viremia/blood , Viremia/pathology , Virus Replication/drug effects
12.
J Exp Med ; 218(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32966579

ABSTRACT

We report that combination bNAb immunotherapy initiated on day 3 post-infection (PI) maintained durable CD8+ T cell-mediated suppression of SHIVAD8 viremia and preinoculation levels of CD4+ T cells in 9 of 13 treated monkeys during nearly 6 yr of observation, as assessed by successive CD8+ T cell-depletion experiments. In an extension of that study, two treatment interventions (bNAbs alone or cART plus bNAbs) beginning on week 2 PI were conducted and conferred controller status to 7 of 12 monkeys that was also dependent on control mediated by CD8+ cells. However, the median time to suppression of plasma viremia following intervention on week 2 was markedly delayed (85 wk) compared with combination bNAb immunotherapy initiated on day 3 (39 wk). In both cases, the principal correlate of virus control was the induction of CD8+ T cellular immunity.


Subject(s)
HIV Infections/therapy , HIV-1/immunology , Immunotherapy , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/immunology , Viremia/therapy , Acute Disease , Animals , CD8-Positive T-Lymphocytes/immunology , Female , HIV Infections/immunology , HIV Infections/pathology , Immunity, Cellular , Macaca mulatta , Male , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Viremia/immunology , Viremia/pathology
13.
Sci Rep ; 10(1): 22286, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33339855

ABSTRACT

Human immunodeficiency virus (HIV) remains incurable due to latent viral reservoirs established in non-activated CD4 T cells that cannot be eliminated via antiretroviral therapy. Current efforts to cure HIV are focused on identifying drugs that will induce viral gene expression in latently infected cells, commonly known as latency reversing agents (LRAs). Some drugs have been shown to reactivate latent HIV but do not cause a reduction in reservoir size. Therefore, finding new LRAs or new combinations or increasing the round of stimulations is needed to cure HIV. However, the effects of these drugs on viral rebound after prolonged treatment have not been evaluated. In a previous clinical trial, antiretroviral therapy intensification with maraviroc for 48 weeks caused an increase in residual viremia and episomal two LTR-DNA circles suggesting that maraviroc could reactivate latent HIV. We amended the initial clinical trial to explore additional virologic parameters in stored samples and to evaluate the time to viral rebound during analytical treatment interruption in three patients. Maraviroc induced an increase in cell-associated HIV RNA during the administration of the drug. However, there was a rapid rebound of viremia after antiretroviral therapy discontinuation. HIV-specific T cell response was slightly enhanced. These results show that maraviroc can reactivate latent HIV in vivo but further studies are required to efficiently reduce the reservoir size.


Subject(s)
HIV Infections/drug therapy , HIV-1/drug effects , Maraviroc/administration & dosage , Viremia/drug therapy , Adult , Animals , Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/adverse effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Female , HIV Infections/blood , HIV Infections/pathology , HIV Infections/virology , HIV-1/pathogenicity , Humans , Male , Maraviroc/adverse effects , Middle Aged , Viral Load/drug effects , Viremia/blood , Viremia/pathology , Viremia/virology , Virus Activation/drug effects , Virus Latency/drug effects , Virus Replication/drug effects
14.
BMC Infect Dis ; 20(1): 758, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33059627

ABSTRACT

BACKGROUND: HIV infection exacerbates the prognosis of HCV infection, with a faster progression of hepatitis. Hepatic fibrosis is the major disruption of the hepatic tissue architecture characterized by anarchic deposition and excess of the extracellular matrix. The objective of this study was to evaluate hepatic fibrosis in HIV/HCV co-infected individuals as compared to HCV mono-infected. METHODS: A total of 97 participants (mean age 60.2 ± 14.3 years and 0.76 male/female sex ratio) was enrolled in a study conducted in Yaoundé, Cameroon from November 2018 to January 2019. Liver fibrosis was assessed by the APRI score (Aspartate Aminotransferase or AST/Platelet Ratio Index) which identifies the stage of fibrosis as classified by the Metavir system (F0 to F4). CD4 counts and plasmatic HIV viral load of HIV/HCV co-infected individuals were determined and the correlation between hepatic fibrosis and immuno-virological status established. Statistical analysis was done using Microsoft Excel 2016 and EpiInfo7 software. RESULTS: A high proportion (63.6%) of HIV/HCV co-infected participants had an abnormal AST level: 73.6 ± 45.8 IU/L as compared to 58.5 ± 39.3 IU/L (59.3%) among HCV mono-infected participants. The frequency of thrombocytopenia was 63.6% with a mean platelet count of 137 ± 50 ×  103 IU/L in HIV/HCV co-infected participants as compared to 176 ± 67 × 103 IU/L in HCV mono-infected participants (38.4%). The progression of hepatic fibrosis in participants with clinically significant fibrosis: F2, F3 and F4 was higher among HIV/HCV co-infected and the mean APRI score was 1.7 ± 1.4 versus 1 ± 0.8 among HCV mono-infected (26.7%). All participants (100%) with detectable HIV viral load had clinically significant fibrosis compared to 33.4% in those with undetectable HIV viral load (p = 0.55). Only 42.9% participants with CD4 >  500 cells/µL had clinically significant fibrosis (p = 0.72) while 100% participants with CD4 <  200 cells/µL had clinically significant fibrosis (p = 0.58). CONCLUSIONS: A high level of AST combined with thrombocytopenia (APRI score > 1.5) is an indicator of hepatic fibrosis in HIV/HCV co-infected individuals. Because of its non-invasive and less costly nature, the APRI score can be a suitable biomarker to monitor hepatic fibrosis in HIV/HCV co-infected individuals in resource constrained settings.


Subject(s)
Aspartate Aminotransferases/blood , Coinfection/virology , HIV Infections/blood , Hepatitis C/blood , Liver Cirrhosis/virology , Platelet Count , Aged , Biomarkers/blood , CD4 Lymphocyte Count , Cameroon , Cross-Sectional Studies , Female , Fibrosis , HIV Infections/virology , Hepatitis C/virology , Humans , Liver Cirrhosis/blood , Male , Middle Aged , Thrombocytopenia/virology , Viral Load , Viremia/complications , Viremia/pathology
15.
PLoS Pathog ; 16(9): e1008853, 2020 09.
Article in English | MEDLINE | ID: mdl-32886726

ABSTRACT

HIV-1 transmission is associated with a severe bottleneck in which a limited number of variants from a pool of genetically diverse quasispecies establishes infection. The IAVI protocol C cohort of discordant couples, female sex workers, other heterosexuals and men who have sex with men (MSM) present varying risks of HIV infection, diverse HIV-1 subtypes and represent a unique opportunity to characterize transmitted/founder viruses (TF) where disease outcome is known. To identify the TF, the HIV-1 repertoire of 38 MSM participants' samples was sequenced close to transmission (median 21 days post infection, IQR 18-41) and assessment of multivariant infection done. Patient derived gag genes were cloned into an NL4.3 provirus to generate chimeric viruses which were characterized for replicative capacity (RC). Finally, an evaluation of how the TF virus predicted disease progression and modified the immune response at both acute and chronic HIV-1 infection was done. There was higher prevalence of multivariant infection compared with previously described heterosexual cohorts. A link was identified between multivariant infection and replicative capacity conferred by gag, whereby TF gag tended to be of lower replicative capacity in multivariant infection (p = 0.02) suggesting an overall lowering of fitness requirements during infection with multiple variants. Notwithstanding, multivariant infection was associated with rapid CD4+ T cell decline and perturbances in the CD4+ T cell and B cell compartments compared to single variant infection, which were reversible upon control of viremia. Strategies aimed at identifying and mitigating multivariant infection could contribute toward improving HIV-1 prognosis and this may involve strategies that tighten the stringency of the transmission bottleneck such as treatment of STI. Furthermore, the sequences and chimeric viruses help with TF based experimental vaccine immunogen design and can be used in functional assays to probe effective immune responses against TF.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Founder Effect , HIV Infections , HIV-1/physiology , Virus Replication , gag Gene Products, Human Immunodeficiency Virus , Acute Disease , Adolescent , Adult , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Female , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/pathology , Humans , Male , Middle Aged , Viremia/genetics , Viremia/immunology , Viremia/pathology , Virus Replication/genetics , Virus Replication/immunology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
16.
Viruses ; 12(9)2020 09 18.
Article in English | MEDLINE | ID: mdl-32962125

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) represents a global health emergency. To improve the understanding of the systemic component of SARS-CoV-2, we investigated if viral load dynamics in plasma and respiratory samples are associated with antibody response and severity of coronavirus disease 2019 (COVID-19). SARS-CoV-2 RNA was found in plasma samples from 14 (44%) out of 32 patients. RNAemia was detected in 5 out of 6 fatal cases. Peak IgG values were significantly lower in mild/moderate than in severe (0.6 (interquartile range, IQR, 0.4-3.2) vs. 11.8 (IQR, 9.9-13.0), adjusted p = 0.003) or critical cases (11.29 (IQR, 8.3-12.0), adjusted p = 0.042). IgG titers were significantly associated with virus Ct (Cycle threshold) value in plasma and respiratory specimens ((ß = 0.4, 95% CI (confidence interval, 0.2; 0.5), p < 0.001 and ß = 0.5, 95% CI (0.2; 0.6), p = 0.002). A classification as severe or a critical case was additionally inversely associated with Ct values in plasma in comparison to mild/moderate cases (ß = -3.3, 95% CI (-5.8; 0.8), p = 0.024 and ß = -4.4, 95% CI (-7.2; 1.6), p = 0.007, respectively). Based on the present data, our hypothesis is that the early stage of SARS-CoV-2 infection is characterized by a primary RNAemia, as a potential manifestation of a systemic infection. Additionally, the viral load in plasma seems to be associated with a worse disease outcome.


Subject(s)
Antibodies, Viral/blood , Betacoronavirus/isolation & purification , Coronavirus Infections/virology , Pneumonia, Viral/virology , RNA, Viral/blood , Aged , Betacoronavirus/genetics , Betacoronavirus/immunology , COVID-19 , Coronavirus Infections/blood , Coronavirus Infections/pathology , Female , Germany/epidemiology , Hospitalization , Humans , Immunoglobulin G/blood , Male , Middle Aged , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/pathology , RNA, Viral/analysis , Respiratory System/virology , SARS-CoV-2 , Severity of Illness Index , Viral Load , Viremia/blood , Viremia/pathology , Viremia/virology
17.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32847848

ABSTRACT

Zika virus (ZIKV) can establish infection in immune privileged sites such as the testes, eye, and placenta. Whether ZIKV infection of white blood cells is required for dissemination of the virus to immune privileged sites has not been definitively shown. To assess whether initial ZIKV replication in myeloid cell populations is critical for dissemination during acute infection, recombinant ZIKVs were generated that could not replicate in these specific cells. ZIKV was cell restricted by insertion of a complementary sequence to a myeloid-specific microRNA in the 3' untranslated region. Following inoculation of a highly sensitive immunodeficient mouse model, crucial immune parameters, such as quantification of leukocyte cell subsets, cytokine and chemokine secretion, and viremia, were assessed. Decreased neutrophil numbers in the spleen were observed during acute infection with myeloid-restricted ZIKV that precluded the generation of viremia and viral dissemination to peripheral organs. Mice inoculated with a nontarget microRNA control ZIKV demonstrated increased expression of key cytokines and chemokines critical for neutrophil and monocyte recruitment and increased neutrophil influx in the spleen. In addition, ZIKV-infected Ly6Chi monocytes were identified in vivo in the spleen. Mice inoculated with myeloid-restricted ZIKV had a decrease in Ly6Chi ZIKV RNA-positive monocytes and a lack of inflammatory cytokine production compared to mice inoculated with control ZIKV.IMPORTANCE Myeloid cells, including monocytes, play a crucial role in immune responses to pathogens. Monocytes have also been implicated as "Trojan horses" during viral infections, carrying infectious virus particles to immune privileged sites and/or to sites protected by physical blood-tissue barriers, such as the blood-testis barrier and the blood-brain barrier. In this study, we found that myeloid cells are crucial to Zika virus (ZIKV) pathogenesis. By engineering ZIKV clones to encode myeloid-specific microRNA target sequences, viral replication was inhibited in myeloid cells by harnessing the RNA interference pathway. Severely immunodeficient mice inoculated with myeloid-restricted ZIKV did not demonstrate clinical signs of disease and survived infection. Furthermore, viral dissemination to peripheral organs was not observed in these mice. Lastly, we identified Ly6Cmid/hi murine monocytes as the major myeloid cell population that disseminates ZIKV.


Subject(s)
Cell Lineage/immunology , Immunocompromised Host , Myeloid Cells/immunology , Viremia/immunology , Virus Replication/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Brain/immunology , Brain/pathology , Brain/virology , Cell Lineage/genetics , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Female , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Male , Mice , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/immunology , Monocytes/immunology , Monocytes/pathology , Monocytes/virology , Myeloid Cells/classification , Myeloid Cells/pathology , Myeloid Cells/virology , Neutrophils/immunology , Neutrophils/pathology , Neutrophils/virology , RNA, Viral/genetics , RNA, Viral/immunology , Signal Transduction , Spleen/immunology , Spleen/pathology , Spleen/virology , Testis/immunology , Testis/pathology , Testis/virology , Viremia/genetics , Viremia/pathology , Viremia/virology , Zika Virus/pathogenicity , Zika Virus Infection/genetics , Zika Virus Infection/pathology , Zika Virus Infection/virology
18.
BMJ Case Rep ; 13(8)2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32816883

ABSTRACT

A 7-year-old boy presented with a constellation of bone pain, a skeletal lesion, and pancytopenia after undergoing allogeneic haematopoietic stem cell transplantation for recurrent acute B-cell lymphoblastic leukaemia. Investigations to rule out leukaemia recurrence were unremarkable. Due to presence of maturation arrest in erythropoiesis with giant pronormoblasts and aberrant intranuclear inclusions on a bone marrow aspirate, parvovirus B19 (PVB-19) staining was completed and confirmed the diagnosis of disseminated PVB-19. Though PVB-19 infection after solid organ transplantation was reported in the literature as early as 1986, acquired PVB-19 viremia presenting with a solitary bone lesion is a novel presentation in paediatrics.


Subject(s)
Hematopoietic Stem Cell Transplantation , Parvoviridae Infections/diagnosis , Parvovirus B19, Human , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Biopsy , Child , Combined Modality Therapy , Diagnosis, Differential , Humans , Male , Pain/virology , Pancytopenia/therapy , Pancytopenia/virology , Parvoviridae Infections/pathology , Parvoviridae Infections/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Recurrence , Viremia/diagnosis , Viremia/pathology , Viremia/therapy
19.
Vet Microbiol ; 246: 108744, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32605751

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) plays a key role in porcine respiratory disease complex modulating the host immune response and favouring secondary bacterial infections. Pulmonary alveolar macrophages (PAMs) are the main cells supporting PRRSV replication, with CD163 as the essential receptor for viral infection. Although interstitial pneumonia is by far the representative lung lesion, suppurative bronchopneumonia is described for PRRSV virulent strains. This research explores the role of several immune markers potentially involved in the regulation of the inflammatory response and sensitisation of lung to secondary bacterial infections by PRRSV-1 strains of different virulence. Conventional pigs were intranasally inoculated with the virulent subtype 3 Lena strain or the low virulent subtype 1 3249 strain and euthanised at 1, 3, 6 and 8 dpi. Lena-infected pigs exhibited more severe clinical signs, macroscopic lung score and viraemia associated with an increase of IL-6 and IFN-γ in sera compared to 3249-infected pigs. Extensive areas of lung consolidation corresponding with suppurative bronchopneumonia were observed in Lena-infected pigs. Lung viral load and PRRSV-N-protein+ cells were always higher in Lena-infected animals. PRRSV-N-protein+ cells were linked to a marked drop of CD163+ macrophages. The number of CD14+ and iNOS+ cells gradually increased along PRRSV-1 infection, being more evident in Lena-infected pigs. The frequency of CD200R1+ and FoxP3+ cells peaked late in both PRRSV-1 strains, with a strong correlation between CD200R1+ cells and lung injury in Lena-infected pigs. These results highlight the role of molecules involved in the earlier and higher extent of lung lesions in piglets infected with the virulent Lena strain, pointing out the activation of routes potentially involved in the restraint of the local inflammatory response.


Subject(s)
Bronchopneumonia/immunology , Inflammation/immunology , Lung/immunology , Lung/pathology , Porcine Reproductive and Respiratory Syndrome/immunology , Acute Disease , Age Factors , Animals , Antibodies, Viral/blood , Bronchopneumonia/virology , Cytokines/blood , Female , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Male , Porcine Reproductive and Respiratory Syndrome/physiopathology , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/pathogenicity , Swine , Viral Load , Viremia/immunology , Viremia/pathology , Virulence
20.
J Neurovirol ; 26(6): 957-960, 2020 12.
Article in English | MEDLINE | ID: mdl-32671809

ABSTRACT

Patients with human immunodeficiency virus (HIV) infection have a decreased risk of developing multiple sclerosis (MS) and MS patients very rarely contract HIV infection. We report on a 35-year-old woman with relapsing-remitting MS, who acquired HIV infection 8 years after MS onset. During 7 years of follow-up without combined antiretroviral therapy (cART), CD4+ counts decreased and HIV viremia increased progressively, but slightly. These trends reverted after starting cART, with optimal viro-immunological control. While the patient had many MS relapses before acquiring HIV infection, she had then only one relapse, shortly after HIV infection, despite irregular or no MS therapy. This case contributes to the discussion about MS and HIV potential interactions and describes for the first time the effects of the MS-targeting drug natalizumab in an HIV-positive patient.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/pathology , Immunologic Factors/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/pathology , Natalizumab/therapeutic use , Viremia/pathology , Adult , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Female , Glatiramer Acetate/therapeutic use , HIV/genetics , HIV/isolation & purification , HIV Infections/diagnostic imaging , HIV Infections/immunology , HIV Infections/virology , Humans , Magnetic Resonance Imaging , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/virology , RNA, Viral/genetics , Viral Load/drug effects , Viremia/diagnostic imaging , Viremia/immunology , Viremia/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...