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










Database
Language
Publication year range
1.
Nat Immunol ; 25(6): 981-993, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811816

ABSTRACT

Viral infection makes us feel sick as the immune system alters systemic metabolism to better fight the pathogen. The extent of these changes is relative to the severity of disease. Whether blood glucose is subject to infection-induced modulation is mostly unknown. Here we show that strong, nonlethal infection restricts systemic glucose availability, which promotes the antiviral type I interferon (IFN-I) response. Following viral infection, we find that IFNγ produced by γδ T cells stimulates pancreatic ß cells to increase glucose-induced insulin release. Subsequently, hyperinsulinemia lessens hepatic glucose output. Glucose restriction enhances IFN-I production by curtailing lactate-mediated inhibition of IRF3 and NF-κB signaling. Induced hyperglycemia constrained IFN-I production and increased mortality upon infection. Our findings identify glucose restriction as a physiological mechanism to bring the body into a heightened state of responsiveness to viral pathogens. This immune-endocrine circuit is disrupted in hyperglycemia, possibly explaining why patients with diabetes are more susceptible to viral infection.


Subject(s)
Blood Glucose , Immunity, Innate , Interferon-gamma , Animals , Interferon-gamma/metabolism , Interferon-gamma/immunology , Mice , Blood Glucose/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Mice, Inbred C57BL , Signal Transduction/immunology , Insulin/metabolism , Insulin/immunology , Mice, Knockout , Hyperglycemia/immunology , Interferon Regulatory Factor-3/metabolism , NF-kappa B/metabolism , Humans , Liver/immunology , Liver/virology , Liver/metabolism , Male
2.
Nat Commun ; 14(1): 6412, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828009

ABSTRACT

Infections in early life can elicit substantially different immune responses and pathogenesis than infections in adulthood. Here, we investigate the consequences of murine cytomegalovirus infection in newborn mice on NK cells. We show that infection severely compromised NK cell maturation and functionality in newborns. This effect was not due to compromised virus control. Inflammatory responses to infection dysregulated the expression of major transcription factors governing NK cell fate, such as Eomes, resulting in impaired NK cell function. Most prominently, NK cells from perinatally infected mice have a diminished ability to produce IFN-γ due to the downregulation of long non-coding RNA Ifng-as1 expression. Moreover, the bone marrow's capacity to efficiently generate new NK cells is reduced, explaining the prolonged negative effects of perinatal infection on NK cells. This study demonstrates that viral infections in early life can profoundly impact NK cell biology, including long-lasting impairment in NK cell functionality.


Subject(s)
Cytomegalovirus Infections , Muromegalovirus , Mice , Animals , Killer Cells, Natural , Cytomegalovirus Infections/genetics
3.
Croat Med J ; 63(2): 126-140, 2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35505646

ABSTRACT

AIM: To determine the predictive value of phosphorylated human epidermal growth factor receptor 2 (pHER2Y1248) status in breast cancer (BC) patients undergoing trastuzumab-based adjuvant therapy. METHODS: Immunohistochemical status of pHER2Y1248, EGFR/HER1, HER3, and HER4 was determined in 124 consecutive HER2-positive BC patients (median age [range]=57 years [49.0-64.0]) treated at the University Hospital for Tumors, Zagreb, between 2008 and 2011. The median follow-up was 84 months (60.0-84.0). Prognostic factors of disease free survival (DFS) rate were evaluated with Kaplan-Meier/log-rank test and Cox regression analysis. RESULTS: pHER2Y1248, HER1, HER3, and HER4 were expressed in 66.1%, 9.7%, 70.2%, and 71.0% of patients, respectively. Disease progression (DP) was observed in 17.1% of pHER2Y1248-positive and 47.6% of pHER2Y1248-negative BCs (P=0.001). Kaplan-Meier analysis showed a worse five-year DFS in pHER2Y1248-negative patients who were older than 60 years (P<0.001) and had positive lymph node status (P<0.001); tumor size >2.0 cm (P<0.001); higher histological grade (P<0.001); HER2E intrinsic subtype (P<0.001), negative hormone receptors (P<0.001); negative HER1 status (P<0.001), positive HER3 (P=0.002); and/or positive HER4 (P=0.002) status. The only negative prognostic factor for five-year DFS in multivariate Cox regression analysis was pHER2Y1248-negative (hazard ratio [HR] 3.6, 95% confidence interval [CI] 1.8-7.2, P<0.001) and lymph node-positive status (HR 3.6, 95% CI 1.3-9.8, P=0.014). CONCLUSION: pHER2Y1248 predicts sensitivity to trastuzumab and a better five-year DFS regardless of any other prognostic parameter. In HER2-positive BC patients. Non-phosphorylated HER2Y1248 is a strong predictor of trastuzumab resistance and a poor DFS.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Disease-Free Survival , Female , Humans , Middle Aged , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Trastuzumab/therapeutic use
4.
Viruses ; 13(12)2021 12 10.
Article in English | MEDLINE | ID: mdl-34960750

ABSTRACT

There is a large unmet need for a prophylactic vaccine against human cytomegalovirus (HCMV) to combat the ubiquitous infection that is ongoing with this pathogen. A vaccination against HCMV could protect immunocompromised patients and prevent birth defects caused by congenital HCMV infections. Moreover, cytomegalovirus (CMV) has a number of features that make it a very interesting vector platform for gene therapy. In both cases, preparation of a highly purified virus is a prerequisite for safe and effective application. Murine CMV (MCMV) is by far the most studied model for HCMV infections with regard to the principles that govern the immune surveillance of CMVs. Knowledge transfer from MCMV and mice to HCMV and humans could be facilitated by better understanding and characterization of the biological and biophysical properties of both viruses. We carried out a detailed investigation of HCMV and MCMV growth kinetics as well as stability under the influence of clarification and different storage conditions. Further, we investigated the possibilities to concentrate and purify both viruses by ultracentrifugation and ion-exchange chromatography. Defective enveloped particles were not separately analyzed; however, the behavior of exosomes was examined during all experiments. The effectiveness of procedures was monitored using CCID50 assay, Nanoparticle tracking analysis, ELISA for host cell proteins, and quantitative PCR for host cell DNA. MCMV generally proved to be more robust in handling. Despite its greater sensitivity, HCMV was efficiently (100% recovery) purified and concentrated by anion-exchange chromatography using QA monolithic support. The majority of the host genomic DNA as well as most of the host cell proteins were removed by this procedure.


Subject(s)
Cytomegalovirus/growth & development , Cytomegalovirus/isolation & purification , Muromegalovirus/growth & development , Muromegalovirus/isolation & purification , Animals , Cell Line , Chromatography, Ion Exchange , Cryopreservation , Exosomes , Humans , Mice , Ultracentrifugation , Virus Cultivation
5.
Front Immunol ; 12: 681380, 2021.
Article in English | MEDLINE | ID: mdl-34168650

ABSTRACT

Viral vectors have emerged as a promising alternative to classical vaccines due to their great potential for induction of a potent cellular and humoral immunity. Cytomegalovirus (CMV) is an attractive vaccine vector due to its large genome with many non-essential immunoregulatory genes that can be easily manipulated to modify the immune response. CMV generates a strong antigen-specific CD8 T cell response with a gradual accumulation of these cells in the process called memory inflation. In our previous work, we have constructed a mouse CMV vector expressing NKG2D ligand RAE-1γ in place of its viral inhibitor m152 (RAE-1γMCMV), which proved to be highly attenuated in vivo. Despite attenuation, RAE-1γMCMV induced a substantially stronger CD8 T cell response to vectored antigen than the control vector and provided superior protection against bacterial and tumor challenge. In the present study, we confirmed the enhanced protective capacity of RAE-1γMCMV as a tumor vaccine vector and determined the phenotypical and functional characteristics of memory CD8 T cells induced by the RAE-1γ expressing MCMV. RNAseq data revealed higher transcription of numerous genes associated with effector-like CD8 T cell phenotype in RAE-1γMCMV immunized mice. CD8 T cells primed with RAE-1γMCMV were enriched in TCF1 negative population, with higher expression of KLRG1 and lower expression of CD127, CD27, and Eomes. These phenotypical differences were associated with distinct functional features as cells primed with RAE-1γMCMV showed inferior cytokine-producing abilities but comparable cytotoxic potential. After adoptive transfer into naive hosts, OT-1 cells induced with both RAE-1γMCMV and the control vector were equally efficient in rejecting established tumors, suggesting the context of latent infection and cell numbers as important determinants of enhanced anti-tumor response following RAE-1γMCMV vaccination. Overall, our results shed new light on the phenotypical and functional distinctness of memory CD8 T cells induced with CMV vector expressing cellular ligand for the NKG2D receptor.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytomegalovirus Vaccines/immunology , Immunologic Memory , NK Cell Lectin-Like Receptor Subfamily K/genetics , Phenotype , Animals , Cancer Vaccines/immunology , Computational Biology/methods , Cytomegalovirus/immunology , Cytotoxicity, Immunologic , Gene Expression Profiling , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Immunophenotyping , Lymphocyte Activation/immunology , Mice , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Transcriptome
6.
Med Microbiol Immunol ; 208(3-4): 487-494, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30923899

ABSTRACT

Cytomegalovirus (CMV) infection is a significant public health problem. Congenital CMV infection is a leading infectious cause of long-term neurodevelopmental sequelae, including mental retardation and sensorineural hearing loss. Immune protection against mouse cytomegalovirus (MCMV) is primarily mediated by NK cells and CD8+ T cells, while CD4+ T cells are not needed for control of MCMV in majority of organs in immunocompetent adult mice. Here, we set out to determine the role of CD4+ T cells upon MCMV infection of newborn mice. We provide evidence that CD4+ T cells are essential for clearance of MCMV infection in brain of neonatal mice and for prevention of recurrence of latent MCMV. In addition, we provide evidence that CD4+ T cells are required for induction and maintenance of tissue-resident memory CD8+ T cells in the brain of mice perinatally infected with MCMV.


Subject(s)
Brain/immunology , Brain/virology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Muromegalovirus/growth & development , Muromegalovirus/immunology , Animals , Animals, Newborn , Disease Models, Animal , Mice
7.
Front Immunol ; 9: 991, 2018.
Article in English | MEDLINE | ID: mdl-29867968

ABSTRACT

The development of a vaccine against human cytomegalovirus (CMV) has been a subject of long-term medical interest. The research during recent years identified CMV as an attractive vaccine vector against infectious diseases and tumors. The immune response to CMV persists over a lifetime and its unique feature is the inflationary T cell response to certain viral epitopes. CMV encodes numerous genes involved in immunoevasion, which are non-essential for virus growth in vitro. The deletion of those genes results in virus attenuation in vivo, which enables us to dramatically manipulate its virulence and the immune response. We have previously shown that the murine CMV (MCMV) expressing RAE-1γ, one of the cellular ligands for the NKG2D receptor, is highly attenuated in vivo but retains the ability to induce a strong CD8+ T cell response. Here, we demonstrate that recombinant MCMV expressing high affinity NKG2D ligand murine UL16 binding protein-like transcript (MULT-1) (MULT-1MCMV) inserted in the place of its viral inhibitor is dramatically attenuated in vivo in a NK cell-dependent manner, both in immunocompetent adult mice and in immunologically immature newborns. MULT-1MCMV was more attenuated than the recombinant virus expressing RAE-1γ. Despite the drastic sensitivity to innate immune control, MULT-1MCMV induced an efficient CD8+ T cell response to viral and vectored antigens. By using in vitro assay, we showed that similar to RAE-1γMCMV, MULT-1 expressing virus provided strong priming of CD8+ T cells. Moreover, MULT-1MCMV was able to induce anti-viral antibodies, which after passing the transplacental barrier protect offspring of immunized mothers from challenge infection. Altogether, this study further supports the concept that CMV expressing NKG2D ligand possesses excellent characteristics to serve as a vaccine or vaccine vector.


Subject(s)
Carrier Proteins/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/immunology , Histocompatibility Antigens Class I/genetics , Muromegalovirus/genetics , Animals , Animals, Newborn , CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/immunology , Cytomegalovirus Vaccines/genetics , Disease Models, Animal , Female , Genetic Vectors/immunology , Histocompatibility Antigens Class I/immunology , Immunity, Maternally-Acquired , Immunocompetence , Killer Cells, Natural/immunology , Membrane Proteins , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology
8.
Eur J Immunol ; 48(6): 950-964, 2018 06.
Article in English | MEDLINE | ID: mdl-29500823

ABSTRACT

Congenital HCMV infection is a leading infectious cause of long-term neurodevelopmental sequelae. Infection of newborn mice with mouse cytomegalovirus (MCMV) intraperitoneally is a well-established model of congenital human cytomegalovirus infection, which best recapitulates the hematogenous route of virus spread to brain and subsequent pathology. Here, we used this model to investigate the role, dynamics, and phenotype of CD8+ T cells in the brain following infection of newborn mice. We show that CD8+ T cells infiltrate the brain and form a pool of tissue-resident memory T cells (TRM cells) that persist for lifetime. Adoptively transferred virus-specific CD8+ T cells provide protection against primary MCMV infection in newborn mice, reduce brain pathology, and remain in the brain as TRM cells. Brain CD8+ TRM cells were long-lived, slowly proliferating cells able to respond to local challenge infection. Importantly, brain CD8+ TRM cells controlled latent MCMV and their depletion resulted in virus reactivation and enhanced inflammation in brain.


Subject(s)
Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/physiology , Muromegalovirus/physiology , T-Lymphocytes, Cytotoxic/immunology , Virus Activation/immunology , Adoptive Transfer , Animals , Animals, Newborn , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Congenital Abnormalities , Disease Models, Animal , Humans , Immunologic Memory , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/transplantation
9.
Microbes Infect ; 20(9-10): 543-551, 2018.
Article in English | MEDLINE | ID: mdl-29287989

ABSTRACT

Human cytomegalovirus (HCMV) is the most common cause of viral infection acquired in utero. Even though the infection has been studied for several decades, immune determinants important for virus control and mechanisms of long-term sequelae caused by infection are still insufficiently characterized. Animal models of congenital HCMV infection provide unique opportunity to study various aspects of human disease. In this review, we summarize current knowledge on the role of immune system in congenital CMV infection, with emphasis on lessons learned from mouse model of congenital CMV infection.


Subject(s)
Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/immunology , Cytomegalovirus/physiology , Adaptive Immunity , Animals , Central Nervous System/immunology , Central Nervous System/virology , Cytomegalovirus/immunology , Disease Models, Animal , Humans , Immunity, Innate
SELECTION OF CITATIONS
SEARCH DETAIL
...