1
|
Otero CE, Petkova S, Ebermann M, Taher H, John N, Hoffmann K, Davalos A, Moström MJ, Gilbride RM, Papen CR, Barber-Axthelm A, Scheef EA, Barfield R, Sprehe LM, Kendall S, Manuel TD, Beechwood T, Nguyen LK, Vande Burgt NH, Chan C, Denton M, Streblow ZJ, Streblow DN, Tarantal AF, Hansen SG, Kaur A, Permar S, Früh K, Hengel H, Malouli D, Kolb P. Rhesus Cytomegalovirus-encoded Fcγ-binding glycoproteins facilitate viral evasion from IgG-mediated humoral immunity. Nat Commun 2025; 16:1200. [PMID: 39885150 PMCID: PMC11782611 DOI: 10.1038/s41467-025-56419-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Human cytomegalovirus (HCMV) encodes four viral Fc-gamma receptors (vFcγRs) that counteract antibody-mediated activation in vitro, but their role in infection and pathogenesis is unknown. To examine their in vivo function in an animal model evolutionarily closely related to humans, we identified and characterized Rh05, Rh152/151 and Rh173 as the complete set of vFcγRs encoded by rhesus CMV (RhCMV). Each one of these proteins displays functional similarities to their prospective HCMV orthologs with respect to antagonizing host FcγR activation in vitro. When RhCMV-naïve male rhesus macaques were infected with vFcγR-deleted RhCMV, peak plasma DNAemia levels and anti-RhCMV antibody responses were comparable to wildtype infections of both male and female animals. However, the duration of plasma DNAemia was significantly shortened in immunocompetent, but not in CD4 + T cell-depleted animals. Since vFcγRs were not required for superinfection of rhesus macaques, we conclude that these proteins can prolong lytic replication during primary infection by evading virus-specific adaptive immune responses, particularly antibodies.
Collapse
Affiliation(s)
- Claire E Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Sophia Petkova
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Ebermann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Nessy John
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Katja Hoffmann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angel Davalos
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Matilda J Moström
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Courtney R Papen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Elizabeth A Scheef
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Lesli M Sprehe
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Savannah Kendall
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Tabitha D Manuel
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Teresa Beechwood
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Linh Khanh Nguyen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Nathan H Vande Burgt
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Zachary J Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Alice F Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, CA, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA.
| | - Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
2
|
Schleiss MR, Crooks CM, Karthigeyan KP, Kruc RM, Otero CE, Wang HY(S, Permar SR, Plotkin SA, Gautam R. Proceedings of the Conference "CMV Vaccine Development-How Close Are We?" (27-28 September 2023). Vaccines (Basel) 2024; 12:1231. [PMID: 39591134 PMCID: PMC11598149 DOI: 10.3390/vaccines12111231] [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: 07/23/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 11/28/2024] Open
Abstract
Congenital cytomegalovirus (cCMV) is the most common infectious cause of disability in children, including sensorineural hearing loss. There is interest in developing a pre-conception vaccine that could confer protective immunity on a woman of child-bearing age, hence resulting in a reduced cCMV disease burden. Other populations, including solid organ transplant (SOT) and hematopoietic stem cell transplant (HSCT) patients, could also benefit from CMV vaccination. To review and discuss vaccines that are in clinical development, a workshop, sponsored by the National Institutes of Health (NIH) and the National Institute of Allergy and Infectious Diseases (NIAID), was empaneled. At this workshop, correlates of protective immunity against CMV, epidemiologic features of CMV transmission, and vaccine platforms in development were reviewed. Representatives from academia, pharma, and the NIH engaged in discussion on the current state-of-the-art in CMV vaccinology. A summary of the presentations from this is provided in this report.
Collapse
Affiliation(s)
- Mark R. Schleiss
- Division of Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | - Chelsea M. Crooks
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Krithika P. Karthigeyan
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Rebecca M. Kruc
- Department of Pediatrics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA;
| | - Claire E. Otero
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Hsuan-Yuan (Sherry) Wang
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, 1300 York Ave Box 65, New York, NY 10065, USA;
| | - Stanley A. Plotkin
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Rajeev Gautam
- Program Officer at Virology Branch, Division of Microbiology and Infectious Diseases, NIAID, NIH, 5601 Fisher’s Lane, Rockville, MD 20892, USA;
| |
Collapse
|
3
|
Andreeva I, Kolb P, Rodon L, Blank N, Lorenz HM, Merkt W. Fcγ-receptor-IIIA bioactivity of circulating and synovial immune complexes in rheumatoid arthritis. RMD Open 2024; 10:e004190. [PMID: 39209371 PMCID: PMC11367361 DOI: 10.1136/rmdopen-2024-004190] [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: 02/04/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Previous technical limitations prevented the proof of Fcγ-receptor (FcγR)-activation by soluble immune complexes (sICs) in patients. FcγRIIIa (CD16) is a risk factor in rheumatoid arthritis (RA). We aimed at determining the presence of CD16-activating sICs in RA and control diseases. METHODS Sera from an exploratory cohort (n=50 patients with RA) and a validation cohort (n=106 patients with RA, 20 patients with psoriasis arthritis (PsA), 22 patients with systemic lupus erythematosus (SLE) and 31 healthy controls) were analysed using a new reporter cell assay. Additionally, 26 synovial fluid samples were analysed, including paired serum/synovial samples. RESULTS For the first time using a reliable and sensitive functional assay, the presence of sICs in RA sera was confirmed. sICs possess an intrinsic capacity to activate CD16 and can be found in both synovial fluid and in blood. In low experimental dilutions, circulating sICs were also detected in a subset of healthy people and in PsA. However, we report a significantly increased frequency of bioactive circulating sICs in RA. While the bioactivity of circulating sICs was low and did not correlate with clinical parameters, synovial sICs were highly bioactive and correlated with serum autoantibody levels. Receiver operator curves indicated that sICs bioactivity in synovial fluid could be used to discriminate immune complex-associated arthritis from non-associated forms. Finally, circulating sICs were more frequently found in SLE than in RA. The degree of CD16 bioactivity showed strong donor-dependent differences, especially in SLE. CONCLUSIONS RA is characterised by the presence of circulating and synovial sICs that can engage and activate CD16.
Collapse
Affiliation(s)
- Ivana Andreeva
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp Kolb
- Institute of Virology, Medical Center—University of Freiburg, Freiburg, Germany
- Albert-Ludwigs-Universitat Freiburg Medizinische Fakultat, Freiburg, Germany
| | - Lea Rodon
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Norbert Blank
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Merkt
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
- Hiller Forschungszentrum, Department of Rheumatology, University Hospital of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
4
|
Otero CE, Petkova S, Ebermann M, Taher H, John N, Hoffmann K, Davalos A, Moström MJ, Gilbride RM, Papen CR, Barber-Axthelm A, Scheef EA, Barfield R, Sprehe LM, Kendall S, Manuel TD, Vande Burgt NH, Chan C, Denton M, Streblow ZJ, Streblow DN, Hansen SG, Kaur A, Permar S, Früh K, Hengel H, Malouli D, Kolb P. Rhesus Cytomegalovirus-encoded Fcγ-binding glycoproteins facilitate viral evasion from IgG-mediated humoral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582371. [PMID: 38464092 PMCID: PMC10925275 DOI: 10.1101/2024.02.27.582371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Human cytomegalovirus (HCMV) encodes four viral Fc-gamma receptors (vFcγRs) that counteract antibody-mediated activation in vitro , but their role in infection and pathogenesis is unknown. To examine the in vivo function of vFcγRs in animal hosts closely related to humans, we identified and characterized vFcγRs encoded by rhesus CMV (RhCMV). We demonstrate that Rh05, Rh152/151 and Rh173 represent the complete set of RhCMV vFcγRs, each displaying functional similarities to their respective HCMV orthologs with respect to antagonizing host FcγR activation in vitro . When RhCMV-naïve rhesus macaques were infected with vFcγR-deleted RhCMV, peak plasma viremia levels and anti-RhCMV antibody responses were comparable to wildtype infections. However, the duration of plasma viremia was significantly shortened in immunocompetent, but not in CD4+ T cell-depleted animals. Since vFcγRs were not required for superinfection, we conclude that vFcγRs delay control by virus-specific adaptive immune responses, particularly antibodies, during primary infection.
Collapse
|
5
|
Semmes EC, Miller IG, Rodgers N, Phan CT, Hurst JH, Walsh KM, Stanton RJ, Pollara J, Permar SR. ADCC-activating antibodies correlate with decreased risk of congenital human cytomegalovirus transmission. JCI Insight 2023; 8:e167768. [PMID: 37427588 PMCID: PMC10371338 DOI: 10.1172/jci.insight.167768] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/23/2023] [Indexed: 07/11/2023] Open
Abstract
Human cytomegalovirus (HCMV) is the most common vertically transmitted infection worldwide, yet there are no vaccines or therapeutics to prevent congenital HCMV (cCMV) infection. Emerging evidence indicates that antibody Fc effector functions may be a previously underappreciated component of maternal immunity against HCMV. We recently reported that antibody-dependent cellular phagocytosis (ADCP) and IgG activation of FcγRI/FcγRII were associated with protection against cCMV transmission, leading us to hypothesize that additional Fc-mediated antibody functions may be important. In this same cohort of HCMV-transmitting (n = 41) and nontransmitting (n = 40) mother-infant dyads, we report that higher maternal sera antibody-dependent cellular cytotoxicity (ADCC) activation is also associated with lower risk of cCMV transmission. We investigated the relationship between ADCC and IgG responses against 9 viral antigens and found that ADCC activation correlated most strongly with sera IgG binding to the HCMV immunoevasin protein UL16. Moreover, we determined that higher UL16-specific IgG binding and FcγRIII/CD16 engagement were associated with the greatest risk reduction in cCMV transmission. Our findings indicate that ADCC-activating antibodies against targets such as UL16 may represent an important protective maternal immune response against cCMV infection that can guide future HCMV correlates studies and vaccine or antibody-based therapeutic development.
Collapse
Affiliation(s)
- Eleanor C. Semmes
- Medical Scientist Training Program, Department of Molecular Genetics and Microbiology, and
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Itzayana G. Miller
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Nicole Rodgers
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Caroline T. Phan
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jillian H. Hurst
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Kyle M. Walsh
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Richard J. Stanton
- Division of Infection and Immunology, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| |
Collapse
|
6
|
Cruz Amaya J, Walcheck B, Smith-Gagen J, Lombardi VC, Hudig D. Detection of Antibody-Dependent Cell-Mediated Cytotoxicity-Supporting Antibodies by NK-92-CD16A Cell Externalization of CD107a: Recognition of Antibody Afucosylation and Assay Optimization. Antibodies (Basel) 2023; 12:44. [PMID: 37489366 PMCID: PMC10366760 DOI: 10.3390/antib12030044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023] Open
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) by natural killer (NK) lymphocytes eliminates cells infected with viruses. Anti-viral ADCC requires three components: (1) antibody; (2) effector lymphocytes with the Fc-IgG receptor CD16A; and (3) viral proteins in infected cell membranes. Fc-afucosylated antibodies bind with greater affinity to CD16A than fucosylated antibodies; individuals' variation in afucosylation contributes to differences in ADCC. Current assays for afucosylated antibodies involve expensive methods. We report an improved bioassay for antibodies that supports ADCC, which encompasses afucosylation. This assay utilizes the externalization of CD107a by NK-92-CD16A cells after antibody recognition. We used anti-CD20 monoclonal antibodies, GA101 WT or glycoengineered (GE), 10% or ~50% afucosylated, and CD20-positive Raji target cells. CD107a increased detection 7-fold compared to flow cytometry to detect Raji-bound antibodies. WT and GE antibody effective concentrations (EC50s) for CD107a externalization differed by 20-fold, with afucosylated GA101-GE more detectable. The EC50s for CD107a externalization vs. 51Cr cell death were similar for NK-92-CD16A and blood NK cells. Notably, the % CD107a-positive cells were negatively correlated with dead Raji cells and were nearly undetectable at high NK:Raji ratios required for cytotoxicity. This bioassay is very sensitive and adaptable to assess anti-viral antibodies but unsuitable as a surrogate assay to monitor cell death after ADCC.
Collapse
Affiliation(s)
- Judith Cruz Amaya
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Bruce Walcheck
- Department of Veterinary and Biological Sciences, Center for Immunology and Masonic Cancer Center, University of Minnesota, 295J AS/VM Building, 1988 Fitch Avenue, Saint Paul, MN 55108, USA
| | - Julie Smith-Gagen
- School of Community Health Sciences, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Vincent C. Lombardi
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Dorothy Hudig
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| |
Collapse
|
7
|
Rosenberg YJ, Ordonez T, Khanwalkar US, Barnette P, Pandey S, Backes IM, Otero CE, Goldberg BS, Crowley AR, Leib DA, Shapiro MB, Jiang X, Urban LA, Lees J, Hessell AJ, Permar S, Haigwood NL, Ackerman ME. Evidence for the Role of a Second Fc-Binding Receptor in Placental IgG Transfer in Nonhuman Primates. mBio 2023; 14:e0034123. [PMID: 36946726 PMCID: PMC10127586 DOI: 10.1128/mbio.00341-23] [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: 02/09/2023] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
Transplacental transfer of maternal antibodies provides the fetus and newborn with passive protection against infectious diseases. While the role of the highly conserved neonatal Fc receptor (FcRn) in transfer of IgG in mammals is undisputed, recent reports have suggested that a second receptor may contribute to transport in humans. We report poor transfer efficiency of plant-expressed recombinant HIV-specific antibodies, including engineered variants with high FcRn affinity, following subcutaneous infusion into rhesus macaques close to parturition. Unexpectedly, unlike those derived from mammalian tissue culture, plant-derived antibodies were essentially unable to cross macaque placentas. This defect was associated with poor Fcγ receptor binding and altered Fc glycans and was not recapitulated in mice. These results suggest that maternal-fetal transfer of IgG across the three-layer primate placenta may require a second receptor and suggest a means of providing maternal antibody treatments during pregnancy while avoiding fetal harm. IMPORTANCE This study compared the ability of several human HIV envelope-directed monoclonal antibodies produced in plants with the same antibodies produced in mammalian cells for their ability to cross monkey and mouse placentas. We found that the two types of antibodies have comparable transfer efficiencies in mice, but they are differentially transferred across macaque placentas, consistent with a two-receptor IgG transport model in primates. Importantly, plant-produced monoclonal antibodies have excellent binding characteristics for human FcRn receptors, permitting desirable pharmacokinetics in humans. The lack of efficient transfer across the primate placenta suggests that therapeutic plant-based antibody treatments against autoimmune diseases and cancer could be provided to the mother while avoiding transfer and preventing harm to the fetus.
Collapse
Affiliation(s)
| | - Tracy Ordonez
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Iara M. Backes
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Claire E. Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | | | - Andrew R. Crowley
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - David A. Leib
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mariya B. Shapiro
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | | | | | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
8
|
Semmes EC, Miller IG, Rodgers N, Phan CT, Hurst JH, Walsh KM, Stanton RJ, Pollara J, Permar SR. ADCC-activating antibodies correlate with protection against congenital human cytomegalovirus infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.15.23287332. [PMID: 36993668 PMCID: PMC10055595 DOI: 10.1101/2023.03.15.23287332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Human cytomegalovirus (HCMV) is the most common vertically transmitted infection worldwide, yet there are no licensed vaccines or therapeutics to prevent congenital HCMV (cCMV) infection. Emerging evidence from studies of natural infection and HCMV vaccine trials indicates that antibody Fc effector functions may defend against HCMV infection. We previously reported that antibody-dependent cellular phagocytosis (ADCP) and IgG activation of FcγRI/FcγRII were associated with reduced risk of cCMV transmission, leading us to hypothesize that other Fc-mediated antibody functions may also contribute to protection. In this same cohort of HCMV transmitting (n = 41) and non-transmitting (n = 40) mother-infant dyads, we found that higher maternal sera antibody-dependent cellular cytotoxicity (ADCC) activation was also associated with decreased risk of cCMV infection. We determined that NK cell-mediated ADCC responses correlated strongly with anti-HCMV IgG FcγRIII/CD16 activation and IgG binding to the HCMV immunoevasin protein UL16. Notably, anti-UL16 IgG binding and engagement of FcγRIII/CD16 were higher in non-transmitting versus transmitting dyads and interacted significantly with ADCC responses. These findings indicate that ADCC-activating antibodies against novel targets such as UL16 may represent an important protective maternal immune response against cCMV infection, which can guide future HCMV correlates studies and vaccine development.
Collapse
|
9
|
Le-Trilling VTK, Jagnjić A, Brizić I, Eilbrecht M, Wohlgemuth K, Rožmanić C, Herdman A, Hoffmann K, Westendorf AM, Hengel H, Jonjić S, Trilling M. Maternal antibodies induced by a live attenuated vaccine protect neonatal mice from cytomegalovirus. NPJ Vaccines 2023; 8:8. [PMID: 36737485 PMCID: PMC9898546 DOI: 10.1038/s41541-023-00602-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Human cytomegalovirus (HCMV) frequently causes congenital infections, resulting in birth defects and developmental disorders. A vaccine is needed, but unavailable. We analyzed the potential of CMV mutants, lacking their STAT2 antagonists to serve as live attenuated vaccine viruses in mice. Infections with attenuated viruses elicited strong ELISA-reactive binding IgG responses and induced neutralizing antibodies as well as antibodies stimulating cellular Fcγ receptors, including the antibody-dependent cellular cytotoxicity (ADCC)-eliciting receptors FcγRIII/CD16 and FcγRIV. Accordingly, vaccinated mice were fully protected against challenge infections. Female mice vaccinated prior to gestation transmitted CMV-specific IgG to their offspring, which protected the progeny from perinatal infections in a mouse model for congenital CMV disease. To define the role of maternal antibodies, female mice either capable or incapable of producing antibodies were vaccinated and subsequently bred to males of the opposite genotype. Challenge infections of the genotypically identical F1 generation revealed the indispensability of maternal antibodies for vaccine-induced protection against cytomegaloviruses.
Collapse
Affiliation(s)
- Vu Thuy Khanh Le-Trilling
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andreja Jagnjić
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ilija Brizić
- grid.22939.330000 0001 2236 1630Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mareike Eilbrecht
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kerstin Wohlgemuth
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carmen Rožmanić
- grid.22939.330000 0001 2236 1630Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Alan Herdman
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Katja Hoffmann
- grid.5963.9Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Astrid M. Westendorf
- grid.5718.b0000 0001 2187 5445Institute for Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hartmut Hengel
- grid.5963.9Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stipan Jonjić
- grid.22939.330000 0001 2236 1630Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mirko Trilling
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
10
|
Irrgang P, Gerling J, Kocher K, Lapuente D, Steininger P, Habenicht K, Wytopil M, Beileke S, Schäfer S, Zhong J, Ssebyatika G, Krey T, Falcone V, Schülein C, Peter AS, Nganou-Makamdop K, Hengel H, Held J, Bogdan C, Überla K, Schober K, Winkler TH, Tenbusch M. Class switch toward noninflammatory, spike-specific IgG4 antibodies after repeated SARS-CoV-2 mRNA vaccination. Sci Immunol 2023; 8:eade2798. [PMID: 36548397 PMCID: PMC9847566 DOI: 10.1126/sciimmunol.ade2798] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RNA vaccines are efficient preventive measures to combat the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. High levels of neutralizing SARS-CoV-2 antibodies are an important component of vaccine-induced immunity. Shortly after the initial two mRNA vaccine doses, the immunoglobulin G (IgG) response mainly consists of the proinflammatory subclasses IgG1 and IgG3. Here, we report that several months after the second vaccination, SARS-CoV-2-specific antibodies were increasingly composed of noninflammatory IgG4, which were further boosted by a third mRNA vaccination and/or SARS-CoV-2 variant breakthrough infections. IgG4 antibodies among all spike-specific IgG antibodies rose, on average, from 0.04% shortly after the second vaccination to 19.27% late after the third vaccination. This induction of IgG4 antibodies was not observed after homologous or heterologous SARS-CoV-2 vaccination with adenoviral vectors. Single-cell sequencing and flow cytometry revealed substantial frequencies of IgG4-switched B cells within the spike-binding memory B cell population [median of 14.4%; interquartile range (IQR) of 6.7 to 18.1%] compared with the overall memory B cell repertoire (median of 1.3%; IQR of 0.9 to 2.2%) after three immunizations. This class switch was associated with a reduced capacity of the spike-specific antibodies to mediate antibody-dependent cellular phagocytosis and complement deposition. Because Fc-mediated effector functions are critical for antiviral immunity, these findings may have consequences for the choice and timing of vaccination regimens using mRNA vaccines, including future booster immunizations against SARS-CoV-2.
Collapse
Affiliation(s)
- Pascal Irrgang
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Juliane Gerling
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany
| | - Katharina Kocher
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Dennis Lapuente
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Philipp Steininger
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Katharina Habenicht
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany
| | - Monika Wytopil
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Stephanie Beileke
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Simon Schäfer
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany
| | - Jahn Zhong
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany
| | - George Ssebyatika
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Luebeck; Luebeck, Germany
| | - Thomas Krey
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Luebeck; Luebeck, Germany
| | - Valeria Falcone
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg; Freiburg, Germany
| | - Christine Schülein
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Antonia Sophia Peter
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Krystelle Nganou-Makamdop
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany
| | - Hartmut Hengel
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg; Freiburg, Germany
| | - Jürgen Held
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany
| | - Klaus Überla
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany.,Corresponding author. (K.S.); (T.H.W.); (M.T.)
| | - Thomas H. Winkler
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany.,Corresponding author. (K.S.); (T.H.W.); (M.T.)
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany.,Corresponding author. (K.S.); (T.H.W.); (M.T.)
| |
Collapse
|
11
|
Krausz M, Mitsuiki N, Falcone V, Komp J, Posadas-Cantera S, Lorenz HM, Litzman J, Wolff D, Kanariou M, Heinkele A, Speckmann C, Häcker G, Hengel H, Gámez-Díaz L, Grimbacher B. Do common infections trigger disease-onset or -severity in CTLA-4 insufficiency? Front Immunol 2022; 13:1011646. [PMID: 36405723 PMCID: PMC9667032 DOI: 10.3389/fimmu.2022.1011646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/17/2022] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Heterozygous mutations in CTLA4 lead to an inborn error of immunity characterized by immune dysregulation and immunodeficiency, known as CTLA-4 insufficiency. Cohort studies on CTLA4 mutation carriers showed a reduced penetrance (around 70%) and variable disease expressivity, suggesting the presence of modifying factors. It is well studied that infections can trigger autoimmunity in humans, especially in combination with a genetic predisposition. METHODS To investigate whether specific infections or the presence of specific persisting pathogens are associated with disease onset or severity in CTLA-4 insufficiency, we have examined the humoral immune response in 13 CTLA4 mutation carriers, seven without clinical manifestation and six with autoimmune manifestations, but without immunoglobulin replacement therapy against cytomegalovirus (CMV), Epstein-Barr virus (EBV), herpes simplex virus 1/2 (HSV 1/2), parvovirus B19 and Toxoplasma gondii. Additionally, we have measured FcγRIII/CD16A activation by EBV-specific IgG antibodies to examine the functional capabilities of immunoglobulins produced by CTLA4 mutation carriers. RESULTS The seroprevalence between affected and unaffected CTLA4 mutation carriers did not differ significantly for the examined pathogens. Additionally, we show here that CTLA4 mutation carriers produce EBV-specific IgG, which are unimpaired in activating FcγRIII/CD16A. CONCLUSIONS Our results show that the investigated pathogens are very unlikely to trigger the disease onset in CTLA-4-insufficient individuals, and their prevalence is not correlated with disease severity or expressivity.
Collapse
Affiliation(s)
- Máté Krausz
- Institute for Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Noriko Mitsuiki
- Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Valeria Falcone
- Institute of Virology, University Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Johanna Komp
- Institute of Medical Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sara Posadas-Cantera
- Institute for Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Institute of Medical Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Jiri Litzman
- Department of Clinical Immunology and Allergology, St. Anne’s University Hospital in Brno and Medical Faculty, Masaryk University, Brno, Czechia
| | - Daniel Wolff
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Maria Kanariou
- Department of Immunology and Histocompatibility, Centre for Primary Immunodeficiencies, “Aghia Sophia” Children’s Hospital, Athens, Greece
| | - Anita Heinkele
- Center for Pediatric Rheumatology, Olgahospital, Stuttgart, Germany
| | - Carsten Speckmann
- Institute for Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Department of Pediatric Hematology and Oncology, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Hengel
- Institute of Virology, University Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura Gámez-Díaz
- Institute for Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,DZIF – German Center for Infection Research, Satellite Center Freiburg, Freiburg, Germany,CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany,RESIST – Cluster of Excellence 2155 to Hannover Medical School, Satellite Center Freiburg, Freiburg, Germany,*Correspondence: Bodo Grimbacher,
| |
Collapse
|
12
|
Ankerhold J, Giese S, Kolb P, Maul-Pavicic A, Voll RE, Göppert N, Ciminski K, Kreutz C, Lother A, Salzer U, Bildl W, Welsink T, Morgenthaler NG, Grawitz AB, Emmerich F, Steinmann D, Huzly D, Schwemmle M, Hengel H, Falcone V. Circulating multimeric immune complexes contribute to immunopathology in COVID-19. Nat Commun 2022; 13:5654. [PMID: 36163132 PMCID: PMC9513013 DOI: 10.1038/s41467-022-32867-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 08/22/2022] [Indexed: 01/08/2023] Open
Abstract
A dysregulated immune response with high levels of SARS-CoV-2 specific IgG antibodies characterizes patients with severe or critical COVID-19. Although a robust IgG response is considered to be protective, excessive triggering of activating Fc-gamma-receptors (FcγRs) could be detrimental and cause immunopathology. Here, we document excessive FcγRIIIA/CD16A activation in patients developing severe or critical COVID-19 but not in those with mild disease. We identify two independent ligands mediating extreme FcγRIIIA/CD16A activation. Soluble circulating IgG immune complexes (sICs) are detected in about 80% of patients with severe and critical COVID-19 at levels comparable to active systemic lupus erythematosus (SLE) disease. FcγRIIIA/CD16A activation is further enhanced by afucosylation of SARS-CoV-2 specific IgG. Utilizing cell-based reporter systems we provide evidence that sICs can be formed prior to a specific humoral response against SARS-CoV-2. Our data suggest a cycle of immunopathology driven by an early formation of sICs in predisposed patients. These findings suggest a reason for the seemingly paradoxical findings of high antiviral IgG responses and systemic immune dysregulation in severe COVID-19. The involvement of circulating sICs in the promotion of immunopathology in predisposed patients opens new possibilities for intervention strategies to mitigate critical COVID-19 progression. During viral infections high levels of antibodies can form soluble immune complexes (sICs) with antigen and trigger Fcγ receptors (FcγR) leading to increased immunopathology. Here the authors measure FcγRs activation by sICs and consider how these may lead to excessive immunopathology during severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jakob Ankerhold
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Sebastian Giese
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Philipp Kolb
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Andrea Maul-Pavicic
- Department of Rheumatology and Clinical Immunology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Nathalie Göppert
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Kevin Ciminski
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Clemens Kreutz
- Institute of Medical Biometry and Statistics, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Achim Lother
- Department of Cardiology and Angiology I, University Heart Center, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Interdisciplinary Medical Intensive Care, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Ulrich Salzer
- Department of Rheumatology and Clinical Immunology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Wolfgang Bildl
- Institute of Physiology II, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Tim Welsink
- InVivo BioTech Services GmbH, Hennigsdorf, Germany
| | | | - Andrea Busse Grawitz
- Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Florian Emmerich
- Institute for Transfusion Medicine and Gene Therapy, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel Steinmann
- Occupational Medical Service, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Daniela Huzly
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Martin Schwemmle
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Hartmut Hengel
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.
| | - Valeria Falcone
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.
| |
Collapse
|
13
|
Chen JL, Fries CN, Berendam SJ, Rodgers NS, Roe EF, Wu Y, Li SH, Jain R, Watts B, Eudailey J, Barfield R, Chan C, Moody MA, Saunders KO, Pollara J, Permar SR, Collier JH, Fouda GG. Self-assembling peptide nanofiber HIV vaccine elicits robust vaccine-induced antibody functions and modulates Fc glycosylation. SCIENCE ADVANCES 2022; 8:eabq0273. [PMID: 36149967 PMCID: PMC9506727 DOI: 10.1126/sciadv.abq0273] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 08/04/2022] [Indexed: 06/16/2023]
Abstract
To develop vaccines for certain key global pathogens such as HIV, it is crucial to elicit both neutralizing and non-neutralizing Fc-mediated effector antibody functions. Clinical evidence indicates that non-neutralizing antibody functions including antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) contribute to protection against several pathogens. In this study, we demonstrated that conjugation of HIV Envelope (Env) antigen gp120 to a self-assembling nanofiber material named Q11 induced antibodies with higher breadth and functionality when compared to soluble gp120. Immunization with Q11-conjugated gp120 vaccine (gp120-Q11) demonstrated higher tier 1 neutralization, ADCP, and ADCC as compared to soluble gp120. Moreover, Q11 conjugation altered the Fc N-glycosylation profile of antigen-specific antibodies, leading to a phenotype associated with increased ADCC in animals immunized with gp120-Q11. Thus, this nanomaterial vaccine strategy can enhance non-neutralizing antibody functions possibly through modulation of immunoglobulin G Fc N-glycosylation.
Collapse
Affiliation(s)
- Jui-Lin Chen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chelsea N. Fries
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Stella J. Berendam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nicole S. Rodgers
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Emily F. Roe
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Shuk Hang Li
- The Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rishabh Jain
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Brian Watts
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Joshua Eudailey
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham NC 27710, USA
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, NC 27707, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham NC 27710, USA
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, NC 27707, USA
| | - M. Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin O. Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joel H. Collier
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
14
|
Iyer RF, Edwards DM, Kolb P, Raué HP, Nelson CA, Epperson ML, Slifka MK, Nolz JC, Hengel H, Fremont DH, Früh K. The secreted protein Cowpox Virus 14 contributes to viral virulence and immune evasion by engaging Fc-gamma-receptors. PLoS Pathog 2022; 18:e1010783. [PMID: 36121874 PMCID: PMC9521928 DOI: 10.1371/journal.ppat.1010783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 09/29/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
The genome of cowpoxvirus (CPXV) could be considered prototypical for orthopoxviridae (OXPV) since it contains many open reading frames (ORFs) absent or lost in other OPXV, including vaccinia virus (VACV). These additional ORFs are non-essential for growth in vitro but are expected to contribute to the broad host range, virulence and immune evasion characteristics of CPXV. For instance, unlike VACV, CPXV encodes proteins that interfere with T cell stimulation, either directly or by preventing antigen presentation or co-stimulation. When studying the priming of naïve T cells, we discovered that CPXV, but not VACV, encodes a secreted factor that interferes with activation and proliferation of naïve CD8+ and CD4+ T cells, respectively, in response to anti-CD3 antibodies, but not to other stimuli. Deletion mapping revealed that the inhibitory protein is encoded by CPXV14, a small secreted glycoprotein belonging to the poxvirus immune evasion (PIE) family and containing a smallpoxvirus encoded chemokine receptor (SECRET) domain that mediates binding to chemokines. We demonstrate that CPXV14 inhibition of antibody-mediated T cell activation depends on the presence of Fc-gamma receptors (FcγRs) on bystander cells. In vitro, CPXV14 inhibits FcγR-activation by antigen/antibody complexes by binding to FcγRs with high affinity and immobilized CPXV14 can trigger signaling through FcγRs, particularly the inhibitory FcγRIIB. In vivo, CPXV14-deleted virus showed reduced viremia and virulence resulting in reduced weight loss and death compared to wildtype virus whereas both antibody and CD8+ T cell responses were increased in the absence of CPXV14. Furthermore, no impact of CPXV14-deletion on virulence was observed in mice lacking the inhibitory FcγRIIB. Taken together our results suggest that CPXV14 contributes to virulence and immune evasion by binding to host FcγRs.
Collapse
Affiliation(s)
- Ravi F. Iyer
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - David M. Edwards
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans-Peter Raué
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Chris A. Nelson
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Megan L. Epperson
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Mark K. Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daved H. Fremont
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| |
Collapse
|
15
|
Semmes EC, Miller IG, Wimberly CE, Phan CT, Jenks JA, Harnois MJ, Berendam SJ, Webster H, Hurst JH, Kurtzberg J, Fouda GG, Walsh KM, Permar SR. Maternal Fc-mediated non-neutralizing antibody responses correlate with protection against congenital human cytomegalovirus infection. J Clin Invest 2022; 132:e156827. [PMID: 35763348 PMCID: PMC9374380 DOI: 10.1172/jci156827] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 06/24/2022] [Indexed: 01/05/2023] Open
Abstract
Human cytomegalovirus (HCMV) is the most common congenital infection and a leading cause of stillbirth, neurodevelopmental impairment, and pediatric hearing loss worldwide. Development of a maternal vaccine or therapeutic to prevent congenital HCMV has been hindered by limited knowledge of the immune responses that protect against HCMV transmission in utero. To identify protective antibody responses, we measured HCMV-specific IgG binding and antiviral functions in paired maternal and cord blood sera from HCMV-seropositive transmitting (n = 41) and non-transmitting (n = 40) mother-infant dyads identified via a large, US-based, public cord blood bank. We found that high-avidity IgG binding to HCMV and antibody-dependent cellular phagocytosis (ADCP) were associated with reduced risk of congenital HCMV infection. We also determined that HCMV-specific IgG activation of FcγRI and FcγRII was enhanced in non-transmitting dyads and that increased ADCP responses were mediated through both FcγRI and FcγRIIA expressed on human monocytes. These findings suggest that engagement of FcγRI/FcγRIIA and Fc effector functions including ADCP may protect against congenital HCMV infection. Taken together, these data can guide future prospective studies on immune correlates against congenital HCMV transmission and inform HCMV vaccine and immunotherapeutic development.
Collapse
Affiliation(s)
- Eleanor C. Semmes
- Medical Scientist Training Program, Department of Molecular Genetics and Microbiology and
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Duke Children’s Health & Discovery Initiative, Duke University, Durham, North Carolina, USA
| | - Itzayana G. Miller
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Weill Cornell School of Medicine, New York, New York, USA
| | - Courtney E. Wimberly
- Duke Children’s Health & Discovery Initiative, Duke University, Durham, North Carolina, USA
- Department of Neurosurgery and
| | - Caroline T. Phan
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jennifer A. Jenks
- Medical Scientist Training Program, Department of Molecular Genetics and Microbiology and
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Melissa J. Harnois
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Stella J. Berendam
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Helen Webster
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jillian H. Hurst
- Duke Children’s Health & Discovery Initiative, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
- Carolinas Cord Blood Bank, Duke University Medical Center, Durham, North Carolina, USA
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Duke Children’s Health & Discovery Initiative, Duke University, Durham, North Carolina, USA
| | - Kyle M. Walsh
- Duke Children’s Health & Discovery Initiative, Duke University, Durham, North Carolina, USA
- Department of Neurosurgery and
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Duke Children’s Health & Discovery Initiative, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Weill Cornell School of Medicine, New York, New York, USA
| |
Collapse
|
16
|
Antibody escape and global spread of SARS-CoV-2 lineage A.27. Nat Commun 2022; 13:1152. [PMID: 35241661 PMCID: PMC8894356 DOI: 10.1038/s41467-022-28766-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/10/2022] [Indexed: 01/07/2023] Open
Abstract
In spring 2021, an increasing number of infections was observed caused by the hitherto rarely described SARS-CoV-2 variant A.27 in south-west Germany. From December 2020 to June 2021 this lineage has been detected in 31 countries. Phylogeographic analyses of A.27 sequences obtained from national and international databases reveal a global spread of this lineage through multiple introductions from its inferred origin in Western Africa. Variant A.27 is characterized by a mutational pattern in the spike gene that includes the L18F, L452R and N501Y spike amino acid substitutions found in various variants of concern but lacks the globally dominant D614G. Neutralization assays demonstrate an escape of A.27 from convalescent and vaccine-elicited antibody-mediated immunity. Moreover, the therapeutic monoclonal antibody Bamlanivimab and partially the REGN-COV2 cocktail fail to block infection by A.27. Our data emphasize the need for continued global monitoring of novel lineages because of the independent evolution of new escape mutations.
Collapse
|
17
|
Zhao S, Grieshaber-Bouyer R, Rao DA, Kolb P, Chen H, Andreeva I, Tretter T, Lorenz HM, Watzl C, Wabnitz G, Tykocinski LO, Merkt W. Effect of JAK Inhibition on the Induction of Proinflammatory HLA-DR+CD90+ Rheumatoid Arthritis Synovial Fibroblasts by Interferon-γ. Arthritis Rheumatol 2022; 74:441-452. [PMID: 34435471 PMCID: PMC9060076 DOI: 10.1002/art.41958] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 06/17/2021] [Accepted: 08/24/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Findings from recent transcriptome analyses of the synovium of patients with rheumatoid arthritis (RA) have revealed that 15-fold expanded HLA-DR+CD90+ synovial fibroblasts potentially act as key mediators of inflammation. The reasons for the expansion of HLA-DR+CD90+ synovial fibroblasts are unclear, but genetic signatures indicate that interferon-γ (IFNγ) plays a central role in the generation of this fibroblast subset. The present study was undertaken to investigate the generation, function and therapeutically intended blockage of HLA-DR+CD90+ synovial fibroblasts. METHODS We combined functional assays using primary human materials and focused bioinformatic analyses of mass cytometry and transcriptomics patient data sets. RESULTS We detected enriched and activated Fcγ receptor type IIIa-positive (CD16+) NK cells in the synovial tissue from patients with active RA. Soluble immune complexes were recognized by CD16 in a newly described reporter cell model, a mechanism that could be contributing to the activation of natural killer (NK) cells in RA. In vitro, NK cell-derived IFNγ induced HLA-DR on CD90+ synovial fibroblasts, leading to an inflammatory, cytokine-secreting HLA-DR+CD90+ phenotype. HLA-DR+CD90+ synovial fibroblasts consecutively activated CD4+ T cells upon receptor crosslinking via superantigens. HLA-DR+CD90+ synovial fibroblasts also activated CD4+ T cells in the absence of superantigens, an effect that was initiated by NK cell-derived IFNγ and that was 4 times stronger in patients with RA compared to patients with osteoarthritis. Finally, JAK inhibition in synovial fibroblasts prevented HLA-DR induction and blocked proinflammatory signals to T cells. CONCLUSION The HLA-DR+CD90+ phenotype represents an activation state of synovial fibroblasts during the process of inflammation in RA that can be induced by IFNγ, likely generated from infiltrating leukocytes such as activated NK cells. The induction of these proinflammatory, interleukin-6-producing, and likely antigen-presenting synovial fibroblasts can be targeted by JAK inhibition.
Collapse
Affiliation(s)
- Shuyang Zhao
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Ricardo Grieshaber-Bouyer
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany,Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Philipp Kolb
- Institute of Virology, University Medical Center, Freiburg, Germany,Faculty of Medicine, Albert-Ludwigs-University Freiburg, Germany
| | - Haizhang Chen
- Institute of Virology, University Medical Center, Freiburg, Germany,Faculty of Medicine, Albert-Ludwigs-University Freiburg, Germany
| | - Ivana Andreeva
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Theresa Tretter
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Watzl
- Leibniz Research Center for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| | - Guido Wabnitz
- Section Molecular Immunology, Institute of Immunology, Heidelberg University, Heidelberg, Germany
| | - Lars-Oliver Tykocinski
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Merkt
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany,Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| |
Collapse
|
18
|
Chen H, Maul‐Pavicic A, Holzer M, Huber M, Salzer U, Chevalier N, Voll RE, Hengel H, Kolb P. Detection and functional resolution of soluble immune complexes by an FcγR reporter cell panel. EMBO Mol Med 2022; 14:e14182. [PMID: 34842342 PMCID: PMC8749491 DOI: 10.15252/emmm.202114182] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
Fc-gamma receptor (FcγR) activation by soluble IgG immune complexes (sICs) represents a major mechanism of inflammation in certain autoimmune diseases such as systemic lupus erythematosus (SLE). A robust and scalable test system allowing for the detection and quantification of sIC bioactivity is missing. We developed a comprehensive reporter cell panel detecting activation of FcγRs. The reporter cell lines were integrated into an assay that enables the quantification of sIC reactivity via ELISA or a faster detection using flow cytometry. This identified FcγRIIA(H) and FcγRIIIA as the most sIC-sensitive FcγRs in our test system. Reaching a detection limit in the very low nanomolar range, the assay proved also to be sensitive to sIC stoichiometry and size reproducing for the first time a complete Heidelberger-Kendall curve in terms of immune receptor activation. Analyzing sera from SLE patients and mouse models of lupus and arthritis proved that sIC-dependent FcγR activation has predictive capabilities regarding severity of SLE disease. The assay provides a sensitive and scalable tool to evaluate the size, amount, and bioactivity of sICs in all settings.
Collapse
Affiliation(s)
- Haizhang Chen
- Institute of VirologyUniversity Medical CenterAlbert‐Ludwigs‐University FreiburgFreiburgGermany
- Faculty of MedicineAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Andrea Maul‐Pavicic
- Department of Rheumatology and Clinical ImmunologyMedical Center – University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Center for Chronic Immunodeficiency (CCI)Medical Center‐University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Martin Holzer
- Institute for Pharmaceutical SciencesAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Magdalena Huber
- Institute of VirologyUniversity Medical CenterAlbert‐Ludwigs‐University FreiburgFreiburgGermany
- Faculty of MedicineAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Ulrich Salzer
- Department of Rheumatology and Clinical ImmunologyMedical Center – University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Nina Chevalier
- Department of Rheumatology and Clinical ImmunologyMedical Center – University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical ImmunologyMedical Center – University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Center for Chronic Immunodeficiency (CCI)Medical Center‐University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Hartmut Hengel
- Institute of VirologyUniversity Medical CenterAlbert‐Ludwigs‐University FreiburgFreiburgGermany
- Faculty of MedicineAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Philipp Kolb
- Institute of VirologyUniversity Medical CenterAlbert‐Ludwigs‐University FreiburgFreiburgGermany
- Faculty of MedicineAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| |
Collapse
|
19
|
The Role of Fc Receptors on the Effectiveness of Therapeutic Monoclonal Antibodies. Int J Mol Sci 2021; 22:ijms22168947. [PMID: 34445651 PMCID: PMC8396266 DOI: 10.3390/ijms22168947] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Since the approval of the first monoclonal antibody (mAb) in 1986, a huge effort has been made to guarantee safety and efficacy of therapeutic mAbs. As of July 2021, 118 mAbs are approved for the European market for a broad range of clinical indications. In order to ensure clinical efficacy and safety aspects, (pre-)clinical experimental approaches evaluate the respective modes of action (MoA). In addition to antigen-specificity including binding affinity and -avidity, MoA comprise Fc-mediated effector functions such as antibody dependent cellular cytotoxicity (ADCC) and the closely related antibody dependent cellular phagocytosis (ADCP). For this reason, a variety of cell-based assays have been established investigating effector functions of therapeutic mAbs with different effector/target-cell combinations and several readouts including Fcγ receptor (FcγR)-mediated lysis, fluorescence, or luminescence. Optimized FcγR-mediated effector functions regarding clinical safety and efficacy are addressed with modification strategies such as point mutations, altered glycosylation patterns, combination of different Fc subclasses (cross isotypes), and Fc-truncation of the mAb. These strategies opened the field for a next generation of therapeutic mAbs. In conclusion, it is of major importance to consider FcγR-mediated effector functions for the efficacy of therapeutic mAbs.
Collapse
|
20
|
León G, Herrera M, Vargas M, Arguedas M, Sánchez A, Segura Á, Gómez A, Solano G, Corrales-Aguilar E, Risner K, Narayanan A, Bailey C, Villalta M, Hernández A, Sánchez A, Cordero D, Solano D, Durán G, Segura E, Cerdas M, Umaña D, Moscoso E, Estrada R, Gutiérrez J, Méndez M, Castillo AC, Sánchez L, Sánchez R, Gutiérrez JM, Díaz C, Alape A. Development and characterization of two equine formulations towards SARS-CoV-2 proteins for the potential treatment of COVID-19. Sci Rep 2021; 11:9825. [PMID: 33972631 PMCID: PMC8110969 DOI: 10.1038/s41598-021-89242-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/21/2021] [Indexed: 02/03/2023] Open
Abstract
In the current global emergency due to SARS-CoV-2 outbreak, passive immunotherapy emerges as a promising treatment for COVID-19. Among animal-derived products, equine formulations are still the cornerstone therapy for treating envenomations due to animal bites and stings. Therefore, drawing upon decades of experience in manufacturing snake antivenom, we developed and preclinically evaluated two anti-SARS-CoV-2 polyclonal equine formulations as potential alternative therapy for COVID-19. We immunized two groups of horses with either S1 (anti-S1) or a mixture of S1, N, and SEM mosaic (anti-Mix) viral recombinant proteins. Horses reached a maximum anti-viral antibody level at 7 weeks following priming, and showed no major adverse acute or chronic clinical alterations. Two whole-IgG formulations were prepared via hyperimmune plasma precipitation with caprylic acid and then formulated for parenteral use. Both preparations had similar physicochemical and microbiological quality and showed ELISA immunoreactivity towards S1 protein and the receptor binding domain (RBD). The anti-Mix formulation also presented immunoreactivity against N protein. Due to high anti-S1 and anti-RBD antibody content, final products exhibited high in vitro neutralizing capacity of SARS-CoV-2 infection, 80 times higher than a pool of human convalescent plasma. Pre-clinical quality profiles were similar among both products, but clinical efficacy and safety must be tested in clinical trials. The technological strategy we describe here can be adapted by other producers, particularly in low- and middle-income countries.
Collapse
Affiliation(s)
- Guillermo León
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - María Herrera
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Mariángela Vargas
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica.
| | - Mauricio Arguedas
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Andrés Sánchez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Álvaro Segura
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Aarón Gómez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Gabriela Solano
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Eugenia Corrales-Aguilar
- Virology-CIET (Research Center for Tropical Diseases), Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Kenneth Risner
- National Center for Biodefense and Infectious Diseases, George Mason University, Virginia, USA
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, George Mason University, Virginia, USA
| | - Charles Bailey
- National Center for Biodefense and Infectious Diseases, George Mason University, Virginia, USA
| | - Mauren Villalta
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Andrés Hernández
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Adriana Sánchez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Daniel Cordero
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Daniela Solano
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Gina Durán
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Eduardo Segura
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Maykel Cerdas
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Deibid Umaña
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Edwin Moscoso
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Ricardo Estrada
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Jairo Gutiérrez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Marcos Méndez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Ana Cecilia Castillo
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Laura Sánchez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Ronald Sánchez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - José María Gutiérrez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Cecilia Díaz
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | - Alberto Alape
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
- Departamento de Bioquímica, Escuela de Medicina, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
21
|
Kolb P, Hoffmann K, Sievert A, Reinhard H, Merce-Maldonado E, Le-Trilling VTK, Halenius A, Gütle D, Hengel H. Human cytomegalovirus antagonizes activation of Fcγ receptors by distinct and synergizing modes of IgG manipulation. eLife 2021; 10:63877. [PMID: 33724188 PMCID: PMC8041466 DOI: 10.7554/elife.63877] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/15/2021] [Indexed: 01/19/2023] Open
Abstract
Human cytomegalovirus (HCMV) is endowed with multiple highly sophisticated immune evasion strategies. This includes the evasion from antibody mediated immune control by counteracting host Fc-gamma receptor (FcγR) mediated immune control mechanisms such as antibody-dependent cellular cytotoxicity (ADCC). We have previously shown that HCMV avoids FcγR activation by concomitant expression of the viral Fc-gamma-binding glycoproteins (vFcγRs) gp34 and gp68. We now show that gp34 and gp68 bind IgG simultaneously at topologically different Fcγ sites and achieve efficient antagonization of host FcγR activation by distinct but synergizing mechanisms. While gp34 enhances immune complex internalization, gp68 acts as inhibitor of host FcγR binding to immune complexes. In doing so, gp68 induces Fcγ accessibility to gp34 and simultaneously limits host FcγR recognition. The synergy of gp34 and gp68 is compelled by the interfering influence of excessive non-immune IgG ligands and highlights conformational changes within the IgG globular chains critical for antibody effector function. Human cytomegalovirus is a type of herpes virus that rarely causes symptoms in healthy people but can cause serious complications in unborn babies and in people with compromised immune systems, such as transplant recipients. The virus has found ways to successfully evade the immune system, and once infected, the body retains the virus for life. It deploys an arsenal of proteins that bind to antibodies, specialized proteins the immune system uses to flag virus-infected cells for destruction. This prevents certain cells of the immune system, the natural killer cells, from recognizing and destroying virus-infected cells. These immune-evading proteins are called viral Fc-gamma receptors, or vFcγRs. While it has been previously shown that these receptors are able to evade the immune system, it remained unknown how exactly they prevent natural killer cells from recognizing infected cells. Now, Kolb et al. show that the cytomegalovirus deploys two vFcγRs called gp34 and gp68, which work together to block natural killer cells. The latter reduces the ability of natural killer cells to bind to antibodies on cytomegalovirus-infected cells. This paves the way for gp34 to pull virus proteins from the surface of the infected cell, making them inaccessible to the immune system. Neither protein fully protects virus-infected cells on its own, but together they are highly effective. The experiments reveal further details about how cytomegalovirus uses two defense mechanisms simultaneously to outmaneuver the immune system. Understanding this two-part viral evasion system may help scientists to develop vaccines or new treatments that can protect vulnerable people from diseases caused by the cytomegalovirus.
Collapse
Affiliation(s)
- Philipp Kolb
- Faculty of Medicine, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Katja Hoffmann
- Faculty of Medicine, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Annika Sievert
- Faculty of Medicine, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Henrike Reinhard
- Institute of Virology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Eva Merce-Maldonado
- Institute of Virology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | - Anne Halenius
- Faculty of Medicine, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Dominique Gütle
- Faculty of Medicine, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Hartmut Hengel
- Faculty of Medicine, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| |
Collapse
|
22
|
Demminger DE, Walz L, Dietert K, Hoffmann H, Planz O, Gruber AD, von Messling V, Wolff T. Adeno-associated virus-vectored influenza vaccine elicits neutralizing and Fcγ receptor-activating antibodies. EMBO Mol Med 2020; 12:e10938. [PMID: 32163240 PMCID: PMC7207162 DOI: 10.15252/emmm.201910938] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022] Open
Abstract
The current seasonal inactivated influenza vaccine protects only against a narrow range of virus strains as it triggers a dominant antibody response toward the hypervariable hemagglutinin (HA) head region. The discovery of rare broadly protective antibodies against conserved regions in influenza virus proteins has propelled research on distinct antigens and delivery methods to efficiently induce broad immunity toward drifted or shifted virus strains. Here, we report that adeno‐associated virus (AAV) vectors expressing influenza virus HA or chimeric HA protected mice against homologous and heterologous virus challenges. Unexpectedly, immunization even with wild‐type HA induced antibodies recognizing the HA‐stalk and activating FcγR‐dependent responses indicating that AAV‐vectored expression balances HA head‐ and HA stalk‐specific humoral responses. Immunization with AAV‐HA partially protected also ferrets against a harsh virus challenge. Results from this study provide a rationale for further clinical development of AAV vectors as influenza vaccine platform, which could benefit from their approved use in human gene therapy.
Collapse
Affiliation(s)
- Daniel E Demminger
- Unit 17-Influenza and Other Respiratory Viruses, Robert Koch Institute, Berlin, Germany
| | - Lisa Walz
- Veterinary Medicine Division, Paul-Ehrlich-Institute, Langen, Germany
| | - Kristina Dietert
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Berlin, Germany
| | - Helen Hoffmann
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| | - Oliver Planz
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| | - Achim D Gruber
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Berlin, Germany
| | | | - Thorsten Wolff
- Unit 17-Influenza and Other Respiratory Viruses, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
23
|
Pérez-Portilla A, Moraru M, Blázquez-Moreno A, Kolb P, Bravo García-Morato M, Ranganath T, Esteso G, Gianelli C, Rodríguez-Pena R, Lozano-Rodríguez R, Torres-Canizales JM, Blish CA, Vales-Gomez M, Hengel H, Vilches C, López-Granados E, Reyburn HT. Identification of the first cases of complete CD16A deficiency: Association with persistent EBV infection. J Allergy Clin Immunol 2020; 145:1288-1292. [PMID: 31953104 DOI: 10.1016/j.jaci.2019.11.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 10/31/2019] [Accepted: 11/27/2019] [Indexed: 10/25/2022]
Affiliation(s)
- A Pérez-Portilla
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - M Moraru
- Department of Immunology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Alfonso Blázquez-Moreno
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Philipp Kolb
- Institute of Virology, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - María Bravo García-Morato
- Clinical Immunology Department, University Hospital La Paz and Lymphocyte Pathophysiology in Immunodeficiencies Group, IdiPAZ Institute for Health Research, Madrid, Spain
| | - Thanmayi Ranganath
- Department of Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Gloria Esteso
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Carla Gianelli
- Clinical Immunology Department, University Hospital La Paz and Lymphocyte Pathophysiology in Immunodeficiencies Group, IdiPAZ Institute for Health Research, Madrid, Spain
| | - Rebeca Rodríguez-Pena
- Clinical Immunology Department, University Hospital La Paz and Lymphocyte Pathophysiology in Immunodeficiencies Group, IdiPAZ Institute for Health Research, Madrid, Spain
| | - Roberto Lozano-Rodríguez
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Juan Manuel Torres-Canizales
- Clinical Immunology Department, University Hospital La Paz and Lymphocyte Pathophysiology in Immunodeficiencies Group, IdiPAZ Institute for Health Research, Madrid, Spain
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Mar Vales-Gomez
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Hartmut Hengel
- Institute of Virology, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carlos Vilches
- Department of Immunology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain.
| | - Eduardo López-Granados
- Clinical Immunology Department, University Hospital La Paz and Lymphocyte Pathophysiology in Immunodeficiencies Group, IdiPAZ Institute for Health Research, Madrid, Spain.
| | - Hugh T Reyburn
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
24
|
De Vlieger D, Hoffmann K, Van Molle I, Nerinckx W, Van Hoecke L, Ballegeer M, Creytens S, Remaut H, Hengel H, Schepens B, Saelens X. Selective Engagement of FcγRIV by a M2e-Specific Single Domain Antibody Construct Protects Against Influenza A Virus Infection. Front Immunol 2019; 10:2920. [PMID: 31921179 PMCID: PMC6921966 DOI: 10.3389/fimmu.2019.02920] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/27/2019] [Indexed: 11/21/2022] Open
Abstract
Lower respiratory tract infections, such as infections caused by influenza A viruses, are a constant threat for public health. Antivirals are indispensable to control disease caused by epidemic as well as pandemic influenza A. We developed a novel anti-influenza A virus approach based on an engineered single-domain antibody (VHH) construct that can selectively recruit innate immune cells to the sites of virus replication. This protective construct comprises two VHHs. One VHH binds with nanomolar affinity to the conserved influenza A matrix protein 2 (M2) ectodomain (M2e). Co-crystal structure analysis revealed that the complementarity determining regions 2 and 3 of this VHH embrace M2e. The second selected VHH specifically binds to the mouse Fcγ Receptor IV (FcγRIV) and was genetically fused to the M2e-specific VHH, which resulted in a bi-specific VHH-based construct that could be efficiently expressed in Pichia pastoris. In the presence of M2 expressing or influenza A virus-infected target cells, this single domain antibody construct selectively activated the mouse FcγRIV. Moreover, intranasal delivery of this bispecific FcγRIV-engaging VHH construct protected wild type but not FcγRIV−/− mice against challenge with an H3N2 influenza virus. These results provide proof of concept that VHHs directed against a surface exposed viral antigen can be readily armed with effector functions that trigger protective antiviral activity beyond direct virus neutralization.
Collapse
Affiliation(s)
- Dorien De Vlieger
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Katja Hoffmann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Inge Van Molle
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium.,VIB-VUB Center for Structural Biology, Brussels, Belgium
| | - Wim Nerinckx
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Marlies Ballegeer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Sarah Creytens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Han Remaut
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium.,VIB-VUB Center for Structural Biology, Brussels, Belgium
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bert Schepens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
25
|
Cerino A, Mantovani S, Mele D, Oliviero B, Varchetta S, Mondelli MU. Human Monoclonal Antibodies as Adjuvant Treatment of Chronic Hepatitis B Virus Infection. Front Immunol 2019; 10:2290. [PMID: 31608071 PMCID: PMC6773823 DOI: 10.3389/fimmu.2019.02290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/10/2019] [Indexed: 01/05/2023] Open
Abstract
Despite the availability of an effective prophylactic vaccine leading to sterilizing immunity, hepatitis B virus (HBV) is responsible for chronic liver disease in more than 250 million individuals, potentially leading to cirrhosis and hepatocellular carcinoma. Antiviral drugs able to completely suppress virus replication are indeed available but they are, by and large, unable to eradicate the virus. Several alternative new treatment approaches are currently being developed but none have so far captured the interest of clinicians for possible clinical development. A constant feature of chronic HBV infection is T-cell exhaustion resulting from persistent exposure to high antigen concentrations as shown by the high expression of programmed cell death protein 1 (PD-1) by HBV-specific CD8 T cells. One way of tackling this problem is to develop HBV-specific neutralizing antibodies that would clear excess envelope proteins from the circulation, allowing for nucleos(t)ide analogs or other antiviral drugs now in preclinical and early clinical development to take advantage of a reconstituted adaptive immunity. Several fully human monoclonal antibodies (mAb) have been developed from HBV-vaccinated and subjects convalescent from acute hepatitis B that show different properties and specificities. It is envisaged that such neutralizing mAb may be used as adjuvant treatment to reduce viral protein load, thus rescuing adaptive immunity in an effort to optimize the effect of antiviral drugs.
Collapse
Affiliation(s)
- Antonella Cerino
- S.C. di Malattie Infettive II - Infettivologia e Immunologia, Dipartimento di Scienze Mediche e Malattie Infettive, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania Mantovani
- S.C. di Malattie Infettive II - Infettivologia e Immunologia, Dipartimento di Scienze Mediche e Malattie Infettive, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Dalila Mele
- S.C. di Malattie Infettive II - Infettivologia e Immunologia, Dipartimento di Scienze Mediche e Malattie Infettive, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Barbara Oliviero
- S.C. di Malattie Infettive II - Infettivologia e Immunologia, Dipartimento di Scienze Mediche e Malattie Infettive, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania Varchetta
- S.C. di Malattie Infettive II - Infettivologia e Immunologia, Dipartimento di Scienze Mediche e Malattie Infettive, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mario U Mondelli
- S.C. di Malattie Infettive II - Infettivologia e Immunologia, Dipartimento di Scienze Mediche e Malattie Infettive, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Dipartimento di Medicina Interna e Terapia Medica, Università di Pavia, Pavia, Italy
| |
Collapse
|
26
|
Lagassé HAD, Hengel H, Golding B, Sauna ZE. Fc-Fusion Drugs Have FcγR/C1q Binding and Signaling Properties That May Affect Their Immunogenicity. AAPS JOURNAL 2019; 21:62. [DOI: 10.1208/s12248-019-0336-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/13/2019] [Indexed: 12/17/2022]
|
27
|
Dudek S, Weißmüller S, Anzaghe M, Miller L, Sterr S, Hoffmann K, Hengel H, Waibler Z. Human Fcγ receptors compete for TGN1412 binding that determines the antibody's effector function. Eur J Immunol 2019; 49:1117-1126. [PMID: 31002172 DOI: 10.1002/eji.201847924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 02/21/2019] [Accepted: 04/09/2019] [Indexed: 11/07/2022]
Abstract
The first-in-human clinical trial of the CD28-specific monoclonal antibody (mAb) TGN1412 resulted in a life-threatening cytokine release syndrome. Although TGN1412 was designed as IgG4, known for weak Fc:Fcγ receptor (FcγR) interactions, these interactions contributed to TGN1412-induced T-cell activation. Using cell lines (TFs) expressing human FcγRI, -IIa, -IIb, or -III, we show that TGN1412 and TGN1412 as IgG1 and IgG2 are bound by FcγRs as it can be deduced from literature. However, upon coculture of TGN1412-decorated T cells with TFs or human primary blood cells, we observed that binding capacities by FcγRs do not correlate with the strength of the mediated effector function. FcγRIIa and FcγRIIb, showing no or very minor binding to TGN1412, mediated strongest T cell proliferation, while high-affinity FcγRI, exhibiting strong TGN1412 binding, mediated hardly any T-cell proliferation. These findings are of biological relevance because we show that FcγRI binds TGN1412, thus prevents binding to FcγRIIa or FcγRIIb, and consequently disables T-cell proliferation. In line with this, FcγRI- FcγRII+ but not FcγRI+ FcγRII+ monocytes mediate TGN1412-induced T-cell proliferation. Collectively, by using TGN1412 as example, our results indicate that binding of monomeric IgG subclasses does not predict the FcγR-mediated effector function, which has major implications for the design of therapeutic mAbs.
Collapse
Affiliation(s)
- Simone Dudek
- Product Testing of Immunological Biopharmaceuticals, Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Sabrina Weißmüller
- Product Testing of Immunological Biopharmaceuticals, Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Martina Anzaghe
- Product Testing of Immunological Biopharmaceuticals, Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Lilija Miller
- Product Testing of Immunological Biopharmaceuticals, Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Sarah Sterr
- Product Testing of Immunological Biopharmaceuticals, Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Katja Hoffmann
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Hengel
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Zoe Waibler
- Product Testing of Immunological Biopharmaceuticals, Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
28
|
Kolb P, Sijmons S, McArdle MR, Taher H, Womack J, Hughes C, Ventura A, Jarvis MA, Stahl-Hennig C, Hansen S, Picker LJ, Malouli D, Hengel H, Früh K. Identification and Functional Characterization of a Novel Fc Gamma-Binding Glycoprotein in Rhesus Cytomegalovirus. J Virol 2019; 93:e02077-18. [PMID: 30487278 PMCID: PMC6364020 DOI: 10.1128/jvi.02077-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/20/2022] Open
Abstract
Receptors recognizing the Fc part of immunoglobulin G (FcγRs) are key determinants in antibody-mediated immune responses. Members of the Herpesviridae interfere with this immune regulatory network by expressing viral FcγRs (vFcγRs). Human cytomegalovirus (HCMV) encodes four distinct vFcγRs that differ with respect to their IgG subtype specificity and their impact on antibody-mediated immune function in vitro The impact of vFcγRs on HCMV pathogenesis and immunomodulation in vivo is not known. The closest evolutionary animal model of HCMV is rhesus CMV (RhCMV) infection of rhesus macaques. To enable the characterization of vFcγR function in this model, we studied IgG binding by RhCMV. We show that lysates of RhCMV-infected cells contain an IgG-binding protein of 30 kDa encoded by the gene Rh05 that is a predicted type I glycoprotein belonging to the RL11 gene family. Upon deletion of Rh05, IgG-Fc binding by RhCMV strain 68-1 is lost, whereas ectopic expression of Rh05 results in IgG binding to transfected cells consistent with Rh05 being a vFcγR. Using a set of reporter cell lines stably expressing human and rhesus FcγRs, we further demonstrate that Rh05 antagonizes host FcγR activation. Compared to Rh05-intact RhCMV, RhCMVΔRh05 showed an increased activation of host FcγR upon exposure of infected cells to IgG from RhCMV-seropositive animals, suggesting that Rh05 protects infected cells from opsonization and IgG-dependent activation of host FcγRs. However, antagonizing host FcγR activation by Rh05 was not required for the establishment and maintenance of infection of RhCMV, even in a seropositive host, as shown by the induction of T cell responses to heterologous antigens expressed by RhCMV lacking the gene region encoding Rh05. In contrast to viral evasion of natural killer cells or T cell recognition, the evasion of antibody-mediated effects does not seem to be absolutely required for infection or reinfection. The identification of the first vFcγR that efficiently antagonizes host FcγR activation in the RhCMV genome will thus permit more detailed studies of this immunomodulatory mechanism in promoting viral dissemination in the presence of natural or vaccine-induced humoral immunity.IMPORTANCE Rhesus cytomegalovirus (RhCMV) offers a unique model for studying human cytomegalovirus (HCMV) pathogenesis and vaccine development. RhCMV infection of nonhuman primates greatly broadened the understanding of mechanisms by which CMVs evade or reprogram T cell and natural killer cell responses in vivo However, the role of humoral immunity and viral modulation of anti-CMV antibodies has not been studied in this model. There is evidence from in vitro studies that HCMVs can evade humoral immunity. By gene mapping and with the help of a novel cell-based reporter assay system we characterized the first RhCMV encoded IgG-Fcγ binding glycoprotein as a potent antagonist of rhesus FcγR activation. We further demonstrate that, unlike evasion of T cell immunity, this viral Fcγ receptor is not required to overcome anti-CMV immunity to establish secondary infections. These findings enable more detailed studies of the in vivo consequences of CMV evasion from IgG responses in nonhuman primate models.
Collapse
Affiliation(s)
- Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Steven Sijmons
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Matthew R McArdle
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Jennie Womack
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Colette Hughes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Abigail Ventura
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Michael A Jarvis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Scott Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| |
Collapse
|
29
|
Wågström P, Yamada-Fowler N, Dahle C, Nilsdotter-Augustinsson Å, Bengnér M, Söderkvist P, Björkander J. Fcγ-receptor polymorphisms associated with clinical symptoms in patients with immunoglobulin G subclass deficiency. Infect Dis (Lond) 2018; 50:853-858. [PMID: 30298768 DOI: 10.1080/23744235.2018.1510183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Immunoglobulin G subclass deficiencies (IgGsd) are associated with recurrent respiratory tract infections. Immunoglobulin substitution therapy may be needed to prevent chronic lung tissue damage but tools for identifying the patients that will benefit from this treatment are still insufficient. Some FcγR polymorphisms seem to predispose for an increased risk for infections. In this study we wanted to evaluate if the FcγR-profile differs between individuals with IgGsd and a control population. METHODS Single nucleotide polymorphisms (SNPs) of FcγRIIa, FcγRIIIa and FcγRIIc in 36 IgGsd patients and 192 controls with similar sex and geographical distribution were analyzed by TaqMan allelic discrimination assay or Sanger sequencing. RESULTS In the IgGsd-group, homozygous frequency for FcγRIIa-R/R131 (low-binding capacity isoform) was higher (p = .03) as well as for non-classical FcγRIIc-ORF (p = .03) and classical FcγRIIc-ORF tended (p = .07) to be more common compared to the controls. There was no difference between the groups regarding FcγRIIIa. CONCLUSION The gene for classical FcγRIIc-ORF tended to be more frequent in individuals with immunoglobulin G subclass deficiency and the genes for non-classical FcγRIIc-ORF as well as low-binding capacity receptor FcγRIIa-R/R131 were more frequent. Further studies on the FcγR polymorphisms may pave way for identifying individuals that will benefit from immunoglobulin substitution.
Collapse
Affiliation(s)
- Per Wågström
- a Department of Infectious Diseases, Ryhov County Hospital, Jönköping and Department of Clinical and Experimental Medicine , Linköping University , Linköping , Sweden
| | - Naomi Yamada-Fowler
- b Department of Clinical and Experimental Medicine , Linköping University, Division of Cell Biology , Linköping , Sweden
| | - Charlotte Dahle
- c Department of Clinical Immunology and Transfusion Medicine and Department of Clinical and Experimental Medicine , Linköping University , Linköping , Sweden
| | - Åsa Nilsdotter-Augustinsson
- d Department of Infectious Diseases and Department of Clinical and Experimental Medicine , Linköping University , Linköping , Sweden
| | - Malin Bengnér
- e Office for Control of Communicable Diseases , Ryhov County Hospital , Jönköping , Sweden
| | - Peter Söderkvist
- b Department of Clinical and Experimental Medicine , Linköping University, Division of Cell Biology , Linköping , Sweden
| | - Janne Björkander
- f Division of Clinical Immunology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences , Linköping University , Sweden
| |
Collapse
|
30
|
Bailey MJ, Broecker F, Leon PE, Tan GS. A Method to Assess Fc-mediated Effector Functions Induced by Influenza Hemagglutinin Specific Antibodies. J Vis Exp 2018. [PMID: 29553549 DOI: 10.3791/56256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Antibodies play a crucial role in coupling the innate and adaptive immune responses against viral pathogens through their antigen binding domains and Fc-regions. Here, we describe how to measure the activation of Fc effector functions by monoclonal antibodies targeting the influenza virus hemagglutinin with the use of a genetically engineered Jurkat cell line expressing an activating type 1 Fc-FcγR. Using this method, the contribution of specific Fc-FcγR interactions conferred by immunoglobulins can be determined using an in vitro assay.
Collapse
Affiliation(s)
- Mark J Bailey
- Department of Microbiology, Icahn School of Medicine at Mount Sinai; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai
| | - Felix Broecker
- Department of Microbiology, Icahn School of Medicine at Mount Sinai
| | - Paul E Leon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai
| | - Gene S Tan
- Department of Infectious Disease, J. Craig Venter Institute;
| |
Collapse
|
31
|
Elias S, Kahlon S, Kotzur R, Kaynan N, Mandelboim O. Obinutuzumab activates FcγRI more potently than other anti-CD20 antibodies in chronic lymphocytic leukemia (CLL). Oncoimmunology 2018; 7:e1428158. [PMID: 29872553 PMCID: PMC5980409 DOI: 10.1080/2162402x.2018.1428158] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/07/2018] [Accepted: 01/09/2018] [Indexed: 11/03/2022] Open
Abstract
Treatment with monoclonal antibodies has revolutionized clinical medicine, especially in the fields of cancer and immunology. One of the oldest antibodies, which is widely used for the treatment of lymphomas and autoimmune diseases, is the anti-CD20 antibody rituximab. In recent years, new antibodies against CD20 have been developed including ofatumumab and obinutuzumab. An important mechanism of action of therapeutic monoclonal antibodies is activation of immune cells via Fc receptors (FcγRs). However, surprisingly, little is known about triggering of FcγRs by different therapeutic antibodies in general and anti-CD20 antibodies in particular. Here we establish a reporter assay to assess whether a particular antibody activates a certain Fc receptor. Using this assay we corroborated previous reports demonstrating obinutuzumab's ability to highly activate FcγRIIIa (CD16a). Importantly, we discovered that obinutuzumab also activates FcγRI (CD64) significantly more than rituximab and ofatumumab in response to chronic lymphocytic leukemia (CLL) cells obtained from patients. Mechanistically we show that this is due to the lack of FcγRIIb-mediated internalization of obinutuzumab following binding to CD20. Moreover, we show that obinutuzumab induces increased phagocytosis by primary macrophages in an FcγRI-dependent manner. Beyond the discovery of a new mechanism of obinutuzumab activity, the reporter assay can be applied to other therapeutic antibodies and may assist in developing antibodies with improved immunological properties.
Collapse
Affiliation(s)
- Shlomo Elias
- Department of Hematology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.,The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Shira Kahlon
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Rebecca Kotzur
- Department of Transfusion Medicine, Institute of Immunogenetics, Hannover Medical School, Hannover, Germany
| | - Noah Kaynan
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
32
|
Intrastructural help: improving the HIV-1 envelope antibody response induced by virus-like particle vaccines. Curr Opin HIV AIDS 2017; 12:272-277. [PMID: 28422791 DOI: 10.1097/coh.0000000000000358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW The importance of IgG Fc-effector functions for the efficacy of HIV vaccines is increasingly recognized. Although different types of vaccines were shown to induce antibodies with different Fc-activities, there is no clear strategy how to raise antibody responses with a desired pattern of Fc-effector functions. Given the central role of T-helper cells in regulating the germinal center reaction and the differentiation of B cells in an antigen-specific manner, the review will discuss whether T-helper cells directed against non-HIV envelope (Env) antigens could be harnessed to improve the HIV-Env antibody response. RECENT FINDINGS Comparing CD4 T-cell responses in HIV-infected individuals with and without neutralizing antibody breadth suggests that robust Gag-specific CD4 T cells may provide important T-cell help to Env-specific B cells. In a murine model, GagPol-specific T-helper cells were shown to provide intrastructural help for HIV-Env-specific antibody responses after immunization with a virus-like particle vaccine. GagPol-specific T-helper cells imprinted the IgG subtype ratio observed for Gag onto the HIV-Env antibody response and modulated the glycosylation pattern of the HIV Env-specific antibodies. SUMMARY Intrastructural help is a promising strategy to improve overall levels and Fc-effector functions of the HIV-Env antibody response.
Collapse
|
33
|
Hierarchical and Redundant Roles of Activating FcγRs in Protection against Influenza Disease by M2e-Specific IgG1 and IgG2a Antibodies. J Virol 2017; 91:JVI.02500-16. [PMID: 28077656 DOI: 10.1128/jvi.02500-16] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 02/01/2023] Open
Abstract
The ectodomain of matrix protein 2 is a universal influenza A virus vaccine candidate that provides protection through antibody-dependent effector mechanisms. Here we compared the functional engagement of Fcγ receptor (FcγR) family members by two M2e-specific monoclonal antibodies (MAbs), MAb 37 (IgG1) and MAb 65 (IgG2a), which recognize a similar epitope in M2e with similar affinities. The binding of MAb 65 to influenza A virus-infected cells triggered all three activating mouse Fcγ receptors in vitro, whereas MAb 37 activated only FcγRIII. The passive transfer of MAb 37 or MAb 65 in wild-type, Fcer1g-/-, Fcgr3-/-, and Fcgr1-/-Fcgr3-/- BALB/c mice revealed the importance of these receptors for protection against influenza A virus challenge, with a clear requirement of FcγRIII for IgG1 MAb 37 being found. We also report that FcγRIV contributes to protection by M2e-specific IgG2a antibodies.IMPORTANCE There is increased awareness that protection by antibodies directed against viral antigens is also mediated by the Fc domain of these antibodies. These Fc-mediated effector functions are often missed in clinical assays, which are used, for example, to define correlates of protection induced by vaccines. The use of antibodies to prevent and treat infectious diseases is on the rise and has proven to be a promising approach in our battle against newly emerging viral infections. It is now also realized that Fcγ receptors significantly enhance the in vivo protective effect of broadly neutralizing antibodies directed against the conserved parts of the influenza virus hemagglutinin. We show here that two M2e-specific monoclonal antibodies with close to identical antigen-binding specificities and affinities have a very different in vivo protective potential that is controlled by their capacity to interact with activating Fcγ receptors.
Collapse
|
34
|
Diab M, Glasner A, Isaacson B, Bar-On Y, Drori Y, Yamin R, Duev-Cohen A, Danziger O, Zamostiano R, Mandelboim M, Jonjic S, Bacharach E, Mandelboim O. NK-cell receptors NKp46 and NCR1 control human metapneumovirus infection. Eur J Immunol 2017; 47:692-703. [DOI: 10.1002/eji.201646756] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/20/2016] [Accepted: 02/08/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Mohammad Diab
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Ariella Glasner
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Batya Isaacson
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Yotam Bar-On
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Yaron Drori
- Central Virology Laboratory, Ministry of Health, Public Health Services; Chaim Sheba Medical Center, Tel-Hashomer; Ramat-Gan Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine; Tel-Aviv University; Tel-Aviv Israel
| | - Rachel Yamin
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Alexandra Duev-Cohen
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Oded Danziger
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Rachel Zamostiano
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Michal Mandelboim
- Central Virology Laboratory, Ministry of Health, Public Health Services; Chaim Sheba Medical Center, Tel-Hashomer; Ramat-Gan Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine; Tel-Aviv University; Tel-Aviv Israel
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine; University of Rijeka; Rijeka Croatia
- Center for Proteomics, Faculty of Medicine; University of Rijeka; Rijeka Croatia
| | - Eran Bacharach
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| |
Collapse
|
35
|
Highly individual patterns of virus-immune IgG effector responses in humans. Med Microbiol Immunol 2016; 205:409-24. [PMID: 27193020 PMCID: PMC5003914 DOI: 10.1007/s00430-016-0457-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/09/2016] [Indexed: 12/25/2022]
Abstract
IgG responses are fundamental to adaptive immunity and document immunological memory of previous pathogen encounter. While specific antigen recognition is mediated by the variable F(ab')2 domain of IgG, various effector functions become activated via the constant Fcγ part bridging IgG-opsonized targets to FcγR-expressing immune effector cells. Traditionally, neutralizing IgG is considered the most appropriate correlate of protective humoral immunity to viruses. However, evidence is increasing that antiviral IgG mediates protection to viruses via activation of FcγRs. Using a test system allowing quantitative detection of virus-immune IgG able to activate FcγRs, sera of healthy individuals and vaccinees were assessed with regard to two prototypical human pathogenic viruses: measles and human cytomegalovirus. Marked differences in the capacity of individuals to generate FcγRI-, FcγRII- and FcγRIII-activating responses were noted. Comparison of FcγR-activating IgG with neutralizing and ELISA IgG concentrations did not correlate for HCMV and only very poorly for MV. Since neither neutralizing IgG nor overall IgG responses faithfully predict the activation of FcγRs, only the simultaneous quantification of IgGs activating defined FcγRs will aid to delineate individual "immunograms" of virus IgG immunity. Such new multiparametric assessment of antiviral IgG qualities could be instrumental in defining correlates of protection and disease progression.
Collapse
|
36
|
Storcksdieck genannt Bonsmann M, Niezold T, Temchura V, Pissani F, Ehrhardt K, Brown EP, Osei-Owusu NY, Hannaman D, Hengel H, Ackerman ME, Streeck H, Nabi G, Tenbusch M, Überla K. Enhancing the Quality of Antibodies to HIV-1 Envelope by GagPol-Specific Th Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:4861-72. [PMID: 26466954 DOI: 10.4049/jimmunol.1501377] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/18/2015] [Indexed: 11/19/2022]
Abstract
The importance of Fc-dependent effector functions of Abs induced by vaccination is increasingly recognized. However, vaccination of mice against HIV envelope (Env) induced a skewed Th cell response leading to Env-specific Abs with reduced effector function. To overcome this bias, GagPol-specific Th cells were harnessed to provide intrastructural help for Env-specific B cells after immunization with virus-like particles containing GagPol and Env. This led to a balanced Env-specific humoral immune response with a more inflammatory Fc glycan profile. The increased quality in the Ab response against Env was confirmed by FcγR activation assays. Because the Env-specific Th cell response was also biased in human vaccinees, intrastructural help is an attractive novel approach to increase the efficacy of prophylactic HIV Env-based vaccines and may also be applicable to other particulate vaccines.
Collapse
Affiliation(s)
| | - Thomas Niezold
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum 44801, Germany
| | - Vladimir Temchura
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum 44801, Germany; Universitätsklinikum Erlangen, Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Franco Pissani
- U.S. Military HIV Research Program, Silver Spring, MD 20910
| | - Katrin Ehrhardt
- Department of Medical Microbiology and Hygiene, Institute of Virology, University Medical Center Freiburg, Freiburg 79106, Germany
| | - Eric P Brown
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755; and
| | | | | | - Hartmut Hengel
- Department of Medical Microbiology and Hygiene, Institute of Virology, University Medical Center Freiburg, Freiburg 79106, Germany
| | | | | | - Ghulam Nabi
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum 44801, Germany
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum 44801, Germany
| | - Klaus Überla
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum 44801, Germany; Universitätsklinikum Erlangen, Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany;
| |
Collapse
|
37
|
Ohlin M, Söderberg-Nauclér C. Human antibody technology and the development of antibodies against cytomegalovirus. Mol Immunol 2015; 67:153-70. [DOI: 10.1016/j.molimm.2015.02.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 02/13/2015] [Accepted: 02/15/2015] [Indexed: 02/08/2023]
|
38
|
Corrales-Aguilar E, Hoffmann K, Hengel H. CMV-encoded Fcγ receptors: modulators at the interface of innate and adaptive immunity. Semin Immunopathol 2014; 36:627-40. [PMID: 25288477 DOI: 10.1007/s00281-014-0448-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 09/03/2014] [Indexed: 12/22/2022]
Abstract
The constant region of IgG antibodies mediates antiviral activities upon engaging host Fcγ receptors (FcγRs) expressed by a variety of immune cells, such as antibody-dependent cellullar cytotoxcity (ADCC) executed by natural killer (NK)cells. Human cytomegalovirus (HCMV) is unique among viruses by encoding also an array of several Fcγ-binding glycoproteins with cell surface disposition and concomitant incorporation into the virion. Evidence is increasing that the virus-encoded Fcγ receptors differ in their Fcγ binding mode but effectively operate as adversaries of host FcγRs since they are able to prevent IgG-mediated triggering of activating host FcγRs, i.e., FcγRI, FcγRIIA, and FcγRIIIA. Here we discuss virus-encoded FcγRs as the first known HCMV inhibitors of IgG-mediated immunity which could account for the limited efficacy of HCMV hyperimmune globulin in clinical settings. A better understanding of their molecular mode of action opens up new perspectives for improving IgG therapies against HCMV disease.
Collapse
Affiliation(s)
- Eugenia Corrales-Aguilar
- Virology-CIET, Faculty of Microbiology, University of Costa Rica, 11501-2060, San José, Costa Rica
| | | | | |
Collapse
|
39
|
Corrales-Aguilar E, Trilling M, Hunold K, Fiedler M, Le VTK, Reinhard H, Ehrhardt K, Mercé-Maldonado E, Aliyev E, Zimmermann A, Johnson DC, Hengel H. Human cytomegalovirus Fcγ binding proteins gp34 and gp68 antagonize Fcγ receptors I, II and III. PLoS Pathog 2014; 10:e1004131. [PMID: 24830376 PMCID: PMC4022731 DOI: 10.1371/journal.ppat.1004131] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 04/03/2014] [Indexed: 12/22/2022] Open
Abstract
Human cytomegalovirus (HCMV) establishes lifelong infection with recurrent episodes of virus production and shedding despite the presence of adaptive immunological memory responses including HCMV immune immunoglobulin G (IgG). Very little is known how HCMV evades from humoral and cellular IgG-dependent immune responses, the latter being executed by cells expressing surface receptors for the Fc domain of IgG (FcγRs). Remarkably, HCMV expresses the RL11-encoded gp34 and UL119-118-encoded gp68 type I transmembrane glycoproteins which bind Fcγ with nanomolar affinity. Using a newly developed FcγR activation assay, we tested if the HCMV-encoded Fcγ binding proteins (HCMV FcγRs) interfere with individual host FcγRs. In absence of gp34 or/and gp68, HCMV elicited a much stronger activation of FcγRIIIA/CD16, FcγRIIA/CD32A and FcγRI/CD64 by polyclonal HCMV-immune IgG as compared to wildtype HCMV. gp34 and gp68 co-expression culminates in the late phase of HCMV replication coinciding with the emergence of surface HCMV antigens triggering FcγRIII/CD16 responses by polyclonal HCMV-immune IgG. The gp34- and gp68-dependent inhibition of HCMV immune IgG was fully reproduced when testing the activation of primary human NK cells. Their broad antagonistic function towards FcγRIIIA, FcγRIIA and FcγRI activation was also recapitulated in a gain-of-function approach based on humanized monoclonal antibodies (trastuzumab, rituximab) and isotypes of different IgG subclasses. Surface immune-precipitation showed that both HCMV-encoded Fcγ binding proteins have the capacity to bind trastuzumab antibody-HER2 antigen complexes demonstrating simultaneous linkage of immune IgG with antigen and the HCMV inhibitors on the plasma membrane. Our studies reveal a novel strategy by which viral FcγRs can compete for immune complexes against various Fc receptors on immune cells, dampening their activation and antiviral immunity. Herpes viruses persist lifelong continuously alternating between latency and virus production and transmission. The latter events occur despite the presence of immune IgG antibodies. IgG acts by neutralization of virions and activation of immune cells bearing one or more surface receptors, called FcγRs, recognizing the constant Fc domain of IgG. Activating FcγRs induce a wide range of immune responses, including antibody dependent cellular cytotoxicity (ADCC) of virus-infected cells by natural killer (NK) cells, cytokine secretion and the uptake of immune complexes to enhance antigen presentation to T cells. We demonstrate that the HCMV glycoproteins RL11/gp34 and UL119-118/gp68 block IgG-mediated activation of FcγRs. A novel reporter cell-based assay was used to test FcγRs individually and assess their relative susceptibility to each antagonist. This approach revealed that gp34 and gp68 block triggering of activating FcγRs, i.e. FcγRI (CD64), FcγRII (CD32A) and FcγRIII (CD16). Co-immunoprecipitation showed the formation of ternary complexes containing IgG, IgG-bound antigen and the viral antagonists on the cell surface. Assigning the redundant abilities of HCMV to hinder IgG effector responses to the viral Fc binding proteins, we discuss gp34 and gp68 as potential culprits which might contribute to the limited efficacy of therapeutic IgG against HCMV.
Collapse
Affiliation(s)
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Katja Hunold
- Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Manuela Fiedler
- Institute for Virology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Vu Thuy Khanh Le
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Henrike Reinhard
- Institute for Virology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Katrin Ehrhardt
- Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Eva Mercé-Maldonado
- Institute for Virology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Enver Aliyev
- Institute for Virology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Albert Zimmermann
- Institute for Virology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - David C. Johnson
- Department of Molecular Microbiology & Immunology, Oregon Health Sciences University, Portland, Oregon, United States of America
| | - Hartmut Hengel
- Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
- * E-mail:
| |
Collapse
|
40
|
Bartholomaeus P, Semmler LY, Bukur T, Boisguerin V, Römer PS, Tabares P, Chuvpilo S, Tyrsin DY, Matskevich A, Hengel H, Castle J, Hünig T, Kalinke U. Cell Contact–Dependent Priming and Fc Interaction with CD32+ Immune Cells Contribute to the TGN1412-Triggered Cytokine Response. THE JOURNAL OF IMMUNOLOGY 2014; 192:2091-8. [DOI: 10.4049/jimmunol.1302461] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|