Publications by authors named "Vanda Juranic-Lisnic"

Group 1 innate lymphoid cells (ILCs) encompass NK cells and ILC1s, which have non-redundant roles in host protection against pathogens and cancer. Despite their circulating nature, NK cells can establish residency in selected tissues during ontogeny, forming a distinct functional subset. The mechanisms that initiate, maintain, and regulate the conversion of NK cells into tissue-resident NK (trNK) cells are currently not well understood.

View Article and Find Full Text PDF
Article Synopsis
  • Cytomegalovirus (CMV) infection can harm newborns, leading to the need for effective treatments to combat both viral and immune damage.
  • Research in a mouse model shows that cellular prion protein (PrP) helps regulate T cell immunity, with PrP-null mice demonstrating better control over CMV through stronger CD8 T cell responses.
  • The study identifies that CMV increases PrP levels via ADAM10, which hinders T cell responses in neonates, and confirms similar downregulation of PrP in human CMV-infected cells, emphasizing the findings' relevance to human health.
View Article and Find Full Text PDF

Cytomegalovirus (CMV), a representative member of the Betaherpesvirinae subfamily of herpesviruses, is common in the human population, but immunocompetent individuals are generally asymptomatic when infected with this virus. However, in immunocompromised individuals and immunologically immature fetuses and newborns, CMV can cause a wide range of often long-lasting morbidities and even death. CMV is not only widespread throughout the population but it is also widespread in its hosts, infecting and establishing latency in nearly all tissues and organs.

View Article and Find Full Text PDF

Congenital human cytomegalovirus (HCMV) infection may cause life-threatening disease and permanent damage to the central nervous system. The mouse model of CMV infection is most commonly used to study mechanisms of infection and pathogenesis. While essential to limit mouse CMV (MCMV) replication, the inflammatory responses, particularly IFNγ and TNFα, cause neurodevelopmental abnormalities.

View Article and Find Full Text PDF

Dendritic cells (DC) play a crucial role in generating and maintaining antiviral immunity. While DC are implicated in the antiviral defense by inducing T cell responses, they can also become infected by Cytomegalovirus (CMV). CMV is not only highly species-specific but also specialized in evading immune protection, and this specialization is in part due to characteristic genes encoded by a given virus.

View Article and Find Full Text PDF
Article Synopsis
  • Infections in newborns trigger different immune responses compared to adults, particularly affecting NK cells during murine cytomegalovirus infection.
  • The infection leads to a significant reduction in NK cell maturation and functionality, primarily due to inflammatory responses disrupting key transcription factors.
  • Ultimately, perinatal infections result in lasting impairments in NK cell function, including decreased IFN-γ production and reduced bone marrow capacity to produce new NK cells.
View Article and Find Full Text PDF

The genomes of both human cytomegalovirus (HCMV) and murine cytomegalovirus (MCMV) were first sequenced over 20 years ago. Similar to HCMV, the MCMV genome had initially been proposed to harbor ≈170 open reading frames (ORFs). More recently, omics approaches revealed HCMV gene expression to be substantially more complex comprising several hundred viral ORFs.

View Article and Find Full Text PDF

Human cytomegalovirus is responsible for morbidity and mortality in immune compromised patients and is the leading viral cause of congenital infection. Virus-encoded microRNAs (miRNAs) represent interesting targets for novel antiviral agents. While many cellular targets that augment productive infection have been identified in recent years, regulation of viral genes such as the major viral immediate early protein 72 (IE72) by hcmv-miR-UL112-1 may contribute to both the establishment and the maintenance of latent infection.

View Article and Find Full Text PDF

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).

View Article and Find Full Text PDF

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.

View Article and Find Full Text PDF

COVID-19 vaccines prevent severe forms of the disease, but do not warrant complete protection against breakthrough infections. This could be due to suboptimal mucosal immunity at the site of virus entry, given that all currently approved vaccines are administered via the intramuscular route. In this study, we assessed humoral and cellular immune responses in BALB/c mice after intranasal and intramuscular immunization with adenoviral vector ChAdOx1-S expressing full-length Spike protein of SARS-CoV-2.

View Article and Find Full Text PDF

During COVID-19 pandemics, the availability of testing has often been a limiting factor during patient admissions into the hospital. To circumvent this problem, we adapted an existing diagnostic assay, Seegene Allplex SARS-CoV-2, into a point-of-care-style direct qPCR (POC dqPCR) assay and implemented it in the Emergency Department of Clinical Hospital Center Rijeka, Croatia. In a 4-month analysis, we tested over 10,000 patients and demonstrated that POC-dqPCR is robust and reliable and can be successfully implemented in emergency departments and similar near-patient settings and can be performed by medical personnel with little prior experience in qPCR.

View Article and Find Full Text PDF

Background: Next Generation Sequencing allows for deep analysis of transcriptional activity in cells and tissues, however it is still a cost intensive method that demands well versed data handling. Reverse transcription quantitative PCR (RT-qPCR) is the most commonly used method to measure gene expression levels, however the information gathered is quite small in comparison to NGS. A newer method called nanoString allows for highly multiplexed gene expression analysis by detecting mRNAs without the use of enzymes for reverse transcription or amplification even for single cells or low input material.

View Article and Find Full Text PDF

Broad tissue tropism of cytomegaloviruses (CMVs) is facilitated by different glycoprotein entry complexes, which are conserved between human CMV (HCMV) and murine CMV (MCMV). Among the wide array of cell types susceptible to the infection, mononuclear phagocytes (MNPs) play a unique role in the pathogenesis of the infection as they contribute both to the virus spread and immune control. CMVs have dedicated numerous genes for the efficient infection and evasion of macrophages and dendritic cells.

View Article and Find Full Text PDF

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.

View Article and Find Full Text PDF
Article Synopsis
  • Viral vectors, particularly Cytomegalovirus (CMV), are promising tools for vaccines as they can effectively stimulate both strong cellular and humoral immunity, with modifications allowing improved immune responses.
  • In a study, a modified CMV vector (RAE-1γMCMV) demonstrated a stronger CD8 T cell response and better protection against tumors and bacterial challenges compared to control vectors, despite being engineered to be less harmful.
  • Analysis of the immune response showed differences in the characteristics and functions of CD8 T cells induced by RAE-1γMCMV, indicating that their ability to respond to tumors is influenced by factors like the context of latent infection and cell populations.
View Article and Find Full Text PDF

Viral infections during pregnancy are a considerable cause of adverse outcomes and birth defects, and the underlying mechanisms are poorly understood. Among those, cytomegalovirus (CMV) infection stands out as the most common intrauterine infection in humans, putatively causing early pregnancy loss. We employed murine CMV as a model to study the consequences of viral infection on pregnancy outcome and fertility maintenance.

View Article and Find Full Text PDF
Article Synopsis
  • Congenital human cytomegalovirus (cHCMV) infection in the brain leads to various neurocognitive issues, and using a mouse model helps study its effects and underlying mechanisms.
  • Researchers found that infection caused increased levels of chemokines CXCL9 and CXCL10, which attract certain immune cells (NK and ILC1) into the brain.
  • These immune cells failed to control the viral infection and instead contributed to harmful inflammation that delayed brain development, but this effect could be countered with anti-IFN-γ antibodies.
View Article and Find Full Text PDF
Article Synopsis
  • Human cytomegalovirus (HCMV) is a major cause of congenital infections affecting the central nervous system, leading to serious long-term developmental issues, which emphasizes the urgent need for a specific vaccine.
  • Animal models, especially murine cytomegalovirus (MCMV) infections in newborn mice, are vital for understanding CMV biology and its effects, as they mimic the human condition.
  • The chapter reviews current research on rodent models of perinatal CMV infection and outlines various methods for studying MCMV infection in newborns, highlighting the immune response and neurological impacts observed.
View Article and Find Full Text PDF
Article Synopsis
  • The study successfully cloned a low-passage strain of murine cytomegalovirus (MCMV) known as G4 as a bacterial artificial chromosome (BAC), enabling better manipulation and understanding of the virus.
  • Unlike lab strains, G4 can replicate effectively in C57BL/6 mice due to a specific gene modification (m157) that alters its interaction with natural killer (NK) cells.
  • Through next-generation sequencing, researchers discovered a mutation affecting salivary gland tropism and identified a novel spliced gene (sgg1.1) whose restoration improved the virus's ability to infect salivary glands, offering new insights into the virus's biology.
View Article and Find Full Text PDF
Article Synopsis
  • The study investigates how the human cytomegalovirus (HCMV) evades the immune response that is activated through type I interferon (IFN) signaling, focusing on the role of the UL35 protein.
  • It was discovered that UL35 inhibits the activation of IFN transcription triggered by DNA and RNA sensors, with experiments showing that cells expressing UL35 have a reduced IFN response compared to those infected with a version of HCMV lacking UL35.
  • The research also highlights that UL35 interacts with the signaling factor TBK1 and demonstrates that while UL35 is modified by O-GlcNAc transferase, this modification is not essential for its function in suppressing the immune response.
View Article and Find Full Text PDF

Cytomegaloviruses (CMVs) are ubiquitous pathogens known to employ numerous immunoevasive strategies that significantly impair the ability of the immune system to eliminate the infected cells. Here, we report that the single mouse CMV (MCMV) protein, m154, downregulates multiple surface molecules involved in the activation and costimulation of the immune cells. We demonstrate that m154 uses its cytoplasmic tail motif, DD, to interfere with the adaptor protein-1 (AP-1) complex, implicated in intracellular protein sorting and packaging.

View Article and Find Full Text PDF

Viruses manipulate cellular signalling by inducing the degradation of crucial signal transducers, usually via the ubiquitin-proteasome pathway. Here, we show that the murine cytomegalovirus (Murid herpesvirus 1) M45 protein induces the degradation of two cellular signalling proteins, the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) essential modulator (NEMO) and the receptor-interacting protein kinase 1 (RIPK1), via a different mechanism: it induces their sequestration as insoluble protein aggregates and subsequently facilitates their degradation by autophagy. Aggregation of target proteins requires a distinct sequence motif in M45, which we termed 'induced protein aggregation motif'.

View Article and Find Full Text PDF

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.

View Article and Find Full Text PDF
Article Synopsis
  • Cytomegaloviruses (CMVs), particularly murine CMV (MCMV), manipulate the immune system, specifically targeting the type I interferon (IFN) response through the protein m152.
  • M152 binds to the stimulator of interferon genes (STING), delaying its movement to the Golgi, which is essential for activating the antiviral IFN signaling.
  • While m152 inhibits STING's ability to trigger IFN-related responses, it does not impact its role in activating NF-κB signaling, allowing MCMV to benefit from early viral replication while counteracting the host's antiviral defenses.
View Article and Find Full Text PDF