1
|
Li Z, Jiang S, Liu W, Yang X, Liu F, Li X, Li J, Yu M, Wei Z, Wang B, Qian D. A promising endeavor against human cytomegalovirus: Predominant epitopes-based recombinant subunit vaccine RH EcIE1/pp65/pp150. Virulence 2025; 16:2497903. [PMID: 40277436 PMCID: PMC12064061 DOI: 10.1080/21505594.2025.2497903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 12/23/2024] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
Human cytomegalovirus (HCMV) is widespread in the population, typically remaining latent. However, it can cause severe morbidity and mortality in transplant patients and immunodeficient individuals. Currently, there is no approved vaccine against HCMV. This study used immunoinformatics methods to predict the predominant T and B-cell epitopes of three key HCMV proteins, including phosphoprotein 65 (pp65), pp150, and immediate-early protein 1 (IE1). Subsequently, we synthesized a recombinant subunit vaccine (RHEcIE1/pp65/pp150) from Escherichia coli, comprising RHEc-1 and RHEc-2. We observed that the RHEcIE1/pp65/pp150 vaccine exhibited high safety and immunogenicity in mice, enhancing a significant upregulation of CD80, CD86, CD40, and MHCII on dendritic cells and macrophages. Additionally, the vaccine activated innate immune responses through the NF-κB signalling pathway, triggering CD4+ and CD8+T cells to secrete tumour necrosis factor (TNF)-α, interferon (IFN)-γ, and interleukin (IL)-2, directing the T-cell response towards Th1. Moreover, it stimulated CD4+T cells to secrete IL-4, IL-6, and IL-10, promoting B-cell immunity. Furthermore, the RHEcIE1/pp65/pp150 vaccine induced the formation of abundant memory cells and high levels of neutralizing antibody titres, conducive to providing long-lasting protection. Taken together, the RHEcIE1/pp65/pp150 vaccine is a promising endeavour against HCMV, and these findings contribute valuable insights to the development of HCMV vaccine candidates.
Collapse
MESH Headings
- Cytomegalovirus Vaccines/immunology
- Cytomegalovirus Vaccines/genetics
- Cytomegalovirus Vaccines/administration & dosage
- Animals
- Vaccines, Subunit/immunology
- Vaccines, Subunit/genetics
- Vaccines, Subunit/administration & dosage
- Cytomegalovirus/immunology
- Cytomegalovirus/genetics
- Mice
- Viral Matrix Proteins/immunology
- Viral Matrix Proteins/genetics
- Cytomegalovirus Infections/prevention & control
- Cytomegalovirus Infections/immunology
- Humans
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Phosphoproteins/immunology
- Phosphoproteins/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, B-Lymphocyte/genetics
- Female
- Immediate-Early Proteins/immunology
- Immediate-Early Proteins/genetics
- Antibodies, Viral/blood
- Mice, Inbred BALB C
- Immunogenicity, Vaccine
Collapse
Affiliation(s)
- Zonghui Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Clinical Laboratory, Chengdu Aerotropolis Asia Heart Hospital, Chengdu, China
| | - Shasha Jiang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Wenxuan Liu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaoli Yang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fengjun Liu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xu Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jun Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Meng Yu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhun Wei
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Dongmeng Qian
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Khodamoradi S, Khodaei F, Mohammadian T, Ferdousi A, Rafiee F. Oxidative stress, inflammation, and apoptosis in Alzheimer's disease associated with HSV-1 and CMV coinfection. Virol J 2025; 22:169. [PMID: 40442743 PMCID: PMC12123989 DOI: 10.1186/s12985-025-02786-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 05/10/2025] [Indexed: 06/02/2025] Open
Abstract
Oxidative stress, inflammation, and apoptosis have been reported to influence cognitive function in patients with Alzheimer's disease (AD), particularly those infected with herpes simplex virus type 1 (HSV-1) or cytomegalovirus (CMV). This study aimed to evaluate the effects of viral infection on oxidative stress markers associated with these pathways in AD patients. A total of 100 adults with mild-to-moderate AD were randomly assigned to a double-blind, placebo-controlled clinical trial and categorized into three groups: AD (uninfected), AD with HSV-1, and AD with CMV. The primary outcomes included changes in serum inflammatory markers (IL-1β and TNF-α), blood antioxidant and oxidative stress markers-glutathione peroxidase (GPx), superoxide dismutase (SOD), malondialdehyde (MDA), reactive oxygen species (ROS), and total antioxidant capacity (TAC), as well as the expression levels of apoptosis-related proteins (BAX and BCL-2). Results showed that, compared to the control group, the AD group exhibited significant alterations in inflammatory and oxidative stress markers. CMV infection led to increased antioxidant enzyme activity and decreased serum inflammatory markers relative to the uninfected AD group. However, there were significant differences in ratio BAX/BCL-2 protein expression between the CMV and HSV-1 groups when compared to the AD group. In conclusion, AD patients infected with HSV-1 or CMV demonstrated distinct alterations in inflammatory, oxidative stress, antioxidant profiles, and apoptosis markers, which may have beneficial implications for circulatory biomarkers and potentially cognitive outcomes in AD.
Collapse
Affiliation(s)
- Sepideh Khodamoradi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Forouzan Khodaei
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Taher Mohammadian
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Atousa Ferdousi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rafiee
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Walter V, Koch A, Hillmann D, Dannehl D, Staebler A, Pfister K, Risch L, Engler T, Brucker S, Janni W, Hartkopf A, Flatz L. Association of CMV status with response to neoadjuvant chemoimmunotherapy in early triple-negative breast cancer. Cancer Lett 2025; 626:217789. [PMID: 40354994 DOI: 10.1016/j.canlet.2025.217789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/15/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Vincent Walter
- Department of Dermatology, University Hospital Tübingen, Germany
| | - André Koch
- Department of Women's Health, University Hospital Tübingen, Germany
| | | | - Dominik Dannehl
- Department of Women's Health, University Hospital Tübingen, Germany
| | | | | | - Lorenz Risch
- Division of Clinical Biochemistry, Medical University Innsbruck, Austrlia
| | - Tobias Engler
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Sara Brucker
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Wolfgang Janni
- Department of Gynecology, University Hospital Ulm, Germany
| | - Andreas Hartkopf
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University Hospital Tübingen, Germany; Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
| |
Collapse
|
4
|
Altadill M, Álvarez I, Ataya M, Heredia G, Alari‐Pahissa E, Muntasell A, Llano M, Fuchs J, Vilches C, Hengel H, Halenius A, López‐Botet M. Human Cytomegalovirus Antigen Presentation by HLA-G in Infected Cells. HLA 2025; 105:e70089. [PMID: 40347012 PMCID: PMC12065092 DOI: 10.1111/tan.70089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/17/2025] [Accepted: 02/08/2025] [Indexed: 05/12/2025]
Abstract
HLA-E and -G class Ib molecules were considered unrelated to viral antigen presentation. HLA-E binds nonamers from the leader sequences of other HLA-I molecules and the human cytomegalovirus (HCMV) UL40 protein, interacting with CD94/NKG2 NK cell receptors. Yet, evidence that HLA-E may present some pathogen-derived peptides to CD8+ T lymphocytes has been reported. By contrast, HLA-G binds a broad spectrum of endogenous sequences but its role in antigen presentation is unknown. An experimental approach was set up to search for HCMV antigens displayed by HLA-G in infected cells. Among the analysed peptidome, 22 sequences corresponding to 16 HCMV molecules were identified; 17 peptides were confirmed to interact in vitro with HLA-G of which 10 displayed characteristic anchor residues. As compared to the response in short-term (6 h) assays to immunodominant IE-1 and pp65 antigens, none of the HLA-G-binding peptides stimulated cytokine production by CD8+ T cells from HCMV-seropositive blood donors (n = 15). Following a 14-day peptide stimulation of PBMC and expansion with IL-2, CD8+ T cells specifically responding to a subset of these viral antigens were detected in some individuals, yet were not restricted by HLA-G in functional assays. A subset of viral peptides did bind to both HLA-G and -E but were not recognised by CD94/NKG2 NK cell receptors. Our results provide the first evidence that HLA-G may display potentially immunogenic viral peptides in HCMV-infected cells, yet do not support their ability to promote HLA-G-restricted CD8+ T cell responses nor to modulate NK cell functions.
Collapse
Affiliation(s)
- Mireia Altadill
- Department of Medicine and Life SciencesUniversity Pompeu FabraBarcelonaSpain
| | - Iñaki Álvarez
- Department of Cell BiologyPhysiology and Immunology, Institute of Biotechnology and Biomedicine, Autonomous University of BarcelonaBellaterraSpain
| | - Michelle Ataya
- Department of Medicine and Life SciencesUniversity Pompeu FabraBarcelonaSpain
| | - Gemma Heredia
- Department of Medicine and Life SciencesUniversity Pompeu FabraBarcelonaSpain
| | | | - Aura Muntasell
- Department of Cell BiologyPhysiology and Immunology, Institute of Biotechnology and Biomedicine, Autonomous University of BarcelonaBellaterraSpain
- Hospital del Mar Research InstituteBarcelonaSpain
| | - Manuel Llano
- Biological Sciences DepartmentThe University of Texas at El PasoEl PasoUSA
| | - Jonas Fuchs
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Carlos Vilches
- Immunogenetics and Histocompatibility Lab, Instituto de Investigación Sanitaria Puerta de Hierro ‐ Segovia de AranaMadridSpain
- Organización Nacional de Trasplantes, Ministerio de SanidadMadridSpain
| | - Hartmut Hengel
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Anne Halenius
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Miguel López‐Botet
- Department of Medicine and Life SciencesUniversity Pompeu FabraBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
| |
Collapse
|
5
|
Fougère Y, Brophy J, Hawkes MT, Lee T, Samson L, Gantt S, Dufour MSK, Renaud C, Dieumegard H, Diallo MA, Canape J, Read S, Bitnun A, Soudeyns H, Kakkar F. Clinical and Immunologic Impact of CMV Coinfection Among Children Living With HIV in Canada. Pediatr Infect Dis J 2025:00006454-990000000-01282. [PMID: 40209769 DOI: 10.1097/inf.0000000000004811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
BACKGROUND Although cytomegalovirus (CMV) disease has been well described among severely immunocompromised children living with HIV (CLWH), the impact of CMV coinfection, is not well understood. The objective of this study was to characterize the clinical and immunologic effects of CMV coinfection in CLWH in Canada. METHODS This is a substudy of the Early Pediatric Initiation, Canada Child Cure Cohort study, which enrolled CLWH in Canada between 2014 and 2018. CMV serostatus was determined at the first (baseline) study visit, and HIV-1 viral load (VL), CMV VL, and lymphocyte subsets were quantified every 3-6 months. For a subset of participants, CD4+ and CD8+ T cell subsets were analyzed using flow cytometry. The clinical outcomes were recorded retrospectively at the baseline visit and prospectively during the study period. RESULTS Of the 225 participants, 85.3% were CMV seropositive (CMV+) and 81% had suppressed HIV VL. While there were no significant differences in clinical outcomes between CMV+ and CMV- children, CMV+ children had lower frequencies of CD4+ T cells, higher frequencies of CD8+ T cells, and lower CD4/CD8 ratio at baseline than CMV- children. Children with CMV+ children also demonstrated a higher frequency of CD4+ effector memory cells, lower CD8+ naïve T cells, and higher frequencies of CD8+ terminally differentiated effector memory cells. These differences remained significant even after adjusting for HIV viral control. CONCLUSIONS CMV coinfection is common among CLWH and is associated with distinct immunological changes despite the effective control of HIV replication with antiretroviral therapy. The long-term implications of these immunological perturbations require further investigation.
Collapse
Affiliation(s)
- Yves Fougère
- From the Unit of Pediatric Infectious Diseases and Vaccinology, Department of Woman Mother and Child, Lausanne University Hospital, Lausanne, Switzerland
- Centre d'infectiologie mère-enfant (CIME), Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Jason Brophy
- Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael T Hawkes
- BC Children's Hospital and Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Terry Lee
- CIHR Pan-Canadian Network for HIV & STBBI Clinical Trials Research (CTN+), Vancouver, British Columbia, Canada
| | - Lindy Samson
- Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - Soren Gantt
- Centre d'infectiologie mère-enfant (CIME), Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology & Immunology
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Mi-Suk Kang Dufour
- Centre d'infectiologie mère-enfant (CIME), Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, Quebec, Canada
- University of California, Berkeley, School of Public Health
| | - Christian Renaud
- Department of Microbiology, Infectiology & Immunology
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Hinatea Dieumegard
- Department of Microbiology, Infectiology & Immunology
- University of California, Berkeley, School of Public Health
| | - Madeleine Aby Diallo
- Department of Microbiology, Infectiology & Immunology
- University of California, Berkeley, School of Public Health
| | - Jade Canape
- Department of Microbiology, Infectiology & Immunology
- University of California, Berkeley, School of Public Health
| | - Stanley Read
- Unité d'immunopathologie virale, Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Ari Bitnun
- Unité d'immunopathologie virale, Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Hugo Soudeyns
- Centre d'infectiologie mère-enfant (CIME), Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology & Immunology
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- University of California, Berkeley, School of Public Health
- Unité d'immunopathologie virale, Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Fatima Kakkar
- Centre d'infectiologie mère-enfant (CIME), Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, Quebec, Canada
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Boquett JA, Sauter J, Schmidt AH, Maiers M, Hollenbach JA. Human leukocyte antigen variation is associated with cytomegalovirus serostatus in healthy individuals. Am J Hum Genet 2025; 112:913-926. [PMID: 40049169 DOI: 10.1016/j.ajhg.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 03/12/2025] Open
Abstract
Cytomegalovirus (CMV) is a common β-herpes virus worldwide with an estimated seroprevalence among the general population of 83%. Primary infection is usually benign; however, CMV can cause severe morbidity in newborns in whom it is acquired congenitally, as well as immunocompromised individuals. Understanding the role of immunogenetic variation in risk for CMV infection can provide insight into the immune control of this ubiquitous pathogen. Here, we evaluated the association of human leukocyte antigen (HLA) genetic variation with CMV seropositivity in more than 518,000 individuals from two independent cohorts. We found three HLA class II alleles (HLA-DRB1∗04:03 with risk; HLA-DRB1∗01:03 and HLA-DRB1∗07:01 with protection) to be significantly associated with CMV serostatus across both cohorts and in multiple population subgroups. Interestingly, HLA-DRB1∗04:03 and HLA-DRB1∗01:03, the alleles with the strongest observed effect, are relatively rare, while common homologous alleles show no association with CMV. We show that these differences are mediated by changes in charge and volume to two key pockets in the peptide-binding groove of the HLA molecule, providing a structural basis for the observed association. Our results provide population-scale evidence for the role of HLA in mediating infection with this ubiquitous human virus and a framework for understanding immunological conditions necessary for efficient viral control.
Collapse
Affiliation(s)
- Juliano A Boquett
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Martin Maiers
- CIBMTR (Center for International Blood and Marrow Transplant Research), NMDP, Minneapolis, MN, USA
| | - Jill A Hollenbach
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Giménez E, Albert E, Colomer E, Pérez A, Montomoli M, Piñana JL, Sanchis I, Górriz JL, Solano C, Navarro D. Evaluation of the VIDAS Cytomegalovirus (CMV)-IGRA for Assessing CMV-Specific T-Cell Responses in Immunocompetent and Immunosuppressed Individuals. J Med Virol 2025; 97:e70355. [PMID: 40232234 DOI: 10.1002/jmv.70355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/03/2025] [Accepted: 04/05/2025] [Indexed: 04/16/2025]
Abstract
We compared the performance of the VIDAS Cytomegalovirus (CMV)-Interferon-gamma release assay (IGRA) with that of laboratory-developed flow cytometry for intracellular cytokine staining (FC-ICS) for the assessment of CMV-specific interferon-gamma (IFN-γ)-producing T-cell responses (CMV-CMI). A total of 147 blood specimens from 78 adult participants were collected: 11 healthy controls, 34 hematological patients (HP), of which 32 had undergone allogeneic hematopoietic cell transplantation (allo-HCT), and 33 Kidney transplant recipients. Of the 147 specimens, 96 tested positive, 24 negative, 25 indeterminate, and 2 were invalid by the VIDAS CMV IGRA. A total of 137 specimens were tested by FC-ICS, of which 107 returned positive results. There were 27 discrepancies across the assays among specimens yielding interpretable results, of which 14 tested VIDAS CMV IGRA-positive/FC-ICS-negative and 12 VIDAS CMV IGRA-negative/FC-ICS-positive. The overall agreement between immunoassays was 78%, and the Kappa coefficient was 0.34 (0.52 for HP). Differences in identifying CMV-infected (CMV IgG-positive) and uninfected participants (CMV IgG-negative) were noticed across both assays. The overall correlation (rho values) between IFN-γ concentrations (IU/ml) measured by the VIDAS CMV IGRA and CMV-specific IFN-γ-producing T-cell frequencies were 0.27 for CD4+ and 0.33 for CD8+ T cells (p = 0.001). In HP, the correlation was stronger (0.48 for CD4+ and 0.49 for CD8+ T cells). In summary, our data lend support to the potential utility of the VIDAS CMV IGRA to assess CMV-CMI in transplant recipients.
Collapse
Affiliation(s)
- Estela Giménez
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Eliseo Albert
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Ester Colomer
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Ariadna Pérez
- Hematology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Marco Montomoli
- Nephrology Service, Hospital Clínico Universitario de Valencia, INCLIVA Health Research Institute, RICORS 2040 RENAL, ISCIII, Valencia, Spain
| | - José Luis Piñana
- Hematology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Irina Sanchis
- Nephrology Service, Hospital Clínico Universitario de Valencia, INCLIVA Health Research Institute, RICORS 2040 RENAL, ISCIII, Valencia, Spain
| | - José Luis Górriz
- Nephrology Service, Hospital Clínico Universitario de Valencia, INCLIVA Health Research Institute, RICORS 2040 RENAL, ISCIII, Valencia, Spain
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | - Carlos Solano
- Hematology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | - David Navarro
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Microbiology School of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
8
|
Olofsson A, Humbert M, Rekha RS, Frankling MH, Lund-Johansen F, Bergman P, Björkhem-Bergman L, Karlsson AC. Adaptive immune responses against common viruses are sustained and functional in end-of-life patients. iScience 2025; 28:112082. [PMID: 40124502 PMCID: PMC11930376 DOI: 10.1016/j.isci.2025.112082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/22/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Viral infections occur with increased frequency in patients in palliative care, impacting their quality of life and increasing mortality rates. Still, the function of the immune system has never been thoroughly studied at the end of life. We investigated virus-specific humoral and cellular immune responses in elderly end-of-life patients (n = 38) and controls (n = 28). Virus-specific T cell responses were characterized using high-parameter flow cytometry, after stimulation with cytomegalovirus (CMV) and human coronavirus OC43 peptides. Although some virus-specific T cells from patients exhibited elevated expression of costimulatory and coinhibitory molecules, their functional profile remained largely intact compared to controls. The expression of the cytotoxic markers Granzyme B, CD107a, and 2B4 on CMV-specific T cells correlated closely with survival time. Significantly, our data demonstrate that both humoral and cellular immunity remain responsive and functional against common viruses in end-of-life patients.
Collapse
Affiliation(s)
- Anna Olofsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marion Humbert
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Rokeya S. Rekha
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Helde Frankling
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Fridtjof Lund-Johansen
- Institute of Clinical Medicine, University of Oslo, Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Immunology, Oslo University Hospital, ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Peter Bergman
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Linda Björkhem-Bergman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Research and Development Unit/Palliative Care, Stockholms Sjukhem, Mariebergsgatan 22, Stockholm, Sweden
| | - Annika C. Karlsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Yoshida K, Misumi M, Hamasaki K, Kyoizumi S, Satoh Y, Tsuruyama T, Uchimura A, Kusunoki Y. High-dose radiation preferentially induces the clonal expansion of hematopoietic progenitor cells over mature T and B cells in mouse bone marrow. Stem Cell Reports 2025; 20:102423. [PMID: 40020684 PMCID: PMC11960520 DOI: 10.1016/j.stemcr.2025.102423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 03/03/2025] Open
Abstract
Radiation induces clonal hematopoiesis (CH) involving high-frequency somatic mutations in hematopoietic cells. However, the effects of radiation on clonal expansion of hematopoietic progenitor cells and lymphocytes remain elusive. Here, we investigate CH mutations and T cell receptor (TCR) and B cell receptor (BCR) sequences within the bone marrow cells of mice 18 months after irradiation (3 Gy) and age-matched controls. Two to six CH mutations were identified in the irradiated mice (N = 5), while only one of the four control mice carried a CH mutation. These CH mutations detected in the bone marrow were also identified in the splenic CD11b+ myeloid cell population. Meanwhile, the cumulative size of the ten largest TCR and BCR clones, as well as their clonality, did not differ significantly between irradiated and control mice. Our findings suggest that radiation preferentially induces clonal expansion of hematopoietic progenitor cells over mature lymphocytes in the bone marrow.
Collapse
Affiliation(s)
- Kengo Yoshida
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan.
| | - Munechika Misumi
- Department of Statistics, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Kanya Hamasaki
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Seishi Kyoizumi
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Yasunari Satoh
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Tatsuaki Tsuruyama
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Arikuni Uchimura
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Yoichiro Kusunoki
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan.
| |
Collapse
|
10
|
De Bartolo A, Angelone T, Rocca C. Elucidating emerging signaling pathways driving endothelial dysfunction in cardiovascular aging. Vascul Pharmacol 2025; 158:107462. [PMID: 39805379 DOI: 10.1016/j.vph.2025.107462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
The risk for developing cardiovascular diseases dramatically increases in older individuals, and aging vasculature plays a crucial role in determining their morbidity and mortality. Aging disrupts endothelial balance between vasodilators and vasoconstrictors, impairing function and promoting pathological vascular remodeling. In this Review, we discuss the impact of key and emerging molecular pathways that transduce aberrant inflammatory signals (i.e., chronic low-grade inflammation-inflammaging), oxidative stress, and mitochondrial dysfunction in aging vascular compartment. We focus on the interplay between these events, which contribute to generating a vicious cycle driving the progressive alterations in vascular structure and function during cardiovascular aging. We also discuss the primary role of senescent endothelial cells and vascular smooth muscle cells, and the potential link between vascular and myeloid cells, in impairing plaque stability and promoting the progression of atherosclerosis. The aim of this summary is to provide potential novel insights into targeting these processes for therapeutic benefit.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
11
|
van den Dijssel J, Konijn VAL, Duurland MC, de Jongh R, Koets L, Veldhuisen B, Raaphorst H, Turksma AW, Freen‐van Heeren JJ, Steenhuis M, Rispens T, van der Schoot CE, van Ham SM, van Lier RAW, van Gisbergen KPJM, ten Brinke A, van de Sandt CE. Age and Latent Cytomegalovirus Infection Do Not Affect the Magnitude of De Novo SARS-CoV-2-Specific CD8 + T Cell Responses. Eur J Immunol 2025; 55:e202451565. [PMID: 40071711 PMCID: PMC11898545 DOI: 10.1002/eji.202451565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 03/15/2025]
Abstract
Immunosenescence, age-related immune dysregulation, reduces immunity upon vaccinations and infections. Cytomegalovirus (CMV) infection results in declining naïve (Tnaïve) and increasing terminally differentiated (Temra) T cell populations, further aggravating immune aging. Both immunosenescence and CMV have been speculated to hamper the formation of protective T-cell immunity against novel or emerging pathogens. The SARS-CoV-2 pandemic presented a unique opportunity to examine the impact of age and/or CMV on the generation of de novo SARS-CoV-2-specific CD8+ T cell responses in 40 younger (22-40 years) and 37 older (50-66 years) convalescent individuals. Heterotetramer combinatorial coding combined with phenotypic markers were used to study 35 SARS-CoV-2 epitope-specific CD8+ T cell populations directly ex vivo. Neither age nor CMV affected SARS-CoV-2-specific CD8+ T cell frequencies, despite reduced total CD8+ Tnaïve cells in older CMV- and CMV+ individuals. Robust SARS-CoV-2-specific central memory CD8+ T (Tcm) responses were detected in younger and older adults regardless of CMV status. Our data demonstrate that immune aging and CMV status did not impact the SARS-CoV-2-specific CD8+ T cell response. However, SARS-CoV-2-specific CD8+ T cells of older CMV- individuals displayed the lowest stem cell memory (Tscm), highest Temra and PD1+ populations, suggesting that age, not CMV, may impact long-term SARS-CoV-2 immunity.
Collapse
Affiliation(s)
- Jet van den Dijssel
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Veronique A. L. Konijn
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Mariël C Duurland
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Rivka de Jongh
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Lianne Koets
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- National Screening Laboratory of SanquinResearch and Laboratory ServicesAmsterdamThe Netherlands
| | - Barbera Veldhuisen
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Immunohematology DiagnosticsSanquin Diagnostic ServicesAmsterdamThe Netherlands
| | | | | | | | - Maurice Steenhuis
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Amsterdam UMC location Vrije Universiteit AmsterdamMolecular Cell Biology and ImmunologyAmsterdamThe Netherlands
| | - C Ellen van der Schoot
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - S. Marieke van Ham
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Swammerdam Institute for Life SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Klaas P. J. M. van Gisbergen
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Physiology and Cancer Programme, Champalimaud ResearchChampalimaud FoundationLisboaPortugal
| | - Anja ten Brinke
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Carolien E. van de Sandt
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Department of Microbiology and ImmunologyUniversity of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| |
Collapse
|
12
|
Hansen SG, Schell JB, Marshall EE, Ojha S, Feltham S, Morrow D, Hughes CM, Gilbride RM, Ford JC, Cleveland-Rubeor HC, McArdle MR, Whitmer T, Barber-Axthelm A, Bochart R, Smedley J, Oswald K, Fast R, Shoemaker R, Kosmider E, Edlefsen PT, Lifson JD, Malouli D, Früh K, Picker LJ. Glycoprotein L-deleted single-cycle rhesus cytomegalovirus vectors elicit MHC-E-restricted CD8+ T cells that protect against SIV. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf104. [PMID: 40420384 DOI: 10.1093/jimmun/vkaf104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/14/2025] [Indexed: 05/28/2025]
Abstract
Strain 68-1 rhesus CMV (RhCMV) vectors induce immune responses that mediate early, complete replication arrest of SIV infection in ∼60% of vaccinated rhesus macaques (RMs). This unique efficacy depends on the ability of these vectors to elicit effector memory (EM)-biased CD8+ T cells recognizing SIV peptides presented by MHC-E, rather than MHC-Ia. These efficacious responses still occurred when spread of the 68-1 vector was impaired by deletion of the viral anti-host intrinsic immunity factor phosphoprotein 71 (pp71), but efficacy was lost with a more stringent attenuation strategy based on destabilization of Rh108, the ortholog of the essential human CMV (HCMV) transcription factor UL79 that is required for late viral gene expression. Although unable to produce infectious progeny (ie single-cycle infection), Rh108-deficient vectors elicited durable, high frequency, EM-biased, SIV-specific CD8+ T-cell responses in RMs, but these responses were MHC-Ia-restricted and therefore non-efficacious. Here, we tested a different single-cycle attenuation strategy based on deletion (Δ) of the glycoprotein L (gL) that is essential for viral entry but allows for late gene expression and viral assembly. ΔgL 68-1 RhCMV/SIV vectors, grown on gL-complementing fibroblasts, were robustly immunogenic at doses above 105 PFU, generating high frequency, EM-biased, SIV-specific CD8+ T-cell responses that were also unconventionally restricted, including the MHC-E restriction associated with efficacy. Indeed, these single-cycle vectors manifested replication arrest efficacy in 70% of vaccinated RMs, further linking MHC-E restriction with efficacy, and demonstrating that 68-1 RhCMV/SIV efficacy does not require vector dissemination within the host.
Collapse
Affiliation(s)
- Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - John B Schell
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Emily E Marshall
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Sohita Ojha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Shana Feltham
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - David Morrow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Colette M Hughes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Julia C Ford
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Hilary C Cleveland-Rubeor
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Matthew R McArdle
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Travis Whitmer
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Rachelle Bochart
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Jeremy Smedley
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Kelli Oswald
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Randy Fast
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Ewelina Kosmider
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Paul T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| |
Collapse
|
13
|
Hundhausen N, Majumder S, Xiao Y, Haeusl SS, Goehler H, Seal R, Chiarolla CM, Rosenwald A, Eyrich M, Cicin-Sain L, Berberich-Siebelt F. NFAT single-deficient murine T cells reduce the risk of aGvHD while controlling cytomegalovirus infection. iScience 2025; 28:111937. [PMID: 40028277 PMCID: PMC11872454 DOI: 10.1016/j.isci.2025.111937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/13/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
NFAT is a family of transcription factors whose activation is inhibited by calcineurin inhibitors (CNIs). In allogeneic hematopoietic stem cell transplantation (allo-HCT), CNIs are employed to prevent and treat graft-versus-host disease (GvHD). Unfortunately, control of cytomegalovirus (CMV), which exacerbates clinical outcomes, is simultaneously lost. Since single NFAT deficiency in T cells ameliorates GvHD in our major mismatch model, we investigated whether protection is maintained during CMV infection. Reassuringly, NFAT-deficient T cells still improved GvHD upon acute CMV infection and after allo-HCT in latently CMV-infected mice, showing reduced proinflammatory and cytotoxic potential. In sharp contrast, CMV-specific NFAT-deficient CD8+ inflated memory T cells expanded more and with higher levels of interferon gamma (IFN-γ) and GzmB expression, effectively controlling CMV. Notably, NFAT-deficient inflated memory T cells could migrate to non-lymphoid tissues and fight CMV. Therefore, CMV infection does not interfere with the protective effect of NFAT inhibition to attenuate GvHD while allowing an anti-CMV response.
Collapse
Affiliation(s)
- Nadine Hundhausen
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Snigdha Majumder
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Yin Xiao
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Sigrun S. Haeusl
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Helen Goehler
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Rishav Seal
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | | | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Matthias Eyrich
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Luka Cicin-Sain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of Helmholtz Centre for Infection Research and Medical School Hannover, Hannover, Germany
| | | |
Collapse
|
14
|
Santos-Peral A, Zaucha M, Nikolova E, Yaman E, Puzek B, Winheim E, Goresch S, Scheck MK, Lehmann L, Dahlstroem F, Karimzadeh H, Thorn-Seshold J, Jia S, Luppa F, Pritsch M, Butt J, Metz-Zumaran C, Barba-Spaeth G, Endres S, Kim-Hellmuth S, Waterboer T, Krug AB, Rothenfusser S. Basal T cell activation predicts yellow fever vaccine response independently of cytomegalovirus infection and sex-related immune variations. Cell Rep Med 2025; 6:101946. [PMID: 39938525 PMCID: PMC11866508 DOI: 10.1016/j.xcrm.2025.101946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/19/2024] [Accepted: 01/15/2025] [Indexed: 02/14/2025]
Abstract
The live-attenuated yellow fever 17D (YF17D) vaccine is a model of acute viral infection that induces long-lasting protective immunity. Among immunocompetent adults, responses to YF17D vary significantly. To understand the sources of this variability, we investigate the influence of sex, age, human leukocyte antigen (HLA) type, and 20 prior infections on basal immune parameters and the cellular and antibody response to YF17D in 250 healthy young individuals. Multivariate regression found that sex and cytomegalovirus (CMV) infection significantly contribute to baseline immune variation but do not affect vaccine responses except for reduced YF17D-specific CD8+ frequencies in CMV-infected males. However, the abundance at baseline of non-specific cytokine-expressing T helper cells in circulation is associated with stronger vaccine responses, a state that smoking favors. Additionally, an elevated baseline level of interferon-stimulated CXCL10 is linked to poorer vaccination outcomes. Altogether, YF17D reactivity is conditioned by the baseline immune status independent of sex and CMV-related variations.
Collapse
Affiliation(s)
- Antonio Santos-Peral
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Magdalena Zaucha
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Elena Nikolova
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Ekin Yaman
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital Munich, Munich, Germany; Institute of Translational Genomics, Department of Computational Health, Helmholtz Munich, Munich, Germany
| | - Barbara Puzek
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital Munich, Munich, Germany; Institute of Translational Genomics, Department of Computational Health, Helmholtz Munich, Munich, Germany
| | - Elena Winheim
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sebastian Goresch
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Magdalena K Scheck
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lisa Lehmann
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Frank Dahlstroem
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Hadi Karimzadeh
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julia Thorn-Seshold
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany; Faculty of Chemistry and Pharmacy, LMU Munich, Munich, Germany
| | - Shenzhi Jia
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Fabian Luppa
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Michael Pritsch
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julia Butt
- Division of Infections and Cancer Epidemiology at the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Camila Metz-Zumaran
- Division of Infections and Cancer Epidemiology at the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Giovanna Barba-Spaeth
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - Stefan Endres
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany; Einheit für Klinische Pharmakologie (EKLiP) Helmholtz Zentrum München German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Sarah Kim-Hellmuth
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital Munich, Munich, Germany; Institute of Translational Genomics, Department of Computational Health, Helmholtz Munich, Munich, Germany
| | - Tim Waterboer
- Division of Infections and Cancer Epidemiology at the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anne B Krug
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Simon Rothenfusser
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany; Einheit für Klinische Pharmakologie (EKLiP) Helmholtz Zentrum München German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
| |
Collapse
|
15
|
Hilt ZT, Reynaldi A, Steinhilber M, Zhang S, Wesnak SP, Smith NL, Davenport MP, Rudd BD. Recent thymic emigrants are preferentially recruited into the memory pool during persistent infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636722. [PMID: 39975271 PMCID: PMC11839080 DOI: 10.1101/2025.02.06.636722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Cytomegalovirus (CMV) leads to a unique phenomenon known as 'memory inflation,' where antigen-specific memory CD8+ T cells continue to accumulate in the peripheral tissues during the latent stage of infection. However, it is still not clear how the inflating pool of memory CD8+ T cells is generated and maintained. In this study, we used murine cytomegalovirus (MCMV) as a model of persistent infection and fate-mapping mice to determine the dynamics of CD8+ T cell recruitment into the memory pool. We found that neonatal exposure to CMV leads to an expansion of newly made CD8+ T cells (recent thymic emigrants, RTEs), which are maintained in the long-lived memory compartment. In contrast, CD8+ T cells made during the latent phase of infection (mature CD8+ T cells) contribute little to the memory pool. We also observed notable phenotypic differences between RTEs and mature cells. Whereas RTEs present at the time of infection gave rise to more effector memory cells, the cells produced later in infection were biased towards becoming central memory cells. Importantly, the preferential recruitment of RTEs into the effector memory pool also occurs during adult exposure to CMV. Collectively, these data demonstrate that persistent infection expands the RTE population, and timing of infection dictates whether neonatal or adult RTEs are 'locked in' to the memory pool.
Collapse
Affiliation(s)
- Zachary T. Hilt
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Megan Steinhilber
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Shide Zhang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Samantha P. Wesnak
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
16
|
Zuber J, Leon J, Déchanet-Merville J, Kaminski H. Belatacept-related cytomegalovirus infection: Advocacy for tailored immunosuppression based on individual assessment of immune fitness. Am J Transplant 2025; 25:277-283. [PMID: 39370115 DOI: 10.1016/j.ajt.2024.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Belatacept, a fusion protein combining cytotoxic T-lymphocyte antigen-4 (CTLA-4) and the Fc region of human IgG1, is increasingly used as a calcineurin inhibitor-sparing regimen in patients with chronic graft dysfunction. Older kidney transplant recipients, particularly from expanded criteria donors, may be switched to belatacept due to poor renal recovery. However, late-onset cytomegalovirus (CMV) reactivation is increasingly reported with this treatment, especially in older patients with graft dysfunction. This suggests a progressive loss of CMV-specific T cell response, potentially driven by T cell exhaustion. Contributing factors include preexisting T cell dysfunction, increased viral antigen exposure, and interference in the PD-L1/PD-1 pathway by belatacept. mTOR inhibitors have shown efficacy in preventing CMV reactivation by reinvigorating CMV-specific T cells. These findings support combining belatacept with mTOR inhibitors in high-risk CMV-seropositive recipients and emphasize the need for personalized immune assessments to guide immunosuppressive strategies.
Collapse
Affiliation(s)
- Julien Zuber
- Département des Maladies du Rein et du Métabolisme, Transplantation et Immunologie Clinique, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, Paris, France; Inserm UMR_S1163, Institut Hospitalo-Universitaire IMAGINE, Université Paris Cité, Paris, France.
| | - Juliette Leon
- Département des Maladies du Rein et du Métabolisme, Transplantation et Immunologie Clinique, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, Paris, France; Inserm UMR_S1163, Institut Hospitalo-Universitaire IMAGINE, Université Paris Cité, Paris, France
| | - Julie Déchanet-Merville
- Université de Bordeaux, CNRS, ImmunoConcEpT UMR_5164, INSERM ERL U1303, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Bordeaux, France
| | - Hannah Kaminski
- Université de Bordeaux, CNRS, ImmunoConcEpT UMR_5164, INSERM ERL U1303, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Bordeaux, France; Département de Néphrologie, Transplantation, Dialyse et Aphérèse, Hôpital Pellegrin, Bordeaux, France.
| |
Collapse
|
17
|
Otero CE, Petkova S, Ebermann M, Taher H, John N, Hoffmann K, Davalos A, Moström MJ, Gilbride RM, Papen CR, Barber-Axthelm A, Scheef EA, Barfield R, Sprehe LM, Kendall S, Manuel TD, Beechwood T, Nguyen LK, Vande Burgt NH, Chan C, Denton M, Streblow ZJ, Streblow DN, Tarantal AF, Hansen SG, Kaur A, Permar S, Früh K, Hengel H, Malouli D, Kolb P. Rhesus Cytomegalovirus-encoded Fcγ-binding glycoproteins facilitate viral evasion from IgG-mediated humoral immunity. Nat Commun 2025; 16:1200. [PMID: 39885150 PMCID: PMC11782611 DOI: 10.1038/s41467-025-56419-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Human cytomegalovirus (HCMV) encodes four viral Fc-gamma receptors (vFcγRs) that counteract antibody-mediated activation in vitro, but their role in infection and pathogenesis is unknown. To examine their in vivo function in an animal model evolutionarily closely related to humans, we identified and characterized Rh05, Rh152/151 and Rh173 as the complete set of vFcγRs encoded by rhesus CMV (RhCMV). Each one of these proteins displays functional similarities to their prospective HCMV orthologs with respect to antagonizing host FcγR activation in vitro. When RhCMV-naïve male rhesus macaques were infected with vFcγR-deleted RhCMV, peak plasma DNAemia levels and anti-RhCMV antibody responses were comparable to wildtype infections of both male and female animals. However, the duration of plasma DNAemia was significantly shortened in immunocompetent, but not in CD4 + T cell-depleted animals. Since vFcγRs were not required for superinfection of rhesus macaques, we conclude that these proteins can prolong lytic replication during primary infection by evading virus-specific adaptive immune responses, particularly antibodies.
Collapse
Affiliation(s)
- Claire E Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Sophia Petkova
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Ebermann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Nessy John
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Katja Hoffmann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angel Davalos
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Matilda J Moström
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Courtney R Papen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Elizabeth A Scheef
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Lesli M Sprehe
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Savannah Kendall
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Tabitha D Manuel
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Teresa Beechwood
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Linh Khanh Nguyen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Nathan H Vande Burgt
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Zachary J Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Alice F Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, CA, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA.
| | - Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
18
|
Bruton J, Hanke T. Exploitation of Unconventional CD8 T-Cell Responses Induced by Engineered Cytomegaloviruses for the Development of an HIV-1 Vaccine. Vaccines (Basel) 2025; 13:72. [PMID: 39852851 PMCID: PMC11769474 DOI: 10.3390/vaccines13010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/26/2025] Open
Abstract
After four decades of intensive research, traditional vaccination strategies for HIV-1 remain ineffective due to HIV-1's extraordinary genetic diversity and complex immune evasion mechanisms. Cytomegaloviruses (CMV) have emerged as a novel type of vaccine vector with unique advantages due to CMV persistence and immunogenicity. Rhesus macaques vaccinated with molecular clone 68-1 of RhCMV (RhCMV68-1) engineered to express simian immunodeficiency virus (SIV) immunogens elicited an unconventional major histocompatibility complex class Ib allele E (MHC-E)-restricted CD8+ T-cell response, which consistently protected over half of the animals against a highly pathogenic SIV challenge. The RhCMV68-1.SIV-induced responses mediated a post-infection replication arrest of the challenge virus and eventually cleared it from the body. These observations in rhesus macaques opened a possibility that MHC-E-restricted CD8+ T-cells could achieve similar control of HIV-1 in humans. The potentially game-changing advantage of the human CMV (HCMV)-based vaccines is that they would induce protective CD8+ T-cells persisting at the sites of entry that would be insensitive to HIV-1 evasion. In the RhCMV68-1-protected rhesus macaques, MHC-E molecules and their peptide cargo utilise complex regulatory mechanisms and unique transport patterns, and researchers study these to guide human vaccine development. However, CMVs are highly species-adapted viruses and it is yet to be shown whether the success of RhCMV68-1 can be translated into an HCMV ortholog for humans. Despite some safety concerns regarding using HCMV as a vaccine vector in humans, there is a vision of immune programming of HCMV to induce pathogen-tailored CD8+ T-cells effective against HIV-1 and other life-threatening diseases.
Collapse
Affiliation(s)
- Joseph Bruton
- Hertford College, University of Oxford, Oxford OX1 3BW, UK;
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
19
|
Permar SR, Schleiss MR, Plotkin SA. A vaccine against cytomegalovirus: how close are we? J Clin Invest 2025; 135:e182317. [PMID: 39744948 DOI: 10.1172/jci182317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
The pursuit of a vaccine against the human cytomegalovirus (HCMV) has been ongoing for more than 50 years. HCMV is the leading infectious cause of birth defects, including damage to the brain, and is a common cause of complications in organ transplantation. The complex biology of HCMV has made vaccine development difficult, but a recent meeting sponsored by the National Institute of Allergy and Infectious Diseases in September of 2023 brought together experts from academia, industry, and federal agencies to discuss progress in the field. The meeting reviewed the status of candidate HCMV vaccines under study and the challenges in clinical trial design in demonstrating efficacy against congenital CMV infection or the reduction of HCMV disease following solid organ transplantation or hematopoietic stem cell transplantation. Discussion in the meeting revealed that, with the numerous candidate vaccines that are under study, it is clear that a safe and effective HCMV vaccine is within reach. Meeting attendees achieved a consensus opinion that even a partially effective vaccine would have a major effect on the global health consequences of HCMV infection.
Collapse
Affiliation(s)
- Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical Center, New York, New York, USA
| | - Mark R Schleiss
- Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Vaxconsult, Doylestown, Pennsylvania, USA
| |
Collapse
|
20
|
Redondo N, Valverde-Manso A, Ruiz-Merlo T, Rodríguez-Goncer I, Parra P, López-Medrano F, González E, Polanco N, San Juan R, Andrés A, Aguado JM, Fernández-Ruiz M. Human Cytomegalovirus Infection Induces Long-Term Changes in the Cytokine Milieu of Kidney Transplant Recipients. J Med Virol 2025; 97:e70178. [PMID: 39831372 DOI: 10.1002/jmv.70178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/28/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
The impact of human cytomegalovirus (HCMV) infection on the mid- and long-term balance between pro-inflammatory and anti-inflammatory cytokines among kidney transplant recipients (KTRs) remains unclear. We measured plasma levels of 12 Th1/Th2-type cytokines (granulocyte-macrophage colony-stimulating factor, interferon-γ, interleukin [IL]-1β, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12p70, IL-13, IL-18 and tumor necrosis factor-α) in a cohort of 290 KTRs at four time points through month 12 after transplantation. Cytokine levels at each point were compared according to the previous documentation of HCMV replication by two approaches: "cumulative exposure" from the time of transplantation and "recent exposure" within the 2-3 months preceding cytokine assessment. Significance levels were Bonferroni-corrected for multiple pairwise comparisons. Plasma levels of IL-6, IL-10, and IL-12p70 at month 1 were significantly increased in KTRs that had experienced HCMV infection during the first 30 days. By month 3, IL-6 and IL-10 remained increased in KTRs with cumulative exposure through day 90. Cumulative exposure to HCMV replication through day 180 was also associated to increased IL-10 levels at month 6. In addition, KTRs with recent HCMV exposure had increased IL-10 levels at months 3 and 6. After multivariable adjustment, cumulative exposure to HCMV infection and/or the area under curve of HCMV DNAemia during the corresponding period were associated to IL-10 levels within the highest quartile at months 1, 3, and 6. Preceding HCMV infection induces sustained changes in the plasma cytokine milieu of KTRs, with elevated IL-6 and IL-10 levels throughout the first 6 months after transplantation.
Collapse
Affiliation(s)
- Natalia Redondo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Andrea Valverde-Manso
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Tamara Ruiz-Merlo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Isabel Rodríguez-Goncer
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Patricia Parra
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Esther González
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Natalia Polanco
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Amado Andrés
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|
21
|
Vijayan K. K. V, De Paris K. Nonhuman primate models of pediatric viral diseases. Front Cell Infect Microbiol 2024; 14:1493885. [PMID: 39691699 PMCID: PMC11649651 DOI: 10.3389/fcimb.2024.1493885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Infectious diseases are the leading cause of death in infants and children under 5 years of age. In utero exposure to viruses can lead to spontaneous abortion, preterm birth, congenital abnormalities or other developmental defects, often resulting in lifelong health sequalae. The underlying biological mechanisms are difficult to study in humans due to ethical concerns and limited sample access. Nonhuman primates (NHP) are closely related to humans, and pregnancy and immune ontogeny in infants are very similar to humans. Therefore, NHP are a highly relevant model for understanding fetal and postnatal virus-host interactions and to define immune mechanisms associated with increased morbidity and mortality in infants. We will discuss NHP models of viruses causing congenital infections, respiratory diseases in early life, and HIV. Cytomegalovirus (CMV) remains the most common cause of congenital defects worldwide. Measles is a vaccine-preventable disease, yet measles cases are resurging. Zika is an example of an emerging arbovirus with devastating consequences for the developing fetus and the surviving infant. Among the respiratory viruses, we will discuss influenza and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We will finish with HIV as an example of a lifelong infection without a cure or vaccine. The review will highlight (i) the impact of viral infections on fetal and infant immune development, (ii) how differences in infant and adult immune responses to infection alter disease outcome, and emphasize the invaluable contribution of pediatric NHP infection models to the design of effective treatment and prevention strategies, including vaccines, for human infants.
Collapse
Affiliation(s)
- Vidya Vijayan K. K.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Center for AIDS Research, University of North Carolina, Chapel Hill, NC, United States
- Children’s Research Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
22
|
Li J, Li X, Liu F, Jiang S, Zhang S, Yu M, Liu W, Li Z, Wang B, Wang Y. Development and characterization of HCMV recombinant subunit vaccines based on T-cell epitopes. Vaccine 2024; 42:126454. [PMID: 39418688 DOI: 10.1016/j.vaccine.2024.126454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Human cytomegalovirus (HCMV), a ubiquitous β-herpes virus, mostly causes asymptomatic infections in adults with healthy immune systems. Due to immunosuppressive therapy, solid organ transplantation (SOT) recipients are at increased risk of HCMV infection. In recent years, the interdisciplinary, filed of immunoinformatics, based on computer science, and modern immunology, has emerged. In this study, we designed three types of recombinant subunit vaccines, which are expressed by the E. coli BL21 strain according to immunoinformatics prediction. Subsequently, we evaluated the innate and cellular immune responses of recombinant subunit vaccines in vivo and/or in vitro. Flow cytometry analysis, revealed that recombinant subunit vaccines enhanced both innate and cellular immune responses in vivo and/or in vitro. We also found that the novel herb adjuvant hesperetin (HES) increased memory T cell inflation. Overall, we developed three types of recombinant subunit vaccines based on HCMV antigen fragments containing multiple T-cell epitopes and assessed the innate and cellular immune responses in vivo and/or in vitro.
Collapse
Affiliation(s)
- Jun Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Xu Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Fengjun Liu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Shasha Jiang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Shuyun Zhang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Meng Yu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Wenxuan Liu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Zonghui Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Bin Wang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China.
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China.
| |
Collapse
|
23
|
Dhital R, Flint K, Kaptsan I, Hegde S, Daloul R, Shimamura M. Virus-specific Th17 Cells Are Induced by Human Cytomegalovirus after Renal Transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1703-1712. [PMID: 39423238 PMCID: PMC11573647 DOI: 10.4049/jimmunol.2300742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
CMV infection and Th17 cells are independently associated with increased risk for late allograft loss after renal transplantation. Although CMV-specific Th17 cells are detectable in animal models and nontransplant clinical populations, evidence linking CMV and Th17 cells after renal transplantation remains unclear. This prospective observational study evaluated a cohort of renal transplant recipients during 12 mo posttransplant to assess the presence of CMV-specific Th17 cells in peripheral blood and their relationship to pretransplant CMV serostatus and CMV DNAemia. CMV-specific Th17 cells were identified among CMV serostatus donor (D)+ and/or recipient (R)+ recipients and expanded during both primary (D+/R-) and reactivated (D+/R+, D-/R+) CMV DNAemia. A subset of CMV-specific Th17 cells coexpressed IFN-γ, indicating a Th1/17 phenotype. These Th17 and Th1/17 cells expressed CCR6, CCR5, activation and terminal differentiation markers (CD95, OX40, HLA-DR, CD57), and a central/effector memory phenotype. CMV-specific Th1/17 cells expressed activating/inhibitory receptors (CD57, 4-1BB, CD160, CTLA-4, PD-1) at higher frequencies than Th17 cells. In contrast, staphylococcal enterotoxin B-induced Th17 cells did not expand during CMV DNAemia, did not differ between CMV serostatus groups over time, expressed CCR6, predominantly coexpressed TNF-α, and had lower expression of activating and inhibitory receptors than pp65-specific Th17 and Th1/17 cells. These data show that CMV-specific Th17 cells expand during episodes of CMV DNAemia among renal transplant recipients, and that these virus-specific Th17 and Th1/17 cells have distinct phenotypes from global circulating Th(1)/17 cells. These results suggest a potential proinflammatory pathway by which CMV-induced Th17 cells may contribute to allograft injury, increasing risk for late allograft loss.
Collapse
Affiliation(s)
- Ravi Dhital
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
| | - Kaitlyn Flint
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
| | - Irina Kaptsan
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
| | - Shweta Hegde
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
| | - Reem Daloul
- Division of Transplant Nephrology, Comprehensive Transplant Center, The Ohio State University, Columbus OH
| | - Masako Shimamura
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
- Division of Pediatric Infectious Diseases, Department of Pediatrics, The Ohio State University, Columbus OH
| |
Collapse
|
24
|
Li Y, Xiao J, Li C, Yang M. Memory inflation: Beyond the acute phase of viral infection. Cell Prolif 2024; 57:e13705. [PMID: 38992867 PMCID: PMC11628752 DOI: 10.1111/cpr.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Memory inflation is confirmed as the most commonly dysregulation of host immunity with antigen-independent manner in mammals after viral infection. By generating large numbers of effector/memory and terminal differentiated effector memory CD8+ T cells with diminished naïve subsets, memory inflation is believed to play critical roles in connecting the viral infection and the onset of multiple diseases. Here, we reviewed the current understanding of memory inflated CD8+ T cells in their distinct phenotypic features that different from exhausted subsets; the intrinsic and extrinsic roles in regulating the formation of memory inflation; and the key proteins in maintaining the expansion and proliferation of inflationary populations. More importantly, based on the evidences from both clinic and animal models, we summarized the potential mechanisms of memory inflation to trigger autoimmune neuropathies, such as Guillain-Barré syndrome and multiple sclerosis; the correlations of memory inflation between tumorigenesis and resistance of tumour immunotherapies; as well as the effects of memory inflation to facilitate vascular disease progression. To sum up, better understanding of memory inflation could provide us an opportunity to beyond the acute phase of viral infection, and shed a light on the long-term influences of CD8+ T cell heterogeneity in dampen host immune homeostasis.
Collapse
Affiliation(s)
- Yanfei Li
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
| | - Jie Xiao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Mu Yang
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
25
|
Pyöriä L, Pratas D, Toppinen M, Simmonds P, Hedman K, Sajantila A, Perdomo M. Intra-host genomic diversity and integration landscape of human tissue-resident DNA virome. Nucleic Acids Res 2024; 52:13073-13093. [PMID: 39436041 PMCID: PMC11602146 DOI: 10.1093/nar/gkae871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/13/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
The viral intra-host genetic diversities and interactions with the human genome during decades of persistence remain poorly characterized. In this study, we analyzed the variability and integration sites of persisting viruses in nine organs from thirteen individuals who died suddenly from non-viral causes. The viruses studied included parvovirus B19, six herpesviruses, Merkel cell (MCPyV) and JC polyomaviruses, totaling 127 genomes. The viral sequences across organs were remarkably conserved within each individual, suggesting that persistence stems from single dominant strains. This indicates that intra-host viral evolution, thus far inferred primarily from immunocompromised patients, is likely overestimated in healthy subjects. Indeed, we detected increased viral subpopulations in two individuals with putative reactivations, suggesting that replication status influences diversity. Furthermore, we identified asymmetrical mutation patterns reflecting selective pressures exerted by the host. Strikingly, our analysis revealed non-clonal viral integrations even in individuals without cancer. These included MCPyV integrations and truncations resembling clonally expanded variants in Merkel cell carcinomas, as well as novel junctions between herpesvirus 6B and mitochondrial sequences, the significance of which remains to be evaluated. Our work systematically characterizes the genomic landscape of the tissue-resident virome, highlighting potential deviations occurring during disease.
Collapse
Affiliation(s)
- Lari Pyöriä
- Department of Virology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, P.O. Box 21, FI-00014, Helsinki, Finland
| | - Diogo Pratas
- Department of Virology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, P.O. Box 21, FI-00014, Helsinki, Finland
- IEETA, Institute of Electronics and Informatics Engineering of Aveiro, and LASI, Intelligent Systems Associate Laboratory, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
- Department of Electronics, Telecommunications and Informatics, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Mari Toppinen
- Department of Forensic Medicine, University of Helsinki, Haartmaninkatu 3, P.O. Box 21, FI-00014, Helsinki, Finland
| | - Peter Simmonds
- Nuffield Department of Medicine, University of Oxford, Peter Medawar Building, South Parks Road, OX1 3SY, Oxford, UK
| | - Klaus Hedman
- Department of Virology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, P.O. Box 21, FI-00014, Helsinki, Finland
| | - Antti Sajantila
- Department of Forensic Medicine, University of Helsinki, Haartmaninkatu 3, P.O. Box 21, FI-00014, Helsinki, Finland
- Forensic Medicine Unit, Finnish Institute for Health and Welfare, Mannerheimintie 166 A, P.O. Box 30, FI-00271, Helsinki, Finland
| | - Maria F Perdomo
- Department of Virology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, P.O. Box 21, FI-00014, Helsinki, Finland
| |
Collapse
|
26
|
Semmes EC, Nettere DR, Nelson AN, Hurst JH, Cain DW, Burt TD, Kurtzberg J, Reeves RK, Coyne CB, Fouda GG, Pollara J, Permar SR, Walsh KM. In utero human cytomegalovirus infection expands NK-like FcγRIII+CD8+ T cells that mediate Fc antibody functions. J Clin Invest 2024; 135:e181342. [PMID: 39531313 PMCID: PMC11684805 DOI: 10.1172/jci181342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Human cytomegalovirus (HCMV) profoundly impacts host T and NK cells across the lifespan, yet how this common congenital infection modulates developing fetal immune cell compartments remains underexplored. Using cord blood from neonates with and without congenital HCMV (cCMV) infection, we identify an expansion of Fcγ receptor III-expressing (FcγRIII-expressing) CD8+ T cells following HCMV exposure in utero. Most FcγRIII+CD8+ T cells express the canonical αβ T cell receptor (TCR), but a proportion express noncanonical γδ TCR. FcγRIII+CD8+ T cells are highly differentiated and have increased expression of NK cell markers and cytolytic molecules. Transcriptional analysis reveals FcγRIII+CD8+ T cells upregulate T-bet and downregulate BCL11B, known transcription factors that govern T/NK cell fate. We show that FcγRIII+CD8+ T cells mediate antibody-dependent IFN-γ production and degranulation against IgG-opsonized target cells, similar to NK cell antibody-dependent cellular cytotoxicity (ADCC). FcγRIII+CD8+ T cell Fc effector functions were further enhanced by IL-15, as has been observed in neonatal NK cells. Our study reveals that FcγRIII+CD8+ T cells elicited in utero by HCMV infection can execute Fc-mediated effector functions bridging cellular and humoral immunity and may be a promising target for antibody-based therapeutics and vaccination in early life.
Collapse
Affiliation(s)
- Eleanor C. Semmes
- Boston Children’s Hospital/Boston Medical Center, Boston, Massachusetts, USA
- Medical Scientist Training Program, and
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Danielle R. Nettere
- Medical Scientist Training Program, and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ashley N. Nelson
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jillian H. Hurst
- Children’s Health and Discovery Initiative
- Division of Infectious Diseases, and
| | - Derek W. Cain
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Trevor D. Burt
- Children’s Health and Discovery Initiative
- Division of Neonatology, Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Children’s Health and Discovery Initiative
- Carolinas Cord Blood Bank, Marcus Center for Cellular Cures, Durham, North Carolina, USA
| | - R. Keith Reeves
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Human Systems Immunology, and
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Carolyn B. Coyne
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Children’s Health and Discovery Initiative
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Children’s Health and Discovery Initiative
- Division of Infectious Diseases, and
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Kyle M. Walsh
- Children’s Health and Discovery Initiative
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| |
Collapse
|
27
|
Rollman TB, Berkebile ZW, Hicks DM, Hatfield JS, Chauhan P, Pravetoni M, Schleiss MR, Milligan GN, Morgan TK, Bierle CJ. CD4+ but not CD8+ T cells are required for protection against severe guinea pig cytomegalovirus infections. PLoS Pathog 2024; 20:e1012515. [PMID: 39495799 PMCID: PMC11563410 DOI: 10.1371/journal.ppat.1012515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/14/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus and the leading cause of infectious disease related birth defects worldwide. How the immune response modulates the risk of intrauterine transmission of HCMV after maternal infection remains poorly understood. Maternal T cells likely play a critical role in preventing infection at the maternal-fetal interface and limiting spread across the placenta, but concerns exist that immune responses to infection may also cause placental dysfunction and adverse pregnancy outcomes. This study investigated the role of CD4+ and CD8+ T cells in a guinea pig model of primary cytomegalovirus infection. Monoclonal antibodies specific to guinea pig CD4 and CD8 were used to deplete T cells in non-pregnant and in pregnant guinea pigs after mid-gestation. CD4+ T cell depletion increased the severity of illness, caused significantly elevated viral loads, and increased the rate of congenital guinea pig cytomegalovirus (GPCMV) infection relative to animals treated with control antibody. CD8+ T cell depletion was comparably well tolerated and did not significantly affect the weight of infected guinea pigs or viral loads in their blood or tissue. However, significantly more viral genomes and transcripts were detected in the placenta and decidua of CD8+ T cell depleted dams post-infection. This study corroborates earlier findings made in nonhuman primates that maternal CD4+ T cells play a critical role in limiting the severity of primary CMV infection during pregnancy while also revealing that other innate and adaptive immune responses can compensate for an absent CD8+ T cell response in α-CD8-treated guinea pigs.
Collapse
Affiliation(s)
- Tyler B. Rollman
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zachary W. Berkebile
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Dustin M. Hicks
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jason S. Hatfield
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Priyanka Chauhan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Marco Pravetoni
- Center for Medication Development for Substance Use Disorders and Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Mark R. Schleiss
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Gregg N. Milligan
- Division of Vaccinology, Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Terry K. Morgan
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Craig J. Bierle
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
28
|
Zhang W, Nilles TL, Bream JH, Li H, Malash E, Langan S, Leng SX, Margolick JB. Breadth and polyfunctionality of T cell responses to human cytomegalovirus in men who have sex with men: relationship with HIV infection and frailty. J Virol 2024; 98:e0116724. [PMID: 39230302 PMCID: PMC11494896 DOI: 10.1128/jvi.01167-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
Cytomegalovirus (CMV)-seropositive adults have large T cell responses to a wide range of CMV proteins; these responses have been associated with chronic inflammation and frailty in people with or without HIV infection. We analyzed the relationships between chronic HIV infection, frailty, and the breadth and polyfunctionality of CD4 and CD8 T cell responses to CMV. Peripheral blood mononuclear cells from 42 men (20 without HIV and 22 with virologically suppressed HIV) in the Multicenter AIDS Cohort Study (MACS) were stimulated with peptide pools spanning 19 CMV open reading frames (ORFs). As measured by flow cytometry and intracellular cytokine staining for IFN-γ, TNF-α, and IL-2, CD8 T cells from men with HIV responded to significantly more CMV ORFs than those from men without HIV. This was primarily due to a broader response to ORFs that are expressed during the late phase of CMV replication. The number of ORFs to which a participant's T cells responded was positively correlated with the sum of all that individual's T cell responses; these correlations were weaker in men with than without HIV. Polyfunctional CMV-specific CD4 responses (production of more than one cytokine) were significantly lower in men with than without HIV. Frailty status did not substantially affect the breadth or magnitude of the CMV-specific T cell responses. These results suggest that immune control of CMV infection is affected more by chronic HIV infection than by frailty. The differences between men with and without HIV were similar to those reported between young and older adults without HIV. IMPORTANCE T cell responses to chronic cytomegalovirus (CMV) infection have significant biological and clinical implications in HIV infection and aging. Here, we systematically analyzed the breadth, magnitude, and polyfunctionality of T cell responses to multiple CMV antigens in men with and without HIV in the Multicenter AIDS Cohort Study (MACS), a longstanding study of the natural and treated history of HIV-1 infection in men who have sex with men. We found that the breadth and polyfunctionality of T cell responses to CMV were different between men with chronic, treated HIV and those without HIV. The reason for these differences is unknown, but these findings suggest that people with treated HIV may have more frequent CMV reactivation than people without HIV. Differences between people with and without HIV also resembled differences reported between young and older adults without HIV, supporting a role for the immune responses to CMV in the aging process.
Collapse
Affiliation(s)
- Weiying Zhang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Tricia L. Nilles
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jay H. Bream
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Huifen Li
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Johns Hopkins Center on Aging and Immune Remodeling, Baltimore, Maryland, USA
| | - Eslam Malash
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Johns Hopkins Center on Aging and Immune Remodeling, Baltimore, Maryland, USA
| | - Susan Langan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sean X. Leng
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Johns Hopkins Center on Aging and Immune Remodeling, Baltimore, Maryland, USA
| | - Joseph B. Margolick
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Malouli D, Taher H, Mansouri M, Iyer RF, Reed J, Papen C, Schell JB, Beechwood T, Martinson T, Morrow D, Hughes CM, Gilbride RM, Randall K, Ford JC, Belica K, Ojha S, Sacha JB, Bimber BN, Hansen SG, Picker LJ, Früh K. Human cytomegalovirus UL18 prevents priming of MHC-E- and MHC-II-restricted CD8 + T cells. Sci Immunol 2024; 9:eadp5216. [PMID: 39392895 PMCID: PMC11797217 DOI: 10.1126/sciimmunol.adp5216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024]
Abstract
Rhesus cytomegalovirus (RhCMV) vectors elicit major histocompatibility complex (MHC)-E-restricted CD8+ T cells that stringently control simian immunodeficiency virus (SIV) in rhesus macaques. These responses require deletion of eight RhCMV chemokine-like open reading frames (ORFs) that are conserved in human cytomegalovirus (HCMV). To determine whether HCMV encodes additional, nonconserved inhibitors of unconventional T cell priming, we inserted 41 HCMV-specific ORFs into a chemokine-deficient strain (68-1 RhCMV). Monitoring of epitope recognition revealed that HCMV UL18 prevented unconventional T cell priming, resulting in MHC-Ia-targeted responses. UL18 is homologous to MHC-I but does not engage T cell receptors and, instead, binds with high affinity to inhibitory leukocyte immunoglobulin-like receptor-1 (LIR-1). UL18 lacking LIR-1 binding no longer interfered with MHC-E-restricted T cell stimulation by RhCMV-infected cells or the induction of unconventionally restricted T cells. Thus, LIR-1 binding needs to be deleted from UL18 of HCMV/HIV vaccines to allow for the induction of protective MHC-E-restricted T cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Scott G. Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
30
|
Watanabe M, Davidson L, Smith P, Castellucio PF, Jergovic M, Uhrlaub JL, Smithey MJ, Fantry LE, Dechambre B, Wilson RC, Knox KC, Ren J, Stowe RP, Weinstock G, Twigg H, Nikolich JŽ. Anti-cytomegalovirus antibody levels stratify human immune profiles across the lifespan. GeroScience 2024; 46:4225-4242. [PMID: 38512581 PMCID: PMC11336022 DOI: 10.1007/s11357-024-01124-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Human cytomegalovirus (hCMV) is a ubiquitous latent persistent herpesvirus infecting 60-90% of the population worldwide. hCMV carriage in immunocompetent people is asymptomatic; thus, hCMV can be considered a component of normative aging. However, hCMV powerfully modulates many features of the immune, and likely other, systems and organs. Questions remain as to how hCMV carriage affects the human host. We used anti-CMV antibody titers as a stratifying criterion to examine the impact of "intensity" of hCMV infection as a potential biomarker of aging, inflammation, and immune homeostasis in a cohort of 247 participants stratified into younger (21-40 years) and older (> 65 years of age) groups. We showed that anti-CMV antibody titers increased with age and directly correlated to increased levels of soluble tumor necrosis factor (sTNFR) I in younger but not older participants. CD8 + cell numbers were reduced in the older group due to the loss in CD8 + T naïve (Tn) cells. In CMV carriers and, in particular, in anti-CMV Ab-high participants, this loss was mitigated or reversed by an increase in the numbers of CD8 + T effector memory (Tem) and T effector memory reexpressing CD45RA (Temra) cells. Analysis of CD38, HLA-DR, and CD57 expression revealed subset (CD4 or CD8)-specific changes that correlated with anti-CMV Ab levels. In addition, anti-CMV Ab levels predicted anti-CMV CD8 T cell responsiveness to different CMV open reading frames (ORFs) selectively in older participants, which correlated to the transcriptional order of expression of specific CMV ORFs. Implications of these results for the potential predictive value of anti-CMV Ab titers during aging are discussed.
Collapse
Affiliation(s)
- Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Lisa Davidson
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Patricia Smith
- Division of Pulmonary Medicine, Department of Medicine, Indiana University College of Medicine, Bloomington, IN, USA
| | - Peter F Castellucio
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mladen Jergovic
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Megan J Smithey
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Lori E Fantry
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Brett Dechambre
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Department of Health Services, Phoenix, AZ, USA
| | - Rachel C Wilson
- Division of Pulmonary Medicine, Department of Medicine, Indiana University College of Medicine, Bloomington, IN, USA
| | - Kenneth C Knox
- Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Jie Ren
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Homer Twigg
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA.
| |
Collapse
|
31
|
Coplen CP, Ashok Sonar S, Nikolich JŽ. Late-life Attenuation of Cytomegalovirus-mediated CD8 T Cell Memory Inflation: Shrinking of the Cytomegalovirus Latency Niche. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:965-970. [PMID: 39150241 PMCID: PMC11463719 DOI: 10.4049/jimmunol.2400113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024]
Abstract
CMV drives the accumulation of virus-specific, highly differentiated CD8 memory T cells (memory inflation [MI]). In mice, MI was shown to directly correlate with the CMV infection dose, yet the CMV-associated CD8 MI plateaus over time. It is unclear how MI is regulated with aging. We infected young mice with 102, 104, and 106 PFU of murine CMV and confirmed that MI magnitude was directly proportional to the infectious dose, reaching a setpoint by midlife. By old age, MI subsided, most prominently in mice infected with 106 PFU, and reached statistical parity between groups in 26-mo-old mice. This corresponded to an age-related loss in lymphatic endothelial cells in lymph nodes, recently shown to be sufficient to drive MI in mice. We propose that MI size and persistence over the lifespan is controlled by the size of the lymphatic endothelial cell niche, whose shrinking leads to reduced MI with aging.
Collapse
Affiliation(s)
- Christopher P. Coplen
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- the University of Arizona Center on Aging, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Sandip Ashok Sonar
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- the University of Arizona Center on Aging, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Janko Ž. Nikolich
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- the University of Arizona Center on Aging, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- Aegis Consortium for Pandemic-free Future, University of Arizona Health Sciences, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
32
|
Mercado NB, Real JN, Kaiserman J, Panagioti E, Cook CH, Lawler SE. Clinical implications of cytomegalovirus in glioblastoma progression and therapy. NPJ Precis Oncol 2024; 8:213. [PMID: 39343770 PMCID: PMC11439950 DOI: 10.1038/s41698-024-00709-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Glioblastoma (GBM) is one of the deadliest brain cancers with a median survival of only 15 months. This poor prognosis has prompted exploration of novel therapeutic targets for GBM patients. Human cytomegalovirus (HCMV) has been implicated in GBM; however, its impact remains poorly defined, and there is conflicting data over the presence of HCMV in tumors. Nonetheless, clinical trials targeting HCMV have shown promising initial data, and evidence suggests that HCMV may negatively impact GBM patient survival by multiple mechanisms including changes in GBM cell behavior and the tumor microenvironment (TME) that potentiate tumor progression as well as therapy-induced virus reactivation. Moreover, HCMV has many effects on host immunity that could impact tumor behavior by altering the TME, which are largely unexplored. The goal of this review is to describe these potential interactions between HCMV and GBM. Better understanding of these processes may allow the development of new therapeutic modalities to improve GBM patient outcomes.
Collapse
Affiliation(s)
- Noe B Mercado
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, US
- The Warren Alpert Medical School, Brown University, Providence, RI, US
| | - Jacqueline N Real
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, US
- The Warren Alpert Medical School, Brown University, Providence, RI, US
| | - Jacob Kaiserman
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, US
- The Warren Alpert Medical School, Brown University, Providence, RI, US
| | - Eleni Panagioti
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Charles H Cook
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Sean E Lawler
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, US.
- The Warren Alpert Medical School, Brown University, Providence, RI, US.
| |
Collapse
|
33
|
Medoro AK, Dhital R, Sánchez PJ, Flint K, Graber B, Pifer T, Crisan R, Ray WC, Phelps CC, Honegger JR, Peng J, Findlen U, Malhotra P, Adunka O, Shimamura M. T cell responses and clinical symptoms among infants with congenital cytomegalovirus infection. JCI Insight 2024; 9:e171029. [PMID: 39315550 PMCID: PMC11457853 DOI: 10.1172/jci.insight.171029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/07/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUNDCongenital cytomegalovirus (cCMV) infection can cause developmental impairment and sensorineural hearing loss (SNHL). To determine the relationship between immune responses to cCMV infection and neurologic sequelae, T cell responses were compared for their connection to clinical symptoms at birth and neurodevelopmental outcomes.METHODSThirty cCMV-infected and 15 uninfected infants were enrolled in a single-center prospective observational case-control study. T cell pp65-specific cytokine responses; CD57, CD28, and PD-1 expression; and memory subsets were compared.RESULTSInfected neonates (73% symptomatic at birth) lacked pp65-specific cytokine-secreting T cells, with elevated frequencies of CD57+, CD28-, and PD-1+CD8+ T cells and effector memory subsets. Though frequencies overlapped between cCMV symptom groups, asymptomatic infants had higher frequencies of CD57+PD-1+CD8+ T cells. Neonates with subsequent developmental delay lacked detectable CMV-specific T cell responses, with patterns resembling those of uninfected infants. Two children with progressive SNHL had high frequencies of PD-1+CD8+ T cells over the first year compared with children without progressive SNHL.CONCLUSIONSimilar to published reports, neonatal viral antigen-specific cytokine-secreting T cell responses were not detected, but overall patterns indicate that globally differentiated memory CD8+ T cell populations were induced by cCMV infection, with higher frequencies of terminally differentiated PD-1+CD8+ T cells potentially associated with asymptomatic infection. In this cohort, a lack of in utero T cell differentiation was associated with developmental delay, and high frequencies of PD-1+CD8+ T cells persisted only in children with progressive SNHL. Further work is needed to define the specificity of these T cells and their mechanistic connection to these outcomes.FUNDINGThis study was funded through an intramural research award at Nationwide Children's Hospital, the Pediatric Infectious Disease Society Fellowship Award funded by Stanley and Susan Plotkin and Sanofi Pasteur, the Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Pichichero Family Foundation Vaccines for Children Initiative Research Award from the Pediatric Infectious Diseases Society Foundation.
Collapse
Affiliation(s)
- Alexandra K. Medoro
- Department of Pediatrics, Division of Infectious Diseases, and
- Department of Pediatrics, Division of Neonatology, Nationwide Children’s Hospital and The Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | - Pablo J. Sánchez
- Department of Pediatrics, Division of Infectious Diseases, and
- Department of Pediatrics, Division of Neonatology, Nationwide Children’s Hospital and The Ohio State University College of Medicine, Columbus, Ohio, USA
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | | | | | - Traci Pifer
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Rachelle Crisan
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - William C. Ray
- Interdisciplinary Graduate Program in Biophysics, The Ohio State University, Columbus Ohio, USA
- IT Research and Innovations Group, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | | | - Jonathan R. Honegger
- Department of Pediatrics, Division of Infectious Diseases, and
- Center for Vaccines and Immunity and
| | - Jing Peng
- Center for Clinical and Translational Science, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Center for Biostatistics, The Ohio State University, Columbus, Ohio, USA
| | | | - Prashant Malhotra
- Department of Otolaryngology, Nationwide Children’s Hospital and The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Oliver Adunka
- Department of Otolaryngology, Nationwide Children’s Hospital and The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Masako Shimamura
- Department of Pediatrics, Division of Infectious Diseases, and
- Center for Vaccines and Immunity and
| |
Collapse
|
34
|
Zamora D, Dasgupta S, Stevens-Ayers T, Edmison B, Winston DJ, Razonable RR, Mehta AK, Lyon GM, Boeckh M, Singh N, Koelle DM, Limaye AP. Cytomegalovirus immunity in high-risk liver transplant recipients following preemptive antiviral therapy versus prophylaxis. JCI Insight 2024; 9:e180115. [PMID: 39099206 PMCID: PMC11457861 DOI: 10.1172/jci.insight.180115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024] Open
Abstract
CMV-specific T cells, NK cells, and neutralizing antibodies (nAbs) were assessed in a randomized trial of CMV prevention with preemptive antiviral therapy (PET) versus prophylactic antiviral therapy (PRO) in donor-seropositive/recipient-seronegative (D+R-) liver transplant recipients (LTxR) at 100 days (end of intervention) and at 6 and 12 months after transplant. The PET group had significantly increased numbers of circulating polyfunctional T cells, NK cells, and nAbs compared with the PRO group at day 100, and several CMV immune parameters remained significantly higher by 12 months after transplant. Among PET recipients, preceding CMV viremia (vs. no preceding viremia) was associated with significantly higher levels of most CMV immune parameters at day 100. Higher numbers of CMV-specific polyfunctional T cells and NKG2C+ NK cells at day 100 were associated with a decreased incidence of CMV disease in multivariable Cox regression. The strongest associations with protection against CMV disease were with increased numbers of CMV-specific polyfunctional CD4+ T cells, CD3negCD56dimCD57negNKG2Cpos cells, and CD3negCD56dimCD57posNKG2Cpos NK cells. Our results suggest that PET is superior to PRO for CMV disease prevention by allowing low-level CMV replication and associated antigen exposure that is promptly controlled by antiviral therapy and facilitates enhanced CMV protective immunity in D+R- LTxR.
Collapse
Affiliation(s)
- Danniel Zamora
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sayan Dasgupta
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Terry Stevens-Ayers
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Bradley Edmison
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Drew J. Winston
- Division of Infectious Diseases, UCLA Medical Center, Los Angeles, California, USA
| | - Raymund R. Razonable
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Aneesh K. Mehta
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - G. Marshall Lyon
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael Boeckh
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Nina Singh
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Transplant Infectious Diseases, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - David M. Koelle
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Global Health and
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Benaroya Research Institute, Seattle, Washington, USA
| | - Ajit P. Limaye
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| |
Collapse
|
35
|
Al-Talib M, Dimonte S, Humphreys IR. Mucosal T-cell responses to chronic viral infections: Implications for vaccine design. Cell Mol Immunol 2024; 21:982-998. [PMID: 38459243 PMCID: PMC11364786 DOI: 10.1038/s41423-024-01140-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/31/2024] [Indexed: 03/10/2024] Open
Abstract
Mucosal surfaces that line the respiratory, gastrointestinal and genitourinary tracts are the major interfaces between the immune system and the environment. Their unique immunological landscape is characterized by the necessity of balancing tolerance to commensal microorganisms and other innocuous exposures against protection from pathogenic threats such as viruses. Numerous pathogenic viruses, including herpesviruses and retroviruses, exploit this environment to establish chronic infection. Effector and regulatory T-cell populations, including effector and resident memory T cells, play instrumental roles in mediating the transition from acute to chronic infection, where a degree of viral replication is tolerated to minimize immunopathology. Persistent antigen exposure during chronic viral infection leads to the evolution and divergence of these responses. In this review, we discuss advances in the understanding of mucosal T-cell immunity during chronic viral infections and how features of T-cell responses develop in different chronic viral infections of the mucosa. We consider how insights into T-cell immunity at mucosal surfaces could inform vaccine strategies: not only to protect hosts from chronic viral infections but also to exploit viruses that can persist within mucosal surfaces as vaccine vectors.
Collapse
Affiliation(s)
- Mohammed Al-Talib
- Systems Immunity University Research Institute/Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
- Bristol Medical School, University of Bristol, 5 Tyndall Avenue, Bristol, BS8 1UD, UK
| | - Sandra Dimonte
- Systems Immunity University Research Institute/Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Ian R Humphreys
- Systems Immunity University Research Institute/Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.
| |
Collapse
|
36
|
Ekman I, Schroderus AM, Vuorinen T, Knip M, Veijola R, Toppari J, Ilonen J, Lempainen J, Kinnunen T. The effect of early life cytomegalovirus infection on the immune profile of children. Clin Immunol 2024; 266:110330. [PMID: 39067678 DOI: 10.1016/j.clim.2024.110330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/05/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Cytomegalovirus (CMV) infection has a life-long impact on the immune system, particularly on memory T cells. However, the effect of early life CMV infection on the phenotype and functionality of T cells in infants and especially longitudinal changes occurring during childhood have not been explored in detail. The phenotype and functionality of peripheral blood CD8+ and CD4+ T cells from children infected with CMV in early life (< 6 months of age) was analyzed using high-dimensional flow cytometry. Samples from CMV IgG-seropositive (CMV+) children were collected at 6 months and 6 years of age and compared to samples from CMV-seronegative (CMV-) children. Early life CMV infection caused multiple alterations within T cells. These include downregulation of CD28 expression and upregulation of CD57 expression within both CD27+ early and CD27- late effector memory CD8+ and CD4+ T-cells at 6 months of age. Of these changes, only alterations within the highly differentiated late effector memory compartment persisted at the age of 6 years. Early life CMV-infection has a distinct impact on developing CD8+ and CD4+ memory T cell compartments. It appears to induce both temporary as well as longer-lasting alterations, which may affect the functionality of the immune system throughout life.
Collapse
Affiliation(s)
- Ilse Ekman
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Mari Schroderus
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tytti Vuorinen
- Institute of Biomedicine, University of Turku, Turku, Finland; Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Mikael Knip
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland; Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Riitta Veijola
- PEDEGO Research Unit, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jorma Toppari
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Research Centre for Integrative Physiology and Pharmacology, and Centre for Population Health Research, InFLAMES Research Flagship, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Johanna Lempainen
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland; Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; ISLAB Laboratory Centre, Kuopio, Finland.
| |
Collapse
|
37
|
Santos LC, Fernandes AMS, Alves IA, Serafini MR, Silva LDSE, de Freitas HF, Leite LCC, Santos CC. Trends in Viral Vector-Based Vaccines for Tuberculosis: A Patent Review (2010-2023). Vaccines (Basel) 2024; 12:876. [PMID: 39204002 PMCID: PMC11359462 DOI: 10.3390/vaccines12080876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Tuberculosis (TB) is an ancient global public health problem. Several strategies have been applied to develop new and more effective vaccines against TB, from attenuated or inactivated mycobacteria to recombinant subunit or genetic vaccines, including viral vectors. This review aimed to evaluate patents filed between 2010 and 2023 for TB vaccine candidates. It focuses on viral vector-based strategies. A search was carried out in Espacenet, using the descriptors "mycobacterium and tuberculosis" and the classification A61K39. Of the 411 patents preliminarily identified, the majority were related to subunit vaccines, with 10 patents based on viral vector platforms selected in this study. Most of the identified patents belong to the United States or China, with a concentration of patent filings between 2013 and 2023. Adenoviruses were the most explored viral vectors, and the most common immunodominant Mycobacterium tuberculosis (Mtb) antigens were present in all the selected patents. The majority of patents were tested in mouse models by intranasal or subcutaneous route of immunization. In the coming years, an increased use of this platform for prophylactic and/or therapeutic approaches for TB and other diseases is expected. Along with this, expanding knowledge about the safety of this technology is essential to advance its use.
Collapse
Affiliation(s)
- Lana C. Santos
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | - Antônio Márcio Santana Fernandes
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | - Izabel Almeida Alves
- Departamento do Medicamento, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil;
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Estado da Bahia, Salvador 41150-000, BA, Brazil
| | - Mairim Russo Serafini
- Departamento de Farmácia, Universidade Federal do Sergipe, São Cristóvão 49100-000, SE, Brazil;
| | - Leandra da Silva e Silva
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | | | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, SP, Brazil;
| | - Carina C. Santos
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil
| |
Collapse
|
38
|
Hamdan TA. The Multifaceted Roles of NK Cells in the Context of Murine Cytomegalovirus and Lymphocytic Choriomeningitis Virus Infections. Immune Netw 2024; 24:e29. [PMID: 39246620 PMCID: PMC11377952 DOI: 10.4110/in.2024.24.e29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 09/10/2024] Open
Abstract
NK cells belong to innate lymphoid cells and able to eliminate infected cells and tumor cells. NK cells play a valuable role in controlling viral infections. Also, they have the potential to shape the adaptive immunity via a unique crosstalk with the different immune cells. Murine models are important tools for delineating the immunological phenomena in viral infection. To decipher the immunological virus-host interactions, two major infection models are being investigated in mice regarding NK cell-mediated recognition: murine cytomegalovirus (MCMV) and lymphocytic choriomeningitis virus (LCMV). In this review, we recapitulate recent findings regarding the multifaceted role of NK cells in controlling LCMV and MCMV infections and outline the exquisite interplay between NK cells and other immune cells in these two settings. Considering that, infections with MCMV and LCMV recapitulates many physiopathological characteristics of human cytomegalovirus infection and chronic virus infections respectively, this study will extend our understanding of NK cells biology in interactions between the virus and its natural host.
Collapse
Affiliation(s)
- Thamer A Hamdan
- Department of Basic Dental Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman 19328, Jordan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan
| |
Collapse
|
39
|
Yang X, Jiang S, Liu F, Li Z, Liu W, Zhang X, Nan F, Li J, Yu M, Wang Y, Wang B. HCMV IE1/IE1mut Therapeutic Vaccine Induces Tumor Regression via Intratumoral Tertiary Lymphoid Structure Formation and Peripheral Immunity Activation in Glioblastoma Multiforme. Mol Neurobiol 2024; 61:5935-5949. [PMID: 38261253 PMCID: PMC11249408 DOI: 10.1007/s12035-024-03937-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 01/07/2024] [Indexed: 01/24/2024]
Abstract
Glioblastoma multiforme (GBM), a highly malignant invasive brain tumor, is associated with poor prognosis and survival and lacks an effective cure. High expression of the human cytomegalovirus (HCMV) immediate early protein 1 (IE1) in GBM tissues is strongly associated with their malignant progression, presenting a novel target for therapeutic strategies. Here, the bioluminescence imaging technology revealed remarkable tumor shrinkage and improved survival rates in a mouse glioma model treated with HCMV IE1/IE1mut vaccine. In addition, immunofluorescence data demonstrated that the treated group exhibited significantly more and larger tertiary lymphoid structures (TLSs) than the untreated group. The presence of TLS was associated with enhanced T cell infiltration, and a large number of proliferating T cells were found in the treated group. Furthermore, the flow cytometry results showed that in the treatment group, cytotoxic T lymphocytes exhibited partial polarization toward effector memory T cells and were activated to play a lethal role in the peripheral immunological organs. Furthermore, a substantial proportion of B cells in the draining lymph nodes expressed CD40 and CD86. Surprisingly, quantitative polymerase chain reaction indicated that a high expression of cytokines, including chemokines in brain tumors and immune tissues, induced the differentiation, development, and chemokine migration of immune cells in the treated group. Our study data demonstrate that IE1 or IE1mut vaccination has a favorable effect in glioma mice models. This study holds substantial implications for identifying new and effective therapeutic targets within GBM.
Collapse
Affiliation(s)
- Xiaoli Yang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Shasha Jiang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fengjun Liu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zonghui Li
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenxuan Liu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xianjuan Zhang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Fulong Nan
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jun Li
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Meng Yu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Bin Wang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
40
|
Metzdorf K, Jacobsen H, Kim Y, Teixeira Alves LG, Kulkarni U, Brdovčak MC, Materljan J, Eschke K, Chaudhry MZ, Hoffmann M, Bertoglio F, Ruschig M, Hust M, Šustić M, Krmpotić A, Jonjić S, Widera M, Ciesek S, Pöhlmann S, Landthaler M, Čičin-Šain L. A single-dose MCMV-based vaccine elicits long-lasting immune protection in mice against distinct SARS-CoV-2 variants. Front Immunol 2024; 15:1383086. [PMID: 39119342 PMCID: PMC11306140 DOI: 10.3389/fimmu.2024.1383086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/11/2024] [Indexed: 08/10/2024] Open
Abstract
Current vaccines against COVID-19 elicit immune responses that are overall strong but wane rapidly. As a consequence, the necessary booster shots have contributed to vaccine fatigue. Hence, vaccines that would provide lasting protection against COVID-19 are needed, but are still unavailable. Cytomegaloviruses (CMVs) elicit lasting and uniquely strong immune responses. Used as vaccine vectors, they may be attractive tools that obviate the need for boosters. Therefore, we tested the murine CMV (MCMV) as a vaccine vector against COVID-19 in relevant preclinical models of immunization and challenge. We have previously developed a recombinant MCMV vaccine vector expressing the spike protein of the ancestral SARS-CoV-2 (MCMVS). In this study, we show that the MCMVS elicits a robust and lasting protection in young and aged mice. Notably, spike-specific humoral and cellular immunity was not only maintained but also even increased over a period of at least 6 months. During that time, antibody avidity continuously increased and expanded in breadth, resulting in neutralization of genetically distant variants, like Omicron BA.1. A single dose of MCMVS conferred rapid virus clearance upon challenge. Moreover, MCMVS vaccination controlled two variants of concern (VOCs), the Beta (B.1.135) and the Omicron (BA.1) variants. Thus, CMV vectors provide unique advantages over other vaccine technologies, eliciting broadly reactive and long-lasting immune responses against COVID-19.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Mice
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Muromegalovirus/immunology
- Muromegalovirus/genetics
- Female
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice, Inbred BALB C
- Humans
- Genetic Vectors
- Immunity, Cellular
- Immunity, Humoral
- Disease Models, Animal
Collapse
Affiliation(s)
- Kristin Metzdorf
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| | - Henning Jacobsen
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| | - Yeonsu Kim
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| | - Luiz Gustavo Teixeira Alves
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Upasana Kulkarni
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| | | | - Jelena Materljan
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Kathrin Eschke
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - M. Zeeshan Chaudhry
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center – Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Federico Bertoglio
- Department of Medical Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Maximilian Ruschig
- Department of Medical Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Michael Hust
- Department of Medical Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Marko Šustić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Astrid Krmpotić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Stipan Jonjić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- German Centre for Infection Research (DZIF), External Partner Site Frankfurt, Frankfurt, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center – Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Luka Čičin-Šain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| |
Collapse
|
41
|
Malouli D, Tiwary M, Gilbride RM, Morrow DW, Hughes CM, Selseth A, Penney T, Castanha P, Wallace M, Yeung Y, Midgett M, Williams C, Reed J, Yu Y, Gao L, Yun G, Treaster L, Laughlin A, Lundy J, Tisoncik-Go J, Whitmore LS, Aye PP, Schiro F, Dufour JP, Papen CR, Taher H, Picker LJ, Früh K, Gale M, Maness NJ, Hansen SG, Barratt-Boyes S, Reed DS, Sacha JB. Cytomegalovirus vaccine vector-induced effector memory CD4 + T cells protect cynomolgus macaques from lethal aerosolized heterologous avian influenza challenge. Nat Commun 2024; 15:6007. [PMID: 39030218 PMCID: PMC11272155 DOI: 10.1038/s41467-024-50345-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
An influenza vaccine approach that overcomes the problem of viral sequence diversity and provides long-lived heterosubtypic protection is urgently needed to protect against pandemic influenza viruses. Here, to determine if lung-resident effector memory T cells induced by cytomegalovirus (CMV)-vectored vaccines expressing conserved internal influenza antigens could protect against lethal influenza challenge, we immunize Mauritian cynomolgus macaques (MCM) with cynomolgus CMV (CyCMV) vaccines expressing H1N1 1918 influenza M1, NP, and PB1 antigens (CyCMV/Flu), and challenge with heterologous, aerosolized avian H5N1 influenza. All six unvaccinated MCM died by seven days post infection with acute respiratory distress, while 54.5% (6/11) CyCMV/Flu-vaccinated MCM survived. Survival correlates with the magnitude of lung-resident influenza-specific CD4 + T cells prior to challenge. These data demonstrate that CD4 + T cells targeting conserved internal influenza proteins can protect against highly pathogenic heterologous influenza challenge and support further exploration of effector memory T cell-based vaccines for universal influenza vaccine development.
Collapse
Affiliation(s)
- Daniel Malouli
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Meenakshi Tiwary
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Roxanne M Gilbride
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - David W Morrow
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Colette M Hughes
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Andrea Selseth
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Toni Penney
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Priscila Castanha
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Megan Wallace
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Yulia Yeung
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | | | - Connor Williams
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Jason Reed
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Yun Yu
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Lina Gao
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Gabin Yun
- Department of Diagnostic Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luke Treaster
- Department of Diagnostic Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Jennifer Tisoncik-Go
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Leanne S Whitmore
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Pyone P Aye
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Faith Schiro
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Jason P Dufour
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Courtney R Papen
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Husam Taher
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Louis J Picker
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Klaus Früh
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, 98195, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Scott G Hansen
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | | | | | - Jonah B Sacha
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA.
| |
Collapse
|
42
|
Riedl A, Bojková D, Tan J, Jeney Á, Larsen PK, Jeney C, Full F, Kalinke U, Ruzsics Z. Construction and Characterization of a High-Capacity Replication-Competent Murine Cytomegalovirus Vector for Gene Delivery. Vaccines (Basel) 2024; 12:791. [PMID: 39066429 PMCID: PMC11281640 DOI: 10.3390/vaccines12070791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
We investigated the basic characteristics of a new murine cytomegalovirus (MCMV) vector platform. Using BAC technology, we engineered replication-competent recombinant MCMVs with deletions of up to 26% of the wild-type genome. To this end, we targeted five gene blocks (m01-m17, m106-m109, m129-m141, m144-m158, and m159-m170). BACs featuring deletions from 18% to 26% of the wild-type genome exhibited delayed virus reconstitution, while smaller deletions (up to 16%) demonstrated reconstitution kinetics similar to those of the wild type. Utilizing an innovative methodology, we introduced large genomic DNA segments, up to 35 kbp, along with reporter genes into a newly designed vector with a potential cloning capacity of 46 kbp (Q4). Surprisingly, the insertion of diverse foreign DNAs alleviated the delayed plaque formation phenotype of Q4, and these large inserts remained stable through serial in vitro passages. With reporter-gene-expressing recombinant MCMVs, we successfully transduced not only mouse cell lines but also non-rodent mammalian cells, including those of human, monkey, bovine, and bat origin. Remarkably, even non-mammalian cell lines derived from chickens exhibited successful transduction.
Collapse
Affiliation(s)
- André Riedl
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Denisa Bojková
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Institute of Medical Virology, Goethe University Frankfurt, University Hospital, 60596 Frankfurt am Main, Germany
| | - Jiang Tan
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Ábris Jeney
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Pia-Katharina Larsen
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Institute for Experimental Infection Research, 30625 Hanover, Germany
| | - Csaba Jeney
- Department of Microsystems Engineering—IMTEK, University of Freiburg, 79110 Freiburg, Germany
| | - Florian Full
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Ulrich Kalinke
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Institute for Experimental Infection Research, 30625 Hanover, Germany
| | - Zsolt Ruzsics
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
43
|
Yuan Q, Fan Z, Huang W, Huo X, Yang X, Ran Y, Chen J, Li H. Human cytomegalovirus UL23 exploits PD-L1 inhibitory signaling pathway to evade T cell-mediated cytotoxicity. mBio 2024; 15:e0119124. [PMID: 38829126 PMCID: PMC11253622 DOI: 10.1128/mbio.01191-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
Human cytomegalovirus (HCMV), a widely prevalent human beta-herpesvirus, establishes lifelong persistence in the host following primary infection. In healthy individuals, the virus is effectively controlled by HCMV-specific T cells and typically exhibits asymptomatic. The T cell immune response plays a pivotal role in combating HCMV infection, while HCMV employs various strategies to counteract it within the host. Previously, we reported that UL23, a tegument protein of HCMV, facilitates viral immune evasion from interferon-gamma (IFN-γ) responses, and it is well known that IFN-γ is mainly derived from T cells. However, the involvement of UL23 in viral immune evasion from T cell-mediated immunity remains unclear. Herein, we present compelling evidence that UL23 significantly enhances viral resistance against T cell-mediated cytotoxicity during HCMV infection from the co-culture assays of HCMV-infected cells with T cells. We found that IFN-γ plays a major role in regulating T cell cytotoxicity mediated by UL23. More interestingly, we demonstrated that UL23 not only regulates the IFN-γ downstream responses but also modulates the IFN-γ secretion by regulating T cell activities. Further experiments indicate that UL23 upregulates the expression and signaling of programmed death ligand 1 (PD-L1), which is responsible for inhibiting multiple aspects of T cell activities, including activation, apoptosis, and IFN-γ secretion, as determined through RNA-seq analysis and inhibitor-blocking experiments, ultimately facilitating viral replication and spread. Our findings highlight the potential role of UL23 as an alternative antagonist in suppressing T cell cytotoxicity and unveil a novel strategy for HCMV to evade T cell immunity. IMPORTANCE T cell immunity is pivotal in controlling primary human cytomegalovirus (HCMV) infection, restricting periodic reactivation, and preventing HCMV-associated diseases. Despite inducing a robust T cell immune response, HCMV has developed sophisticated immune evasion mechanisms that specifically target T cell responses. Although numerous studies have been conducted on HCMV-specific T cells, the primary focus has been on the impact of HCMV on T cell recognition via major histocompatibility complex molecules. Our studies show for the first time that HCMV exploits the programmed death ligand 1 (PD-L1) inhibitory signaling pathway to evade T cell immunity by modulating the activities of T cells and thereby blocking the secretion of IFN-γ, which is directly mediated by HCMV-encoded tegument protein UL23. While PD-L1 has been extensively studied in the context of tumors and viruses, its involvement in HCMV infection and viral immune evasion is rarely reported. We observed an upregulation of PD-L1 in normal cells during HCMV infection and provided strong evidence supporting its critical role in UL23-induced inhibition of T cell-mediated cytotoxicity. The novel strategy employed by HCMV to manipulate the inhibitory signaling pathway of T cell immune activation for viral evasion through its encoded protein offers valuable insights for the understanding of HCMV-mediated T cell immunomodulation and developing innovative antiviral treatment strategies.
Collapse
Affiliation(s)
- Qin Yuan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
- Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhaosong Fan
- Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Wenqiang Huang
- Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiaoping Huo
- Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiaoping Yang
- Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yanhong Ran
- Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jun Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Hongjian Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
- Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
44
|
Mancebo FJ, Nuévalos M, Lalchandani J, Martín Galiano AJ, Fernández-Ruiz M, Aguado JM, García-Ríos E, Pérez-Romero P. Cytomegalovirus UL44 protein induces a potent T-cell immune response in mice. Antiviral Res 2024; 227:105914. [PMID: 38759930 DOI: 10.1016/j.antiviral.2024.105914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
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.
Collapse
Affiliation(s)
- Francisco J Mancebo
- National Center for Microbiology, Instituto de Salud Carlos III. Majadahonda, Madrid, Spain
| | - Marcos Nuévalos
- National Center for Microbiology, Instituto de Salud Carlos III. Majadahonda, Madrid, Spain
| | - Jaanam Lalchandani
- National Center for Microbiology, Instituto de Salud Carlos III. Majadahonda, Madrid, Spain
| | | | - Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre', Instituto de Investigación Biomédica Hospital "12 de Octubre' (imas12) Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre', Instituto de Investigación Biomédica Hospital "12 de Octubre' (imas12) Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Estéfani García-Ríos
- Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain.
| | - Pilar Pérez-Romero
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
45
|
Kar R, Chattopadhyay S, Sharma A, Sharma K, Sinha S, Arimbasseri GA, Patil VS. Single-cell transcriptomic and T cell antigen receptor analysis of human cytomegalovirus (hCMV)-specific memory T cells reveals effectors and pre-effectors of CD8 +- and CD4 +-cytotoxic T cells. Immunology 2024; 172:420-439. [PMID: 38501302 PMCID: PMC7616077 DOI: 10.1111/imm.13783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
Latent human cytomegalovirus (hCMV) infection can pose a serious threat of reactivation and disease occurrence in immune-compromised individuals. Although T cells are at the core of the protective immune response to hCMV infection, a detailed characterization of different T cell subsets involved in hCMV immunity is lacking. Here, in an unbiased manner, we characterized over 8000 hCMV-reactive peripheral memory T cells isolated from seropositive human donors, at a single-cell resolution by analysing their single-cell transcriptomes paired with the T cell antigen receptor (TCR) repertoires. The hCMV-reactive T cells were highly heterogeneous and consisted of different developmental and functional memory T cell subsets such as, long-term memory precursors and effectors, T helper-17, T regulatory cells (TREGs) and cytotoxic T lymphocytes (CTLs) of both CD4 and CD8 origin. The hCMV-specific TREGs, in addition to being enriched for molecules known for their suppressive functions, showed enrichment for the interferon response signature gene sets. The hCMV-specific CTLs were of two types, the pre-effector- and effector-like. The co-clustering of hCMV-specific CD4-CTLs and CD8-CTLs in both pre-effector as well as effector clusters suggest shared transcriptomic signatures between them. The huge TCR clonal expansion of cytotoxic clusters suggests a dominant role in the protective immune response to CMV. The study uncovers the heterogeneity in the hCMV-specific memory T cells revealing many functional subsets with potential implications in better understanding of hCMV-specific T cell immunity. The data presented can serve as a knowledge base for designing vaccines and therapeutics.
Collapse
Affiliation(s)
- Raunak Kar
- Immunogenomics Lab, National Institute of Immunology, New Delhi, Delhi, India
| | | | - Anjali Sharma
- Department of Transfusion Medicine and Blood Bank, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, Delhi, India
| | - Kirti Sharma
- Immunogenomics Lab, National Institute of Immunology, New Delhi, Delhi, India
| | - Shreya Sinha
- Immunogenomics Lab, National Institute of Immunology, New Delhi, Delhi, India
| | | | - Veena S. Patil
- Immunogenomics Lab, National Institute of Immunology, New Delhi, Delhi, India
| |
Collapse
|
46
|
Andres-Martin F, James C, Catalfamo M. IL-27 expression regulation and its effects on adaptive immunity against viruses. Front Immunol 2024; 15:1395921. [PMID: 38966644 PMCID: PMC11222398 DOI: 10.3389/fimmu.2024.1395921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
IL-27, a member of the IL-6/IL-12 cytokine superfamily, is primarily secreted by antigen presenting cells, specifically by dendric cells, macrophages and B cells. IL-27 has antiviral activities and modulates both innate and adaptive immune responses against viruses. The role of IL-27 in the setting of viral infections is not well defined and both pro-inflammatory and anti-inflammatory functions have been described. Here, we discuss the latest advancements in the role of IL-27 in several viral infection models of human disease. We highlight important aspects of IL-27 expression regulation, the critical cell sources at different stages of the infection and their impact in cell mediated immunity. Lastly, we discuss the need to better define the antiviral and modulatory (pro-inflammatory vs anti-inflammatory) properties of IL-27 in the context of human chronic viral infections.
Collapse
Affiliation(s)
| | | | - Marta Catalfamo
- Department of Microbiology Immunology, Georgetown University School of Medicine, Washington, DC, United States
| |
Collapse
|
47
|
Gerke C, Bauersfeld L, Schirmeister I, Mireisz CNM, Oberhardt V, Mery L, Wu D, Jürges CS, Spaapen RM, Mussolino C, Le-Trilling VTK, Trilling M, Dölken L, Paster W, Erhard F, Hofmann M, Schlosser A, Hengel H, Momburg F, Halenius A. Multimodal HLA-I genotype regulation by human cytomegalovirus US10 and resulting surface patterning. eLife 2024; 13:e85560. [PMID: 38900146 PMCID: PMC11189632 DOI: 10.7554/elife.85560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
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) β2m-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.
Collapse
Affiliation(s)
- Carolin Gerke
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
- Spemann Graduate School of Biology and Medicine (SGBM), University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
| | - Liane Bauersfeld
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Ivo Schirmeister
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Chiara Noemi-Marie Mireisz
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of WürzburgWürzburgGermany
| | - Valerie Oberhardt
- Faculty of Medicine, University of FreiburgFreiburgGermany
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Medical Center University of FreiburgFreiburgGermany
| | - Lea Mery
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Di Wu
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | | | - Robbert M Spaapen
- Department of Immunopathology, Sanquin ResearchAmsterdamNetherlands
- Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Claudio Mussolino
- Faculty of Medicine, University of FreiburgFreiburgGermany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center University of FreiburgFreiburgGermany
- Center for Chronic Immunodeficiency, Medical Center University of FreiburgFreiburgGermany
| | | | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-EssenEssenGermany
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital EssenEssenGermany
| | - Lars Dölken
- Institute for Virology and Immunobiology, University of WürzburgWürzburgGermany
- Institute of Virology, Hannover Medical SchoolHannoverGermany
| | - Wolfgang Paster
- St. Anna Children’s Cancer Research Institute (CCRI)ViennaAustria
| | - Florian Erhard
- Institute for Virology and Immunobiology, University of WürzburgWürzburgGermany
| | - Maike Hofmann
- Faculty of Medicine, University of FreiburgFreiburgGermany
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Medical Center University of FreiburgFreiburgGermany
| | - Andreas Schlosser
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of WürzburgWürzburgGermany
| | - Hartmut Hengel
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Frank Momburg
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center, National Center for Tumor Diseases (NCT), Heidelberg University HospitalHeidelbergGermany
| | - Anne Halenius
- Institute of Virology, Medical Center University of FreiburgFreiburgGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| |
Collapse
|
48
|
Marri L, Contini P, Ivaldi F, Schiavi C, Magnani O, Vassallo C, Guastalla A, Traversone N, Angelini C, Del Zotto G, De Maria A, De Palma R. Evaluation of Frequency of CMV Replication and Disease Complications Reveals New Cellular Defects and a Time Dependent Pattern in CVID Patients. J Clin Immunol 2024; 44:142. [PMID: 38847943 PMCID: PMC11161436 DOI: 10.1007/s10875-024-01744-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
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.
Collapse
Affiliation(s)
- Luca Marri
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
- Unit of Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Paola Contini
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
| | - Federico Ivaldi
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
| | - Chiara Schiavi
- Unit of Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Ottavia Magnani
- Unit of Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Vassallo
- Unit of Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Guastalla
- Unit of Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Noemi Traversone
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
- Unit of Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudia Angelini
- Institute for Applied Mathematics "Mauro Picone", National Research Council, Naples, Italy
| | - Genny Del Zotto
- Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Andrea De Maria
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy.
- Infections in Immunocompromised Host Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Raffaele De Palma
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy.
- Unit of Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
49
|
Santamorena MM, Tischer-Zimmermann S, Bonifacius A, Mireisz CNM, Costa B, Khan F, Kulkarni U, Lauruschkat CD, Sampaio KL, Stripecke R, Blasczyk R, Maecker-Kolhoff B, Kraus S, Schlosser A, Cicin-Sain L, Kalinke U, Eiz-Vesper B. Engineered HCMV-infected APCs enable the identification of new immunodominant HLA-restricted epitopes of anti-HCMV T-cell immunity. HLA 2024; 103:e15541. [PMID: 38923358 DOI: 10.1111/tan.15541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 06/28/2024]
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.
Collapse
Affiliation(s)
- Maria Michela Santamorena
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - Chiara Noemi-Marie Mireisz
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Wuerzburg, Wuerzburg, Germany
| | - Bibiana Costa
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Fawad Khan
- Immune Ageing and Chronic Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Upasana Kulkarni
- Immune Ageing and Chronic Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | - Renata Stripecke
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Center for Molecular Medicine Cologne (CMMC), Institute of Translational Immuno-oncology, Cologne, Germany
- German Center for Infections Research (DZIF) Bonn-Cologne, Cologne, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
| | - Britta Maecker-Kolhoff
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
- German Center for Infections Research (DZIF) Bonn-Cologne, Cologne, Germany
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Sabrina Kraus
- Department of Internal Medicine II, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Andreas Schlosser
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Wuerzburg, Wuerzburg, Germany
| | - Luka Cicin-Sain
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
- Immune Ageing and Chronic Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence - Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Cluster of Excellence - Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| |
Collapse
|
50
|
Albright ER, Kalejta RF. cGAS-STING-TBK1 Signaling Promotes Valproic Acid-Responsive Human Cytomegalovirus Immediate-Early Transcription during Infection of Incompletely Differentiated Myeloid Cells. Viruses 2024; 16:877. [PMID: 38932169 PMCID: PMC11209474 DOI: 10.3390/v16060877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Repression of human cytomegalovirus (HCMV) immediate-early (IE) gene expression is a key regulatory step in the establishment and maintenance of latent reservoirs. Viral IE transcription and protein accumulation can be elevated during latency by treatment with histone deacetylase inhibitors such as valproic acid (VPA), rendering infected cells visible to adaptive immune responses. However, the latency-associated viral protein UL138 inhibits the ability of VPA to enhance IE gene expression during infection of incompletely differentiated myeloid cells that support latency. UL138 also limits the accumulation of IFNβ transcripts by inhibiting the cGAS-STING-TBK1 DNA-sensing pathway. Here, we show that, in the absence of UL138, the cGAS-STING-TBK1 pathway promotes both IFNβ accumulation and VPA-responsive IE gene expression in incompletely differentiated myeloid cells. Inactivation of this pathway by either genetic or pharmacological inhibition phenocopied UL138 expression and reduced VPA-responsive IE transcript and protein accumulation. This work reveals a link between cytoplasmic pathogen sensing and epigenetic control of viral lytic phase transcription and suggests that manipulation of pattern recognition receptor signaling pathways could aid in the refinement of MIEP regulatory strategies to target latent viral reservoirs.
Collapse
Affiliation(s)
- Emily R. Albright
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin—Madison, Madison, WI 53706, USA;
| | | |
Collapse
|