Cytomegalovirus Immunity 2021

A special issue of Viruses (ISSN 1999-4915). This special issue belongs to the section "Viral Immunology, Vaccines, and Antivirals".

Deadline for manuscript submissions: closed (15 December 2021) | Viewed by 35693

Special Issue Editor


E-Mail Website
Guest Editor
CHU Ste-Justine Research Centre, Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, QC H3T 1C5, Canada
Interests: cytomegalovirus (CMV); congenital infection; viral genomics; immunology; vaccine development; mathematical modeling

Special Issue Information

Dear Colleagues,

Cytomegalovirus (CMV) infects most of the world’s population, and has evolved to persist for the life of the infected host. Although most immunocompetent individuals do not suffer overtly from CMV infection, an enormous burden of morbidity and mortality occurs in congenitally infected children and in people of all ages with impaired immunity, in whom viral replication is poorly controlled. Innate, humoral, and cellular immune responses have all been found to contribute to the control of CMV infection. Importantly, however, natural immunity to CMV is incomplete and does not prevent reinfection or transmission from mother to fetus. Moreover, evidence indicates that CMV infection can modulate immune responses to other infections and to vaccination, and may contribute to immune senescence in the elderly. A better understanding of immune correlates of protection against CMV infection, disease, and vertical transmission are needed, with the ultimate goal of developing an effective CMV vaccine. Advances in the field of CMV immunology of particular interest include natural killer cell memory, adoptive virus-specific T-cell therapies, memory T-cell inflation, and the role of strain-specific antibodies. This Issue will explore novel aspects of CMV immunology and their impacts on the control of infection and disease.

Dr. Soren Gantt
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Viruses is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cytomegalovirus
  • immunology
  • NK cell
  • T cell
  • antibody
  • memory
  • congenital infection
  • reinfection
  • immune modulation
  • transplantation
  • vaccine

Published Papers (13 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

17 pages, 3475 KiB  
Article
Inclusion of the Guinea Pig Cytomegalovirus Pentameric Complex in a Live Virus Vaccine Aids Efficacy against Congenital Infection but Is Not Essential for Improving Maternal and Neonatal Outcomes
by Mark R. Schleiss, Claudia Fernández-Alarcón, Nelmary Hernandez-Alvarado, Jian Ben Wang, Adam P. Geballe and Michael A. McVoy
Viruses 2021, 13(12), 2370; https://doi.org/10.3390/v13122370 - 26 Nov 2021
Cited by 4 | Viewed by 1813
Abstract
The development of a vaccine against congenital human cytomegalovirus (HCMV) infection is a major priority. The pentameric complex (PC) of virion envelope proteins gH, gL, UL128, UL130, and UL131A is a key vaccine target. To determine the importance of immunity to the homologous [...] Read more.
The development of a vaccine against congenital human cytomegalovirus (HCMV) infection is a major priority. The pentameric complex (PC) of virion envelope proteins gH, gL, UL128, UL130, and UL131A is a key vaccine target. To determine the importance of immunity to the homologous PC encoded by guinea pig cytomegalovirus (GPCMV) in preventing congenital CMV, PC-intact and PC-deficient live-attenuated vaccines were generated and directly compared for immunogenicity and efficacy against vertical transmission in a vertical transmission model. A virulent PC-intact GPCMV (PC/intact) was modified by galK mutagenesis either to abrogate PC expression (PC/null; containing a frame-shift mutation in GP129, homolog of UL128) or to delete genes encoding three MHC Class I homologs and a protein kinase R (PKR) evasin while retaining the PC (3DX/Δ145). Attenuated vaccines were compared to sham immunization in a two-dose preconception subcutaneous inoculation regimen in GPCMV seronegative Hartley guinea pigs. Vaccines induced transient, low-grade viremia in 5/12 PC/intact-, 2/12 PC/null-, and 1/11 3DX/Δ145-vaccinated animals. Upon completion of the two-dose vaccine series, ELISA titers for the PC/intact group (geometic mean titer (GMT) 13,669) were not significantly different from PC/null (GMT 8127) but were significantly higher than for the 3DX/Δ145 group (GMT 6185; p < 0.01). Dams were challenged with salivary gland-adapted GPCMV in the second trimester. All vaccines conferred protection against maternal viremia. Newborn weights were significantly lower in sham-immunized controls (84.5 ± 2.4 g) compared to PC/intact (96 ± 2.3 g), PC/null (97.6 ± 1.9 g), or 3DX/Δ145 (93 ± 1.7) pups (p < 0.01). Pup mortality in sham-immunized controls was 29/40 (73%) and decreased to 1/44 (2.3%), 2/46 (4.3%), or 4/40 (10%) in PC/intact, PC/null, or 3DX/Δ145 groups, respectively (all p < 0.001 compared to control). Congenital GPCMV transmission occurred in 5/44 (11%), 16/46 (35%), or 29/38 (76%) of pups in PC/intact, PC/null, or 3DX/Δ145 groups, versus 36/40 (90%) in controls. For infected pups, viral loads were lower in pups born to vaccinated dams compared to controls. Sequence analysis demonstrated that infected pups in the vaccine groups had salivary gland-adapted GPCMV and not vaccine strain-specific sequences, indicating that congenital transmission was due to the challenge virus and not vaccine virus. We conclude that inclusion of the PC in a live, attenuated preconception vaccine improves immunogenicity and reduces vertical transmission, but PC-null vaccines are equal to PC-intact vaccines in reducing maternal viremia and protecting against GPCMV-related pup mortality. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

14 pages, 2461 KiB  
Article
Cytomegalovirus Immunity, Inflammation and Cognitive Abilities in the Elderly
by Jacqueline Hesson, Neva Fudge and Michael Grant
Viruses 2021, 13(11), 2321; https://doi.org/10.3390/v13112321 - 21 Nov 2021
Viewed by 1859
Abstract
Reducing the socioeconomic toll from age-related physical and mental morbidities requires better understanding of factors affecting healthy aging. While many environmental, lifestyle, and genetic factors affect healthy aging, this study addressed the influence of cytomegalovirus (CMV) infection and immunity on age-related inflammation and [...] Read more.
Reducing the socioeconomic toll from age-related physical and mental morbidities requires better understanding of factors affecting healthy aging. While many environmental, lifestyle, and genetic factors affect healthy aging, this study addressed the influence of cytomegalovirus (CMV) infection and immunity on age-related inflammation and cognitive abilities. Healthy adults 70–90 years old were recruited into a prospective study investigating relationships between anti-CMV immunity, markers of inflammation, baseline measures of cognitive ability, and changes in cognitive ability over 18 months. Humoral and cellular responses against CMV, levels of inflammatory markers, and cognitive abilities were measured at study entry, with measurement of cognitive abilities repeated 18 months later. CMV-seropositive and -seronegative sub-groups were compared, and relationships between anti-CMV immunity, markers of inflammation, and cognitive ability were assessed. Twenty-eight of 39 participants were CMV-seropositive, and two had CMV-specific CD8+ T cell responses indicative of CMV immune memory inflation. No significant differences for markers of inflammation or measures of cognitive ability were observed between groups, and cognitive scores changed little over 18 months. Significant correlations between markers of inflammation and cognitive scores with interconnection between anti-CMV antibody levels, fractalkine, cognitive ability, and depression scores suggest areas of focus for future studies. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

22 pages, 6307 KiB  
Article
Characterization of Adaptive-like γδ T Cells in Ugandan Infants during Primary Cytomegalovirus Infection
by Jessica Tuengel, Sanya Ranchal, Alexandra Maslova, Gurpreet Aulakh, Maria Papadopoulou, Sibyl Drissler, Bing Cai, Cetare Mohsenzadeh-Green, Hugo Soudeyns, Sara Mostafavi, Peter van den Elzen, David Vermijlen, Laura Cook and Soren Gantt
Viruses 2021, 13(10), 1987; https://doi.org/10.3390/v13101987 - 03 Oct 2021
Cited by 7 | Viewed by 3381
Abstract
Gamma-delta (γδ) T cells are unconventional T cells that help control cytomegalovirus (CMV) infection in adults. γδ T cells develop early in gestation, and a fetal public γδ T cell receptor (TCR) clonotype is detected in congenital CMV infections. However, age-dependent γδ T [...] Read more.
Gamma-delta (γδ) T cells are unconventional T cells that help control cytomegalovirus (CMV) infection in adults. γδ T cells develop early in gestation, and a fetal public γδ T cell receptor (TCR) clonotype is detected in congenital CMV infections. However, age-dependent γδ T cell responses to primary CMV infection are not well-understood. Flow cytometry and TCR sequencing was used to comprehensively characterize γδ T cell responses to CMV infection in a cohort of 32 infants followed prospectively from birth. Peripheral blood γδ T cell frequencies increased during infancy, and were higher among CMV-infected infants relative to uninfected. Clustering analyses revealed associations between CMV infection and activation marker expression on adaptive-like Vδ1 and Vδ3, but not innate-like Vγ9Vδ2 γδ T cell subsets. Frequencies of NKG2C+CD57+ γδ T cells were temporally associated with the quantity of CMV shed in saliva by infants with primary infection. The public γδ TCR clonotype was only detected in CMV-infected infants <120 days old and at lower frequencies than previously described in fetal infections. Our findings support the notion that CMV infection drives age-dependent expansions of specific γδ T cell populations, and provide insight for novel strategies to prevent CMV transmission and disease. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

24 pages, 3045 KiB  
Article
Multiple Autonomous Cell Death Suppression Strategies Ensure Cytomegalovirus Fitness
by Pratyusha Mandal, Lynsey N. Nagrani, Liliana Hernandez, Anita Louise McCormick, Christopher P. Dillon, Heather S. Koehler, Linda Roback, Emad S. Alnemri, Douglas R. Green and Edward S. Mocarski
Viruses 2021, 13(9), 1707; https://doi.org/10.3390/v13091707 - 27 Aug 2021
Cited by 6 | Viewed by 3368
Abstract
Programmed cell death pathways eliminate infected cells and regulate infection-associated inflammation during pathogen invasion. Cytomegaloviruses encode several distinct suppressors that block intrinsic apoptosis, extrinsic apoptosis, and necroptosis, pathways that impact pathogenesis of this ubiquitous herpesvirus. Here, we expanded the understanding of three cell [...] Read more.
Programmed cell death pathways eliminate infected cells and regulate infection-associated inflammation during pathogen invasion. Cytomegaloviruses encode several distinct suppressors that block intrinsic apoptosis, extrinsic apoptosis, and necroptosis, pathways that impact pathogenesis of this ubiquitous herpesvirus. Here, we expanded the understanding of three cell autonomous suppression mechanisms on which murine cytomegalovirus relies: (i) M38.5-encoded viral mitochondrial inhibitor of apoptosis (vMIA), a BAX suppressor that functions in concert with M41.1-encoded viral inhibitor of BAK oligomerization (vIBO), (ii) M36-encoded viral inhibitor of caspase-8 activation (vICA), and (iii) M45-encoded viral inhibitor of RIP/RHIM activation (vIRA). Following infection of bone marrow-derived macrophages, the virus initially deflected receptor-interacting protein kinase (RIPK)3-dependent necroptosis, the most potent of the three cell death pathways. This process remained independent of caspase-8, although suppression of this apoptotic protease enhances necroptosis in most cell types. Second, the virus deflected TNF-mediated extrinsic apoptosis, a pathway dependent on autocrine TNF production by macrophages that proceeds independently of mitochondrial death machinery or RIPK3. Third, cytomegalovirus deflected BCL-2 family protein-dependent mitochondrial cell death through combined TNF-dependent and -independent signaling even in the absence of RIPK1, RIPK3, and caspase-8. Furthermore, each of these cell death pathways dictated a distinct pattern of cytokine and chemokine activation. Therefore, cytomegalovirus employs sequential, non-redundant suppression strategies to specifically modulate the timing and execution of necroptosis, extrinsic apoptosis, and intrinsic apoptosis within infected cells to orchestrate virus control and infection-dependent inflammation. Virus-encoded death suppressors together hold control over an intricate network that upends host defense and supports pathogenesis in the intact mammalian host. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

15 pages, 669 KiB  
Article
Cytomegalovirus Infection Impairs the Mobilization of Tissue-Resident Innate Lymphoid Cells into the Peripheral Blood Compartment in Response to Acute Exercise
by Eunhan Cho, Bailey Theall, James Stampley, Joshua Granger, Neil M. Johannsen, Brian A. Irving and Guillaume Spielmann
Viruses 2021, 13(8), 1535; https://doi.org/10.3390/v13081535 - 03 Aug 2021
Cited by 1 | Viewed by 1875
Abstract
Circulating immune cell numbers and phenotypes are impacted by high-intensity acute bouts of exercise and infection history with the latent herpesviruses cytomegalovirus (CMV). In particular, CMV infection history impairs the exercise-induced mobilization of cytotoxic innate lymphoid cells 1 (ILC1) cells, also known as [...] Read more.
Circulating immune cell numbers and phenotypes are impacted by high-intensity acute bouts of exercise and infection history with the latent herpesviruses cytomegalovirus (CMV). In particular, CMV infection history impairs the exercise-induced mobilization of cytotoxic innate lymphoid cells 1 (ILC1) cells, also known as NK cells, in the blood. However, it remains unknown whether exercise and CMV infection modulate the mobilization of traditionally tissue-resident non-cytotoxic ILCs into the peripheral blood compartment. To address this question, 22 healthy individuals with or without CMV (20–35 years—45% CMVpos) completed 30 min of cycling at 70% VO2 max, and detailed phenotypic analysis of circulating ILCs was performed at rest and immediately post-exercise. We show for the first time that a bout of high-intensity exercise is associated with an influx of ILCs that are traditionally regarded as tissue-resident. In addition, this is the first study to highlight that latent CMV infection blunts the exercise-response of total ILCs and progenitor ILCs (ILCPs). These promising data suggest that acute exercise facilitates the circulation of certain ILC subsets, further advocating for the improvements in health seen with exercise by enhancing cellular mobilization and immunosurveillance, while also highlighting the indirect deleterious effects of CMV infection in healthy adults. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

20 pages, 2254 KiB  
Article
A Fully Protective Congenital CMV Vaccine Requires Neutralizing Antibodies to Viral Pentamer and gB Glycoprotein Complexes but a pp65 T-Cell Response Is Not Necessary
by K. Yeon Choi and Alistair McGregor
Viruses 2021, 13(8), 1467; https://doi.org/10.3390/v13081467 - 27 Jul 2021
Cited by 6 | Viewed by 3202
Abstract
A vaccine against congenital cytomegalovirus infection is a high priority. Guinea pig cytomegalovirus (GPCMV) is the only congenital CMV small animal model. GPCMV encodes essential glycoprotein complexes for virus entry (gB, gH/gL/gO, gM/gN) including a pentamer complex (gH/gL/GP129/GP131/GP133 or PC) for endocytic cell [...] Read more.
A vaccine against congenital cytomegalovirus infection is a high priority. Guinea pig cytomegalovirus (GPCMV) is the only congenital CMV small animal model. GPCMV encodes essential glycoprotein complexes for virus entry (gB, gH/gL/gO, gM/gN) including a pentamer complex (gH/gL/GP129/GP131/GP133 or PC) for endocytic cell entry. The cohorts for protection against congenital CMV are poorly defined. Neutralizing antibodies to the viral glycoprotein complexes are potentially more important than an immunodominant T-cell response to the pp65 protein. In GPCMV, GP83 (pp65 homolog) is an evasion factor, and the GP83 mutant GPCMV has increased sensitivity to type I interferon. Although GP83 induces a cell-mediated response, a GP83-only-based vaccine strategy has limited efficacy. GPCMV attenuation via GP83 null deletion mutant in glycoprotein PC positive or negative virus was evaluated as live-attenuated vaccine strains (GP83dPC+/PC-). Vaccinated animals induced antibodies to viral glycoprotein complexes, and PC+ vaccinated animals had sterilizing immunity against wtGPCMV challenge. In a pre-conception vaccine (GP83dPC+) study, dams challenged mid-2nd trimester with wtGPCMV had complete protection against congenital CMV infection without detectable virus in pups. An unvaccinated control group had 80% pup transmission rate. Overall, gB and PC antibodies are key for protection against congenital CMV infection, but a response to pp65 is not strictly necessary. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Graphical abstract

10 pages, 250 KiB  
Article
Low Maternal Immunoglobulin G Avidity and Single Parity as Adverse Implications of Human Cytomegalovirus Vertical Transmission in Pregnant Women with Immunoglobulin M Positivity
by Masatoki Kaneko, Junsuke Muraoka, Kazumi Kusumoto and Toshio Minematsu
Viruses 2021, 13(5), 866; https://doi.org/10.3390/v13050866 - 09 May 2021
Cited by 3 | Viewed by 1748
Abstract
Human cytomegalovirus (CMV) is the leading cause of neurological sequelae in infants. Understanding the risk factors of primary CMV infection is crucial in establishing preventive strategies. Thus, we conducted a retrospective cohort study to identify risk factors of vertical transmission among pregnant women [...] Read more.
Human cytomegalovirus (CMV) is the leading cause of neurological sequelae in infants. Understanding the risk factors of primary CMV infection is crucial in establishing preventive strategies. Thus, we conducted a retrospective cohort study to identify risk factors of vertical transmission among pregnant women with immunoglobulin (Ig) M positivity. The study included 456 pregnant women with IgM positivity. Information on age, parity, occupation, clinical signs, IgM levels, and IgG avidity index (AI) was collected. The women were divided into infected and non-infected groups. The two groups showed significant differences in IgM level, IgG AI, number of women with low IgG AI, clinical signs, and number of pregnant women with single parity. In the multiple logistic regression analysis, pregnant women with single parity and low IgG AI were independent predictors. Among 40 women who tested negative for IgG antibody in their previous pregnancy, 20 showed low IgG AI in their current pregnancy. Among the 20 women, 4 had vertical transmission. These results provide better understanding of the risk factors of vertical transmission in pregnant women with IgM positivity. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
11 pages, 888 KiB  
Article
CMV-Specific Cell-Mediated Immunity in Immunocompetent Adults with Primary CMV Infection: A Case Series and Review of the Literature
by Angela Chiereghin, Gabriella Verucchi and Tiziana Lazzarotto
Viruses 2021, 13(5), 816; https://doi.org/10.3390/v13050816 - 01 May 2021
Cited by 4 | Viewed by 1950
Abstract
Cytomegalovirus-specific cell-mediated immunity (CMV-CMI) in actively infected healthy immunocompetent hosts has been poorly investigated. Conversely, correlates of maternal protective immunity for the fetus after primary infection in pregnancy continue to be studied. The kinetics and magnitude of CMV-specific CMI in immunocompetent primary CMV-infected [...] Read more.
Cytomegalovirus-specific cell-mediated immunity (CMV-CMI) in actively infected healthy immunocompetent hosts has been poorly investigated. Conversely, correlates of maternal protective immunity for the fetus after primary infection in pregnancy continue to be studied. The kinetics and magnitude of CMV-specific CMI in immunocompetent primary CMV-infected adults are described. A literature review on CMV-CMI in primarily infected pregnant women and its correlation to the risk of vertical virus transmission is included. Immunological measurements after infection were performed by enzyme-linked ImmunoSPOT assay enumerating IFN-γ secreting CMV-specific T cells, at a single cell level, upon in vitro stimulation with viral antigens. Simultaneously, serological and virological profiles of infected patients were investigated. Patients displayed mild-to-moderate clinical and laboratory profiles for infection, and all showed positive EliSpot results in the early stage of infection (<20 days after onset). The virus-CMI was strong in the majority of patients (58.8%) in which the lowest CMV-DNAemia levels (<300 copies/mL) were detected. Significantly higher viral loads were observed in patients with weak CMV-CMI at the same time-point post-infection (up to 15,104 copies/mL; p < 0.001). T cell response magnitudes to IE-1 and pp65-UL83 peptides were overlapping and stable over time. In these case series, the early presence of CMV-CMI was probably pivotal in controlling viral replication and led to spontaneous viral clearance. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

16 pages, 5907 KiB  
Article
Detection of Genotype-Specific Antibody Responses to Glycoproteins B and H in Primary and Non-Primary Human Cytomegalovirus Infections by Peptide-Based ELISA
by Federica Zavaglio, Loretta Fiorina, Nicolás M. Suárez, Chiara Fornara, Marica De Cicco, Daniela Cirasola, Andrew J. Davison, Giuseppe Gerna and Daniele Lilleri
Viruses 2021, 13(3), 399; https://doi.org/10.3390/v13030399 - 03 Mar 2021
Cited by 4 | Viewed by 2006
Abstract
Background: Strain-specific antibodies to human cytomegalovirus (HCMV) glycoproteins B and H (gB and gH) have been proposed as a potential diagnostic tool for identifying reinfection. We investigated genotype-specific IgG antibody responses in parallel with defining the gB and gH genotypes of the infecting [...] Read more.
Background: Strain-specific antibodies to human cytomegalovirus (HCMV) glycoproteins B and H (gB and gH) have been proposed as a potential diagnostic tool for identifying reinfection. We investigated genotype-specific IgG antibody responses in parallel with defining the gB and gH genotypes of the infecting viral strains. Methods: Subjects with primary (n = 20) or non-primary (n = 25) HCMV infection were studied. The seven gB (gB1-7) and two gH (gH1-2) genotypes were determined by real-time PCR and whole viral genome sequencing, and genotype-specific IgG antibodies were measured by a peptide-based enzyme-linked immunosorbent assay (ELISA). Results: Among subjects with primary infection, 73% (n = 8) infected by gB1-HCMV and 63% (n = 5) infected by gB2/3-HCMV had genotype-specific IgG antibodies to gB (gB2 and gB3 are similar in the region tested). Peptides from the rarer gB4-gB7 genotypes had nonspecific antibody responses. All subjects infected by gH1-HCMV and 86% (n = 6) infected by gH2-HCMV developed genotype-specific responses. Among women with non-primary infection, gB and gH genotype-specific IgG antibodies were detected in 40% (n = 10) and 80% (n = 20) of subjects, respectively. Conclusions: Peptide-based ELISA is capable of detecting primary genotype-specific IgG responses to HCMV gB and gH, and could be adopted for identifying reinfections. However, about half of the subjects did not have genotype-specific IgG antibodies to gB. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

Review

Jump to: Research

14 pages, 300 KiB  
Review
Cytomegalovirus Infections in Children with Primary and Secondary Immune Deficiencies
by Caroline M. Bateman, Alison Kesson, Madeleine Powys, Melanie Wong and Emily Blyth
Viruses 2021, 13(10), 2001; https://doi.org/10.3390/v13102001 - 05 Oct 2021
Cited by 14 | Viewed by 3571
Abstract
Cytomegalovirus (CMV) is a human herpes virus that causes significant morbidity and mortality in immunosuppressed children. CMV primary infection causes a clinically mild disease in healthy children, usually in early childhood; the virus then utilises several mechanisms to establish host latency, which allows [...] Read more.
Cytomegalovirus (CMV) is a human herpes virus that causes significant morbidity and mortality in immunosuppressed children. CMV primary infection causes a clinically mild disease in healthy children, usually in early childhood; the virus then utilises several mechanisms to establish host latency, which allows for periodic reactivation, particularly when the host is immunocompromised. It is this reactivation that is responsible for the significant morbidity and mortality in immunocompromised children. We review CMV infection in the primary immunodeficient host, including early identification of these infants by newborn screening to allow for CMV infection prevention strategies. Furthermore, clinical CMV is discussed in the context of children treated with secondary immunodeficiency, particularly paediatric cancer patients and children undergoing haematopoietic stem cell transplant (HSCT). Treatments for CMV are highlighted and include CMV immunotherapy. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
10 pages, 2482 KiB  
Review
Consequence of Histoincompatibility beyond GvH-Reaction in Cytomegalovirus Disease Associated with Allogeneic Hematopoietic Cell Transplantation: Change of Paradigm
by Matthias J. Reddehase, Rafaela Holtappels and Niels A. W. Lemmermann
Viruses 2021, 13(8), 1530; https://doi.org/10.3390/v13081530 - 03 Aug 2021
Cited by 10 | Viewed by 2570
Abstract
Hematopoietic cell (HC) transplantation (HCT) is the last resort to cure hematopoietic malignancies that are refractory to standard therapies. Hematoablative treatment aims at wiping out tumor cells as completely as possible to avoid leukemia/lymphoma relapse. This treatment inevitably co-depletes cells of hematopoietic cell [...] Read more.
Hematopoietic cell (HC) transplantation (HCT) is the last resort to cure hematopoietic malignancies that are refractory to standard therapies. Hematoablative treatment aims at wiping out tumor cells as completely as possible to avoid leukemia/lymphoma relapse. This treatment inevitably co-depletes cells of hematopoietic cell lineages, including differentiated cells that constitute the immune system. HCT reconstitutes hematopoiesis and thus, eventually, also antiviral effector cells. In cases of an unrelated donor, that is, in allogeneic HCT, HLA-matching is performed to minimize the risk of graft-versus-host reaction and disease (GvHR/D), but a mismatch in minor histocompatibility antigens (minor HAg) is unavoidable. The transient immunodeficiency in the period between hematoablative treatment and reconstitution by HCT gives latent cytomegalovirus (CMV) the chance to reactivate from latently infected donor HC or from latently infected organs of the recipient, or from both. Clinical experience shows that HLA and/or minor-HAg mismatches increase the risk of complications from CMV. Recent results challenge the widespread, though never proven, view of a mechanistic link between GvHR/D and CMV. Instead, new evidence suggests that histoincompatibility promotes CMV disease by inducing non-cognate transplantation tolerance that inhibits an efficient reconstitution of high-avidity CD8+ T cells capable of recognizing and resolving cytopathogenic tissue infection. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

18 pages, 3931 KiB  
Review
Common Polymorphisms in the Glycoproteins of Human Cytomegalovirus and Associated Strain-Specific Immunity
by Hsuan-Yuan Wang, Sarah M. Valencia, Susanne P. Pfeifer, Jeffrey D. Jensen, Timothy F. Kowalik and Sallie R. Permar
Viruses 2021, 13(6), 1106; https://doi.org/10.3390/v13061106 - 09 Jun 2021
Cited by 16 | Viewed by 3241
Abstract
Human cytomegalovirus (HCMV), one of the most prevalent viruses across the globe, is a common cause of morbidity and mortality for immunocompromised individuals. Recent clinical observations have demonstrated that mixed strain infections are common and may lead to more severe disease progression. This [...] Read more.
Human cytomegalovirus (HCMV), one of the most prevalent viruses across the globe, is a common cause of morbidity and mortality for immunocompromised individuals. Recent clinical observations have demonstrated that mixed strain infections are common and may lead to more severe disease progression. This clinical observation illustrates the complexity of the HCMV genome and emphasizes the importance of taking a population-level view of genotypic evolution. Here we review frequently sampled polymorphisms in the glycoproteins of HCMV, comparing the variable regions, and summarizing their corresponding geographic distributions observed to date. The related strain-specific immunity, including neutralization activity and antigen-specific cellular immunity, is also discussed. Given that these glycoproteins are common targets for vaccine design and anti-viral therapies, this observed genetic variation represents an important resource for future efforts to combat HCMV infections. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

13 pages, 799 KiB  
Review
Revisiting the Role of γδ T Cells in Anti-CMV Immune Response after Transplantation
by Ahmed Gaballa, Faisal Alagrafi, Michael Uhlin and Arwen Stikvoort
Viruses 2021, 13(6), 1031; https://doi.org/10.3390/v13061031 - 29 May 2021
Cited by 8 | Viewed by 2986
Abstract
Gamma delta (γδ) T cells form an unconventional subset of T lymphocytes that express a T cell receptor (TCR) consisting of γ and δ chains. Unlike conventional αβ T cells, γδ T cells share the immune signature of both the innate and the [...] Read more.
Gamma delta (γδ) T cells form an unconventional subset of T lymphocytes that express a T cell receptor (TCR) consisting of γ and δ chains. Unlike conventional αβ T cells, γδ T cells share the immune signature of both the innate and the adaptive immunity. These features allow γδ T cells to act in front-line defense against infections and tumors, rendering them an attractive target for immunotherapy. The role of γδ T cells in the immune response to cytomegalovirus (CMV) has been the focus of intense research for several years, particularly in the context of transplantation, as CMV reactivation remains a major cause of transplant-related morbidity and mortality. Therefore, a better understanding of the mechanisms that underlie CMV immune responses could enable the design of novel γδ T cell-based therapeutic approaches. In this regard, the advent of next-generation sequencing (NGS) and single-cell TCR sequencing have allowed in-depth characterization of CMV-induced TCR repertoire changes. In this review, we try to shed light on recent findings addressing the adaptive role of γδ T cells in CMV immunosurveillance and revisit CMV-induced TCR reshaping in the era of NGS. Finally, we will demonstrate the favorable and unfavorable effects of CMV reactive γδ T cells post-transplantation. Full article
(This article belongs to the Special Issue Cytomegalovirus Immunity 2021)
Show Figures

Figure 1

Back to TopTop