Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.581
Filter
1.
Arch Virol ; 169(8): 157, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38969819

ABSTRACT

Viruses use various strategies and mechanisms to deal with cells and proteins of the immune system that form a barrier against infection. One of these mechanisms is the encoding and production of viral microRNAs (miRNAs), whose function is to regulate the gene expression of the host cell and the virus, thus creating a suitable environment for survival and spreading viral infection. miRNAs are short, single-stranded, non-coding RNA molecules that can regulate the expression of host and viral proteins, and due to their non-immunogenic nature, they are not eliminated by the cells of the immune system. More than half of the viral miRNAs are encoded and produced by Orthoherpesviridae family members. Human cytomegalovirus (HCMV) produces miRNAs that mediate various processes in infected cells to contribute to HCMV pathogenicity, including immune escape, viral latency, and cell apoptosis. Here, we discuss which cellular and viral proteins or cellular pathways and processes these mysterious molecules target to evade immunity and support viral latency in infected cells. We also discuss current evidence that their function of bypassing the host's innate and adaptive immune system is essential for the survival and multiplication of the virus and the spread of HCMV infection.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Immune Evasion , MicroRNAs , Virus Latency , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Virus Latency/genetics , MicroRNAs/genetics , Humans , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/immunology , RNA, Viral/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Gene Expression Regulation, Viral
2.
Virol J ; 21(1): 149, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956615

ABSTRACT

OBJECTIVES: This study aimed to characterize incidences of CMV reactivations within one year post-allo-SCT and identify risk factors for CMV second reactivation episode in population with high seropositivity where first CMV reactivation episode deemed to be high. METHODS: This retrospective cohort study analyzed data from 359 allo-SCT patients aged 14 and older admitted to a tertiary academic hospital. Data on demographic and clinical factors, CMV serostatus, conditioning regimens, graft-versus-host disease prophylaxis, engraftment time, and CMV reactivations were collected. RESULTS: First and second CMV reactivations occurred in 88.9% and 18.4% of post-allo-SCT patients respectively. Patients were stratified into two groups based on primary disease necessitating allo-SCT, patients with malignant (Group 1) and non-malignant (Group 2) hematological disease. Factors associated with the second reactivation included cord blood as a stem cell source, human leukocyte antigen mismatch, acute graft-versus-host disease, and hematological malignancies. Patients with non-malignant hematological disease displayed better outcomes, including a higher rate of spontaneous clearance of first CMV reactivation (70% versus 49.4%) and lower rates of second CMV reactivation (9.6% versus 31%) than those with malignant hematological disease. The one-year overall survival rate was 87.7% (95.5% in non-malignant hematological disease and 78.13% in malignant hematological disease). CONCLUSION: Our findings are concordant with previous local study in regard to high rate of first CMV reactivation post-allo-SCT. It appears that patients with nonmalignant hematological disease had better outcomes, such as lower second CMV reactivation and higher survival rates compared to patients with malignant hematological disease. Further investigation is needed to identify other factors affecting recurrent CMV reactivations in allo-SCT in patients with malignant hematological disease.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Transplantation, Homologous , Virus Activation , Humans , Male , Female , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/epidemiology , Middle Aged , Adult , Retrospective Studies , Young Adult , Cytomegalovirus/immunology , Adolescent , Risk Factors , Aged , Transplantation, Homologous/adverse effects , Recurrence , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/adverse effects , Incidence
3.
J Clin Immunol ; 44(6): 142, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847943

ABSTRACT

PURPOSE: Common Variable Immunodeficiency (CVID) is characterized by hypogammaglobulinemia and failure of specific antibody production due to B-cell defects. However, studies have documented various T-cell abnormalities, potentially linked to viral complications. The frequency of Cytomegalovirus (CMV) replication in CVID cohorts is poorly studied. To address this gap in knowledge, we set up an observational study with the objectives of identifying CVID patients with active viraemia (CMV, Epstein-Barr virus (EBV)), evaluating potential correlations with immunophenotypic characteristics, clinical outcome, and the dynamic progression of clinical phenotypes over time. METHODS: 31 CVID patients were retrospectively analysed according to viraemia, clinical and immunologic characteristics. 21 patients with non CVID humoral immunodeficiency were also evaluated as control. RESULTS: Active viral replication of CMV and/or EBV was observed in 25% of all patients. CMV replication was detected only in CVID patients (16%). CVID patients with active viral replication showed reduced HLA-DR+ NK counts when compared with CMV-DNA negative CVID patients. Viraemic patients had lower counts of LIN-DNAMbright and LIN-CD16+ inflammatory lymphoid precursors which correlated with NK-cell subsets. Analysis of the dynamic progression of CVID clinical phenotypes over time, showed that the initial infectious phenotype progressed to complicated phenotypes with time. All CMV viraemic patients had complicated disease. CONCLUSION: Taken together, an impaired production of inflammatory precursors and NK activation is present in CVID patients with active viraemia. Since "Complicated" CVID occurs as a function of disease duration, there is need for an accurate evaluation of this aspect to improve classification and clinical management of CVID patients.


Subject(s)
Common Variable Immunodeficiency , Cytomegalovirus Infections , Cytomegalovirus , Herpesvirus 4, Human , Virus Replication , Humans , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/complications , Male , Female , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Adult , Middle Aged , Herpesvirus 4, Human/physiology , Herpesvirus 4, Human/immunology , Retrospective Studies , Killer Cells, Natural/immunology , Young Adult , Viremia/immunology , Epstein-Barr Virus Infections/immunology , Immunophenotyping , Aged , Adolescent
4.
Curr Opin Infect Dis ; 37(4): 264-269, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38861267

ABSTRACT

PURPOSE OF REVIEW: Discuss the recent evidence on cytomegalovirus (CMV) serology in allogeneic hematopoeic cell transplant (HCT) recipients. RECENT FINDINGS: Whereas the role CMV-specific cellular mediated immunity has recently emerged as an important factor of CMV DNAemia posttransplant, the value of CMV serology has remained unchanged through decades, associated with donor selection and posttransplant prophylactic and monitoring strategies. In this review, we describe and discuss the emerging reports on the association between the magnitude of pretransplant CMV immunoglobulin G (IgG) titer and the posttransplant incidence of CMV DNAemia, as CMV IgG titer could become an additional tool in CMV risk assessment in the future. SUMMARY: Pretransplant recipient CMV serology may have significant implications in posttransplant CMV reactivation in allogeneic HCT recipients.


Subject(s)
Antibodies, Viral , Cytomegalovirus Infections , Cytomegalovirus , Hematopoietic Stem Cell Transplantation , Transplantation, Homologous , Humans , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Cytomegalovirus/immunology , Transplantation, Homologous/adverse effects , Antibodies, Viral/blood , Immunoglobulin G/blood , Serologic Tests/methods , DNA, Viral/blood
5.
Transpl Int ; 37: 12963, 2024.
Article in English | MEDLINE | ID: mdl-38868358

ABSTRACT

Cytomegalovirus (CMV) infection detrimentally influences graft survival in kidney transplant recipients, with the risk primarily determined by recipient and donor serostatus. However, recipient CD8+ T cells play a crucial role in CMV control. The optimal preventive strategy (prophylaxis vs. pre-emptive treatment), particularly for seropositive (intermediate risk) recipients, remains uncertain. We investigated CD8+ T cell subpopulation dynamics and CMV occurrence (DNAemia ≥ 100 IU/mL) in 65 kidney transplant recipients, collecting peripheral blood mononuclear cells before (T1) and 1 year after transplantation (T2). Comparing the two timepoints, we found an increase in granulocyte, monocyte and CD3+CD8+ T cells numbers, while FoxP3+CD25+, LAG-3+ and PD-1+ frequencies were reduced at T2. CMV DNAemia occurred in 33 recipients (55.8%) during the first year. Intermediate risk patients were disproportionally affected by posttransplant CMV (N = 29/45, 64.4%). Intermediate risk recipients developing CMV after transplantation exhibited lower leukocyte, monocyte, and granulocyte counts and higher FoxP3+CD25+ frequencies in CD3+CD8+ T cells pre-transplantation compared to patients staying CMV negative. Pre-transplant FoxP3+CD25+ in CD3+CD8+ T cells had the best discriminatory potential for CMV infection prediction within the first year after transplantation (AUC: 0.746). The FoxP3+CD25+ CD3+CD8+ T cell subset may aid in selecting intermediate risk kidney transplant recipients for CMV prophylaxis.


Subject(s)
CD8-Positive T-Lymphocytes , Cytomegalovirus Infections , Forkhead Transcription Factors , Interleukin-2 Receptor alpha Subunit , Kidney Transplantation , Humans , Kidney Transplantation/adverse effects , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Female , Male , CD8-Positive T-Lymphocytes/immunology , Middle Aged , Forkhead Transcription Factors/metabolism , Adult , Interleukin-2 Receptor alpha Subunit/metabolism , Aged , CD3 Complex/metabolism , Cytomegalovirus/immunology , Risk Factors , Transplant Recipients , Graft Survival/immunology
6.
Viral Immunol ; 37(5): 259-265, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38848306

ABSTRACT

Cytomegalovirus (CMV) has long been thought to have an association with glioblastoma multiforme (GBM), although the exact role of CMV and any subsequent implications for treatment have yet to be fully understood. This study addressed whether IGH complementarity determining region-3 (CDR3)-CMV protein chemical complementarity, with IGH CDR3s representing both tumor resident and blood-sourced IGH recombinations, was associated with overall survival (OS) distinctions. IGH recombination sequencing reads were obtained from (a) the Clinical Proteomic Tumor Analysis Consortium, tumor RNAseq files; and (b) the cancer genome atlas, blood exome-derived files. The Adaptive Match web tool was used to calculate chemical complementarity scores (CSs) based on hydrophobic interactions, and those scores were used to group GBM cases and assess survival probabilities. We found a higher OS probability for cases whose hydrophobic IGH CDR3-CMV protein chemical complementarity scores (Hydro CSs) were in the upper 50th percentile for several CMV proteins, including UL99 and UL123, as well as for CSs based on known B cell epitopes representing these proteins. We also identified multiple immune signature genes, including CD79A and TNFRSF17, for which higher RNA expression was associated with higher Hydro CSs. Results were consistent with the idea that stronger immunoglobulin responses to CMV are associated with better OS probabilities for GBM.


Subject(s)
Complementarity Determining Regions , Cytomegalovirus Infections , Cytomegalovirus , Glioblastoma , Viral Proteins , Humans , Glioblastoma/mortality , Glioblastoma/genetics , Glioblastoma/virology , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Cytomegalovirus Infections/mortality , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Viral Proteins/genetics , Viral Proteins/immunology , Immunoglobulin Heavy Chains/genetics , Female , Middle Aged , Male , Survival Analysis , Aged , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics
8.
Clin Lab ; 70(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38868867

ABSTRACT

BACKGROUND: Both humoral and cell-mediated immunity of the patient affected by multiple myeloma (MM) are impaired; thus, infection is the main cause of the onset of symptoms and death caused by MM. Bortezomib is a first-line drug approved for patients with multiple myeloma (MM) and has significantly increased their overall survival. However, bortezomib-induced peripheral neuropathy (PN) remains a significant side effect that has led to its discontinuation in some patients. Guillain-Barre syndrome (GBS) is thought to be related to immune damage, and most patients have cytomegalovirus (CMV), Epstein-Barr virus (EBV), or mycoplasma infection before onset. Cases of GBS secondary to MM are rare. METHODS: We provide a case of GBS caused by cytomegalovirus infection after MM treatment, and briefly review the existing literature. RESULTS: Secondary GBS after MM. This patient received active treatment. The clinical symptoms are gradually improving. CONCLUSIONS: The use of bortezomib has the risk of reactivating the virus. It is more about the reactivation of hep-atitis B virus. Nonetheless, cytomegalovirus and Epstein-Barr virus shall have our attention. Patients with MM need to monitor CMV, regularly, especially during the treatment of bortezomib. At the same time, they also need to closely monitor the symptoms and signs of the nervous system to guard against the occurrence of GBS.


Subject(s)
Bortezomib , Cytomegalovirus Infections , Guillain-Barre Syndrome , Multiple Myeloma , Female , Humans , Middle Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Bortezomib/adverse effects , Cytomegalovirus/immunology , Cytomegalovirus/drug effects , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/virology , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/drug therapy , Guillain-Barre Syndrome/etiology , Multiple Myeloma/drug therapy , Multiple Myeloma/complications
9.
Methods Mol Biol ; 2813: 295-308, 2024.
Article in English | MEDLINE | ID: mdl-38888785

ABSTRACT

Identification and characterization of CD8+ T-cells is important to determine their role in protecting and clearing viral infections. Here we provide details of the peptide-MHC (pMHC) tetramers-based approach to identify antigen-specific T-cells in human and murine samples. This method provides ex vivo quantification and functional characterization of T-cells reactive to specific viral antigens derived from CMV and rotavirus in human blood and in murine intestinal lamina propria samples, respectively.


Subject(s)
Antigens, Viral , CD8-Positive T-Lymphocytes , Rotavirus , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , Antigens, Viral/immunology , Rotavirus/immunology , Cytomegalovirus/immunology , Virus Diseases/immunology , Virus Diseases/virology , Epitopes, T-Lymphocyte/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/virology
11.
Elife ; 132024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900146

ABSTRACT

Human leucocyte antigen class I (HLA-I) molecules play a central role for both NK and T-cell responses that prevent serious human cytomegalovirus (HCMV) disease. To create opportunities for viral spread, several HCMV-encoded immunoevasins employ diverse strategies to target HLA-I. Among these, the glycoprotein US10 is so far insufficiently studied. While it was reported that US10 interferes with HLA-G expression, its ability to manipulate classical HLA-I antigen presentation remains unknown. In this study, we demonstrate that US10 recognizes and binds to all HLA-I (HLA-A, -B, -C, -E, -G) heavy chains. Additionally, impaired recruitment of HLA-I to the peptide loading complex was observed. Notably, the associated effects varied significantly dependending on HLA-I genotype and allotype: (i) HLA-A molecules evaded downregulation by US10, (ii) tapasin-dependent HLA-B molecules showed impaired maturation and cell surface expression, and (iii) ß2m-assembled HLA-C, in particular HLA-C*05:01 and -C*12:03, and HLA-G were strongly retained in complex with US10 in the endoplasmic reticulum. These genotype-specific effects on HLA-I were confirmed through unbiased HLA-I ligandome analyses. Furthermore, in HCMV-infected fibroblasts inhibition of overlapping US10 and US11 transcription had little effect on HLA-A, but induced HLA-B antigen presentation. Thus, the US10-mediated impact on HLA-I results in multiple geno- and allotypic effects in a so far unparalleled and multimodal manner.


During a viral infection, the immune system must discriminate between healthy and infected cells to selectively kill infected cells. Healthy cells have different types of molecules known collectively as HLA-I on their surface. These molecules present small fragments of proteins from the cell, called antigens, to patrolling immune cells, known as CTLs or natural killer cells. While CTLs ignore antigens from human proteins (which indicate the cell is healthy), they can bind to and recognize antigens from viral proteins, which triggers them to activate immune responses that kill the infected cell. However, some viruses can prevent infected cells from presenting HLA-I molecules on their surfaces as a strategy to evade the immune system. Natural killer cells have evolved to overcome this challenge. They bind to the HLA-I molecules themselves, which causes them to remain inactive. However, if the HLA-I molecules are missing, the NK cells can more easily switch on and kill the target cell. The human cytomegalovirus is a common virus that causes lifelong infection in humans. Although it rarely causes illness in healthy individuals, it can be life-threatening to newborn babies and for individuals with weakened immune systems. One human cytomegalovirus protein known as US10 was previously found to bind to HLA-I without reducing the levels of these molecules on the surface of the cell. However, its precise role remained unclear. Gerke et al. used several biochemical and cell biology approaches to investigate whether US10 manipulates the quality of the three types of HLA-I, which could impact both CTL and NK cell recognition. The experiments showed that US10 acted differently on the various kinds of HLA-I. To one type, it bound strongly within the cell and prevented it from reaching the surface. US10 also prevented another type of HLA-I from maturing properly and presenting antigens but did not affect the third type of HLA-I. These findings suggest that US10 interferes with the ability of different HLA-I types to present antigens in specific ways. Further research is needed to measure how US10 activity affects immune cells, which may ultimately aid the development of new therapies against human cytomegalovirus and other similar viruses.


Subject(s)
Cytomegalovirus , Histocompatibility Antigens Class I , Humans , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Genotype , Viral Proteins/genetics , Viral Proteins/metabolism , Protein Binding , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Gene Expression Regulation , Antigen Presentation/genetics
12.
Rev Med Virol ; 34(4): e2560, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38866595

ABSTRACT

Immunosenescence (IS) occurs as a natural outcome of ageing and may be described as a decline in immune system flexibility and adaptability to sufficiently respond to new, foreign antigens. Potential factors that may precipitate IS include persistent herpesvirus infections, such as cytomegalovirus (CMV). Here, we conducted a review of the literature evaluating the potential association between CMV and IS. Twenty-seven epidemiologic studies that included direct comparisons between CMV-seropositive and CMV-seronegative immunocompetent individuals were analysed. The majority of these studies (n = 20) were conducted in European populations. The strength of evidence supporting a relationship between CMV, and various IS-associated immunologic endpoints was assessed. T-cell population restructuring was the most prominently studied endpoint, described in 21 studies, most of which reported a relationship between CMV and reduced CD4:CD8 T-cell ratio or modified CD8+ T-cell levels. Telomere length (n = 4) and inflammageing (n = 3) were less frequently described in the primary literature, and the association of these endpoints with CMV and IS was less pronounced. An emergent trend from our review is the potential effect modification of the CMV-IS relationship with both sex and age, indicating the importance of considering various effector variables when evaluating associations between CMV and IS. Our analysis revealed plausible mechanisms that may underlie the larger epidemiologic trends seen in the literature that support the indirect effect of CMV on IS. Future studies are needed to clarify CMV-associated and IS-associated immunologic endpoints, as well as in more diverse global and immunocompromised populations.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Immunosenescence , Humans , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus/immunology , Observational Studies as Topic
13.
Rev Med Virol ; 34(4): e2558, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38878003

ABSTRACT

Cytomegalovirus (CMV) infection poses significant risks in allogeneic haematopoietic stem cell transplant (allo-HSCT) recipients. Despite advances in antiviral therapies, issues such as drug resistance, side effects, and inadequate immune reconstitution remain. This systematic review and meta-analysis aim to evaluate the efficacy and safety of adoptive cell therapy (ATC) in managing CMV infections in allo-HSCT recipients. Adhering to preferred reporting items for systematic reviews and meta-analyses guidelines, we conducted a comprehensive database search through July 2023. A systematic review and meta-analysis were conducted on studies involving HSCT patients with CMV infections treated with ATC. The primary outcome was the response rate to ATC, and secondary outcomes included adverse events associated with ATC. The Freeman-Tukey transformation was applied for analysis. In the meta-analysis of 40 studies involving 953 participants, ATC achieved an overall integrated response rate of 90.16%, with a complete response of 82.59% and a partial response of 22.95%. ATC source, HLA matching, steroid intake, and age group markedly influenced response rates. Donor-derived T-cell treatments exhibited a higher response rate (93.66%) compared to third-party sources (88.94%). HLA-matched patients demonstrated a response rate of 92.90%, while mismatched patients had a lower rate. Children showed a response rate of 83.40%, while adults had a notably higher rate of 98.46%. Adverse events were minimal, with graft-versus-host disease occurring in 24.32% of patients. ATC shows promising response rates in treating CMV infections post-HSCT, with an acceptable safety profile. However, to establish its efficacy conclusively and compare it with other antiviral treatments, randomised controlled trials are essential. Further research should prioritise such trials over observational and one-arm studies to provide robust evidence for clinical decision-making.


Subject(s)
Cytomegalovirus Infections , Hematopoietic Stem Cell Transplantation , T-Lymphocytes , Humans , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/therapy , Cytomegalovirus Infections/virology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocytes/immunology , Treatment Outcome , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Cytomegalovirus/immunology , Transplantation, Homologous/adverse effects
14.
HLA ; 103(6): e15541, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923358

ABSTRACT

Complications due to HCMV infection or reactivation remain a challenging clinical problem in immunocompromised patients, mainly due to insufficient or absent T-cell functionality. Knowledge of viral targets is crucial to improve monitoring of high-risk patients and optimise antiviral T-cell therapy. To expand the epitope spectrum, genetically-engineered dendritic cells (DCs) and fibroblasts were designed to secrete soluble (s)HLA-A*11:01 and infected with an HCMV mutant lacking immune evasion molecules (US2-6 + 11). More than 700 HLA-A*11:01-restricted epitopes, including more than 50 epitopes derived from a broad range of HCMV open-reading-frames (ORFs) were identified by mass spectrometry and screened for HLA-A*11:01-binding using established prediction tools. The immunogenicity of the 24 highest scoring new candidates was evaluated in vitro in healthy HLA-A*11:01+/HCMV+ donors. Thus, four subdominant epitopes and one immunodominant epitope, derived from the anti-apoptotic protein UL36 and ORFL101C (A11SAL), were identified. Their HLA-A*11:01 complex stability was verified in vitro. In depth analyses revealed highly proliferative and cytotoxic memory T-cell responses against A11SAL, with T-cell responses comparable to the immunodominant HLA-A*02:01-restricted HCMVpp65NLV epitope. A11SAL-specific T cells were also detectable in vivo in immunosuppressed transplant patients and shown to be effective in an in vitro HCMV-infection model, suggesting their crucial role in inhibiting viral replication and improvement of patient's outcome. The developed in vitro pipeline is the first to utilise genetically-engineered DCs to identify naturally presented immunodominant HCMV-derived epitopes. It therefore offers advantages over in silico predictions, is transferable to other HLA alleles, and will significantly expand the repertoire of viral targets to improve therapeutic options.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Dendritic Cells , Epitopes, T-Lymphocyte , Immunodominant Epitopes , Humans , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Immunodominant Epitopes/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A11 Antigen/immunology , HLA-A11 Antigen/genetics , Fibroblasts/immunology , Fibroblasts/virology , Antigen-Presenting Cells/immunology
15.
Front Immunol ; 15: 1397483, 2024.
Article in English | MEDLINE | ID: mdl-38915409

ABSTRACT

Background: Cytomegalovirus (CMV) reactivation is a significant concern following allogeneic stem cell transplantation. While previous research has highlighted the anti-CMV reactivation effect of γδ T cells in immunocompromised transplant patients, their characterization in recipients at high risk of CMV reactivation remains limited. Methods: This study focused on D+/R+ recipients (where both donor and recipient are CMV seropositive) at high risk of CMV reactivation. We analyzed 28 patients who experienced CMV recurrence within 100 days post-allogeneic hematopoietic stem cell transplantation, along with 36 matched recipients who did not experience CMV recurrence. Clinical data from both groups were compared, and risk factors for CMV reactivation were identified. Additionally, CMV viral load was measured, and flow cytometric analysis was conducted to assess changes in peripheral blood γδ T cell proportions, subpopulation distribution, and differentiation status. We also analyzed the CDR3 repertoire of the TCR δ chain in different γδ T cell subsets. Functional analysis was performed by measuring the lysis of CMV-infected cells upon stimulation. Results: CMV reactivation post-transplantation was associated with acute graft-versus-host disease (aGvHD) and reactivation of non-CMV herpesviruses. Notably, CMV reactivation led to sustained expansion of γδ T cells, primarily within the Vδ2neg γδ T cell subpopulation, with a trend toward differentiation from Naive to effector memory cells. Analysis of the δ chain CDR3 repertoire revealed a delay in the reconstitution of clonal diversity in Vδ2neg γδ T cells following CMV reactivation, while Vδ2pos T cells remained unaffected. Upon stimulation with CMV-infected MRC5 cells, the Vδ2neg γδ T cell subpopulation emerged as the primary effector cell group producing IFN-γ and capable of lysing CMV-infected cells. Moreover, our findings suggest that NKG2D is not necessary involved in Vδ2neg γδ T cell-mediated anti-CMV cytotoxicity. Conclusion: This study provides novel insights into the role of γδ T cells in the immune response to CMV reactivation in transplantation recipients at high risk of CMV infection. Specifically, the Vδ2neg γδ T cell subpopulation appears to be closely associated with CMV reactivation, underscoring their potential role in controlling infection and reflecting CMV reactivation in HSCT patients.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Hematopoietic Stem Cell Transplantation , Receptors, Antigen, T-Cell, gamma-delta , Transplantation, Homologous , Virus Activation , Humans , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Male , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Virus Activation/immunology , Female , Adult , Middle Aged , Hematopoietic Stem Cell Transplantation/adverse effects , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Graft vs Host Disease/immunology , Young Adult , Memory T Cells/immunology , Aged
16.
Antiviral Res ; 227: 105914, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38759930

ABSTRACT

Due to the severity of CMV infection in immunocompromised individuals the development of a vaccine has been declared a priority. However, despite the efforts made there is no yet a vaccine available for clinical use. We designed an approach to identify new CMV antigens able to inducing a broad immune response that could be used in future vaccine formulations. We have used serum samples from 28 kidney transplant recipients, with a previously acquired CMV-specific immune response to identify viral proteins that were recognized by the antibodies present in the patient serum samples by Western blot. A band of approximately 45 kDa, identified as UL44, was detected by most serum samples. UL44 immunogenicity was tested in BALB/c mice that received three doses of the UL44-pcDNA DNA vaccine. UL44 elicited both, a strong antibody response and CMV-specific cellular response. Using bioinformatic analysis we demonstrated that UL44 is a highly conserved protein and contains epitopes that are able to activate CD8 lymphocytes of the most common HLA alleles in the world population. We constructed a UL44 ORF deletion mutant virus that produced no viral progeny, suggesting that UL44 is an essential viral protein. In addition, other authors have demonstrated that UL44 is one of the most abundant viral proteins after infection and have suggested an essential role of UL44 in viral replication. Altogether, our data suggests that UL44 is a potent antigen, and favored by its abundance, it may be a good candidate to include in a vaccine formulation.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Mice, Inbred BALB C , Viral Proteins , Animals , Mice , Humans , Cytomegalovirus/immunology , Cytomegalovirus/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Viral Proteins/immunology , Viral Proteins/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Female , Cytomegalovirus Vaccines/immunology , Cytomegalovirus Vaccines/administration & dosage , T-Lymphocytes/immunology , Antigens, Viral/immunology , Kidney Transplantation , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular
17.
Cytokine ; 180: 156654, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38810501

ABSTRACT

Persistent infections with human cytomegalovirus (HCMV) affect the hosts' immune system and have been linked with chronic inflammation and cardiovascular disease. These effects may be influenced by a HCMV-encoded homologue of the anti-inflammatory cytokine, IL-10 (cmvIL-10). To assess this, we quantitated cmvIL-10 in plasma from renal transplant recipients (RTR) and healthy adults. Detectable levels of cmvIL-10 associated with seropositivity in RTR, but were found in some seronegative healthy adults. RTR with detectable cmvIL-10 had elevated interferon-γ T-cell responses to HCMV antigens, whilst cmvIL-10 in healthy adults associated with reduced populations of terminally-differentiated T-cells - a known "footprint" of HCMV. Plasma cmvIL-10 associated with lower VCAM-1 levels in healthy adults. The data suggest cmvIL-10 may suppress seroconversion and/or reduce the footprint of HCMV in healthy adults. This appears to be subverted in RTR by their high burden of HCMV and/or immune dysregulation associated with transplantation. A role for cmvIL-10 in protection of vascular health is discussed.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Interleukin-10 , Kidney Transplantation , Vascular Cell Adhesion Molecule-1 , Humans , Cytomegalovirus/immunology , Adult , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Male , Middle Aged , Female , Interferon-gamma/metabolism , T-Lymphocytes/immunology , Aged
18.
J Surg Res ; 299: 129-136, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754251

ABSTRACT

INTRODUCTION: Cytomegalovirus (CMV) infection is associated with a poor prognosis after lung transplantation, and donor and recipient CMV serostatus is a risk factor for reactivation. CMV prophylaxis is commonly administered in the first year following transplantation to reduce CMV infection; however, the risk factors for long-term reactivation remain unclear. We investigated the timing and risk factors of CMV infection after prophylactic administration. METHODS: This study was a retrospective review of the institutional lung transplantation database from June 2014 to June 2022. Data on patient characteristics, pretransplantation laboratory values, postoperative outcomes, and CMV infection were collected. Donor CMV-IgG-positive and recipient CMV-IgG-negative groups were defined as the CMV mismatch group. RESULTS: During the study period, 257 patients underwent lung transplantation and received a prophylactic dose of valganciclovir hydrochloride for up to 1 y. CMV infection was detected in 69 patients (26.8%): 40 of 203 (19.7%) in the non-CMV mismatch group and 29 of 54 (53.7%) in the CMV mismatch group (P < 0.001). CMV infection after prophylaxis occurred at a median of 425 and 455 d in the CMV mismatch and non-CMV mismatch groups, respectively (P = 0.07). Multivariate logistic regression analysis revealed that preoperative albumin level (odds ratio [OR] = 0.39, P = 0.04), CMV mismatch (OR = 15.7, P < 0.001), and donor age (OR = 1.05, P = 0.009) were significantly associated with CMV infection. CONCLUSIONS: CMV mismatch may have increased the risk of CMV infection after lung transplantation, which decreased after prophylaxis. In addition to CMV mismatch, low preoperative albumin level and donor age were independent predictors of CMV infection.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Lung Transplantation , Humans , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/diagnosis , Male , Female , Retrospective Studies , Middle Aged , Lung Transplantation/adverse effects , Adult , Risk Factors , Antiviral Agents/therapeutic use , Antiviral Agents/administration & dosage , Recurrence , Valganciclovir/therapeutic use , Valganciclovir/administration & dosage , Aged , Cytomegalovirus/immunology , Cytomegalovirus/isolation & purification , Postoperative Complications/prevention & control , Postoperative Complications/epidemiology
19.
PLoS Pathog ; 20(5): e1012058, 2024 May.
Article in English | MEDLINE | ID: mdl-38768227

ABSTRACT

Viral disruption of innate immune signaling is a critical determinant of productive infection. The Human Cytomegalovirus (HCMV) UL26 protein prevents anti-viral gene expression during infection, yet the mechanisms involved are unclear. We used TurboID-driven proximity proteomics to identify putative UL26 interacting proteins during infection to address this issue. We find that UL26 forms a complex with several immuno-regulatory proteins, including several STAT family members and various PIAS proteins, a family of E3 SUMO ligases. Our results indicate that UL26 prevents STAT phosphorylation during infection and antagonizes transcriptional activation induced by either interferon α (IFNA) or tumor necrosis factor α (TNFα). Additionally, we find that the inactivation of PIAS1 sensitizes cells to inflammatory stimulation, resulting in an anti-viral transcriptional environment similar to ΔUL26 infection. Further, PIAS1 is important for HCMV cell-to-cell spread, which depends on the presence of UL26, suggesting that the UL26-PIAS1 interaction is vital for modulating intrinsic anti-viral defense.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Protein Inhibitors of Activated STAT , Viral Proteins , Humans , Cytomegalovirus/immunology , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Gene Expression Regulation, Viral , Immunity, Innate
20.
Transpl Infect Dis ; 26(3): e14290, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38708941

ABSTRACT

BACKGROUND: Cytomegalovirus-specific T-cell-mediated immunity (CMV-CMI) protects from CMV infection in allogeneic hematopoietic cell transplantation (allo-HCT), but to date, there is no validated measure of CMV immunity for this population. METHODS: In this prospective, observational, pilot study, CMV T-cell responses were evaluated monthly and at onset of graft-versus-host disease (GVHD) or CMV infection in CMV-seropositive allo-HCT recipients using a commercial flow cytometry assay, the CMV inSIGHT T-Cell Immunity Panel (CMV-TCIP). The primary endpoint was the time to first positive CMV-TCIP, defined as percentage of interferon-γ-producing CD4+ or CD8+ CMV-specific T cells >0.2%. Letermovir was prescribed from day +10 to ≥100. RESULTS: Twenty-eight allo-HCT recipients were enrolled. The median time to first positive CMV-TCIP result was earlier for CD4+ (60 days [interquartile range, IQR 33‒148]) than for CD8+ T cells (96 days [IQR 33‒155]) and longer for haploidentical and mismatched transplant recipients (77 and 96 days, respectively) than for matched donors (45 and 33 days, respectively). CD4+ and CD8+ CMV-CMI recovery was sustained in 10/10 (100%) and 10/11 (91%) patients, respectively, without GVHD, whereas CD4+ and/or CD8+ CMV-CMI was lost in 4/6 and 2/6 patients, respectively, with GVHD requiring steroids. As a predictor of clinically significant CMV infection in patients with low-level CMV reactivation, the sensitivity and negative predictive value of CMV-TCIP were 90% and 87.5%, respectively, for CD4+ CMV-TCIP and 66.7% and 62.5%, respectively, for CD8+ CMV-TCIP. CONCLUSIONS: There was significant variability in time to CMV-CMI recovery post-HCT, with slower recovery after haploidentical and mismatched HCT. CD4+ CMV-CMI may protect against CS-CMVi, but immunity may be lost with GVHD diagnosis and treatment.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cytomegalovirus Infections , Cytomegalovirus , Flow Cytometry , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Transplantation, Homologous , Humans , Pilot Projects , Hematopoietic Stem Cell Transplantation/adverse effects , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/prevention & control , Middle Aged , Male , Cytomegalovirus/immunology , Prospective Studies , Female , Flow Cytometry/methods , Adult , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Transplantation, Homologous/adverse effects , Aged , Immunity, Cellular , Antiviral Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...