Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Viruses ; 14(9)2022 09 04.
Article in English | MEDLINE | ID: mdl-36146773

ABSTRACT

Studies assessing the dynamics and duration of antibody responses following SARS-CoV-2 infection or vaccination are an invaluable tool for vaccination schedule planning, assessment of risk groups and management of pandemics. In this study, we developed and employed ELISA assays to analyze the humoral responses to Nucleocapsid and Spike proteins in vaccinated health-care workers (HCW) and critically ill COVID-19 patients. Sera of more than 1000 HCWs and critically ill patients from the Clinical Hospital Center Rijeka were tested across a one-year period, encompassing the spread of major SARS-CoV-2 variants of concern (VOCs). We observed 97% of seroconversion in HCW cohort as well as sustained anti-Spike antibody response in vaccinees for more than 6 months. In contrast, the infection-induced anti-Nucleocapsid response was waning significantly in a six-month period. Furthermore, a substantial decrease in vaccinees' anti-Spike antibodies binding to Spike protein of Omicron VOC was also observed. Critically ill COVID-19 patients had higher levels of anti-Spike and anti-Nucleocapsid antibodies compared to HCWs. No significant differences in anti-Spike and anti-Nucleocapsid antibody levels between the critically ill COVID-19 patients that were on non-invasive oxygen supplementation and those on invasive ventilation support were observed. However, stronger anti-Spike, but not anti-Nucleocapsid, antibody response correlated with a better disease outcome in the cohort of patients on invasive ventilation support. Altogether, our results contribute to the growing pool of data on humoral responses to SARS-CoV-2 infection and vaccination.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Viral , Antibody Formation , COVID-19/prevention & control , Cohort Studies , Critical Illness , Croatia , Health Personnel , Humans , Nucleocapsid Proteins , Spike Glycoprotein, Coronavirus
2.
Viruses ; 14(8)2022 07 28.
Article in English | MEDLINE | ID: mdl-36016282

ABSTRACT

Herpes simplex virus 1 (HSV-1) expresses a large number of miRNAs, and their function is still not completely understood. In addition, HSV-1 has been found to deregulate host miRNAs, which adds to the complexity of the regulation of efficient virus replication. In this study, we comprehensively addressed the deregulation of host miRNAs by massive-parallel sequencing. We found that only miRNAs expressed from a single cluster, miR-183/96/182, are reproducibly deregulated during productive infection. These miRNAs are predicted to regulate a great number of potential targets involved in different cellular processes and have only 33 shared targets. Among these, members of the FoxO family of proteins were identified as potential targets for all three miRNAs. However, our study shows that the upregulated miRNAs do not affect the expression of FoxO proteins, moreover, these proteins were upregulated in HSV-1 infection. Furthermore, we show that the individual FoxO proteins are not required for efficient HSV-1 replication. Taken together, our results indicate a complex and redundant response of infected cells to the virus infection that is efficiently inhibited by the virus.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , MicroRNAs , Herpes Simplex/genetics , Herpesvirus 1, Human/physiology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Up-Regulation , Virus Replication
3.
Life (Basel) ; 11(8)2021 Aug 22.
Article in English | MEDLINE | ID: mdl-34440603

ABSTRACT

Murine cytomegalovirus (MCMV) initiates the stepwise establishment of the pre-assembly compartment (pre-AC) in the early phase of infection by the expansion of the early endosome (EE)/endosomal recycling compartment (ERC) interface and relocation of the Golgi complex. We depleted Vps34-derived phosphatidylinositol-3-phosphate (PI(3)P) at EEs by VPS34-IN1 and inhibited PI(3)P-associated functions by overexpression of 2xFYVE- and p40PX PI(3)P-binding modules to assess the role of PI(3)P-dependent EE domains in the pre-AC biogenesis. We monitored the accumulation of Rab10 and Evectin-2 in the inner pre-AC and the relocation of GM130-positive cis-Golgi organelles to the outer pre-AC by confocal microscopy. Although PI(3)P- and Vps34-positive endosomes build a substantial part of pre-AC, the PI(3)P depletion and the inhibition of PI(3)P-associated functions did not prevent the establishment of infection and progression through the early phase. The PI(3)P depletion in uninfected and MCMV-infected cells rapidly dispersed PI(3)P-bond proteins and reorganized EEs, including ablation of EE-to-ERC transport and relocation of Rab11 endosomes. The PI(3)P depletion one hour before pre-AC initiation and overexpression of 2xFYVE and p40PX domains neither prevented Rab10- and Evectin-2 accumulation, nor Golgi unlinking and relocation. These data demonstrate that PI(3)P-dependent functions, including the Rab11-dependent EE-to-ERC route, are dispensable for pre-AC initiation. Nevertheless, the virus growth was drastically reduced in PI(3)P-depleted cells, indicating that PI(3)P-associated functions are essential for the late phase of infection.

4.
Microorganisms ; 8(6)2020 May 26.
Article in English | MEDLINE | ID: mdl-32466380

ABSTRACT

The rapid activation of pattern recognition receptor (PRR)-mediated type I interferon (IFN) signaling is crucial for the host response to infection. In turn, human cytomegalovirus (HCMV) must evade this potent response to establish life-long infection. Here, we reveal that the HCMV tegument protein UL35 antagonizes the activation of type I IFN transcription downstream of the DNA and RNA sensors cGAS and RIG-I, respectively. We show that ectopic expression of UL35 diminishes the type I IFN response, while infection with a recombinant HCMV lacking UL35 induces an elevated type I IFN response compared to wildtype HCMV. With a series of luciferase reporter assays and the analysis of signaling kinetics upon HCMV infection, we observed that UL35 downmodulates PRR signaling at the level of the key signaling factor TANK-binding kinase 1 (TBK1). Finally, we demonstrate that UL35 and TBK1 co-immunoprecipitate when co-expressed in HEK293T cells. In addition, we show that a previously reported cellular binding partner of UL35, O-GlcNAc transferase (OGT), post-translationally GlcNAcylates UL35, but that this modification is not required for the antagonizing effect of UL35 on PRR signaling. In summary, we have identified UL35 as the first HCMV protein to antagonize the type I IFN response at the level of TBK1, thereby enriching our understanding of how this important herpesvirus escapes host immune responses.

5.
J Exp Med ; 216(8): 1809-1827, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31142589

ABSTRACT

CMVs efficiently target MHC I molecules to avoid recognition by cytotoxic T cells. However, the lack of MHC I on the cell surface renders the infected cell susceptible to NK cell killing upon missing self recognition. To counter this, mouse CMV (MCMV) rescues some MHC I molecules to engage inhibitory Ly49 receptors. Here we identify a new viral protein, MATp1, that is essential for MHC I surface rescue. Rescued altered-self MHC I molecules show increased affinity to inhibitory Ly49 receptors, resulting in inhibition of NK cells despite substantially reduced MHC I surface levels. This enables the virus to evade recognition by licensed NK cells. During evolution, this novel viral immune evasion mechanism could have prompted the development of activating NK cell receptors that are specific for MATp1-modified altered-self MHC I molecules. Our study solves a long-standing conundrum of how MCMV avoids recognition by NK cells, unravels a fundamental new viral immune evasion mechanism, and demonstrates how this forced the evolution of virus-specific activating MHC I-restricted Ly49 receptors.


Subject(s)
Herpesviridae Infections/immunology , Histocompatibility Antigens Class I/metabolism , Immune Evasion/immunology , Killer Cells, Natural/immunology , Muromegalovirus/metabolism , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Viral Proteins/metabolism , Animals , Antigens, Ly/genetics , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Fibroblasts/metabolism , Herpesviridae Infections/virology , Histocompatibility Antigens Class I/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics
6.
Immunol Lett ; 189: 40-47, 2017 09.
Article in English | MEDLINE | ID: mdl-28414184

ABSTRACT

Cytomegaloviruses (CMVs) have dedicated a large portion of their genome towards immune evasion targeting many aspects of the host immune system, particularly NK cells. However, the host managed to cope with the infection by developing multiple mechanisms to recognize viral threat and counterattack it, thus illustrating never-ending evolutionary interplay between CMV and its host. In this review, we will focus on several mechanisms of NK cell evasion by mouse CMV (MCMV), the role of host inhibitory and activating Ly49 receptors involved in the virus control and acquisition of adaptive features by NK cells as a consequence of MCMV infection.


Subject(s)
Herpesviridae Infections/immunology , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Animals , Autoantigens/immunology , Cytotoxicity, Immunologic , Evolution, Molecular , Histocompatibility Antigens Class I/metabolism , Host-Pathogen Interactions , Humans , Immune Evasion , Mice , NK Cell Lectin-Like Receptor Subfamily A/genetics
7.
Curr Opin Virol ; 15: 9-18, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26208082

ABSTRACT

NK cells play an important role in the control of viral infections. Cytomegaloviruses have played a big part in the accumulation of current knowledge describing how NK cells recognize and eliminate infected cells. The interference with these functions at multiple levels may not only play a role in the control of primary infections or reactivations but can also impact other arms of the immune system and leave a long-lasting stable imprint on the NK cell population. These imprints may, in turn, modify how we respond to other infections. Understanding these processes will allow us to design better diagnostic approaches and new treatment options through manipulation of our immune responses and the viruses themselves.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Animals , Cytomegalovirus Infections/virology , Humans , Immunity, Innate , Mice
8.
Methods Mol Biol ; 1119: 289-310, 2014.
Article in English | MEDLINE | ID: mdl-24639229

ABSTRACT

Human cytomegalovirus (HCMV) is a leading viral cause of congenital infections in the central nervous system (CNS) and may result in severe long-term sequelae. High rates of sequelae following congenital HCMV infection and insufficient antiviral therapy in the perinatal period make the development of an HCMV-specific vaccine a high priority of modern medicine. Due to species specificity of HCMV, animal models are frequently used to study CMV pathogenesis. Studies of murine cytomegalovirus (MCMV) infections of adult mice have served a major role as a model of CMV biology and pathogenesis, while MCMV infection of newborn mice has been successfully used as a model of perinatal CMV infection. Newborn mice infected with MCMV have high levels of viremia during which the virus establishes productive infection in most organs, coupled with a strong inflammatory response. Productive infection in the brain parenchyma during early postnatal period leads to an extensive non-necrotizing multifocal widespread encephalitis characterized by infiltration of components of both innate and adaptive immunity. As a result, impairment in postnatal development of mouse cerebellum leads to long-term motor and sensor disabilities. This chapter summarizes current findings of rodent models of perinatal CMV infection and describes methods for analysis of perinatal MCMV infection in newborn mice.


Subject(s)
Central Nervous System/virology , Cytomegalovirus Infections/genetics , Molecular Biology/methods , Muromegalovirus/genetics , Animals , Animals, Newborn/virology , Central Nervous System/pathology , Cerebellum/pathology , Cerebellum/virology , Cytomegalovirus Infections/virology , Disease Models, Animal , Humans , Mice , Muromegalovirus/immunology , Muromegalovirus/pathogenicity
9.
PLoS Pathog ; 8(8): e1002901, 2012.
Article in English | MEDLINE | ID: mdl-22952450

ABSTRACT

Little is known about the role of viral genes in modulating host cytokine responses. Here we report a new functional role of the viral encoded IE1 protein of the murine cytomegalovirus in sculpting the inflammatory response in an acute infection. In time course experiments of infected primary macrophages (MΦs) measuring cytokine production levels, genetic ablation of the immediate-early 1 (ie1) gene results in a significant increase in TNFα production. Intracellular staining for cytokine production and viral early gene expression shows that TNFα production is highly associated with the productively infected MΦ population of cells. The ie1- dependent phenotype of enhanced MΦ TNFα production occurs at both protein and RNA levels. Noticeably, we show in a series of in vivo infection experiments that in multiple organs the presence of ie1 potently inhibits the pro-inflammatory cytokine response. From these experiments, levels of TNFα, and to a lesser extent IFNß, but not the anti-inflammatory cytokine IL10, are moderated in the presence of ie1. The ie1- mediated inhibition of TNFα production has a similar quantitative phenotype profile in infection of susceptible (BALB/c) and resistant (C57BL/6) mouse strains as well as in a severe immuno-ablative model of infection. In vitro experiments with infected macrophages reveal that deletion of ie1 results in increased sensitivity of viral replication to TNFα inhibition. However, in vivo infection studies show that genetic ablation of TNFα or TNFRp55 receptor is not sufficient to rescue the restricted replication phenotype of the ie1 mutant virus. These results provide, for the first time, evidence for a role of IE1 as a regulator of the pro-inflammatory response and demonstrate a specific pathogen gene capable of moderating the host production of TNFα in vivo.


Subject(s)
Gene Expression Regulation, Viral/genetics , Herpesviridae Infections/immunology , Immediate-Early Proteins/genetics , Muromegalovirus/genetics , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Cytokines/metabolism , DNA Replication , DNA, Viral/genetics , Female , Herpesviridae Infections/virology , Immediate-Early Proteins/metabolism , Liver/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/growth & development , Muromegalovirus/physiology , Phenotype , Signal Transduction , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
10.
J Virol ; 85(12): 6065-76, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21471238

ABSTRACT

The global transcriptional program of murine cytomegalovirus (MCMV), involving coding, noncoding, and antisense transcription, remains unknown. Here we report an oligonucleotide custom microarray platform capable of measuring both coding and noncoding transcription on a genome-wide scale. By profiling MCMV wild-type and immediate-early mutant strains in fibroblasts, we found rapid activation of the transcriptome by 6.5 h postinfection, with absolute dependency on ie3, but not ie1 or ie2, for genomic programming of viral gene expression. Evidence is also presented to show, for the first time, genome-wide noncoding and bidirectional transcription at late stages of MCMV infection.


Subject(s)
Fibroblasts/virology , Gene Expression Profiling , Gene Expression Regulation, Viral , Muromegalovirus/metabolism , Viral Proteins/metabolism , Animals , Genome, Viral , Herpesviridae Infections/virology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Muromegalovirus/genetics , Muromegalovirus/pathogenicity , Mutation , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Time Factors , Viral Proteins/genetics
11.
Clin Exp Metastasis ; 28(5): 451-62, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21442355

ABSTRACT

Galectin-3, a ß galactoside-binding lectin, plays an important role in the processes relevant to tumorigenesis such as malignant cell transformation, invasion and metastasis. We have investigated whether deletion of Galectin-3 in the host affects the metastasis of B16F1 malignant melanoma. Galectin-3-deficient (Gal-3(-/-)) mice are more resistant to metastatic malignant melanoma as evaluated by number and size of metastatic colonies in the lung. In vitro assays showed lower number of attached malignant cells in the tissue section derived from Gal-3(-/-) mice. Furthermore, lack of Galectin-3 correlates with higher serum levels of IFN-γ and IL-17 in tumor bearing hosts. Interestingly, spleens of Gal-3(-/-) mice have lower number of Foxp3(+) T cells after injection of B16F1 melanoma cells. Finally, we found that while CD8(+) T cell and adherent cell cytotoxicity were similar, there was greater cytotoxic activity of splenic NK cells of Gal-3(-/-) mice compared with "wild-type" (Gal-3( +/+ )) mice. Despite the reduction in total number of CD3ε(-)NK1.1(+), Gal-3(-/-) mice constitutively have a significantly higher percentage of effective cytotoxic CD27(high)CD11b(high) NK cells as well as the percentage of immature CD27(high)CD11b(low) NK cells. In contrast, CD27(low)CD11b(high) less functionally exhausted NK cells and NK cells bearing inhibitory KLRG1 receptor were more numerous in Gal-3( +/+ ) mice. It appears that lack of Galectin-3 affects tumor metastasis by at least two independent mechanisms: by a decrease in binding of melanoma cells onto target tissue and by enhanced NK-mediated anti-tumor response suggesting that Galectin-3 may be considered as therapeutic target.


Subject(s)
Galectin 3/deficiency , Galectin 3/genetics , Gene Deletion , Killer Cells, Natural/immunology , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis/genetics , Animals , Cell Adhesion , Female , Galectin 3/metabolism , Male , Melanoma/immunology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/immunology
12.
J Clin Invest ; 120(12): 4532-45, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21099111

ABSTRACT

Human CMV (HCMV) is a major cause of morbidity and mortality in both congenitally infected and immunocompromised individuals. Development of an effective HCMV vaccine would help protect these vulnerable groups. NK group 2, member D (NKG2D) is a potent activating receptor expressed by cells of the innate and adaptive immune systems. Its importance in HCMV immune surveillance is indicated by the elaborative evasion mechanisms evolved by the virus to avoid NKG2D. In order to study this signaling pathway, we engineered a recombinant mouse CMV expressing the high-affinity NKG2D ligand RAE-1γ (RAE-1γMCMV). Expression of RAE-1γ by MCMV resulted in profound virus attenuation in vivo and lower latent viral DNA loads. RAE-1γMCMV infection was efficiently controlled by immunodeficient hosts, including mice lacking type I interferon receptors or immunosuppressed by sublethal γ-irradiation. Features of MCMV infection in neonates were also diminished. Despite tight innate immune control, RAE-1γMCMV infection elicited strong and long-lasting protective immunity. Maternal RAE-1γMCMV immunization protected neonatal mice from MCMV disease via placental transfer of antiviral Abs. Despite strong selective pressure, the RAE-1γ transgene did not exhibit sequence variation following infection. Together, our results indicate that use of a recombinant virus encoding the ligand for an activating NK cell receptor could be a powerful approach to developing a safe and immunogenic HCMV vaccine.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/immunology , Muromegalovirus/genetics , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology , Animals , Animals, Newborn , Base Sequence , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , DNA Primers/genetics , Female , Genetic Engineering , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Humans , Immunity, Maternally-Acquired , Killer Cells, Natural/immunology , Ligands , Mice , Mice, Inbred BALB C , Pregnancy , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
13.
Curr Opin Microbiol ; 13(4): 530-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20558100

ABSTRACT

Since their discovery, our understanding of NK cells has evolved from branding them marginal innate immunity cells to key players in anti-viral and anti-tumor immunity. Importance of NK cells in control of various viral infections is perhaps best illustrated by the existence of plethora of viral mechanisms aimed to modulate their function. These mechanisms include not only virally encoded immunoevasion proteins but also viral miRNA. Moreover, the evidence has been accumulated supporting the role of viral immunoevasion of NK cells in viral pathogenesis in vivo.


Subject(s)
Killer Cells, Natural/immunology , Virus Diseases/virology , Viruses/immunology , Animals , Humans , Immune Evasion , Killer Cells, Natural/virology , MicroRNAs/immunology , MicroRNAs/metabolism , Proteins/immunology , Proteins/metabolism , RNA, Viral/immunology , RNA, Viral/metabolism , Receptors, Natural Killer Cell/immunology , Receptors, Natural Killer Cell/metabolism , Virus Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...