1
|
Graninger M, Endmayr V, Kühner LM, Berger SM, Rommer P, Klotz S, Gelpi E, Vietzen H, Höftberger R, Puchhammer‐Stöckl E. Association Between NK Cell Genetic Variants and the Development of Long COVID Associated- and Prepandemic Small Fiber Neuropathy. J Med Virol 2024; 96:e70091. [PMID: 39610330 PMCID: PMC11605371 DOI: 10.1002/jmv.70091] [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: 07/25/2024] [Revised: 10/18/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024]
Abstract
Long coronavirus disease 2019 (COVID) (LC) symptoms including pain and autonomic dysfunction are in some patients associated with small-fiber neuropathy (SFN). The pathomechanisms underlying SFN are mostly unclear. Natural killer (NK) cells play a crucial role in immune regulation, viral clearance and nerve metabolism. The aim of this study was to identify associations between development of small-fiber dysfunction dependent and independent of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and human genetic markers associated with specific NK cell functions. The genetic markers assessed in all cohorts included: FCGR3A, IGHG1, HLA-E, NKG2C, and rs9916629. Genotyping was performed using TaqMan assays, Sanger sequencing and touchdown polymerase chain reaction. We assessed human cytomegalovirus (HCMV) IgG serostatus in all participants, and screened for anti-neuronal, anti-glial and anti-ganglioside autoantibodies in both patient cohorts. We included 50 LC patients with newly-emerged symptoms of small-fiber dysfunction after SARS-CoV-2 infection, 27 prepandemic SFN patients and 320 control persons. Markers associated with low NKG2C response, that is, deletion of the NKG2C gene and lack of prior HCMV infection (IgG seronegativity), occurred significantly more frequently in prepandemic SFN patients compared to LC patients and controls (p = 0.0109 and 0.0005, respectively). In conclusion, markers of impaired NKG2C pathways are associated with prepandemic SFN, but not with Long COVID-associated small-fiber dysfunction.
Collapse
Affiliation(s)
| | - Verena Endmayr
- Department of Neurology, Division of Neuropathology and NeurochemistryMedical University of ViennaViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaViennaAustria
| | - Laura M. Kühner
- Center for VirologyMedical University of ViennaViennaViennaAustria
| | - Sarah M. Berger
- Center for VirologyMedical University of ViennaViennaViennaAustria
| | - Paulus Rommer
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaViennaAustria
- Department of NeurologyMedical University of ViennaViennaViennaAustria
| | - Sigrid Klotz
- Department of Neurology, Division of Neuropathology and NeurochemistryMedical University of ViennaViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaViennaAustria
| | - Ellen Gelpi
- Department of Neurology, Division of Neuropathology and NeurochemistryMedical University of ViennaViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaViennaAustria
| | - Hannes Vietzen
- Center for VirologyMedical University of ViennaViennaViennaAustria
| | - Romana Höftberger
- Department of Neurology, Division of Neuropathology and NeurochemistryMedical University of ViennaViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaViennaAustria
| | | |
Collapse
|
2
|
Slein MD, Backes IM, Kelkar NS, Garland CR, Khanwalkar US, Sholukh AM, Johnston CM, Leib DA, Ackerman ME. Improving antibody-mediated protection against HSV infection by eliminating interactions with the viral Fc receptor gE/gI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624598. [PMID: 39605495 PMCID: PMC11601663 DOI: 10.1101/2024.11.20.624598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Herpes simplex virus (HSV) encodes surface glycoproteins that are host defense evasion molecules, allowing the virus to escape immune clearance. In addition to their role in neuropathogenesis and cell-cell spread, glycoproteins E and I (gE/gI) form a viral Fc receptor (vFcR) for most subclasses and allotypes of human IgG and promote evasion of humoral immune responses. While monoclonal antibodies (mAbs) protect mice from neonatal HSV (nHSV) infections, the impact of the vFcR on mAb-mediated protection by binding to IgG is unknown. Using HSV-1 with intact and ablated gE-mediated IgG Fc binding, and Fc-engineered antibodies with modified ability to interact with gE/gI, we investigated the role of the vFcR in viral pathogenesis and mAb-mediated protection from nHSV. The gD-specific human mAb HSV8 modified to lack binding to gE exhibited enhanced neutralization and in vivo protection compared to its native IgG1 form. This improved protection by the engineered mAbs was dependent on the presence of the vFcR. Human IgG3 allotypes lacking vFcR binding also exhibited enhanced antiviral activity in vivo, suggesting that vaccines that robustly induce IgG3 responses could show enhanced protection. suggesting the value of vaccination strategies that robustly induce this subclass. Lastly, analysis of longitudinal responses to acute primary genital infection in humans raised the possibility that unlike most viruses, HSV may exhibited slow induction of IgG3. In summary, this study demonstrates that mAbs lacking the ability to interact with the vFcR can exhibit improved protection from HSV-offering new prospects for antibody-based interventions.
Collapse
Affiliation(s)
- Matthew D. Slein
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Iara M. Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Callaghan R. Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | | | - Christine M. Johnston
- Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Departments of Medicine and Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104, USA
| | - David A. Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
3
|
Asenjo J, Moraru M, Al‐Akioui‐Sanz K, Altadill M, Muntasell A, López‐Botet M, Vilches C. NKG2C Sequence Polymorphism Modulates the Expansion of Adaptive NK Cells in Response to Human CMV. HLA 2024; 104:e15764. [PMID: 39581700 PMCID: PMC11586157 DOI: 10.1111/tan.15764] [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: 04/20/2024] [Revised: 10/23/2024] [Accepted: 11/02/2024] [Indexed: 11/26/2024]
Abstract
A subpopulation of NK cells with distinctive phenotype and function differentiates and expands specifically in response to infection by human cytomegalovirus (HCMV). A hallmark of these adaptive NK cells is their increased expression levels of the activating CD94/NKG2C receptor for HLA-E, and lack of expression of its inhibitory homologue CD94/NKG2A. Their frequency is highly variable in HCMV+ individuals, and the basis for such differences is only partially understood. Here, we explore the possible influence of sequence polymorphism of the NKG2C (or KLRC2) gene on the expansion of NKG2C+NKG2A- NK cells in healthy HCMV-seropositive donors. Our results show a significant association of greater proportions of adaptive NK cells with allele NKG2C*02. This is defined by two amino acid substitutions in comparison with the most prevalent allele, NKG2C*01, and associates with additional sequence polymorphisms in noncoding regions. Furthermore, we demonstrate consistently higher mRNA levels of NKG2C*02 in heterozygous individuals co-expressing this allele in combination with NKG2C*01 or *03. This predominance is independent of polymorphisms in the promoter and 3' UTRs and is appreciated also in HCMV-seronegative donors. In summary, although additional factors are most likely implicated in the variable expansion of NKG2C+NKG2A- NK cells in response to HCMV, our results demonstrate that host immunogenetics, in particular NKG2C diversity, influences the magnitude of such response.
Collapse
Affiliation(s)
- Judit Asenjo
- Immunogenetics & Histocompatibility LabInstituto de Investigación Sanitaria Puerta de Hierro – Segovia de AranaMajadahondaSpain
| | - Manuela Moraru
- Immunogenetics & Histocompatibility LabInstituto de Investigación Sanitaria Puerta de Hierro – Segovia de AranaMajadahondaSpain
| | - Karima Al‐Akioui‐Sanz
- Immunogenetics & Histocompatibility LabInstituto de Investigación Sanitaria Puerta de Hierro – Segovia de AranaMajadahondaSpain
| | | | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autonòma de BarcelonaBellaterraSpain
| | - Miguel López‐Botet
- University Pompeu FabraBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
| | - Carlos Vilches
- Immunogenetics & Histocompatibility LabInstituto de Investigación Sanitaria Puerta de Hierro – Segovia de AranaMajadahondaSpain
- Organización Nacional de TrasplantesMinisterio de SanidadMadridSpain
| |
Collapse
|
4
|
Pandey JP, Baglio F, Mancuso R, Guerini FR, Cabinio M, Isernia S, Clerici M, Agostini S. Biomarkers of neural integrity and immunoglobulin genes influence neurodegeneration in Alzheimer's disease. J Neurol Sci 2024; 464:123167. [PMID: 39142084 PMCID: PMC11347077 DOI: 10.1016/j.jns.2024.123167] [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: 03/13/2024] [Revised: 06/26/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024]
Abstract
Compelling evidence has been presented in favor of herpes simplex virus type 1 (HSV1) being one of the causative agents of Alzheimer's disease (AD). The success of HSV1 as a pathogen relates to its sophisticated strategies to evade host immunosurveillance. One strategy involves encoding a decoy Fcγ receptor (FcγR) that thwarts the Fcγ-mediated effector functions, such as antibody-dependent cellular cytotoxicity (ADCC), a potent host immunosurveillance mechanism against virally infected cells. The decoy FcγR binds to antibodies of all IgG subclasses, except IgG3; therefore, IgG3 would be expected to play an important role in viral clearance by neutralization and ADCC, and thus contribute to protection from HSV1-spurred diseases. Previous studies have shown significant association between anti-HSV1 IgG3 antibodies and cortical thinning of the areas of the brain typically altered in AD and also targeted by HSV1. The aim of the present investigation was to determine whether GM (γ marker) 5 and GM 21 allotypes, hereditary allelic determinants expressed on IgG3, together with brain biomarkers of neural integrity, contributed to neurodegeneration-as measured by mini-mental state examination (MMSE) score-in patients with AD. Multiple regression analyses showed that the homozygous GM 5/5 genotype, preserved right hippocampus, and right insula thickness were associated with higher MMSE scores (p < 0.001), whereas the opposite pattern and GM 5/21 genotype were associated with worse clinical profiles. Influence of GM 5/21-expressing IgG3 antibodies on the ADCC of HSV1-infected neurons could, at least partially, explain these results.
Collapse
Affiliation(s)
- Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
| | | | | | | | - Monia Cabinio
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Sara Isernia
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | | |
Collapse
|
5
|
Vázquez-Coto D, Kimball C, Albaiceta GM, Amado-Rodríguez L, García-Clemente M, Gómez J, Coto E, Pandey JP. Immunoglobulin genes and severity of COVID-19. Immunogenetics 2024; 76:213-217. [PMID: 38602517 PMCID: PMC11087305 DOI: 10.1007/s00251-024-01341-z] [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: 02/08/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
There is tremendous interindividual and interracial variability in the outcome of SARS-CoV-2 infection, suggesting the involvement of host genetic factors. Here, we investigated whether IgG allotypes GM (γ marker) 3 and GM 17, genetic markers of IgG1, contributed to the severity of COVID-19. IgG1 plays a pivotal role in response against SARS-CoV-2 infection. We also investigated whether these GM alleles synergistically/epistatically with IGHG3 and FCGR2A alleles-which have been previously implicated in COVID-19-modulated the extent of COVID-19 severity. The study population consisted of 316 COVID-19 patients who needed treatment in the intensive care unit of Hospital Universitario Central de Asturias. All individuals were genotyped for GM 3/17, IGHG3 hinge length, and FCGR2A rs1801274 A/G polymorphisms. Among the 316 critical patients, there were 86 deaths. The risk of death among critical patients was significantly higher in subjects with GM 17 (IgG1) and short hinge length (IgG3). GM 17-carriers were at almost three-fold higher risk of death than non-carriers (p < 0.001; OR = 2.86, CI 1.58-5.16). Subjects with short hinge length of IgG3 had a two-fold higher risk of death than those with medium hinge length (p = 0.01; OR = 2.16, CI 1.19-3.90). GM 3/3 and IGHG3 (MM) genotypes were less frequent among death vs. survivors (9% vs 36%, p < 0.001) and associated with protective effect (OR = 0.18, 95% CI = 0.08-0.39). This is the first report implicating IgG1 allotypes in COVID-19-spurred death. It needs to be replicated in an independent study population.
Collapse
Affiliation(s)
- Daniel Vázquez-Coto
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, Oviedo, Spain
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain
| | - Christine Kimball
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Guillermo M Albaiceta
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central Asturias, Oviedo, Spain
- Universidad de Oviedo, Oviedo, Spain
- CIBER-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Laura Amado-Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central Asturias, Oviedo, Spain
- Universidad de Oviedo, Oviedo, Spain
- CIBER-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Marta García-Clemente
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, Oviedo, Spain
- Universidad de Oviedo, Oviedo, Spain
- Neumología, Hospital Universitario Central Asturias, Oviedo, Spain
| | - Juan Gómez
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, Oviedo, Spain
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain
| | - Eliecer Coto
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, Oviedo, Spain.
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain.
- Universidad de Oviedo, Oviedo, Spain.
| | - Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
6
|
Tittarelli A, Pereda C, Gleisner MA, López MN, Flores I, Tempio F, Lladser A, Achour A, González FE, Durán-Aniotz C, Miranda JP, Larrondo M, Salazar-Onfray F. Long-Term Survival and Immune Response Dynamics in Melanoma Patients Undergoing TAPCells-Based Vaccination Therapy. Vaccines (Basel) 2024; 12:357. [PMID: 38675738 PMCID: PMC11053591 DOI: 10.3390/vaccines12040357] [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: 12/05/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer vaccines present a promising avenue for treating immune checkpoint blockers (ICBs)-refractory patients, fostering immune responses to modulate the tumor microenvironment. We revisit a phase I/II trial using Tumor Antigen-Presenting Cells (TAPCells) (NCT06152367), an autologous antigen-presenting cell vaccine loaded with heat-shocked allogeneic melanoma cell lysates. Initial findings showcased TAPCells inducing lysate-specific delayed-type hypersensitivity (DTH) reactions, correlating with prolonged survival. Here, we extend our analysis over 15 years, categorizing patients into short-term (<36 months) and long-term (≥36 months) survivors, exploring novel associations between clinical outcomes and demographic, genetic, and immunologic parameters. Notably, DTHpos patients exhibit a 53.1% three-year survival compared to 16.1% in DTHneg patients. Extended remissions are observed in long-term survivors, particularly DTHpos/M1cneg patients. Younger age, stage III disease, and moderate immune events also benefit short-term survivors. Immunomarkers like increased C-type lectin domain family 2 member D on CD4+ T cells and elevated interleukin-17A were detected in long-term survivors. In contrast, toll-like receptor-4 D229G polymorphism and reduced CD32 on B cells are associated with reduced survival. TAPCells achieved stable long remissions in 35.2% of patients, especially M1cneg/DTHpos cases. Conclusions: Our study underscores the potential of vaccine-induced immune responses in melanoma, emphasizing the identification of emerging biological markers and clinical parameters for predicting long-term remission.
Collapse
Affiliation(s)
- Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile;
| | - Cristian Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - María A. Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Mercedes N. López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Iván Flores
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Fabián Tempio
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Alvaro Lladser
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580702, Chile;
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 8580702, Chile
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden;
- Division of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Fermín E. González
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, Universidad de Chile, Santiago 8380000, Chile;
| | - Claudia Durán-Aniotz
- Latin American Brain Health Institute (BrainLat), Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibañez, Santiago 7941169, Chile;
| | | | - Milton Larrondo
- Banco de Sangre, Hospital Clínico de la Universidad de Chile, Santiago 8380453, Chile;
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden;
- Division of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|
7
|
Purcell RA, Aurelia LC, Esterbauer R, Allen LF, Bond KA, Williamson DA, Trevillyan JM, Trubiano JA, Juno JJ, Wheatley AK, Davenport MP, Nguyen THO, Kedzierska K, Kent SJ, Selva KJ, Chung AW. Immunoglobulin G genetic variation can confound assessment of antibody levels via altered binding to detection reagents. Clin Transl Immunology 2024; 13:e1494. [PMID: 38433763 PMCID: PMC10902689 DOI: 10.1002/cti2.1494] [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: 12/18/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 03/05/2024] Open
Abstract
Objectives Amino acid variations across more than 30 immunoglobulin (Ig) allotypes may introduce structural changes that influence recognition by anti-Ig detection reagents, consequently confounding interpretation of antibody responses, particularly in genetically diverse cohorts. Here, we assessed a panel of commercial monoclonal anti-IgG1 clones for capacity to universally recognise two dominant IgG1 haplotypes (G1m-1,3 and G1m1,17). Methods Four commercial monoclonal anti-human IgG1 clones were assessed via ELISAs and multiplex bead-based assays for their ability to bind G1m-1,3 and G1m1,17 IgG1 variants. Detection antibodies were validated against monoclonal IgG1 allotype standards and tested for capacity to recognise antigen-specific plasma IgG1 from G1m-1,3 and G1m1,17 homozygous and heterozygous SARS-CoV-2 BNT162b2 vaccinated (n = 28) and COVID-19 convalescent (n = 44) individuals. An Fc-specific pan-IgG detection antibody corroborated differences between hinge- and Fc-specific anti-IgG1 responses. Results Hinge-specific anti-IgG1 clone 4E3 preferentially bound G1m1,17 compared to G1m-1,3 IgG1. Consequently, SARS-CoV-2 Spike-specific IgG1 levels detected in G1m1,17/G1m1,17 BNT162b2 vaccinees appeared 9- to 17-fold higher than in G1m-1,3/G1m-1,3 vaccinees. Fc-specific IgG1 and pan-IgG detection antibodies equivalently bound G1m-1,3 and G1m1,17 IgG1 variants, and detected comparable Spike-specific IgG1 levels between haplotypes. IgG1 responses against other human coronaviruses and influenza were similarly poorly detected by 4E3 anti-IgG1 in G1m-1,3/G1m-1,3 subjects. Conclusion Anti-IgG1 clone 4E3 confounds assessment of antibody responses in clinical cohorts owing to bias towards detection of G1m1,17 IgG1 variants. Validation of anti-Ig clones should include evaluation of binding to relevant antibody variants, particularly as the role of immunogenetics upon humoral immunity is increasingly explored in diverse populations.
Collapse
Affiliation(s)
- Ruth A Purcell
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - L Carissa Aurelia
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Robyn Esterbauer
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Lilith F Allen
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Katherine A Bond
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
- Victorian Infectious Diseases Reference Laboratory (VIDRL)The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Deborah A Williamson
- Victorian Infectious Diseases Reference Laboratory (VIDRL)The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Infectious DiseasesThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Janine M Trevillyan
- Department of Infectious DiseasesThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
- Centre for Antibiotic Allergy and Research, Department of Infectious DiseasesAustin HealthHeidelbergVICAustralia
| | - Jason A Trubiano
- Centre for Antibiotic Allergy and Research, Department of Infectious DiseasesAustin HealthHeidelbergVICAustralia
- Department of MedicineUniversity of MelbourneParkvilleVICAustralia
- Department of Infectious DiseasesPeter MacCallum Cancer CentreMelbourneVICAustralia
- National Centre for Infections in CancerPeter MacCallum Cancer CentreMelbourneVICAustralia
| | - Jennifer J Juno
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Adam K Wheatley
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | | | - Thi HO Nguyen
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Katherine Kedzierska
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Stephen J Kent
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Health, Central Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Kevin John Selva
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Amy W Chung
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| |
Collapse
|
8
|
Damelang T, Brinkhaus M, van Osch TLJ, Schuurman J, Labrijn AF, Rispens T, Vidarsson G. Impact of structural modifications of IgG antibodies on effector functions. Front Immunol 2024; 14:1304365. [PMID: 38259472 PMCID: PMC10800522 DOI: 10.3389/fimmu.2023.1304365] [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: 09/29/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Immunoglobulin G (IgG) antibodies are a critical component of the adaptive immune system, binding to and neutralizing pathogens and other foreign substances. Recent advances in molecular antibody biology and structural protein engineering enabled the modification of IgG antibodies to enhance their therapeutic potential. This review summarizes recent progress in both natural and engineered structural modifications of IgG antibodies, including allotypic variation, glycosylation, Fc engineering, and Fc gamma receptor binding optimization. We discuss the functional consequences of these modifications to highlight their potential for therapeutical applications.
Collapse
Affiliation(s)
- Timon Damelang
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Sanquin Research, Department of Immunopathology, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Maximilian Brinkhaus
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Thijs L. J. van Osch
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Janine Schuurman
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Aran F. Labrijn
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Theo Rispens
- Sanquin Research, Department of Immunopathology, Amsterdam, Netherlands
| | - Gestur Vidarsson
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
9
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
10
|
Graninger M, Vietzen H, Puchhammer-Stöckl E. Association between human genetic variants affecting the host NK cell response and the development of herpes simplex virus type 1 encephalitis. J Med Virol 2023; 95:e28759. [PMID: 37212301 DOI: 10.1002/jmv.28759] [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: 02/08/2023] [Revised: 03/22/2023] [Accepted: 04/13/2023] [Indexed: 05/23/2023]
Abstract
Herpes simplex virus encephalitis (HSE) is a rare complication of herpes simplex virus type 1 (HSV-1) infection or reactivation. It is so far unclear why only few patients develop HSE. As natural killer (NK) cells provide an important defense against HSV-1, we investigated whether there is an association between distinct human genetic variants associated with the host NK cell response and HSE. Forty-nine adult patients with confirmed HSE and 247 matched controls were analyzed for the distribution of the following genotypes: CD16A (FcγRIIIA) V/F and IGHG1 G1m3/17, both influencing antibody-dependent cellular cytotoxicity; HLA-E*0101/*0103, associated with NK cell activation; and SLFN13 rs9916629C/T associated with NK cell response. Homozygous HLA-E*0101:0101 and HLA-E*0103:0103 variants as well as the rs9916629CC genotype were overrepresented in HSE patients compared to controls (p ≤ 0.001). Notably, cooccurrence of the homozygous HLA-E*0101 and rs9916629CC genotypes was present in 19% of patients but totally absent in controls (p ≤ 0.0001). Distribution of CD16A and IGHG1 variants did not differ between patients and controls. Our data show that the rare combination of HLA-E*0101:0101 and rs9916629CC is significantly associated with HSE. Possibly, these genetic variations could be useful as clinical markers predicting HSE prognosis and helping to adapt the treatment of HSE in the individual patient.
Collapse
Affiliation(s)
| | - Hannes Vietzen
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
11
|
Crowley AR, Richardson SI, Tuyishime M, Jennewein M, Bailey MJ, Lee J, Alter G, Ferrari G, Morris L, Ackerman ME. Functional consequences of allotypic polymorphisms in human immunoglobulin G subclasses. Immunogenetics 2023; 75:1-16. [PMID: 35904629 PMCID: PMC9845132 DOI: 10.1007/s00251-022-01272-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/07/2022] [Indexed: 01/21/2023]
Abstract
Heritable polymorphisms within the human IgG locus, collectively termed allotypes, have often been linked by statistical associations, but rarely mechanistically, to a wide range of disease states. One potential explanation for these associations is that IgG allotype alters host cell receptors' affinity for IgG, dampening or enhancing an immune response depending on the nature of the change and the receptors. In this work, a panel of allotypic antibody variants were evaluated using multiplexed, label-free biophysical methods and cell-based functional assays to determine what effect, if any, human IgG polymorphisms have on antibody function. While we observed several differences in FcγR affinity among allotypes, there was little evidence of dramatically altered FcγR-based effector function or antigen recognition activity associated with this aspect of genetic variability.
Collapse
Affiliation(s)
- Andrew R Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Simone I Richardson
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, 2131, Gauteng, South Africa
- MRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Marina Tuyishime
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | | | - Meredith J Bailey
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, 2131, Gauteng, South Africa
- MRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA.
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA.
| |
Collapse
|
12
|
López-Botet M, De Maria A, Muntasell A, Della Chiesa M, Vilches C. Adaptive NK cell response to human cytomegalovirus: Facts and open issues. Semin Immunol 2023; 65:101706. [PMID: 36542944 DOI: 10.1016/j.smim.2022.101706] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/28/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Human cytomegalovirus (HCMV) infection exerts broad effects on the immune system. These include the differentiation and persistent expansion of a mature NK cell subset which displays a characteristic phenotypic and functional profile hallmarked by expression of the HLA-E-specific CD94/NKG2C activating receptor. Based on our experience and recent advances in the field, we overview the adaptive features of the NKG2C+ NK cell response, discussing observations and open questions on: (a) the mechanisms and influence of viral and host factors; (b) the existence of other NKG2C- NK cell subsets sharing adaptive features; (c) the development and role of adaptive NKG2C+ NK cells in the response to HCMV in hematopoietic and solid organ transplant patients; (d) their relation with other viral infections, mainly HIV-1; and (e) current perspectives for their use in adoptive immunotherapy of cancer.
Collapse
Affiliation(s)
- Miguel López-Botet
- Hospital del Mar Medical Research Institute (IMIM). Barcelona, Spain; Department of Medicine and Life Sciences. Univ. Pompeu Fabra. Barcelona, Spain.
| | - Andrea De Maria
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Health Sciences, University of Genoa, Genoa, Italy.
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM). Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Spain; Universitat Autònoma de Barcelona, Barcelona, Spain.
| | | | - Carlos Vilches
- Immunogenetics & Histocompatibility Lab, Instituto de Investigación Sanitaria Puerta de Hierro - Segovia de Arana, Majadahonda, Madrid, Spain.
| |
Collapse
|
13
|
Backes IM, Leib DA, Ackerman ME. Monoclonal antibody therapy of herpes simplex virus: An opportunity to decrease congenital and perinatal infections. Front Immunol 2022; 13:959603. [PMID: 36016956 PMCID: PMC9398215 DOI: 10.3389/fimmu.2022.959603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022] Open
Abstract
The fetal/neonatal period represents both a unique window of opportunity for interventions as well as vulnerability to a number of viral infections. While Herpesviruses such as herpes simplex virus (HSV) are highly prevalent and typically of little consequence among healthy adults, they are among the most consequential infections of early life. Despite treatment with antiviral drugs, neonatal HSV (nHSV) infections can still result in significant mortality and lifelong neurological morbidity. Fortunately, newborns in our pathogen-rich world inherit some of the protection provided by the maternal immune system in the form of transferred antibodies. Maternal seropositivity, resulting in placental transfer of antibodies capable of neutralizing virus and eliciting the diverse effector functions of the innate immune system are associated with dramatically decreased risk of nHSV. Given this clear epidemiological evidence of reduced risk of infection and its sequelae, we present what is known about the ability of monoclonal antibody therapies to treat or prevent HSV infection and explore how effective antibody-based interventions in conjunction with antiviral therapy might reduce early life mortality and long-term morbidity.
Collapse
Affiliation(s)
- Iara M Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - David A Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | | |
Collapse
|
14
|
Pandey JP, Agostini S, Namboodiri AM, Mancuso R, Guerini FR, Meloni M, Costa AS. Immunoglobulin γ chain allotypes and humoral immunity to HSV1 in Parkinson's disease. J Neuroimmunol 2022; 371:577948. [DOI: 10.1016/j.jneuroim.2022.577948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/06/2022] [Accepted: 08/03/2022] [Indexed: 11/26/2022]
|
15
|
van Tilburg SJ, Jacobs BC, Ooijevaar-de Heer P, Fokkink WJR, Huizinga R, Vidarsson G, Rispens T. Novel approach to monitor intravenous immunoglobulin pharmacokinetics in humans using polymorphic determinants in IgG1 constant domains. Eur J Immunol 2021; 52:609-617. [PMID: 34854474 DOI: 10.1002/eji.202149653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/20/2021] [Indexed: 12/12/2022]
Abstract
Clinical efficacy of intravenous immunoglobulin treatment (IVIg) is related to its pharmacokinetic (PK) profile. Its usual evaluation, by measuring serum total IgG levels, is imprecise, because IVIg cannot be distinguished from endogenous IgG. We developed ELISAs to specifically monitor the PK of IVIg using the polymorphic determinants G1m(a), G1m(x), and G1m(f). The specificity of the IgG1 allotype assays was sufficient to determine IVIg concentrations as low as 0.1 mg/mL in sera from individuals not expressing the respective markers. IVIg was quantified in posttreatment serum from patients with Guillain-Barré syndrome (GBS) by measuring IgG1 allotypes not expressed endogenously. After serotyping, 27/28 GBS patients were found eligible for IVIg monitoring using one or two genetic markers. In 17 cases, IVIg levels could be determined by both anti-G1m(a) and anti-G1m(x) measurement, showing significant correlation. Longitudinal monitoring of IVIg PK in seven GBS patients showed potential differences in clearance of total IgG versus IVIg-derived IgG, highlighting that total IgG measurements may not accurately reflect IVIg PK. To summarize, anti-IgG1 allotype assays can discriminate between endogenous IgG and therapeutic polyclonal IgG. These assays will be an important tool to better understand the variability in IVIg PK and treatment response of all patients treated with IVIg.
Collapse
Affiliation(s)
- Sander J van Tilburg
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Bart C Jacobs
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Pleuni Ooijevaar-de Heer
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Willem-Jan R Fokkink
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ruth Huizinga
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Ataya M, Redondo-Pachón D, Llinàs-Mallol L, Yélamos J, Alari-Pahissa E, Pérez-Sáez MJ, Altadill M, Raïch-Regué D, Vilches C, Pascual J, Crespo M, López-Botet M. Long-Term Evolution of the Adaptive NKG2C + NK Cell Response to Cytomegalovirus Infection in Kidney Transplantation: An Insight on the Diversity of Host-Pathogen Interaction. THE JOURNAL OF IMMUNOLOGY 2021; 207:1882-1890. [PMID: 34470855 DOI: 10.4049/jimmunol.2100055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 08/01/2021] [Indexed: 11/19/2022]
Abstract
Human CMV infection is frequent in kidney transplant recipients (KTR). Pretransplant Ag-specific T cells and adaptive NKG2C+ NK cells associate with reduced incidence of infection in CMV+ KTR. Expansions of adaptive NKG2C+ NK cells were reported in posttransplant CMV-infected KTR. To further explore this issue, NKG2C+ NK, CD8+, and TcRγδ T cells were analyzed pretransplant and at different time points posttransplant for ≥24 mo in a cohort of CMV+ KTR (n = 112), stratified according to CMV viremia detection. In cryopreserved samples from a subgroup (n = 49), adaptive NKG2C+ NK cell markers and T cell subsets were compared after a longer follow-up (median, 56 mo), assessing the frequencies of CMV-specific T cells and viremia at the last time point. Increased proportions of NKG2C+ NK, CD8+, and TcRγδ T cells were detected along posttransplant evolution in viremia(+) KTR. However, the individual magnitude and kinetics of the NKG2C+ NK response was variable and only exceptionally detected among viremia(-) KTR, presumably reflecting subclinical viral replication events. NKG2C+ expansions were independent of KLRC2 zygosity and associated with higher viral loads at diagnosis; no relation with other clinical parameters was perceived. Increased proportions of adaptive NKG2C+ NK cells (CD57+, ILT2+, FcεRIγ-) were observed after resolution of viremia long-term posttransplant, coinciding with increased CD8+ and Vδ2- γδ T cells; at that stage CMV-specific T cells were comparable to viremia(-) cases. These data suggest that adaptive NKG2C+ NK cells participate with T cells to restore CMV replication control, although their relative contribution cannot be discerned.
Collapse
Affiliation(s)
| | - Dolores Redondo-Pachón
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | | | - José Yélamos
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Immunology Laboratory, Department of Pathology, Hospital del Mar, Barcelona, Spain; and
| | | | - María J Pérez-Sáez
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | | | - Dàlia Raïch-Regué
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain
| | - Carlos Vilches
- Immunogenetics-HLA, Instituto de Investigación Sanitaria Puerta de Hierro Segovia de Arana, Majadahonda, Madrid, Spain
| | - Julio Pascual
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Marta Crespo
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Miguel López-Botet
- Universitat Pompeu Fabra, Barcelona, Spain; .,Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Immunology Laboratory, Department of Pathology, Hospital del Mar, Barcelona, Spain; and
| |
Collapse
|
17
|
Pandey JP, Namboodiri AM, Nietert PJ, Knowles MR, Pace RG, Pier GB. Immunoglobulin GM and KM allotypes are associated with antibody responses to Pseudomonas aeruginosa antigens in chronically infected cystic fibrosis patients. J Cyst Fibros 2021; 20:1080-1084. [PMID: 34246573 DOI: 10.1016/j.jcf.2021.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Chronic infection with Pseudomonas aeruginosa (P. aeruginosa) is a leading cause of death in patients with cystic fibrosis (CF). Immunobiology of P. aeruginosa infection is complex and not well understood. Chronically infected CF patients generate high levels of antibodies to P. aeruginosa, but this response does not lead to clinical improvement. Therefore, additional studies aimed at identification and understanding of the host factors that influence naturally occurring immune responses to P. aeruginosa are needed. In this investigation, we evaluated the contribution of immunoglobulin GM (γ marker) and KM (κ marker) allotypes to the antibody responses to P. aeruginosa lipopolysaccharide (LPS) O1, O6, O11, and alginate antigens and the broadly-conserved surface polysaccharide expressed by many microbial pathogens, poly-N-acetyl-D-glucosamine (PNAG), in 58 chronically infected CF patients. METHODS IgG1 markers GM 3 and 17 and IgG2 markers GM 23- and 23+ were determined by a pre-designed TaqMan® genotyping assay. The κ chain determinants KM 1 and 3 were characterized by PCR-RFLP. Antibodies to the LPS O antigens, alginate, and PNAG were measured by an ELISA. RESULTS Several significant associations were noted with KM alleles. Particular KM 1/3 genotypes were individually and epistatically (with GM 3/17) associated with the level of IgG antibodies to O1, O11, alginate, and PNAG antigens. CONCLUSIONS Immunoglobulin GM and KM genotypes influence the magnitude of humoral immunity to LPS O, alginate, and PNAG antigens. These results, if confirmed in a larger study population, will be helpful in devising novel immunotherapeutic approaches against P. aeruginosa.
Collapse
Affiliation(s)
- Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States.
| | - Aryan M Namboodiri
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Paul J Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Michael R Knowles
- Department of Medicine, Marsico Lung Institute and Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Rhonda G Pace
- Department of Medicine, Marsico Lung Institute and Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Gerald B Pier
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
18
|
Moraru M, Perez-Portilla A, Al-Akioui Sanz K, Blazquez-Moreno A, Arnaiz-Villena A, Reyburn HT, Vilches C. FCGR Genetic Variation in Two Populations From Ecuador Highlands-Extensive Copy-Number Variation, Distinctive Distribution of Functional Polymorphisms, and a Novel, Locally Common, Chimeric FCGR3B/A (CD16B/A) Gene. Front Immunol 2021; 12:615645. [PMID: 34108956 PMCID: PMC8183472 DOI: 10.3389/fimmu.2021.615645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Fcγ receptors (FcγR), cell-surface glycoproteins that bind antigen-IgG complexes, control both humoral and cellular immune responses. The FCGR locus on chromosome 1q23.3 comprises five homologous genes encoding low-affinity FcγRII and FcγRIII, and displays functionally relevant polymorphism that impacts on human health. Recurrent events of non-allelic homologous recombination across the FCGR locus result in copy-number variation of ~82.5 kbp-long fragments known as copy-number regions (CNR). Here, we characterize a recently described deletion that we name CNR5, which results in loss of FCGR3A, FCGR3B, and FCGR2C, and generation of a recombinant FCGR3B/A gene. We show that the CNR5 recombination spot lies at the beginning of the third FCGR3 intron. Although the FCGR3B/A-encoded hybrid protein CD16B/A reaches the plasma membrane in transfected cells, its possible natural expression, predictably restricted to neutrophils, could not be demonstrated in resting or interferon γ-stimulated cells. As the CNR5-deletion was originally described in an Ecuadorian family from Llano Grande (an indigenous community in North-Eastern Quito), we characterized the FCGR genetic variation in two populations from the highlands of Ecuador. Our results reveal that CNR5-deletion is relatively frequent in Llano Grande (5 carriers out of 36 donors). Furthermore, we found a high frequency of two strong-phagocytosis variants: the FCGR3B-NA1 haplotype and the CNR1 duplication, which translates into an increased FCGR3B and FCGR2C copy-number. CNR1 duplication was particularly increased in Llano Grande, 77.8% of the studied sample carrying at least one such duplication. In contrast, an extended haplotype CD16A-176V – CD32C-ORF+2B.2 – CD32B-2B.4 including strong activating and inhibitory FcγR variants was absent in Llano Grande and found at a low frequency (8.6%) in Ecuador highlands. This particular distribution of FCGR polymorphism, possibly a result of selective pressures, further confirms the importance of a comprehensive, joint analysis of all genetic variations in the locus and warrants additional studies on their putative clinical impact. In conclusion, our study confirms important ethnic variation at the FCGR locus; it shows a distinctive FCGR polymorphism distribution in Ecuador highlands; provides a molecular characterization of a novel CNR5-deletion associated with CD16A and CD16B deficiency; and confirms its presence in that population.
Collapse
Affiliation(s)
- Manuela Moraru
- Immunogenetics & Histocompatibility Lab, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, Majadahonda, Spain
| | - Adriana Perez-Portilla
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Karima Al-Akioui Sanz
- Immunogenetics & Histocompatibility Lab, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, Majadahonda, Spain
| | - Alfonso Blazquez-Moreno
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | | | - Hugh T Reyburn
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Carlos Vilches
- Immunogenetics & Histocompatibility Lab, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, Majadahonda, Spain
| |
Collapse
|
19
|
Quatrini L, Della Chiesa M, Sivori S, Mingari MC, Pende D, Moretta L. Human NK cells, their receptors and function. Eur J Immunol 2021; 51:1566-1579. [PMID: 33899224 PMCID: PMC9292411 DOI: 10.1002/eji.202049028] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/20/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
NK cells are cytotoxic components of innate lymphoid cells (ILC) that provide a first line of defense against viral infections and contribute to control tumor growth and metastasis. Their function is finely regulated by an array of HLA-specific and non-HLA-specific inhibitory and activating receptors which allow to discriminate between healthy and altered cells. Human NK cells gained a major attention in recent years because of the important progresses in understanding their biology and of some promising data in tumor therapy. In this review, we will outline well-established issues of human NK cells and discuss some of the open questions, debates, and recent advances regarding their origin, differentiation, and tissue distribution. Newly defined NK cell specializations, including the impact of inhibitory checkpoints on their function, their crosstalk with other cell types, and the remarkable adaptive features acquired in response to certain virus infections will also be discussed.
Collapse
Affiliation(s)
- Linda Quatrini
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Simona Sivori
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Immunology Laboratory, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela Pende
- Immunology Laboratory, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
20
|
Tso FY, Lidenge SJ, Poppe LK, Peña PB, Privatt SR, Bennett SJ, Ngowi JR, Mwaiselage J, Belshan M, Siedlik JA, Raine MA, Ochoa JB, Garcia-Diaz J, Nossaman B, Buckner L, Roberts WM, Dean MJ, Ochoa AC, West JT, Wood C. Presence of antibody-dependent cellular cytotoxicity (ADCC) against SARS-CoV-2 in COVID-19 plasma. PLoS One 2021; 16:e0247640. [PMID: 33661923 PMCID: PMC7932539 DOI: 10.1371/journal.pone.0247640] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/10/2021] [Indexed: 12/28/2022] Open
Abstract
Background Neutralizing-antibody (nAb) is the major focus of most ongoing COVID-19 vaccine trials. However, nAb response against SARS-CoV-2, when present, decays rapidly. Given the myriad roles of antibodies in immune responses, it is possible that antibodies could also mediate protection against SARS-CoV-2 via effector mechanisms such as antibody-dependent cellular cytotoxicity (ADCC), which we sought to explore here. Methods Plasma of 3 uninfected controls and 20 subjects exposed to, or recovering from, SARS-CoV-2 infection were collected from U.S. and sub-Saharan Africa. Immunofluorescence assay was used to detect the presence of SARS-CoV-2 specific IgG antibodies in the plasma samples. SARS-CoV-2 specific neutralizing capability of these plasmas was assessed with SARS-CoV-2 spike pseudotyped virus. ADCC activity was assessed with a calcein release assay. Results SARS-CoV-2 specific IgG antibodies were detected in all COVID-19 subjects studied. All but three COVID-19 subjects contained nAb at high potency (>80% neutralization). Plasma from 19/20 of COVID-19 subjects also demonstrated strong ADCC activity against SARS-CoV-2 spike glycoprotein, including two individuals without nAb against SARS-CoV-2. Conclusion Both neutralizing and non-neutralizing COVID-19 plasmas can mediate ADCC. Our findings argue that evaluation of potential vaccines against SARS-CoV-2 should include investigation of the magnitude and durability of ADCC, in addition to nAb.
Collapse
Affiliation(s)
- For Yue Tso
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Salum J. Lidenge
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Lisa K. Poppe
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Phoebe B. Peña
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Sara R. Privatt
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Sydney J. Bennett
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - John R. Ngowi
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Julius Mwaiselage
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Michael Belshan
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- Department of Medical Microbiology & Immunology, Creighton University, Omaha, NE, United States of America
| | - Jacob A. Siedlik
- Department of Exercise Science and Pre-Health Professions, Creighton University, Omaha, NE, United States of America
| | - Morgan A. Raine
- Department of Medical Microbiology & Immunology, Creighton University, Omaha, NE, United States of America
| | - Juan B. Ochoa
- Department of Surgery, Ochsner Medical Center, New Orleans, LA, United States of America
| | - Julia Garcia-Diaz
- Department of Internal Medicine Ochsner Medical Center, New Orleans, LA, United States of America
| | - Bobby Nossaman
- Department of Internal Medicine Ochsner Medical Center, New Orleans, LA, United States of America
| | - Lyndsey Buckner
- Department of Internal Medicine Ochsner Medical Center, New Orleans, LA, United States of America
| | - W. Mark Roberts
- Department of Internal Medicine Ochsner Medical Center, New Orleans, LA, United States of America
| | - Matthew J. Dean
- Louisiana State University Cancer Center, New Orleans, LA, United States of America
| | - Augusto C. Ochoa
- Louisiana State University Cancer Center, New Orleans, LA, United States of America
- Department of Pediatrics LSU Health, New Orleans, LA, United States of America
| | - John T. West
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Charles Wood
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- * E-mail:
| |
Collapse
|
21
|
Pandey JP, Kothera RT, Liu S, Costa AS, Mancuso R, Agostini S. Immunoglobulin Genes and Immunity to HSV1 in Alzheimer's Disease. J Alzheimers Dis 2020; 70:917-924. [PMID: 31306125 DOI: 10.3233/jad-190265] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Although genome-wide association studies (GWAS) of late-onset Alzheimer's disease (AD) have identified numerous genes that influence the risk for disease, the majority of the genetic variance of AD remains uncharacterized. Furthermore, current GWAS, despite their name, do not evaluate all genes in the human genome. One such gene complex is immunoglobulin GM (γ marker) genes on chromosome 14. GM genes are excellent candidate genes for AD because they influence immunity to herpes simplex virus type 1 (HSV1), which has been implicated in AD pathology by an increasing number of reports. The aim of this investigation was to determine if particular GM genotypes were associated with AD and mild cognitive impairment (MCI), and whether they contributed to the interindividual differences in the level of anti-HSV1 IgG antibodies. A total of 141 HSV1 seropositive individuals-56 AD patients, 48 MCI individuals, and 37 sex- and age-matched healthy controls-were characterized for GM alleles 3, 17, and 23. The homozygosity for the GM 3 allele was significantly associated with MCI (p = 0.025). GM 3/17 heterozygous AD patients had significantly higher levels of anti-HSV1 antibodies than the healthy controls expressing the same genotype (p = 0.0004). Among MCI subjects, the GM 3/17 genotype was associated with significantly higher level of anti-HSV1 antibodies as compared to the GM 17/17 homozygous genotype (pc = 0.040). Among AD patients, the GM 23+/-genotype was significantly associated with anti-HSV1 antibody responses (pc = 0.025). These results suggest that GM genes could act as potential unifiers of the genetic and viral etiology of AD.
Collapse
Affiliation(s)
- Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Ronald T Kothera
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Shufeng Liu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | |
Collapse
|
22
|
Poppe LK, Wood C, West JT. The Presence of Antibody-Dependent Cell Cytotoxicity-Mediating Antibodies in Kaposi Sarcoma-Associated Herpesvirus-Seropositive Individuals Does Not Correlate with Disease Pathogenesis or Progression. THE JOURNAL OF IMMUNOLOGY 2020; 205:2742-2749. [PMID: 32998986 DOI: 10.4049/jimmunol.2000489] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/04/2020] [Indexed: 01/29/2023]
Abstract
Although the immune response is likely to play a pivotal role in controlling Kaposi sarcoma (KS)-associated herpesvirus (KSHV) and preventing disease development, the exact factors responsible for that control remain ill defined. T cell responses are weak and variable, and neutralizing Abs are more frequently detected in individuals with KS. This suggests a potential role for nonneutralizing Abs, which to date have been largely uninvestigated. Ab-dependent cell cytotoxicity (ADCC) is a common effector function for nonneutralizing Abs and is known to play a protective role in other herpesvirus infections; yet, ADCC has never been investigated in the context of KSHV infection. In this study, we provide, to our knowledge, the first evidence that anti-KSHV Abs are capable of mediating ADCC responses against infected human cells undergoing lytic reactivation. ADCC activity significantly higher than seronegative controls was detected in 24 of 68 KSHV-seropositive individuals tested. However, ADCC responses were not associated with KS development or progression. ADCC activity was also found to be independent of HIV status, sex, age, KSHV Ab titer, and KSHV-neutralizing activity. Nevertheless, additional investigations into effector cell function between KS and asymptomatic individuals are needed to determine whether ADCC has a role in preventing KS.
Collapse
Affiliation(s)
- Lisa K Poppe
- Nebraska Center for Virology, Lincoln, NE 68583.,School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583; and
| | - Charles Wood
- Nebraska Center for Virology, Lincoln, NE 68583.,School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583; and.,Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - John T West
- Nebraska Center for Virology, Lincoln, NE 68583; .,Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68583
| |
Collapse
|
23
|
Costa AS, Agostini S, Guerini FR, Mancuso R, Clerici M, Pandey JP. Relation between FCGRIIB rs1050501 and HSV-1 specific IgG antibodies in Alzheimer's disease. J Transl Med 2020; 18:325. [PMID: 32859213 PMCID: PMC7455989 DOI: 10.1186/s12967-020-02495-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/20/2020] [Indexed: 01/08/2023] Open
Abstract
Background Alzheimer’s Disease (AD) is a chronic neurodegenerative disorder characterized by extracellular plaques, intracellular neurofibrillary tangles and neuronal loss in the central nervous system (CNS). Pathogens are suspected to have a role in the development of AD; herpes simplex virus type 1 (HSV-1), in particular, is suggested to be a risk factor for the disease. The gamma receptor for the Fc portion of IgG molecules (FCGRs) plays a crucial role in regulating immune responses, and among FCGRs, FCGRIIB is endowed with an inhibitory function. Notably, the rs1050501 polymorphism of FCGRIIB gene associates with autoimmune diseases and with neuronal uptake and interneuronal accumulation of amyloid beta in animal AD models. Methods Genotype and allelic distribution of ApoE4 and FCGRIIB rs1050501 were evaluated in a case–control population of 225 AD patients, 93 MCI individuals and 201 sex and age matched healthy controls (HC). HSV-1 total IgG titers and IgG subclasses were detected and quantified in a subgroup of the main study population by ELISA. Results Genotype and allelic distribution of FCGRIIB was comparable in the study population. HSV-1-specific antibody titers were significantly higher in AD and MCI compared to HC (p < 0.01 for both); IgG3 titers, in particular, were increased in MCI compared to AD (p = 0.04). Analyses of possible correlations between the FCGRIIB rs1050501 genotype polymorphism and IgG subclasses showed that the presence of IgG3 was more frequent in MCI carrying the FCGRIIB TT (94.1%) compared to those carrying the CT genotype (63.6%) (p = 0.03). Conclusion Results herein show an association between humoral immune response against HSV-1 and FCGRIIB rs1050501 genetic variation in the first stage of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milano, Milan, Italy
| | - Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
24
|
Warrender AK, Kelton W. Beyond Allotypes: The Influence of Allelic Diversity in Antibody Constant Domains. Front Immunol 2020; 11:2016. [PMID: 32973808 PMCID: PMC7461860 DOI: 10.3389/fimmu.2020.02016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/24/2020] [Indexed: 01/25/2023] Open
Abstract
Polymorphic diversity in antibody constant domains has long been defined by allotypic motifs that cross react with the sera of other individuals. Improvements in sequencing technologies have led to the discovery of a large number of new allelic sequences that underlie this diversity. Many of the point mutations lie outside traditional allotypic motifs suggesting they do not elicit immunogenic responses. As antibodies play an important role in immune defense and biotechnology, understanding how this newly resolved diversity influences the function of antibodies is important. This review investigates the current known diversity of antibody alleles at a protein level for each antibody isotype as well as the kappa and lambda light chains. We focus on evidence emerging for how these mutations perturb antibody interactions with antigens and Fc receptors that are critical for function, as well as the influence this might have on the use of antibodies as therapeutics and reagents.
Collapse
Affiliation(s)
| | - William Kelton
- Te Huataki Waiora School of Health, The University of Waikato, Hamilton, New Zealand
| |
Collapse
|
25
|
Pandey JP, Olsson J, Weidung B, Kothera RT, Johansson A, Eriksson S, Hallmans G, Elgh F, Lövheim H. An Ig γ Marker Genotype Is a Strong Risk Factor for Alzheimer Disease, Independent of Apolipoprotein E ε4 Genotype. THE JOURNAL OF IMMUNOLOGY 2020; 205:1318-1322. [PMID: 32709662 DOI: 10.4049/jimmunol.2000351] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/26/2020] [Indexed: 01/17/2023]
Abstract
Increasing evidence implicates HSV type 1 (HSV1) in the pathogenesis of late-onset Alzheimer disease (AD). HSV1 has evolved highly sophisticated strategies to evade host immunosurveillance. One strategy involves encoding a decoy Fcγ receptor (FcγR), which blocks Fc-mediated effector functions, such as Ab-dependent cellular cytotoxicity. Ig γ marker (GM) allotypes, encoded by highly polymorphic IGHG genes on chromosome 14q32, modulate this immunoevasion strategy, and thus may act as effect modifiers of the HSV1-AD association. In this nested case-control human study, 365 closely matched case-control pairs-whose blood was drawn on average 9.6 y before AD diagnosis-were typed for GM alleles by a TaqMan genotyping assay. APOE genotype and a genetic risk score based on nine additional previously known AD risk genes (ABCA7, BIN1, CD33, CLU, CR1, EPHA1, MS4A4E, NECTIN2, and PICALM) were extracted from a genome-wide association study analysis. Antiviral Abs were measured by ELISA. Conditional logistic regression models were applied. The distribution of GM 3/17 genotypes differed significantly between AD cases and controls, with higher frequency of GM 17/17 homozygotes in AD cases as compared with controls (19.8 versus 10.7%, p = 0.001). The GM 17/17 genotype was associated with a 4-fold increased risk of AD (odds ratio 4.142, p < 0.001). In conclusion, the results of this study demonstrate that Ig GM 17/17 genotype contributes to the risk of later AD development, independent of apolipoprotein ε4 genotype and other AD risk genes, and explain, at least in part, why every HSV1-infected person is not equally likely to develop HSV1-associated AD.
Collapse
Affiliation(s)
- Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425;
| | - Jan Olsson
- Division of Virology, Department of Clinical Microbiology, Umeå University, 901 85 Umeå, Sweden
| | - Bodil Weidung
- Division of Geriatric Medicine, Department of Public Health and Caring Sciences, Uppsala University, 751 05 Uppsala, Sweden
| | - Ronald T Kothera
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Anders Johansson
- Department of Odontology, Umeå University, Umeå, 901 85, Sweden.,Division of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden
| | - Sture Eriksson
- Division of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.,Division of Geriatric Medicine, Department of Community Medicine and Rehabilitation, Umeå University, 901 85 Umeå, Sweden; and
| | - Göran Hallmans
- Division of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden
| | - Fredrik Elgh
- Division of Virology, Department of Clinical Microbiology, Umeå University, 901 85 Umeå, Sweden
| | - Hugo Lövheim
- Division of Geriatric Medicine, Department of Community Medicine and Rehabilitation, Umeå University, 901 85 Umeå, Sweden; and.,Wallenberg Centre for Molecular Medicine, Umeå University, 901 85 Umeå, Sweden
| |
Collapse
|
26
|
Cortés DA, Aguilar MC, Ríos HA, Rodríguez FJ, Montes KV, Gómez-Marín JE, de-la-Torre A. Severe acute multi-systemic failure with bilateral ocular toxoplasmosis in immunocompetent patients from urban settings in Colombia: Case reports. Am J Ophthalmol Case Rep 2020; 18:100661. [PMID: 32195446 PMCID: PMC7078491 DOI: 10.1016/j.ajoc.2020.100661] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 10/28/2019] [Accepted: 03/09/2020] [Indexed: 01/22/2023] Open
Abstract
Propose To report two cases of severe acute multi-systemic failure with bilateral ocular toxoplasmosis in immunocompetent patients from urban settings in Colombia. Observations We report two immunocompetent male patients aged 44- and 67-years-old who, despite not having visited the Amazonian region in Colombia, had severe bilateral posterior uveitis and extensive-bilateral macular lesions and multiple organ failure that required admission to an intensive care unit. Toxoplasma gondii was positive by PCR assay in vitreous humor samples. Patients were treated with intravitreal clindamycin and dexamethasone in addition to systemic treatment with trimethoprim-sulfamethoxazole. In both patients, infection by atypical strains was confirmed; in one case by serotyping and in another one by genotyping (ROP 18 virulent allele). After 2 and 4 months of treatment respectively, the patients showed improvement of the posterior uveitis and its systemic manifestations. However, there was no significant visual acuity improvement due to bilateral extensive macular involvement. Conclusions and importance Clinicians should be aware that toxoplasmosis originating from South America could be associated with severe acute multisystemic and intraocular bilateral involvement, even in patients with no history of exposure to jungle environments.
Collapse
Affiliation(s)
- Diana Alejandra Cortés
- Fundación Oftalmológica Nacional, Uveitis and Retina Services, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Cl. 50 #13 - 50, Bogotá, Colombia
| | - María Camila Aguilar
- Fundación Oftalmológica Nacional, Uveitis and Retina Services, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Cl. 50 #13 - 50, Bogotá, Colombia
| | - Hernán Andres Ríos
- Fundación Oftalmológica Nacional, Uveitis and Retina Services, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Cl. 50 #13 - 50, Bogotá, Colombia
| | - Francisco José Rodríguez
- Fundación Oftalmológica Nacional, Uveitis and Retina Services, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Cl. 50 #13 - 50, Bogotá, Colombia
| | - Kelly Verónica Montes
- Fundación Oftalmológica Nacional, Uveitis and Retina Services, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Cl. 50 #13 - 50, Bogotá, Colombia
| | - Jorge Enrique Gómez-Marín
- GEPAMOL Group, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Carrera 15 calle 12 norte Armenia, Quindío, Colombia
| | - Alejandra de-la-Torre
- Fundación Oftalmológica Nacional, Uveitis and Retina Services, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Cl. 50 #13 - 50, Bogotá, Colombia.,Neuroscience Research group (NeURos), Escuela de Medicina y Ciencias de la Salud, Unidad de Inmunología, Universidad del Rosario, Carrera 24 # 63 C 69, Bogotá, Colombia
| |
Collapse
|
27
|
Gyurova IE, Ali A, Waggoner SN. Natural Killer Cell Regulation of B Cell Responses in the Context of Viral Infection. Viral Immunol 2019; 33:334-341. [PMID: 31800366 DOI: 10.1089/vim.2019.0129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Secretion of both neutralizing and nonneutralizing virus-specific antibodies by B cells is a key component of immune control of many virus infections and a critical benchmark of successful preventative vaccines. Natural killer (NK) cells also play a vital role in antiviral immune defense via cytolytic elimination of infected cells and production of proinflammatory antiviral cytokines. Accumulating evidence points to multifaceted crosstalk between NK cells and antiviral B cell responses that can determine virus elimination, pathogenesis of infection, and efficacy of vaccine-elicited protection. These outcomes are a result of both positive and negative influences of NK cells on the B cell responses, as well as canonical antiviral killing of infected B cells. On one hand, NK cell-derived cytokines such as interferon-gamma (IFN-γ) may promote B cell activation and enhance immunoglobulin production. In contrast, NK cell immunoregulatory killing of CD4 T cells can limit affinity maturation in germinal centers resulting in weak infection or vaccine induction of antiviral neutralizing antibodies. In this review, we will discuss these and other dueling contributions of NK cells to B cell responses during virus infection or vaccination.
Collapse
Affiliation(s)
- Ivayla E Gyurova
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ayad Ali
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati, Cincinnati, Ohio, USA.,Graduate Program in Immunology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stephen N Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati, Cincinnati, Ohio, USA.,Graduate Program in Immunology, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Di Bona D, Pandey JP, Aiello A, Bilancia M, Candore G, Caruso C, Colomba C, Duro G, Ligotti ME, Macchia L, Rizzo S, Accardi G. The immunoglobulin γ marker 17 allotype and KIR/HLA genes prevent the development of chronic hepatitis B in humans. Immunology 2019; 159:178-182. [PMID: 31613998 DOI: 10.1111/imm.13133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/18/2019] [Accepted: 10/09/2019] [Indexed: 01/02/2023] Open
Abstract
Hepatitis B virus (HBV) infection causes a self-limiting disease in most individuals. However, < 10% of infected subjects develop a chronic disease. Genetic host variability of polymorphic genes at the interface of innate and acquired immunity, such as killer immunoglobulin-like receptors (KIR), their human leucocyte antigen (HLA) and IgG allotypes (GM), could explain this different clinical picture. We previously showed a protective role of the KIR2DL3 gene for the development of chronic hepatitis B (CHB), and a detrimental role of the KIR ligand groups, HLA-A-Bw4 and HLA-C2. We have expanded the previous analysis genotyping patients for GM23 and GM3/17 allotypes. The comparison of the patients with CHB with those who resolved HBV infection showed that the presence of GM17 allele virtually eliminated the risk of developing CHB (OR, 0·03; 95% CI, 0·004-0·16; P < 0·0001). In addition, the combination of GM17, KIR2DL3, HLA-A-Bw4 and HLA-C2 was highly sensitive to predict the outcome of HBV infection.
Collapse
Affiliation(s)
- Danilo Di Bona
- Dipartimento dell'Emergenza e dei Trapianti d'Organo, Università di Bari Aldo Moro, Bari, Italy
| | - Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Anna Aiello
- Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Massimo Bilancia
- Dipartmento Jonico in Sistemi Giuridici ed Economici del Mediterraneo: società, ambiente, culture, Università di Bari Aldo Moro, Bari, Italy
| | - Giuseppina Candore
- Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Calogero Caruso
- Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Claudia Colomba
- Dipartimento di Scienze per la Promozione della Salute e Materno-Infantile 'G. D'Alessandro', Università di Palermo, Palermo, Italy
| | - Giovanni Duro
- Istituto di Biomedicina ed Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Mattia Emanuela Ligotti
- Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Luigi Macchia
- Dipartimento dell'Emergenza e dei Trapianti d'Organo, Università di Bari Aldo Moro, Bari, Italy
| | - Sergio Rizzo
- Unità Operativa di Medicina Trasfusionale, AOUP, Palermo, Italy
| | - Giulia Accardi
- Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| |
Collapse
|
29
|
Della Chiesa M, De Maria A, Muccio L, Bozzano F, Sivori S, Moretta L. Human NK Cells and Herpesviruses: Mechanisms of Recognition, Response and Adaptation. Front Microbiol 2019; 10:2297. [PMID: 31636622 PMCID: PMC6788305 DOI: 10.3389/fmicb.2019.02297] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/20/2019] [Indexed: 12/01/2022] Open
Abstract
NK cells contribute to early defenses against viruses through their inborn abilities that include sensing of PAMPs and inflammatory signals such as cytokines or chemokines, recognition, and killing of infected cells through activating surface receptors engagement. Moreover, they support adaptive responses via Ab-dependent mechanisms, triggered by CD16, and DC editing. Their fundamental role in anti-viral responses has been unveiled in patients with NK cell deficiencies suffering from severe Herpesvirus infections. Notably, these infections, often occurring as primary infections early in life, can be efficiently cleared by NK, T, and B cells in healthy hosts. Herpesviruses however, generate a complicated balance with the host immune system through their latency cycle moving between immune control and viral reactivation. This lifelong challenge has contributed to the development of numerous evasion mechanisms by Herpesviruses, many of which devoted to elude NK cell surveillance from viral reactivations rather than primary infections. This delicate equilibrium can be altered in proportions of healthy individuals promoting virus reactivation and, more often, in immunocompromised subjects. However, the constant stimulus provided by virus-host interplay has also favored NK-cell adaptation to Herpesviruses. During anti-HCMV responses, NK cells can reshape their receptor repertoire and function, through epigenetic remodeling, and acquire adaptive traits such as longevity and clonal expansion abilities. The major mechanisms of recognition and effector responses employed by NK cells against Herpesviruses, related to their genomic organization will be addressed, including those allowing NK cells to generate memory-like responses. In addition, the mechanisms underlying virus reactivation or control will be discussed.
Collapse
Affiliation(s)
- Mariella Della Chiesa
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Andrea De Maria
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,Department of Health Sciences (DISSAL), School of Medical and Pharmaceutical Sciences University of Genoa, Genoa, Italy.,Clinica Malattie Infettive, Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Letizia Muccio
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Federica Bozzano
- Laboratory of Tumor Immunology, Department of Immunology, IRCCS Ospedale Bambino Gesù, Rome, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Lorenzo Moretta
- Laboratory of Tumor Immunology, Department of Immunology, IRCCS Ospedale Bambino Gesù, Rome, Italy
| |
Collapse
|
30
|
Jenks JA, Goodwin ML, Permar SR. The Roles of Host and Viral Antibody Fc Receptors in Herpes Simplex Virus (HSV) and Human Cytomegalovirus (HCMV) Infections and Immunity. Front Immunol 2019; 10:2110. [PMID: 31555298 PMCID: PMC6742691 DOI: 10.3389/fimmu.2019.02110] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/21/2019] [Indexed: 12/01/2022] Open
Abstract
Herpesvirus infections are a leading cause of neurodevelopmental delay in newborns and end-organ disease in immunocompromised patients. One leading strategy to reduce the disease burden of herpesvirus infections such as herpes simplex virus (HSV) and human cytomegalovirus (HCMV) is to prevent primary acquisition by vaccination, yet vaccine development remains hampered by limited understanding of immune correlates of protection against infection. Traditionally, vaccine development has aimed to increase antibody titers with neutralizing function, which involves the direct binding of antibodies to viral particles. However, recent research has explored the numerous other responses that can be mediated by engagement of the antibody constant region (Fc) with Fc receptors (FcR) present on immune cells or with complement molecules. These functions include antiviral responses such as antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Uniquely, herpesviruses encode FcR that can act as distractor receptors for host antiviral IgG, thus enabling viral evasion of host defenses. This review focuses on the relative roles of neutralizing and non-neutralizing functions antibodies that target herpesvirus antigens for HSV and HCMV, as well as the roles of Fc-FcR interactions for both host defenses and viral escape.
Collapse
Affiliation(s)
- Jennifer A Jenks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States
| | - Matthew L Goodwin
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States
| | - Sallie R Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States.,Department of Pediatrics, Children's Health and Discovery Institute, Durham, NC, United States
| |
Collapse
|
31
|
Muntasell A, Servitja S, Cabo M, Bermejo B, Pérez-Buira S, Rojo F, Costa-García M, Arpí O, Moraru M, Serrano L, Tusquets I, Martínez MT, Heredia G, Vera A, Martínez-García M, Soria L, Comerma L, Santana-Hernández S, Eroles P, Rovira A, Vilches C, Lluch A, Albanell J, López-Botet M. High Numbers of Circulating CD57 + NK Cells Associate with Resistance to HER2-Specific Therapeutic Antibodies in HER2 + Primary Breast Cancer. Cancer Immunol Res 2019; 7:1280-1292. [PMID: 31189644 DOI: 10.1158/2326-6066.cir-18-0896] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/11/2019] [Accepted: 06/10/2019] [Indexed: 11/16/2022]
Abstract
Natural killer (NK) cells can orchestrate effective antitumor immunity. The presence of tumor-infiltrating NK cells in diagnostic biopsies predicts pathologic complete response (pCR) to HER2-specific therapeutic antibodies in patients with primary breast cancer. Here, we analyzed whether diversity in circulating NK cells might influence tumor infiltration and HER2-specific therapeutic antibody efficacy. We found that numbers of circulating CD57+ NK cells inversely correlated with pCR to HER2-specific antibody treatment in patients with primary breast cancer independently of age, traditional clinicopathologic factors, and CD16A 158F/V genotype. This association was uncoupled from the expression of other NK-cell receptors, the presence of adaptive NK cells, or changes in major T-cell subsets, reminiscent of cytomegalovirus-induced immunomodulation. NK-cell activation against trastuzumab-coated HER2+ breast cancer cells was comparable in patients with high and low proportions of CD57+ NK cells. However, circulating CD57+ NK cells displayed decreased CXCR3 expression and CD16A-induced IL2-dependent proliferation in vitro Presence of CD57+ NK cells was reduced in breast tumor-associated infiltrates as compared with paired peripheral blood samples, suggesting deficient homing, proliferation, and/or survival of NK cells in the tumor niche. Indeed, numbers of circulating CD57+ were inversely related to tumor-infiltrating NK-cell numbers. Our data reveal that NK-cell differentiation influences their antitumor potential and that CD57+ NK cells may be a biomarker useful for tailoring HER2 antibody-based therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Aura Muntasell
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.
| | - Sònia Servitja
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Mariona Cabo
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Begoña Bermejo
- Department of Oncology, Hospital Clinico de Valencia-CIBERONC, Valencia, Spain
| | - Sandra Pérez-Buira
- Department of Pathology, IIS "Fundacion Jimenez Diaz University Hospital," Madrid, Spain
| | - Federico Rojo
- Department of Pathology, IIS "Fundacion Jimenez Diaz University Hospital," Madrid, Spain
| | | | - Oriol Arpí
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Manuela Moraru
- HLA-Immunogenetics Department, Instituto Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Laia Serrano
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Ignasi Tusquets
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | - Andrea Vera
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - María Martínez-García
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Laura Soria
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Laura Comerma
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Sara Santana-Hernández
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Pilar Eroles
- Department of Oncology, Hospital Clinico de Valencia-CIBERONC, Valencia, Spain.,Biomedical Research Institute, INCLIVA, Valencia, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Carlos Vilches
- HLA-Immunogenetics Department, Instituto Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Ana Lluch
- Department of Oncology, Hospital Clinico de Valencia-CIBERONC, Valencia, Spain.,Biomedical Research Institute, INCLIVA, Valencia, Spain.,Universitat de Valencia, Valencia, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain. .,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Miguel López-Botet
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
32
|
van Erp EA, Luytjes W, Ferwerda G, van Kasteren PB. Fc-Mediated Antibody Effector Functions During Respiratory Syncytial Virus Infection and Disease. Front Immunol 2019; 10:548. [PMID: 30967872 PMCID: PMC6438959 DOI: 10.3389/fimmu.2019.00548] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/28/2019] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract infections and hospitalization in infants under 1 year of age and there is currently no market-approved vaccine available. For protection against infection, young children mainly depend on their innate immune system and maternal antibodies. Traditionally, antibody-mediated protection against viral infections is thought to be mediated by direct binding of antibodies to viral particles, resulting in virus neutralization. However, in the case of RSV, virus neutralization titers do not provide an adequate correlate of protection. The current lack of understanding of the mechanisms by which antibodies can protect against RSV infection and disease or, alternatively, contribute to disease severity, hampers the design of safe and effective vaccines against this virus. Importantly, neutralization is only one of many mechanisms by which antibodies can interfere with viral infection. Antibodies consist of two structural regions: a variable fragment (Fab) that mediates antigen binding and a constant fragment (Fc) that mediates downstream effector functions via its interaction with Fc-receptors on (innate) immune cells or with C1q, the recognition molecule of the complement system. The interaction with Fc-receptors can lead to killing of virus-infected cells through a variety of immune effector mechanisms, including antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Antibody-mediated complement activation may lead to complement-dependent cytotoxicity (CDC). In addition, both Fc-receptor interactions and complement activation can exert a broad range of immunomodulatory functions. Recent studies have emphasized the importance of Fc-mediated antibody effector functions in both protection and pathogenesis for various infectious agents. In this review article, we aim to provide a comprehensive overview of the current knowledge on Fc-mediated antibody effector functions in the context of RSV infection, discuss their potential role in establishing the balance between protection and pathogenesis, and point out important gaps in our understanding of these processes. Furthermore, we elaborate on the regulation of these effector functions on both the cellular and humoral side. Finally, we discuss the implications of Fc-mediated antibody effector functions for the rational design of safe and effective vaccines and monoclonal antibody therapies against RSV.
Collapse
Affiliation(s)
- Elisabeth A. van Erp
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Radboud Center for Infectious Diseases, Nijmegen, Netherlands
| | - Willem Luytjes
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Gerben Ferwerda
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Radboud Center for Infectious Diseases, Nijmegen, Netherlands
| | - Puck B. van Kasteren
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
33
|
Puca AA, Ferrario A, Maciag A, Accardi G, Aiello A, Gambino CM, Candore G, Caruso C, Namboodiri AM, Pandey JP. Association of immunoglobulin GM allotypes with longevity in long-living individuals from Southern Italy. IMMUNITY & AGEING 2018; 15:26. [PMID: 30450119 PMCID: PMC6219083 DOI: 10.1186/s12979-018-0134-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/19/2018] [Indexed: 01/04/2023]
Abstract
Background The aim of this study was to analyse the role of GM allotypes, i.e. the hereditary antigenic determinants expressed on immunoglobulin polypeptide chains, in the attainment of longevity. The role played by immunoglobulin allotypes in the control of immune responses is well known as well as the role of an efficient immune response in longevity achievement. So, it is conceivable that particular GM allotypes may contribute to the generation of an efficient immune response that supports successful ageing, hence longevity. Methods In order to show if GM allotypes play a role in the achievement of longevity, we typed the DNA of 95 Long-living individuals (LLIs) and 96 young control individuals (YCs) from South Italy for GM3/17 and GM23+/− alleles. Results To demonstrate the role of GM allotypes in the attainment of longevity we compared genotype and allele frequencies of GM allotypes between LLIs and YCs. A global chi-square test (3 × 2) shows that the distribution of genotypes at the GM 3/17 locus is highly significantly different in LLIs from that observed in YCs (p < 0.0001). The 2 × 2 chi-square test shows that the carriers of the GM3 allele contribute to this highly significant difference. Accordingly, GM3 allele is significantly overrepresented in LLIs. No significant differences were instead observed regarding GM23 allele. Conclusion These preliminary results show that GM3 allotype is significantly overrepresented in LLIs. To best of our knowledge, this is the first study performed to assess the role of GM allotypes in longevity. So, it should be necessary to verify the data in a larger sample of individuals to confirm GM role in the attainment of longevity.
Collapse
Affiliation(s)
- Annibale A Puca
- 1Department of Medicine and Surgery, University of Salerno, Baronissi, Italy.,2IRCCS MultiMedica, Milan, Italy
| | | | | | - Giulia Accardi
- 3Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy
| | - Anna Aiello
- 3Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy
| | - Caterina Maria Gambino
- 3Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy
| | - Giuseppina Candore
- 3Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy
| | - Calogero Caruso
- 3Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy
| | - Aryan M Namboodiri
- 4Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Janardan P Pandey
- 4Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425 USA
| |
Collapse
|
34
|
Parsons MS, Chung AW, Kent SJ. Importance of Fc-mediated functions of anti-HIV-1 broadly neutralizing antibodies. Retrovirology 2018; 15:58. [PMID: 30134945 PMCID: PMC6103878 DOI: 10.1186/s12977-018-0438-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/09/2018] [Indexed: 01/11/2023] Open
Abstract
Anti-HIV-1 broadly neutralizing antibodies (BnAbs) exhibit an impressive capacity to protect against chimeric SIV-HIV (SHIV) challenges in macaques and potently reduce viremia in both SHIV-infected macaques and HIV-1-infected humans. There is a body of evidence suggesting Fc-mediated functions of anti-HIV-1 binding antibodies are important in protecting from infection and controlling viremia. The degree to which the efficacy of BnAbs is assisted by Fc-mediated functions is of great interest. Challenge experiments with the older generation BnAb b12 showed that mutating the Fc region to abrogate Fcγ receptor binding reduced protective efficacy in macaques. Similar data have been generated with newer BnAbs using murine models of HIV-1. In addition, the degree to which therapeutically administered BnAbs reduce viremia suggests that elimination of infected cells through Fc-mediated functions may contribute to their efficacy. Fc-mediated functions that eliminate infected cells may be particularly important for challenge systems involving cell-associated virus. Herein we review data regarding the importance of Fc-mediated functions of BnAbs in mediating protective immunity and control of viremia.
Collapse
Affiliation(s)
- Matthew S Parsons
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria, Australia.
| | - Amy W Chung
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria, Australia. .,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Victoria, Australia. .,Melbourne Sexual Health Centre, Alfred Hospital, Monash University Central Clinical School, Victoria, Australia.
| |
Collapse
|
35
|
Muccio L, Falco M, Bertaina A, Locatelli F, Frassoni F, Sivori S, Moretta L, Moretta A, Della Chiesa M. Late Development of FcεRγ neg Adaptive Natural Killer Cells Upon Human Cytomegalovirus Reactivation in Umbilical Cord Blood Transplantation Recipients. Front Immunol 2018; 9:1050. [PMID: 29868012 PMCID: PMC5968376 DOI: 10.3389/fimmu.2018.01050] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/27/2018] [Indexed: 01/25/2023] Open
Abstract
In human natural killer (NK) cells, human cytomegalovirus (HCMV) has been shown to be a driving force capable of inducing the expansion of a highly differentiated NKG2C+CD57+ subset, persisting over time in both HCMV+ healthy subjects and umbilical cord blood transplantation (UCBT) recipients experiencing HCMV viral reactivation. In HCMV+ healthy subjects, such expanded NK-cells are characterized by epigenetic modifications that modulate their phenotypic and functional characteristics. In particular, an enhanced ADCC activity is detectable in NK cells lacking the signaling protein FcεRγ. Timing and mechanisms involved in the acquisition of HCMV-induced, adaptive-like features by NK cells are currently unknown. In this study, we investigated the de novo acquisition of several adaptive features in NK cells developing after UCBT by monitoring NK-cell differentiation for at least 2 years after transplant. In UCBT recipients experiencing HCMV reactivation, a rapid phenotypic reconfiguration occurred resulting in the expected expansion of CD56dim NKG2C+CD57+ NK cells. However, while certain HCMV-driven adaptive hallmarks, including high KIR, LILRB1, CD2 and low/negative NKG2A, Siglec-7, and CD161 expression, were acquired early after UCBT (namely by month 6), downregulation of the signaling protein FcεRγ was detected at a later time interval (i.e., by month 12). This feature characterized only a minor fraction of the HCMV-imprinted NKG2C+CD57+ CD56dim NK cell subset, while it was detectable in higher proportions of CD57+ NK cells lacking NKG2C. Interestingly, in patients developing a hyporesponsive CD56-CD16bright NK-cell subset, FcεRγ downregulation occurred in these cells earlier than in CD56dim NK cells. Our data suggest that the acquisition of a fully "adaptive" profile requires signals that may lack in UCBT recipients and/or longer time is needed to obtain a stable epigenetic reprogramming. On the other hand, we found that both HCMV-induced FcεRγneg and FcεRγ+ NK cells from these patients, display similar CD107a degranulation and IFN-γ production capabilities in response to different stimuli, thus indicating that the acquisition of specialized effector functions can be achieved before the "adaptation" to HCMV is completed. Our study provides new insights in the process leading to the generation of different adaptive NK-cell subsets and may contribute to develop new approaches for their employment as novel immunotherapeutic tools.
Collapse
Affiliation(s)
- Letizia Muccio
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy
| | - Michela Falco
- IRCCS Istituto Giannina Gaslini, Dipartimento dei Laboratori di Ricerca, Genova, Italy
| | - Alice Bertaina
- IRCCS Ospedale Pediatrico Bambino Gesù, Dipartimento di Oncoematologia e Terapia Cellulare e Genica, Rome, Italy
| | - Franco Locatelli
- IRCCS Ospedale Pediatrico Bambino Gesù, Dipartimento di Oncoematologia e Terapia Cellulare e Genica, Rome, Italy.,Dipartimento di Scienze Pediatriche, Università degli Studi di Pavia, Pavia, Italy
| | - Francesco Frassoni
- IRCCS Istituto Giannina Gaslini, Dipartimento dei Laboratori di Ricerca, Genova, Italy
| | - Simona Sivori
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy.,Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova, Genova, Italy
| | - Lorenzo Moretta
- IRCCS Ospedale Pediatrico Bambin Gesù, Area di Ricerca Immunologica, Rome, Italy
| | - Alessandro Moretta
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy.,Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova, Genova, Italy
| | - Mariella Della Chiesa
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy.,Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova, Genova, Italy
| |
Collapse
|
36
|
Suárez-Fueyo A, Bradley SJ, Katsuyama T, Solomon S, Katsuyama E, Kyttaris VC, Moulton VR, Tsokos GC. Downregulation of CD3ζ in NK Cells from Systemic Lupus Erythematosus Patients Confers a Proinflammatory Phenotype. THE JOURNAL OF IMMUNOLOGY 2018; 200:3077-3086. [PMID: 29602774 DOI: 10.4049/jimmunol.1700588] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 03/06/2018] [Indexed: 01/14/2023]
Abstract
Cytotoxic function and cytokine profile of NK cells are compromised in patients with systemic lupus erythematosus (SLE). CD3ζ, an important molecule for NK cell activation, is downregulated in SLE T cells and contributes to their altered function. However, little is known about the role of CD3ζ in SLE NK cells. We studied CD3ζ levels and its contribution to cytotoxic, degranulation, and cytokine production capacity of NK cells from patients with SLE. Furthermore, we studied the human NK cell line, NKL, in which manipulation of CD3ζ levels was achieved using small interfering RNA and NK cells from Rag2 mice deficient in CD3ζ. We found reduced CD3ζ expression in NK cells from SLE patients independent of disease activity. Downregulation of CD3ζ expression in NK cells is mediated, at least in part, by Caspase 3, the activity of which is higher in NK cells from patients with SLE compared with NK cells from healthy donors. CD3ζ levels correlated inversely with natural cytotoxicity and the percentage of cells capable of producing the proinflammatory cytokines IFN-γ and TNF. In contrast, CD3ζ levels showed a direct correlation with levels of Ab-dependent cellular cytotoxicity. Experiments performed in CD3ζ-silenced NKL and CD3ζ-deficient NK cells from Rag2 mice confirmed the dependence of NK cell function on CD3ζ levels. Our results demonstrate a differential role for CD3ζ in natural cytotoxicity and Ab-dependent cellular cytotoxicity. We conclude that downregulated CD3ζ confers a proinflammatory phenotype to SLE NK cells and contributes to their altered function in patients with SLE.
Collapse
Affiliation(s)
- Abel Suárez-Fueyo
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Sean J Bradley
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Takayuki Katsuyama
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Sarah Solomon
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Eri Katsuyama
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Vasileios C Kyttaris
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Vaishali R Moulton
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
37
|
Pandey JP. HIV-1 Control and Immunoglobulin Genes. J Infect Dis 2018; 217:1170. [PMID: 29346629 DOI: 10.1093/infdis/jiy021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/12/2017] [Indexed: 11/12/2022] Open
Affiliation(s)
- Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston
| |
Collapse
|
38
|
Chung AW, Alter G. Systems serology: profiling vaccine induced humoral immunity against HIV. Retrovirology 2017; 14:57. [PMID: 29268769 PMCID: PMC5740944 DOI: 10.1186/s12977-017-0380-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/27/2017] [Indexed: 02/04/2023] Open
Abstract
The results of the RV144 HIV vaccine, in combination with several recent non-human primate vaccine studies continue to highlight the potentially protective role of non-neutralizing Fc functional antibodies in HIV vaccine design. For many currently licensed vaccines, assays that detect antigen-specific antibody titers or neutralization levels have been used as a correlate of protection. However, antibodies can confer protection through multiple other mechanisms beyond neutralization, or mechanisms which are not dependent on total antibody titers. Alternative strategies that allow us to further understand the precise mechanisms by which antibodies confer protection against HIV and other infectious pathogens is vitally important for the development of future vaccines. Systems serology aims to comprehensively survey a diverse array of antibody features and functions, in order to simultaneously examine the mechanisms behind and distinguish the most important antibody features required for protection, thus identifying key targets for future experimental vaccine testing. This review will focus on the technical aspects required for the application of Systems serology and summarizes the recent advances provided by application of this systemic analytical approach.
Collapse
Affiliation(s)
- Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, 792 Elizabeth St., Melbourne, VIC, 3000, Australia.
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard University, 400 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
39
|
Di Bona D, Accardi G, Aiello A, Bilancia M, Candore G, Colomba C, Caruso C, Duro G, Gambino CM, Macchia L, Pandey JP. Association between γ marker, human leucocyte antigens and killer immunoglobulin-like receptors and the natural course of human cytomegalovirus infection: a pilot study performed in a Sicilian population. Immunology 2017; 153:523-531. [PMID: 29067686 DOI: 10.1111/imm.12855] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/07/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022] Open
Abstract
Natural killer (NK) cells provide a major defence against human cytomegalovirus (HCMV) infection through the interaction of their surface receptors, including the activating and inhibitory killer immunoglobulin-like receptors (KIRs), and human leucocyte antigen (HLA) class I molecules. Also γ marker (GM) allotypes, able to influence the NK antibody-dependent cell-mediated cytotoxicity, appear to be involved in the immunological control of virus infections, including HCMV. In some cases, their contribution requires epistatic interaction with other genes of the immune system, such as HLA. In the present report, with the aim of gaining insight into the immune mechanisms controlling HCMV, we have studied the possible associations among humoral and NK responses, and HCMV infections. In a previous study we assessed whether the KIR and HLA repertoire might influence the risk of developing symptomatic (n = 60) or asymptomatic (n = 60) disease after primary HCMV infection in the immunocompetent host. In the present study, the immunocompetent patients with primary symptomatic HCMV infection were genotyped for GM3/17 and GM23 allotypes, along with the 60 participants with a previous asymptomatic infection as controls. Notwithstanding the presence of missing data record, advanced missing data recovery techniques were able to show that individuals carrying the GM23 allotypes, both homozygous and heterozygous, GM17/17, HLA-C2 and Bw4T KIR-ligand groups are associated with the risk of developing symptomatic infection. Our findings on the role of both cellular and humoral immunity in the control of HCMV infection should be of value in guiding efforts to reduce HCMV-associated health complications in the elderly, including immunosenescence, and in transplantation.
Collapse
Affiliation(s)
- Danilo Di Bona
- Dipartimento dell'Emergenza e dei Trapianti d'Organo, Università di Bari Aldo Moro, Bari, Italy
| | - Giulia Accardi
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Anna Aiello
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Massimo Bilancia
- Dipartmento Jonico in Sistemi Giuridici ed Economici del Mediterraneo: società, ambiente, culture, Università di Bari Aldo Moro, Bari, Italy
| | - Giuseppina Candore
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Claudia Colomba
- Dipartimento di Scienze per la Promozione della Salute e Materno-Infantile "G. D'Alessandro", Università di Palermo, Palermo, Italy
| | - Calogero Caruso
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Giovanni Duro
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Caterina M Gambino
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Luigi Macchia
- Dipartimento dell'Emergenza e dei Trapianti d'Organo, Università di Bari Aldo Moro, Bari, Italy
| | - Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, USA
| |
Collapse
|
40
|
Muntasell A, Cabo M, Servitja S, Tusquets I, Martínez-García M, Rovira A, Rojo F, Albanell J, López-Botet M. Interplay between Natural Killer Cells and Anti-HER2 Antibodies: Perspectives for Breast Cancer Immunotherapy. Front Immunol 2017; 8:1544. [PMID: 29181007 PMCID: PMC5694168 DOI: 10.3389/fimmu.2017.01544] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/30/2017] [Indexed: 01/16/2023] Open
Abstract
Overexpression of the human epidermal growth factor receptor 2 (HER2) defines a subgroup of breast tumors with aggressive behavior. The addition of HER2-targeted antibodies (i.e., trastuzumab, pertuzumab) to chemotherapy significantly improves relapse-free and overall survival in patients with early-stage and advanced disease. Nonetheless, considerable proportions of patients develop resistance to treatment, highlighting the need for additional and co-adjuvant therapeutic strategies. HER2-specific antibodies can trigger natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity and indirectly enhance the development of tumor-specific T cell immunity; both mechanisms contributing to their antitumor efficacy in preclinical models. Antibody-dependent NK cell activation results in the release of cytotoxic granules as well as the secretion of pro-inflammatory cytokines (i.e., IFNγ and TNFα) and chemokines. Hence, NK cell tumor suppressive functions include direct cytolytic killing of tumor cells as well as the regulation of subsequent antitumor adaptive immunity. Albeit tumors with gene expression signatures associated to the presence of cytotoxic lymphocyte infiltrates benefit from trastuzumab-based treatment, NK cell-related biomarkers of response/resistance to HER2-specific therapeutic antibodies in breast cancer patients remain elusive. Several variables, including (i) the configuration of the patient NK cell repertoire; (ii) tumor molecular features (i.e., estrogen receptor expression); (iii) concomitant therapeutic regimens (i.e., chemotherapeutic agents, tyrosine kinase inhibitors); and (iv) evasion mechanisms developed by progressive breast tumors, have been shown to quantitatively and qualitatively influence antibody-triggered NK cell responses. In this review, we discuss possible interventions for restoring/enhancing the therapeutic activity of HER2 therapeutic antibodies by harnessing NK cell antitumor potential through combinatorial approaches, including immune checkpoint blocking/stimulatory antibodies, cytokines and toll-like receptor agonists.
Collapse
Affiliation(s)
- Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Mariona Cabo
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Sonia Servitja
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Department of Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Ignasi Tusquets
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Department of Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - María Martínez-García
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Department of Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Ana Rovira
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Department of Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | | | - Joan Albanell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Department of Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain.,Univ. Pompeu Fabra, Barcelona, Spain
| | - Miguel López-Botet
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Univ. Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
41
|
Sung AP, Tang JJJ, Guglielmo MJ, Redelman D, Smith-Gagen J, Bateman L, Hudig D. An improved method to quantify human NK cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) per IgG FcR-positive NK cell without purification of NK cells. J Immunol Methods 2017; 452:63-72. [PMID: 29113954 DOI: 10.1016/j.jim.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022]
Abstract
Natural killer (NK) lymphocyte ADCC supports anti-viral protection and monoclonal antibody (mAb) anti-tumor therapies. To predict in vivo ADCC therapeutic responses of different individuals, measurement of both ADCC cellular lytic capacity and their NK cellular receptor recognition of antibodies on 'target' cells are needed, using clinically available amounts of blood. Twenty ml of blood provides sufficient peripheral blood mononuclear cells (PBMCs) for the new assay for lytic capacity described here and for an antibody EC50 assay for Fc-receptor recognition. For the lytic capacity assay, we employed flow cytometry to quantify the CD16A IgG Fc-receptor positive NK effector cells from PBMCs to avoid loss of NKs during isolation. Targets were 51Cr-labeled Daudi B cells pretreated with excess obinutuzumab type 2 anti-CD20 mAb and washed; remaining free mAb was insufficient to convert B cells in the PBMCs into 'targets'. We calculated: the percentage Daudis killed at a 1:1 ratio of CD16A-positive NK cells to Daudis (CX1:1); lytic slopes; and ADCC50 lytic units. Among 27 donors, we detected wide ranges in CX1:1 (16-73% targets killed) and in lytic slopes. Slope variations prevented application of lytic units. We recommend CX1:1 to compare individuals' ADCC capacity. CX1:1 was similar for purified NK cells vs. PBMCs and independent of CD16A V & F genotypes and antibody EC50s. With high mAb bound onto targets and the high affinity of obinutuzumab Fc for CD16A, CX1:1 measurements discern ADCC lytic capacity rather than antibody recognition. This assay allows ADCC to be quantified without NK cell isolation and avoids distortion associated with lytic units.
Collapse
Affiliation(s)
- Alexander P Sung
- University of Nevada Reno School of Medicine, Department of Microbiology and Immunology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Jennifer J-J Tang
- University of Nevada Reno School of Medicine, Department of Microbiology and Immunology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Michael J Guglielmo
- University of Nevada Reno School of Medicine, Department of Microbiology and Immunology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Doug Redelman
- University of Nevada Reno School of Medicine, Department of Physiology and Cell Biology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Julie Smith-Gagen
- University of Nevada Reno School of Medicine, School of Community Health Sciences Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Lucinda Bateman
- Bateman Horne Center, Salt Lake City, UT 84102, United States
| | - Dorothy Hudig
- University of Nevada Reno School of Medicine, Department of Microbiology and Immunology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States.
| |
Collapse
|
42
|
Paul P, Picard C, Sampol E, Lyonnet L, Di Cristofaro J, Paul-Delvaux L, Lano G, Nicolino-Brunet C, Ravis E, Collart F, Dignat-George F, Dussol B, Sabatier F, Mouly-Bandini A. Genetic and Functional Profiling of CD16-Dependent Natural Killer Activation Identifies Patients at Higher Risk of Cardiac Allograft Vasculopathy. Circulation 2017; 137:1049-1059. [PMID: 29097449 DOI: 10.1161/circulationaha.117.030435] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/19/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cardiac transplantation is an effective therapy for end-stage heart failure. Because cardiac allograft vasculopathy (CAV) is the major cause of late mortality after heart transplant (HT), there is a need to identify markers that reflect inflammatory or cytotoxic immune mechanisms contributing to its onset. Noninvasive and early stratification of patients at risk remains a challenge for adapting individualized therapy. The CD16 (Fc-gamma receptor 3A [FCGR3A]) receptor was recently identified as a major determinant of antibody-mediated natural killer (NK) cell activation in HT biopsies; however, little is known about the role of CD16 in promoting allograft vasculopathy. This study aimed to investigate whether markers that reflect CD16-dependent circulating NK cell activation may identify patients at higher risk of developing CAV after HT. METHODS Blood samples were collected from 103 patients undergoing routine coronarography angiography for CAV diagnosis (median 5 years since HT). Genomic and phenotypic analyses of FCGR3A/CD16 Fc-receptor profiles were compared in CAV-positive (n=52) and CAV-free patients (n=51). The levels of CD16 expression and rituximab-dependent cell cytotoxic activity of peripheral NK cells in HT recipients were evaluated using a noninvasive NK-cellular humoral activation test. RESULTS Enhanced levels of CD16 expression and antibody-dependent NK cell cytotoxic function of HT recipients were associated with the FCGR3A-VV genotype. The frequency of the FCGR3A-VV genotype was significantly higher in the CAV+ group (odds ratio, 3.9; P=0.0317) than in the CAV- group. The FCGR3A-VV genotype was identified as an independent marker correlated with the presence of CAV at the time of coronary angiography by using multivariate logistic regression models. The FCGR3A-VV genotype was also identified as a baseline-independent predictor of CAV risk (odds ratio, 4.7; P=0.023). CONCLUSIONS This study unravels a prominent role for the CD16-dependent NK cell activation pathway in the complex array of factors that favor the progression of transplant arteriosclerosis. It highlights the clinical potential of a noninvasive evaluation of FCGR3A/CD16 in the early stratification of CAV risk. The recognition of CD16 as a major checkpoint that controls immune surveillance may promote the design of individualized NK cell-targeted therapies to limit vascular damage in highly responsive sensitized patients. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT01569334.
Collapse
Affiliation(s)
- Pascale Paul
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.). .,INSERM, Aix-Marseille Université (AMU), VRCM, UMR-1076, France (P.P., F.D.G., F.S.)
| | - Christophe Picard
- Immunogenetic Laboratory, Établissement Français du Sang, Marseille, France (C.P.).,Aix Marseille Université, Etablissement Français du Sang, CNRS, UMR 7268 ADÉS, France (C.P., J.D.C.)
| | - Emmanuelle Sampol
- Assistance Publique-Hôpitaux Marseille (AP-HM), Pharmacokinetics Department, France (E.S.)
| | - Luc Lyonnet
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.)
| | - Julie Di Cristofaro
- Assistance Publique-Hôpitaux Marseille (AP-HM), Pharmacokinetics Department, France (E.S.)
| | - Louise Paul-Delvaux
- ENSAE-Paristech, Data Science & Statistics Department, Paris, France (L.P.-D.)
| | - Guillaume Lano
- Assistance Publique-Hôpitaux Marseille (AP-HM), Aix Marseille Univ, Center for Clinical Investigation, France (G.L., B.D.)
| | - Corinne Nicolino-Brunet
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.)
| | - Eleonore Ravis
- Assistance Publique Hopitaux de Marseille (AP-HM), Adult Cardiac Surgery Department, France (E.R., F.C., A.M.B.)
| | - Frederic Collart
- Assistance Publique Hopitaux de Marseille (AP-HM), Adult Cardiac Surgery Department, France (E.R., F.C., A.M.B.)
| | - Francoise Dignat-George
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.).,INSERM, Aix-Marseille Université (AMU), VRCM, UMR-1076, France (P.P., F.D.G., F.S.)
| | - Bertrand Dussol
- Assistance Publique-Hôpitaux Marseille (AP-HM), Aix Marseille Univ, Center for Clinical Investigation, France (G.L., B.D.)
| | - Florence Sabatier
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.).,INSERM, Aix-Marseille Université (AMU), VRCM, UMR-1076, France (P.P., F.D.G., F.S.)
| | - Annick Mouly-Bandini
- Assistance Publique Hopitaux de Marseille (AP-HM), Adult Cardiac Surgery Department, France (E.R., F.C., A.M.B.)
| |
Collapse
|
43
|
Pupuleku A, Costa-García M, Farré D, Hengel H, Angulo A, Muntasell A, López-Botet M. Elusive Role of the CD94/NKG2C NK Cell Receptor in the Response to Cytomegalovirus: Novel Experimental Observations in a Reporter Cell System. Front Immunol 2017; 8:1317. [PMID: 29114247 PMCID: PMC5660692 DOI: 10.3389/fimmu.2017.01317] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/29/2017] [Indexed: 11/13/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection promotes the differentiation and persistent expansion of a mature NK cell subset, which displays high surface levels of the activating CD94/NKG2C NK cell receptor, together with additional distinctive phenotypic and functional features. The mechanisms underlying the development of adaptive NK cells remain uncertain but some observations support the involvement of a cognate interaction of CD94/NKG2C with ligand(s) displayed by HCMV-infected cells. To approach this issue, the heterodimer and its adaptor (DAP12) were expressed in the human Jurkat leukemia T cell line; signaling was detected by transfection of a reporter plasmid encoding for Luciferase (Luc) under NFAT/AP1-dependent control. Engagement of the receptor by solid-phase bound CD94- or NKG2C-specific monoclonal antibodies (mAbs) triggered Luc expression. Moreover, reporter activation was detectable upon interaction with HLA-E+ 721.221 (.221-AEH) cells, as well as with 721.221 cells incubated with synthetic peptides, which stabilized surface expression of endogenous HLA-E; the response was specifically antagonized by soluble NKG2C- and HLA-E-specific mAbs. By contrast, activation of Jurkat-NKG2C+ was undetectable upon interaction with Human Fetal Foreskin Fibroblasts (HFFF) infected with HCMV laboratory strains (i.e., AD169, Towne), regardless of their differential ability to preserve surface HLA-E expression. On the other hand, infection with two clinical isolates or with the endotheliotropic TB40/E strain triggered Jurkat-NKG2C+ activation; yet, this response was not inhibited by blocking mAbs and was independent of CD94/NKG2C expression. The results are discussed in the framework of previous observations supporting the hypothetical existence of specific ligand(s) for CD94/NKG2C in HCMV-infected cells.
Collapse
Affiliation(s)
- Aldi Pupuleku
- Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Marcel Costa-García
- Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Domènec Farré
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Hartmut Hengel
- Institute of Virology, Albert Ludwigs University of Freiburg, Freiburg, Germany.,Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Ana Angulo
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Miguel López-Botet
- Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|
44
|
Immunoglobulin genotypes and cognitive functions in schizophrenia. Immunogenetics 2017; 70:67-72. [PMID: 28936707 DOI: 10.1007/s00251-017-1030-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/14/2017] [Indexed: 01/10/2023]
Abstract
Exposure to neurotropic viruses, such as herpes simplex virus type 1 and human cytomegalovirus, has been reported to be associated with cognitive impairment in schizophrenia. These viruses have evolved highly sophisticated strategies for decreasing the efficacy of the host immune response and interfering with viral clearance. Particular immunoglobulin GM (γ marker) genotypes modulate these viral immunoevasion strategies, influence antibody responsiveness to viral proteins, and are also associated with susceptibility to schizophrenia, providing an excellent rationale for determining their possible involvement in the cognitive functions in this highly heritable neurodevelopmental disorder. In this investigation, we assessed the cognitive functions (verbal memory, working memory, motor speed, verbal fluency, attention and processing speed, and executive function) in 145 patients with schizophrenia and characterized their DNA for several GM and KM (κ marker) alleles. Particular KM and GM genotypes were significantly associated with verbal memory and attention and processing speed scores, respectively (P = 0.01 and 0.001). Epistatic effects of GM and KM genotypes on attention and processing speed, verbal fluency, and motor speed were also noted (P = 0.031, 0.047, 0.003). These results, for the first time, show that hitherto understudied immunoglobulin GM and KM genotypes-individually and epistatically-contribute to the magnitude of interindividual variability in the cognitive functions in patients with schizophrenia. Additional studies involving these highly polymorphic genes of the immune system are needed.
Collapse
|
45
|
López-Montañés M, Alari-Pahissa E, Sintes J, Martínez-Rodríguez JE, Muntasell A, López-Botet M. Antibody-Dependent NK Cell Activation Differentially Targets EBV-Infected Cells in Lytic Cycle and Bystander B Lymphocytes Bound to Viral Antigen-Containing Particles. THE JOURNAL OF IMMUNOLOGY 2017. [PMID: 28630095 DOI: 10.4049/jimmunol.1601574] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
NK cells have been reported to respond against EBV-infected B cells in the lytic cycle and to control the viral infection involving IFN-γ secretion. Early reports proposed a role for NK cell Ab-dependent cellular cytotoxicity (ADCC) triggered via FcγR-IIIA (CD16) in the response to EBV. In the current study, we revisited this issue, showing that serum from EBV+ individuals triggered vigorous NK cell degranulation and cytokine production (i.e., TNF-α and IFN-γ) against EBV-infected cells, enhancing NK cell activation. The effect was preferentially directed against cells in the lytic phase and was associated with surface expression of the gp350/220 envelope Ag. In contrast, binding of gp350+ particles, released by EBV-infected cells, to B cell lines or autologous primary B lymphocytes also promoted specific Ab-dependent NK cell degranulation and TNF-α production but induced minimal IFN-γ secretion. In that case, target cell damage appeared marginal compared with the effect of a control anti-CD20 Ab (rituximab) at concentrations that triggered similar NK cell activation, indicating that cell-associated gp350+ particles may divert the cytolytic machinery, impairing its direct action on the plasma membrane. These observations support that Ab-dependent NK cell activation plays an important role in the control of EBV, enhancing NK cell effector functions against infected B cells in the lytic cycle. In contrast, the data reveal that gp350+ particles bound to bystander B cells trigger Ab-dependent NK cell degranulation and TNF-α but not cytotoxicity or IFN-γ production, potentially favoring the progression of viral infection.
Collapse
Affiliation(s)
| | | | - Jordi Sintes
- Hospital del Mar Medical Research Institute, Barcelona 08003, Spain; and
| | | | - Aura Muntasell
- Hospital del Mar Medical Research Institute, Barcelona 08003, Spain; and
| | - Miguel López-Botet
- University Pompeu Fabra, Barcelona 08003, Spain; .,Hospital del Mar Medical Research Institute, Barcelona 08003, Spain; and.,Department of Immunology, Hospital del Mar, Barcelona 08003, Spain
| |
Collapse
|
46
|
Rahe MC, Murtaugh MP. Effector mechanisms of humoral immunity to porcine reproductive and respiratory syndrome virus. Vet Immunol Immunopathol 2017; 186:15-18. [PMID: 28413045 DOI: 10.1016/j.vetimm.2017.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/03/2017] [Accepted: 02/13/2017] [Indexed: 11/28/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) continues to afflict swine nearly 30 years after it was first discovered as the causative agent of "mystery swine disease". Immunological tools of vaccination and exposure to virulent viruses have not succeeded in achieving control and prevention of PRRSV. Humoral immunity, mediated by antibodies, is a hallmark of anti-viral immunity, but little is known about the effector mechanisms of humoral immunity against PRRSV. It is essential to understand the immunological significance of antibody functions, including recently described broadly neutralizing antibodies and potential non-neutralizing activities, in the immune response to PRRSV. Here, we review recent research from PRRSV and other host-pathogen interactions to inform novel routes of exploration into PRRSV humoral immunity which may be important for identifying the immunological correlates of protection against PRRSV infection.
Collapse
Affiliation(s)
- Michael C Rahe
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1971 Commonwealth Avenue, St. Paul, MN, 55108 USA.
| | - Michael P Murtaugh
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1971 Commonwealth Avenue, St. Paul, MN, 55108 USA
| |
Collapse
|
47
|
López-Botet M, Vilches C, Redondo-Pachón D, Muntasell A, Pupuleku A, Yélamos J, Pascual J, Crespo M. Dual Role of Natural Killer Cells on Graft Rejection and Control of Cytomegalovirus Infection in Renal Transplantation. Front Immunol 2017; 8:166. [PMID: 28261220 PMCID: PMC5311043 DOI: 10.3389/fimmu.2017.00166] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/02/2017] [Indexed: 12/20/2022] Open
Abstract
Allograft rejection constitutes a major complication of solid organ transplantation requiring prophylactic/therapeutic immunosuppression, which increases susceptibility of patients to infections and cancer. Beyond the pivotal role of alloantigen-specific T cells and antibodies in the pathogenesis of rejection, natural killer (NK) cells may display alloreactive potential in case of mismatch between recipient inhibitory killer-cell immunoglobulin-like receptors (KIRs) and graft HLA class I molecules. Several studies have addressed the impact of this variable in kidney transplant with conflicting conclusions; yet, increasing evidence supports that alloantibody-mediated NK cell activation via FcγRIIIA (CD16) contributes to rejection. On the other hand, human cytomegalovirus (HCMV) infection constitutes a risk factor directly associated with the rate of graft loss and reduced host survival. The levels of HCMV-specific CD8+ T cells have been reported to predict the risk of posttransplant infection, and KIR-B haplotypes containing activating KIR genes have been related with protection. HCMV infection promotes to a variable extent an adaptive differentiation and expansion of a subset of mature NK cells, which display the CD94/NKG2C-activating receptor. Evidence supporting that adaptive NKG2C+ NK cells may contribute to control the viral infection in kidney transplant recipients has been recently obtained. The dual role of NK cells in the interrelation of HCMV infection with rejection deserves attention. Further phenotypic, functional, and genetic analyses of NK cells may provide additional insights on the pathogenesis of solid organ transplant complications, leading to the development of biomarkers with potential clinical value.
Collapse
Affiliation(s)
- Miguel López-Botet
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Immunology, Hospital del Mar, Barcelona, Spain; Univ. Pompeu Fabra, Barcelona, Spain
| | - Carlos Vilches
- Immunogenetics-Histocompatibility, Instituto de Investigación Sanitaria Puerta de Hierro , Majadahonda , Spain
| | - Dolores Redondo-Pachón
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM) , Barcelona , Spain
| | | | - José Yélamos
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Immunology, Hospital del Mar, Barcelona, Spain
| | - Julio Pascual
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Marta Crespo
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Nephrology, Hospital del Mar, Barcelona, Spain
| |
Collapse
|
48
|
Abstract
Natural killer (NK) cells play a critical role in viral immunity. In the setting of HIV infection, epidemiologic and functional evidence support a role for NK cells in both protection from new infection and in viral control. Specifically, NK cells directly mediate immune pressure leading to virus evolution, and NK cell receptor genotypic profiles, clonal repertoires, and functional capacity have all been implicated in virus containment. In addition, indirect NK cell-mediated antibody-dependent cellular cytotoxicity has been linked to vaccine-induced protective immunity against HIV infection. With recent advances in our understanding of NK cell deficiency, development, memory-like responses, and editing of the adaptive immune system, the opportunities to direct and exploit NK cell antiviral immunity to target HIV have exponentially grown. In this review, we seek to highlight the intersections between discoveries in basic NK cell biology and the challenges of HIV chronic infection, vaccine development, and cure/eradication strategies.
Collapse
Affiliation(s)
- Eileen Scully
- />Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, 400 Technology Square, Cambridge, MA 02139 USA
- />Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02130 USA
| | - Galit Alter
- />Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, 400 Technology Square, Cambridge, MA 02139 USA
| |
Collapse
|
49
|
Stanfield BA, Pahar B, Chouljenko VN, Veazey R, Kousoulas KG. Vaccination of rhesus macaques with the live-attenuated HSV-1 vaccine VC2 stimulates the proliferation of mucosal T cells and germinal center responses resulting in sustained production of highly neutralizing antibodies. Vaccine 2016; 35:536-543. [PMID: 28017425 DOI: 10.1016/j.vaccine.2016.12.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/28/2016] [Accepted: 12/12/2016] [Indexed: 02/08/2023]
Abstract
We have shown that the live-attenuated HSV-1 VC2 vaccine strain with mutations in glycoprotein K (gK) and the membrane protein UL20 is unable to establish latency in vaccinated animals and produces a robust immune response capable of completely protecting mice against lethal vaginal HSV-1 or HSV-2 infections. To better understand the immune response generated by vaccination with VC2, we tested its ability to elicit immune responses in rhesus macaques. Vaccinated animals showed no signs of disease and developed increasing HSV-1 and HSV-2 reactive IgG1 after two booster vaccinations, while IgG subtypes IgG2 and IgG3 remained at low to undetectable levels. All vaccinated animals produced high levels of cross protective neutralizing antibodies. Flow cytometry analysis of cells isolated from draining lymph nodes showed that VC2 vaccination stimulated significant increases in plasmablast (CD27highCD38high) and mature memory (CD21-IgM-) B cells. T cell analysis on cells isolated from draining lymph node biopsies demonstrated a statistically significant increase in proliferating (Ki67+) follicular T helper cells and regulatory CXCR5+ CD8+ cytotoxic T cells. Analysis of plasma isolated two weeks post vaccination showed significant increases in circulating CXCL13 indicating increased germinal center activity. Cells isolated from vaginal biopsy samples collected over the course of the study exhibited vaccination-dependent increases in proliferating (Ki67+) CD4+ and CD8+ T cell populations. These results suggest that intramuscular vaccination with the live-attenuated HSV-1 VC2 vaccine strain can stimulate robust IgG1 antibody responses that persist for >250days post vaccination. In addition, vaccination lead to the maturation of B cells into plasmablast and mature memory B cells, the expansion of follicular T helper cells, and affects in the mucosal immune responses. These data suggest that the HSV VC2 vaccine induces potent immune responses that could help define correlates of protection towards developing an efficacious HSV-1/HSV-2 vaccine in humans.
Collapse
Affiliation(s)
- Brent A Stanfield
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Bapi Pahar
- Department of Comparative Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA 70433, United States
| | - Vladimir N Chouljenko
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Ronald Veazey
- Department of Comparative Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA 70433, United States
| | - Konstantin G Kousoulas
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States.
| |
Collapse
|
50
|
Phadke VK, Friedman-Moraco RJ, Quigley BC, Farris AB, Norvell JP. Concomitant herpes simplex virus colitis and hepatitis in a man with ulcerative colitis: Case report and review of the literature. Medicine (Baltimore) 2016; 95:e5082. [PMID: 27759636 PMCID: PMC5079320 DOI: 10.1097/md.0000000000005082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Herpesvirus infections often complicate the clinical course of patients with inflammatory bowel disease; however, invasive disease due to herpes simplex virus is distinctly uncommon. METHODS We present a case of herpes simplex virus colitis and hepatitis, review all the previously published cases of herpes simplex virus colitis, and discuss common clinical features and outcomes. We also discuss the epidemiology, clinical manifestations, diagnosis, and management of herpes simplex virus infections, focusing specifically on patients with inflammatory bowel disease. RESULTS A 43-year-old man with ulcerative colitis, previously controlled with an oral 5-aminosalicylic agent, developed symptoms of a colitis flare that did not respond to treatment with systemic corticosteroid therapy. One week later he developed orolabial ulcers and progressive hepatic dysfunction, with markedly elevated transaminases and coagulopathy. He underwent emergent total colectomy when imaging suggested bowel micro-perforation. Pathology from both the colon and liver was consistent with herpes simplex virus infection, and a viral culture of his orolabial lesions and a serum polymerase chain reaction assay also identified herpes simplex virus. He was treated with systemic antiviral therapy and made a complete recovery. CONCLUSIONS Disseminated herpes simplex virus infection with concomitant involvement of the colon and liver has been reported only 3 times in the published literature, and to our knowledge this is the first such case in a patient with inflammatory bowel disease. The risk of invasive herpes simplex virus infections increases with some, but not all immunomodulatory therapies. Optimal management of herpes simplex virus in patients with inflammatory bowel disease includes targeted prophylactic therapy for patients with evidence of latent infection, and timely initiation of antiviral therapy for those patients suspected to have invasive disease.
Collapse
MESH Headings
- Adult
- Colitis/complications
- Colitis/diagnosis
- Colitis/virology
- Colitis, Ulcerative/complications
- Colitis, Ulcerative/diagnosis
- Colonoscopy
- DNA, Viral/analysis
- Hepatitis, Viral, Human/complications
- Hepatitis, Viral, Human/diagnosis
- Hepatitis, Viral, Human/virology
- Herpes Simplex/complications
- Herpes Simplex/diagnosis
- Herpes Simplex/virology
- Humans
- Male
- Simplexvirus/genetics
- Tomography, X-Ray Computed
Collapse
Affiliation(s)
- Varun K. Phadke
- Division of Infectious Diseases, Emory University School of Medicine
- Correspondence: Varun K. Phadke, 49 Jesse Hill Jr. Drive, Atlanta 30303, GA (e-mail: )
| | | | - Brian C. Quigley
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Emory University Hospital
| | - Alton B. Farris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Emory University Hospital
| | - J. P. Norvell
- Division of Digestive Diseases, Emory University School of Medicine
- Emory Transplant Center, Atlanta, GA
| |
Collapse
|