1
|
Neyrinck-Leglantier D, Tamagne M, Ben Rayana R, Many S, Pinheiro MK, Delorme AS, Andrieu M, Boilard E, Cognasse F, Hamzeh-Cognasse H, Perez-Patrigeon S, Lelievre JD, Pirenne F, Gallien S, Vingert B. Remodeling of immune system functions by extracellular vesicles. Front Immunol 2025; 16:1549107. [PMID: 40181981 PMCID: PMC11966064 DOI: 10.3389/fimmu.2025.1549107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/17/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The treatment of chronic viral infections can often bring viral replication under control. However, chronic immune activation persists and can lead to the development of comorbid conditions, such as cardiovascular disease and cancer. This is particularly true for people living with HIV (PLWH), who have significantly more extracellular vesicles from membrane budding, also called plasma microparticles (MPs), than healthy individuals (HDs), and a much more immunomodulatory phenotype. We hypothesized that the number and phenotypic heterogeneity of MPs can trigger a functional remodeling of immune responses in PLWH, preventing full immune restoration. Methods We investigated the rapid impact of three types of MPs - derived from membrane budding in platelets (CD41a+ PMPs), monocytes (CD14+ MMPs) and lymphocytes (CD3+ LMPs) in the plasma of PLWH or HDs-on four cell types (CD4+ and CD8+T lymphocytes, monocytes and DCs). Results These investigations of the short multiple interactions and functions of MPs with these cells revealed an increase in the secretion of cytokines such as IFNg, IL2, IL6, IL12, IL17 and TNFa by the immune cells studied following interactions with MPs. We show that this functional remodeling of immune cells depends not only on the number, but also on the phenotype of MPs. Conclusion These data suggest that the large numbers of MPs and their impact on functional remodeling in PLWH may be incompatible with the effective control of chronic infections, potentially leading to chronic immune activation and the onset of comorbid diseases.
Collapse
Affiliation(s)
- Deborah Neyrinck-Leglantier
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Marie Tamagne
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Raida Ben Rayana
- Service de Maladies Infectieuses et Immunologie Clinique, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Souganya Many
- Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique (CNRS) UMR8104, Université Paris-Cité, Paris, France
| | - Marion Klea Pinheiro
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Adèle Silane Delorme
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Muriel Andrieu
- Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique (CNRS) UMR8104, Université Paris-Cité, Paris, France
| | - Eric Boilard
- Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France
- Univ Jean Monnet, Mines Saint-Étienne, INSERM, U1059 Sainbiose, Saint-Étienne, France
| | - Hind Hamzeh-Cognasse
- Univ Jean Monnet, Mines Saint-Étienne, INSERM, U1059 Sainbiose, Saint-Étienne, France
| | | | - Jean-Daniel Lelievre
- Service de Maladies Infectieuses et Immunologie Clinique, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - France Pirenne
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Sébastien Gallien
- Service de Maladies Infectieuses et Immunologie Clinique, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Benoît Vingert
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| |
Collapse
|
2
|
Vasileiou S, Kuvalekar M, Velazquez Y, Watanabe A, Leen AM, Gilmore SA. Phenotypic and functional characterization of posoleucel, a multivirus-specific T cell therapy for the treatment and prevention of viral infections in immunocompromised patients. Cytotherapy 2024; 26:869-877. [PMID: 38597860 DOI: 10.1016/j.jcyt.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/09/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Deficits in T cell immunity translate into increased risk of severe viral infection in recipients of solid organ and hematopoietic cell transplants. Thus, therapeutic strategies that employ the adoptive transfer of virus-specific T cells are being clinically investigated to treat and prevent viral diseases in these highly immunocompromised patients. Posoleucel is an off-the-shelf multivirus-specific T cell investigational product for the treatment and prevention of infections due to adenovirus, BK virus, cytomegalovirus, Epstein-Barr virus, human herpesvirus 6 or JC virus. METHODS Herein we perform extensive characterization of the phenotype and functional profile of posoleucel to illustrate the cellular properties that may contribute to its in vivo activity. RESULTS AND CONCLUSIONS Our results demonstrate that posoleucel is enriched for central and effector memory CD4+ and CD8+ T cells with specificity for posoleucel target viruses and expressing a broad repertoire of T cell receptors. Antigen-driven upregulation of cell-surface molecules and production of cytokine and effector molecules indicative of proliferation, co-stimulation, and cytolytic potential demonstrate the specificity of posoleucel and its potential to mount a broad, polyfunctional, and effective Th1-polarized antiviral response upon viral exposure. We also show the low risk for off-target and nonspecific effects as evidenced by the enrichment of posoleucel in memory T cells, low frequency of naive T cells, and lack of demonstrated alloreactivity in vitro. The efficacy of posoleucel is being explored in four placebo-controlled clinical trials in transplant recipients to treat and prevent viral infections (NCT05179057, NCT05305040, NCT04390113, NCT04605484).
Collapse
Affiliation(s)
- Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX; AlloVir, Inc., Waltham, MA
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX; AlloVir, Inc., Waltham, MA
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX; AlloVir, Inc., Waltham, MA
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX; AlloVir, Inc., Waltham, MA
| | | |
Collapse
|
3
|
Reiter L, Greffrath J, Zidel B, Ostrowski M, Gommerman J, Madhi SA, Tran R, Martin-Orozco N, Panicker RKG, Cooper C, Pastrak A. Comparable safety and non-inferior immunogenicity of the SARS-CoV-2 mRNA vaccine candidate PTX-COVID19-B and BNT162b2 in a phase 2 randomized, observer-blinded study. Sci Rep 2024; 14:5365. [PMID: 38438427 PMCID: PMC10912344 DOI: 10.1038/s41598-024-55320-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/22/2024] [Indexed: 03/06/2024] Open
Abstract
In the aftermath of the COVID-19 pandemic, the evolution of the SARS-CoV-2 into a seasonal pathogen along with the emergence of new variants, underscores the need for dynamic and adaptable responses, emphasizing the importance of sustained vaccination strategies. This observer-blind, double-dummy, randomized immunobridging phase 2 study (NCT05175742) aimed to compare the immunogenicity induced by two doses of 40 μg PTX-COVID19-B vaccine candidate administered 28 days apart, with the response induced by two doses of 30 µg Pfizer-BioNTech COVID-19 vaccine (BNT162b2), administered 21 days apart, in Nucleocapsid-protein seronegative adults 18-64 years of age. Both vaccines were administrated via intramuscular injection in the deltoid muscle. Two weeks after the second dose, the neutralizing antibody (NAb) geometric mean titer ratio and seroconversion rate met the non-inferiority criteria, successfully achieving the primary immunogenicity endpoints of the study. PTX-COVID19-B demonstrated similar safety and tolerability profile to BNT162b2 vaccine. The lowest NAb response was observed in subjects with low-to-undetectable NAb at baseline or no reported breakthrough infection. Conversely, participants who experienced breakthrough infections during the study exhibited higher NAb titers. This study also shows induction of cell-mediated immune (CMI) responses by PTX-COVID19-B. In conclusion, the vaccine candidate PTX-COVID19-B demonstrated favourable safety profile along with immunogenicity similar to the active comparator BNT162b2 vaccine.
Collapse
Affiliation(s)
- Lawrence Reiter
- Providence Therapeutics Holdings Inc., 120-8832 Blackfoot Trail SE, Calgary, AB, T2J 3J1, Canada
| | - Johann Greffrath
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bian Zidel
- Malton Medical Center, 6870 Goreway Dr., Mississauga, ON, L4V 1P1, Canada
| | - Mario Ostrowski
- Department of Medicine, Immunology, University of Toronto, Medical Sciences Building, Rm 6271. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Jennifer Gommerman
- Department of Immunology, Temerty Faculty of Medicine, 1 King's College Circle, Rm. 7233, Toronto, ON, M5S 1A8, Canada
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Richard Tran
- Providence Therapeutics Holdings Inc., 120-8832 Blackfoot Trail SE, Calgary, AB, T2J 3J1, Canada
| | - Natalia Martin-Orozco
- Providence Therapeutics Holdings Inc., 120-8832 Blackfoot Trail SE, Calgary, AB, T2J 3J1, Canada
| | | | - Curtis Cooper
- The Ottawa Hospital Viral Hepatitis Program, Division of Infectious Diseases, Department of Medicine, The Ottawa Hospital, University of Ottawa, 75 Laurier Ave. East, Ottawa, ON, K1N 6N5, Canada
| | - Aleksandra Pastrak
- Providence Therapeutics Holdings Inc., 120-8832 Blackfoot Trail SE, Calgary, AB, T2J 3J1, Canada.
| |
Collapse
|
4
|
Puente-Massaguer E, Vasilev K, Beyer A, Loganathan M, Francis B, Scherm MJ, Arunkumar GA, González-Domínguez I, Zhu X, Wilson IA, Coughlan L, Sun W, Palese P, Krammer F. Chimeric hemagglutinin split vaccines elicit broadly cross-reactive antibodies and protection against group 2 influenza viruses in mice. SCIENCE ADVANCES 2023; 9:eadi4753. [PMID: 37703367 PMCID: PMC10499326 DOI: 10.1126/sciadv.adi4753] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/11/2023] [Indexed: 09/15/2023]
Abstract
Seasonal influenza virus vaccines are effective when they are well matched to circulating strains. Because of antigenic drift/change in the immunodominant hemagglutinin (HA) head domain, annual vaccine reformulations are necessary to maintain a match with circulating strains. In addition, seasonal vaccines provide little to no protection against newly emerging pandemic strains. Sequential vaccination with chimeric HA (cHA) constructs has been proven to direct the immune response toward the immunosubdominant but more conserved HA stalk domain. In this study, we show that immunization with group 2 cHA split vaccines in combination with the CpG 1018 adjuvant elicits broadly cross-reactive antibodies against all group 2 HAs, as well as systemic and local antigen-specific T cell responses. Antibodies elicited after sequential vaccination are directed to conserved regions of the HA such as the stalk and the trimer interface and also to the N2 neuraminidase (NA). Immunized mice were fully protected from challenge with a broad panel of influenza A viruses.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kirill Vasilev
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Annika Beyer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Benjamin Francis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael J. Scherm
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | | | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
5
|
Nemirov K, Authié P, Souque P, Moncoq F, Noirat A, Blanc C, Bourgine M, Majlessi L, Charneau P. Preclinical proof of concept of a tetravalent lentiviral T-cell vaccine against dengue viruses. Front Immunol 2023; 14:1208041. [PMID: 37654495 PMCID: PMC10466046 DOI: 10.3389/fimmu.2023.1208041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/17/2023] [Indexed: 09/02/2023] Open
Abstract
Dengue virus (DENV) is responsible for approximately 100 million cases of dengue fever annually, including severe forms such as hemorrhagic dengue and dengue shock syndrome. Despite intensive vaccine research and development spanning several decades, a universally accepted and approved vaccine against dengue fever has not yet been developed. The major challenge associated with the development of such a vaccine is that it should induce simultaneous and equal protection against the four DENV serotypes, because past infection with one serotype may greatly increase the severity of secondary infection with a distinct serotype, a phenomenon known as antibody-dependent enhancement (ADE). Using a lentiviral vector platform that is particularly suitable for the induction of cellular immune responses, we designed a tetravalent T-cell vaccine candidate against DENV ("LV-DEN"). This vaccine candidate has a strong CD8+ T-cell immunogenicity against the targeted non-structural DENV proteins, without inducing antibody response against surface antigens. Evaluation of its protective potential in the preclinical flavivirus infection model, i.e., mice knockout for the receptor to the type I IFN, demonstrated its significant protective effect against four distinct DENV serotypes, based on reduced weight loss, viremia, and viral loads in peripheral organs of the challenged mice. These results provide proof of concept for the use of lentiviral vectors for the development of efficient polyvalent T-cell vaccine candidates against all DENV serotypes.
Collapse
Affiliation(s)
- Kirill Nemirov
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Papayanni PG, Koukoulias K, Kuvalekar M, Watanabe A, Velazquez Y, Ramos CA, Leen AM, Vasileiou S. T cell immune profiling of respiratory syncytial virus for the development of a targeted immunotherapy. Br J Haematol 2023; 202:874-878. [PMID: 37323051 DOI: 10.1111/bjh.18933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/17/2023]
Abstract
Respiratory syncytial virus (RSV)-associated viral infections are a major public health problem affecting the immunologically naïve/compromised populations. Given the RSV-associated morbidity and the limited treatment options, we sought to characterize the cellular immune response to RSV to develop a targeted T cell therapy for off-the-shelf administration to immunocompromised individuals. Here we report on the immunological profiling, as well as manufacturing, characterization and antiviral properties of these RSV-targeted T cells. A randomized, phase 1/2 clinical trial evaluating their safety and activity in haematopoietic stem cell transplant recipients as an off-the-shelf multi-respiratory virus-directed product is currently underway (NCT04933968, https://clinicaltrials.gov).
Collapse
Affiliation(s)
- Penelope Georgia Papayanni
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - Kiriakos Koukoulias
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - Carlos A Ramos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
7
|
Vasileiou S, Hill L, Kuvalekar M, Workineh AG, Watanabe A, Velazquez Y, Lulla S, Mooney K, Lapteva N, Grilley BJ, Heslop HE, Rooney CM, Brenner MK, Eagar TN, Carrum G, Grimes KA, Leen AM, Lulla P. Allogeneic, off-the-shelf, SARS-CoV-2-specific T cells (ALVR109) for the treatment of COVID-19 in high-risk patients. Haematologica 2023; 108:1840-1850. [PMID: 36373249 PMCID: PMC10316279 DOI: 10.3324/haematol.2022.281946] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/31/2022] [Indexed: 07/22/2023] Open
Abstract
Defects in T-cell immunity to SARS-CoV-2 have been linked to an increased risk of severe COVID-19 (even after vaccination), persistent viral shedding and the emergence of more virulent viral variants. To address this T-cell deficit, we sought to prepare and cryopreserve banks of virus-specific T cells, which would be available as a partially HLA-matched, off-the-shelf product for immediate therapeutic use. By interrogating the peripheral blood of healthy convalescent donors, we identified immunodominant and protective T-cell target antigens, and generated and characterized polyclonal virus-specific T-cell lines with activity against multiple clinically important SARS-CoV-2 variants (including 'delta' and 'omicron'). The feasibility of making and safely utilizing such virus-specific T cells clinically was assessed by administering partially HLA-matched, third-party, cryopreserved SARS-CoV-2-specific T cells (ALVR109) in combination with other antiviral agents to four individuals who were hospitalized with COVID-19. This study establishes the feasibility of preparing and delivering off-the-shelf, SARS-CoV-2-directed, virus-specific T cells to patients with COVID-19 and supports the clinical use of these products outside of the profoundly immune compromised setting (ClinicalTrials.gov number, NCT04401410).
Collapse
Affiliation(s)
- Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX.
| | - LaQuisa Hill
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Aster G Workineh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Suhasini Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Kimberly Mooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Bambi J Grilley
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Todd N Eagar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - George Carrum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Kevin A Grimes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| |
Collapse
|
8
|
Employing T-Cell Memory to Effectively Target SARS-CoV-2. Pathogens 2023; 12:pathogens12020301. [PMID: 36839573 PMCID: PMC9967959 DOI: 10.3390/pathogens12020301] [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: 01/04/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
Well-trained T-cell immunity is needed for early viral containment, especially with the help of an ideal vaccine. Although most severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected convalescent cases have recovered with the generation of virus-specific memory T cells, some cases have encountered T-cell abnormalities. The emergence of several mutant strains has even threatened the effectiveness of the T-cell immunity that was established with the first-generation vaccines. Currently, the development of next-generation vaccines involves trying several approaches to educate T-cell memory to trigger a broad and fast response that targets several viral proteins. As the shaping of T-cell immunity in its fast and efficient form becomes important, this review discusses several interesting vaccine approaches to effectively employ T-cell memory for efficient viral containment. In addition, some essential facts and future possible consequences of using current vaccines are also highlighted.
Collapse
|
9
|
Smith‐Norowitz TA, Shidid S, Norowitz YM, Kohlhoff S. CD4<sup>+</sup> T effector memory cell responses in Chlamydia pneumoniae-stimulated peripheral blood mononuclear cells in nonasthmatic subjects. Immun Inflamm Dis 2022; 10:e726. [PMID: 36301027 PMCID: PMC9609443 DOI: 10.1002/iid3.726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
Chlamydia pneumoniae (C. pneumoniae) is a gram-negative intracellular bacterium that causes respiratory infection in humans, including subjects with or without asthma. C. pneumoniae activates cells (e.g., monocytes/macrophages) in vitro, and produces cytokines that may contribute to inflammatory responses observed in asthma. Immunological differences exist between subjects with or without asthma, with regard to host responses to C. pneumoniae. The heterogeneity and subsequent diverse pathophysiology of asthma can be better understood by analyzing the repertoire of T-cell subpopulations; the most common distinction between different asthma endotypes includes cytokines produced by CD4<sup>+</sup> cells (T helper (Th)2 high vs. Th2 low).
Collapse
Affiliation(s)
- Tamar A. Smith‐Norowitz
- Department of Pediatrics, Division of Infectious DiseasesState University of New York Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Sarah Shidid
- Department of Pediatrics, Division of Infectious DiseasesState University of New York Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Yitzchok M. Norowitz
- Department of Pediatrics, Division of Infectious DiseasesState University of New York Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Stephan Kohlhoff
- Department of Pediatrics, Division of Infectious DiseasesState University of New York Downstate Health Sciences UniversityBrooklynNew YorkUSA
| |
Collapse
|
10
|
A role for CD4 + helper cells in HIV control and progression. AIDS 2022; 36:1501-1510. [PMID: 35730394 DOI: 10.1097/qad.0000000000003296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE It remains unclear why HIV persists in most untreated individuals, and why a small minority of individuals can control the virus, either spontaneously or after an early treatment. Striking differences have been discovered between patient cohorts in CD4 + T-cell avidity but not in CD8 + T-cell avidity. The present work has the aim to explain the diverse outcome of infection and identify the key virological and immunological parameters predicting the outcome. DESIGN AND METHOD A mathematical model informed by these experiments and taking into account the details of HIV virology is developed. RESULTS The model predicts an arms race between viral dissemination and the proliferation of HIV-specific CD4 + helper cells leading to one of two states: a low-viremia state (controller) or a high-viremia state (progressor). Helper CD4 + cells with a higher avidity favor virus control. The parameter segregating spontaneous and posttreatment controllers is the infectivity difference between activated and resting CD4 + T cells. The model is shown to have a better connection to experiment than a previous model based on T-cell 'exhaustion'. CONCLUSION Using the model informed by patient data, the timing of antiretroviral therapy can be optimized.
Collapse
|
11
|
Exercise in Cold Weather for COVID-19-Recovered Individuals (CRI). Asian J Sports Med 2022. [DOI: 10.5812/asjsm-127151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The sedentary lifestyle caused by the COVID-19 quarantine has resulted in a devastating threat to human health due to stress and anxiety. Although infected individuals must stop exercising, exercise is not prohibited when without symptoms and complications. Whereas exercise can be effective in immune system reinforcement during the prevention, recovery, and post-recovery stages, COVID-19-recovered Individuals (CRI) must exercise under accurate considerations. Objectives: This study aimed to study exercise in cold weather for the CRI. Methods: This article overviews how different exercises affect the immune system. PubMed, Science Direct, and Google Scholar Databases and keywords including cold weather, COVID-19, immune system, and combined exercise were used to access scientific articles. Results: Recent reports show that different sports and exercises significantly improve COVID-19 symptoms, although there are many discrepancies among researchers in prescribing exercise programs (various training protocols, duration, and intensity). Also, CRI should avoid exercise in cold weather due to breathing complications Conclusions: Based on the present study, regular exercises (aerobic, resistance, and combined) with moderate intensity improve COVID-19 symptoms and the immune system.
Collapse
|
12
|
Perdiguero B, Asbach B, Gómez CE, Köstler J, Barnett SW, Koutsoukos M, Weiss DE, Cristillo AD, Foulds KE, Roederer M, Montefiori DC, Yates NL, Ferrari G, Shen X, Sawant S, Tomaras GD, Sato A, Fulp WJ, Gottardo R, Ding S, Heeney JL, Pantaleo G, Esteban M, Wagner R. Early and Long-Term HIV-1 Immunogenicity Induced in Macaques by the Combined Administration of DNA, NYVAC and Env Protein-Based Vaccine Candidates: The AUP512 Study. Front Immunol 2022; 13:939627. [PMID: 35935978 PMCID: PMC9354927 DOI: 10.3389/fimmu.2022.939627] [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: 05/09/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
To control HIV infection there is a need for vaccines to induce broad, potent and long-term B and T cell immune responses. With the objective to accelerate and maintain the induction of substantial levels of HIV-1 Env-specific antibodies and, at the same time, to enhance balanced CD4 and CD8 T cell responses, we evaluated the effect of concurrent administration of MF59-adjuvanted Env protein together with DNA or NYVAC vectors at priming to establish if early administration of Env leads to early induction of antibody responses. The primary goal was to assess the immunogenicity endpoint at week 26. Secondary endpoints were (i) to determine the quality of responses with regard to RV144 correlates of protection and (ii) to explore a potential impact of two late boosts. In this study, five different prime/boost vaccination regimens were tested in rhesus macaques. Animals received priming immunizations with either NYVAC or DNA alone or in combination with Env protein, followed by NYVAC + protein or DNA + protein boosts. All regimens induced broad, polyfunctional and well-balanced CD4 and CD8 T cell responses, with DNA-primed regimens eliciting higher response rates and magnitudes than NYVAC-primed regimens. Very high plasma binding IgG titers including V1/V2 specific antibodies, modest antibody-dependent cellular cytotoxicity (ADCC) and moderate neutralization activity were observed. Of note, early administration of the MF59-adjuvanted Env protein in parallel with DNA priming leads to more rapid elicitation of humoral responses, without negatively affecting the cellular responses, while responses were rapidly boosted after repeated immunizations, indicating the induction of a robust memory response. In conclusion, our findings support the use of the Env protein component during priming in the context of an heterologous immunization regimen with a DNA and/or NYVAC vector as an optimized immunization protocol against HIV infection.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII ), Madrid, Spain
| | - Benedikt Asbach
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Carmen E. Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII ), Madrid, Spain
| | - Josef Köstler
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | | | - Marguerite Koutsoukos
- Department of Product Development, GlaxoSmithKline (GSK) Vaccines, Rixensart, Belgium
| | - Deborah E. Weiss
- Department of Immunobiology, Advanced BioScience Laboratories (ABL) Inc., Rockville, MD, United States
| | - Anthony D. Cristillo
- Department of Immunobiology, Advanced BioScience Laboratories (ABL) Inc., Rockville, MD, United States
| | - Kathryn E. Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - David C. Montefiori
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Nicole L. Yates
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Guido Ferrari
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Xiaoying Shen
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Sheetal Sawant
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Alicia Sato
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - William J. Fulp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Biomedical Data Sciences, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Translational Data Science, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Song Ding
- EuroVacc Foundation EuroVacc Programme Coordinator, Lausanne, Switzerland
| | - Jonathan L. Heeney
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII ), Madrid, Spain
- *Correspondence: Mariano Esteban, ; Ralf Wagner,
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
- *Correspondence: Mariano Esteban, ; Ralf Wagner,
| |
Collapse
|
13
|
Caputo MB, Elias J, Cesar G, Alvarez MG, Laucella SA, Albareda MC. Role of the Complement System in the Modulation of T-Cell Responses in Chronic Chagas Disease. Front Cell Infect Microbiol 2022; 12:910854. [PMID: 35846776 PMCID: PMC9282465 DOI: 10.3389/fcimb.2022.910854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/26/2022] [Indexed: 01/19/2023] Open
Abstract
Chagas disease, caused by the intracellular pathogen Trypanosoma cruzi, is the parasitic disease with the greatest impact in Latin America and the most common cause of infectious myocarditis in the world. The immune system plays a central role in the control of T. cruzi infection but at the same time needs to be controlled to prevent the development of pathology in the host. It has been shown that persistent infection with T. cruzi induces exhaustion of parasite-specific T cell responses in subjects with chronic Chagas disease. The continuous inflammatory reaction due to parasite persistence in the heart also leads to necrosis and fibrosis. The complement system is a key element of the innate immune system, but recent findings have also shown that the interaction between its components and immune cell receptors might modulate several functions of the adaptive immune system. Moreover, the findings that most of immune cells can produce complement proteins and express their receptors have led to the notion that the complement system also has non canonical functions in the T cell. During human infection by T. cruzi, complement activation might play a dual role in the acute and chronic phases of Chagas disease; it is initially crucial in controlling parasitemia and might later contributes to the development of symptomatic forms of Chagas disease due to its role in T-cell regulation. Herein, we will discuss the putative role of effector complement molecules on T-cell immune exhaustion during chronic human T. cruzi infection.
Collapse
Affiliation(s)
- María Belén Caputo
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Josefina Elias
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Gonzalo Cesar
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - María Gabriela Alvarez
- Chagas Section, Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - Susana Adriana Laucella
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
- Chagas Section, Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - María Cecilia Albareda
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| |
Collapse
|
14
|
Kroemer M, Boullerot L, Ramseyer M, Spehner L, Barisien C, Gravelin E, Renaudin A, Cognasse F, Gallian P, Hermine O, Lacombe K, Tiberghien P, Adotévi O. The Quality of Anti-SARS-CoV-2 T Cell Responses Predicts the Neutralizing Antibody Titer in Convalescent Plasma Donors. Front Public Health 2022; 10:816848. [PMID: 35372242 PMCID: PMC8965758 DOI: 10.3389/fpubh.2022.816848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022] Open
Abstract
Convalescent plasma therapy has been described as an attractive approach to treat critically ill patients with COVID-19 (Coronavirus disease 2019). The selection of convalescent plasma donors (CPD) is commonly based on neutralizing antibody titer. A better understanding of the quality of immune responses following COVID-19 will enable the optimization of convalescent donors' selection in convalescent plasma programs. The involvement of SARS-CoV-2 specific T cells in the induction and persistence of high affinity anti-SARS-CoV-2 neutralizing antibody is still poorly investigated. In this study, 115 CPD who presented SARS-CoV-2 and who were eligible for plasma donation were included. Comprehensive analysis of T cells together with humoral responses were performed in regards of sex, age and blood group type. High frequency of T cell responses against SARS-CoV-2 related protein such as spike glycoprotein (80.0%), nucleocapsid (NCAP) (70.4%) and membrane protein (VME1) (74.8%) were detected in CPD by ex vivo IFN-γ and TNF-α ELISpot assays. Among CPD responders, most exhibited poly-specific T cell responses (75%) defined by the ability to mount responses against at least two SARS-CoV-2 antigens. We found a positive correlation between the magnitude and the poly-specificity of anti-SARS-CoV-2 T cell responses in CPD. Notably, both the magnitude and poly-specificity of SARS-CoV-2 T cell responses were highly correlated with neutralizing antibody titer in CPD. The present study highlights that the poly-specificity and strength of SARS-CoV-2 specific T cell responses predicts neutralizing antibody titer following COVID-19. These observations show the interest to combine T cell assays and antibody titer for the selection of CPD and to a latter extend to assess COVID-19 vaccine efficacy in at-risk patients.
Collapse
Affiliation(s)
- Marie Kroemer
- INSERM, EFS BFC, UMR1098, RIGHT, University of Burgundy Franche-Comte, Besançon, France
- Department of Pharmacy, University Hospital of Besançon, Besançon, France
| | - Laura Boullerot
- INSERM, EFS BFC, UMR1098, RIGHT, University of Burgundy Franche-Comte, Besançon, France
- INSERM CIC1431, Clinical Investigation Center in Biotherapy, Biomonitoring Plateform, Besançon, France
| | - Mélanie Ramseyer
- INSERM CIC1431, Clinical Investigation Center in Biotherapy, Biomonitoring Plateform, Besançon, France
| | - Laurie Spehner
- INSERM, EFS BFC, UMR1098, RIGHT, University of Burgundy Franche-Comte, Besançon, France
- Department of Medical Oncology, Biotechnology and Immune-Oncology Platforme, University Hospital of Besançon, Besançon, France
| | | | - Eleonore Gravelin
- INSERM CIC1431, Clinical Investigation Center in Biotherapy, Biomonitoring Plateform, Besançon, France
| | - Adeline Renaudin
- INSERM CIC1431, Clinical Investigation Center in Biotherapy, Biomonitoring Plateform, Besançon, France
| | - Fabrice Cognasse
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Pierre Gallian
- Établissement Français du Sang, La Plaine St-Denis, France
- UMR “Unité des Virus Emergents”, Aix-Marseille Université - IRD 190 - INSERM 1207 - IRBA - EFS - IHU Méditerranée Infection, Marseille, France
| | - Olivier Hermine
- Department of Hematology, Necker Hospital, Paris, France
- Institut Imagine, INSERM UMR1183, Paris University, Paris, France
| | - Karine Lacombe
- Infectious Diseases Department, Saint-Antoine Hospital, AP-HP, Paris, France
- INSERM IPLESP, AP-HP, Sorbonne University, Paris, France
| | - Pierre Tiberghien
- INSERM, EFS BFC, UMR1098, RIGHT, University of Burgundy Franche-Comte, Besançon, France
- Établissement Français du Sang, La Plaine St-Denis, France
| | - Olivier Adotévi
- INSERM, EFS BFC, UMR1098, RIGHT, University of Burgundy Franche-Comte, Besançon, France
- INSERM CIC1431, Clinical Investigation Center in Biotherapy, Biomonitoring Plateform, Besançon, France
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- *Correspondence: Olivier Adotévi
| |
Collapse
|
15
|
Preservation of lymphocyte functional fitness in perinatally-infected and treated HIV+ pediatric patients displaying sub-optimal viral control. COMMUNICATIONS MEDICINE 2022; 2. [PMID: 35434722 PMCID: PMC9012494 DOI: 10.1038/s43856-022-00085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Host–pathogen dynamics associated with HIV infection are quite distinct in children versus adults. We interrogated the functional fitness of the lymphocyte responses in two cohorts of perinatally infected HIV+ pediatric subjects with early anti-retroviral therapy (ART) initiation but divergent patterns of virologic control. We hypothesized that sub-optimal viral control would compromise immune functional fitness.
Methods
The immune responses in the two HIV+ cohorts (n = 6 in each cohort) were benchmarked against the responses measured in age-range matched, uninfected healthy control subjects (n = 11) by utilizing tests for normality, and comparison [the Kruskal–Wallis test, and the two-tailed Mann–Whitney U test (where appropriate)]. Lymphocyte responses were examined by intra-cellular cytokine secretion, degranulation assays as well as phosflow. A subset of these data were further queried by an automated clustering algorithm. Finally, we evaluated the humoral immune responses to four childhood vaccines in all three cohorts.
Results
We demonstrate that contrary to expectations pediatric HIV+ patients with sub-optimal viral control display no significant deficits in immune functional fitness. In fact, the patients that display better virologic control lack functional Gag-specific T cell responses and compared to healthy controls they display signaling deficits and an enrichment of mitogen-stimulated CD3 negative and positive lymphocyte clusters with suppressed cytokine production.
Conclusions
These results highlight the immune resilience in HIV+ children on ART with sub-optimal viral control. With respect to HIV+ children on ART with better viral control, our data suggest that this cohort might potentially benefit from targeted interventions that might mitigate cell-mediated immune functional quiescence.
Collapse
|
16
|
Mangare C, Tischer-Zimmermann S, Bonifacius A, Riese SB, Dragon AC, Blasczyk R, Maecker-Kolhoff B, Eiz-Vesper B. Variances in Antiviral Memory T-Cell Repertoire of CD45RA- and CD62L-Depleted Lymphocyte Products Reflect the Need of Individual T-Cell Selection Strategies to Reduce the Risk of GvHD while Preserving Antiviral Immunity in Adoptive T-Cell Therapy. Transfus Med Hemother 2022; 49:30-43. [PMID: 35221866 PMCID: PMC8832244 DOI: 10.1159/000516284] [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: 09/10/2020] [Accepted: 04/01/2021] [Indexed: 04/03/2025] Open
Abstract
INTRODUCTION Viral infections and reactivations still remain a cause of morbidity and mortality after hematopoietic stem cell transplantation due to immunodeficiency and immunosuppression. Transfer of unmanipulated donor-derived lymphocytes (DLI) represents a promising strategy for improving cellular immunity but carries the risk of graft versus host disease (GvHD). Depleting alloreactive naïve T cells (TN) from DLIs was implemented to reduce the risk of GvHD induction while preserving antiviral memory T-cell activity. Here, we compared two TN depletion strategies via CD45RA and CD62L expression and investigated the presence of antiviral memory T cells against human adenovirus (AdV) and Epstein-Barr virus (EBV) in the depleted fractions in relation to their functional and immunophenotypic characteristics. METHODS T-cell responses against ppEBV_EBNA1, ppEBV_Consensus and ppAdV_Hexon within TN-depleted (CD45RA-/CD62L-) and TN-enriched (CD45RA+/CD62L+) fractions were quantified by interferon-gamma (IFN-γ) ELISpot assay after short- and long-term in vitro stimulation. T-cell frequencies and immunophenotypic composition were assessed in all fractions by flow cytometry. Moreover, alloimmune T-cell responses were evaluated by mixed lymphocyte reaction. RESULTS According to differences in the phenotype composition, antigen-specific T-cell responses in CD45RA- fraction were up to 2 times higher than those in the CD62L- fraction, with the highest increase (up to 4-fold) observed after 7 days for ppEBV_EBNA1-specific T cells. The CD4+ effector memory T cells (TEM) were mainly responsible for EBV_EBNA1- and AdV_Hexon-specific T-cell responses, whereas the main functionally active T cells against ppEBV_Consensus were CD8+ central memory T cells (TCM) and TEM. Moreover, comparison of both depletion strategies indicated that alloreactivity in CD45RA- was lower than that in CD62L- fraction. CONCLUSION Taken together, our results indicate that CD45RA depletion is a more suitable strategy for generating TN-depleted products consisting of memory T cells against ppEBV_EBNA1 and ppAdV_Hexon than CD62L in terms of depletion effectiveness, T-cell functionality and alloreactivity. To maximally exploit the beneficial effects mediated by antiviral memory T cells in TN-depleted products, depletion methods should be selected individually according to phenotype composition and CD4/CD8 antigen restriction. TN-depleted DLIs may improve the clinical outcome in terms of infections, GvHD, and disease relapse if selection of pathogen-specific donor T cells is not available.
Collapse
Affiliation(s)
- Caroline Mangare
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sebastian B. Riese
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Anna Christina Dragon
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Zheng R, Li Y, Chen D, Su J, Han N, Chen H, Ning Z, Xiao M, Zhao M, Zhu B. Changes of Host Immunity Mediated by IFN-γ + CD8 + T Cells in Children with Adenovirus Pneumonia in Different Severity of Illness. Viruses 2021; 13:v13122384. [PMID: 34960654 PMCID: PMC8708941 DOI: 10.3390/v13122384] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 01/14/2023] Open
Abstract
The host immunity of patients with adenovirus pneumonia in different severity of illness is unclear. This study compared the routine laboratory tests and the host immunity of human adenovirus (HAdV) patients with different severity of illness. A co-cultured cell model in vitro was established to verify the T cell response in vitro. Among 140 patients with confirmed HAdV of varying severity, the number of lymphocytes in the severe patients was significantly reduced to 1.91 × 109/L compared with the healthy control (3.92 × 109/L) and the mild patients (4.27 × 109/L). The levels of IL-6, IL-10, and IFN-γ in patients with adenovirus pneumonia were significantly elevated with the severity of the disease. Compared with the healthy control (20.82%) and the stable patients (33.96%), the percentage of CD8+ T cells that produced IFN-γ increased to 56.27% in the progressing patients. Adenovirus infection increased the percentage of CD8+ T and CD4+ T cells that produce IFN-γ in the co-culture system. The hyperfunction of IFN-γ+ CD8+ T cells might be related to the severity of adenovirus infection. The in vitro co-culture cell model could also provide a usable cellular model for subsequent experiments.
Collapse
MESH Headings
- Adenovirus Infections, Human/genetics
- Adenovirus Infections, Human/immunology
- Adenovirus Infections, Human/pathology
- Adenovirus Infections, Human/virology
- Adenoviruses, Human/genetics
- Adenoviruses, Human/physiology
- CD8-Positive T-Lymphocytes/microbiology
- Child
- Child, Preschool
- Female
- Humans
- Infant
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interleukin-10/genetics
- Interleukin-10/immunology
- Interleukin-6/genetics
- Interleukin-6/immunology
- Lymphocyte Count
- Male
- Patient Acuity
- Pneumonia, Viral/genetics
- Pneumonia, Viral/immunology
- Pneumonia, Viral/pathology
- Pneumonia, Viral/virology
Collapse
|
18
|
|
19
|
Zykova AA, Blokhina EA, Stepanova LA, Shuklina MA, Tsybalova LM, Kuprianov VV, Ravin NV. Nanoparticles based on artificial self-assembling peptide and displaying M2e peptide and stalk HA epitopes of influenza A virus induce potent humoral and T-cell responses and protect against the viral infection. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 39:102463. [PMID: 34583058 DOI: 10.1016/j.nano.2021.102463] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
The extracellular domain of the M2 protein (M2e) and conserved region of the second subunit of the hemagglutinin (HA2) could be used for the development of broad-spectrum vaccines against influenza A. Here we obtained and characterized recombinant mosaic proteins containing tandem copies of M2e and HA2 fused to an artificial self-assembling peptide (SAP). The inclusion of SAP peptides in the fusion proteins enabled their self-assembly in vitro into spherical particles with a size of 30-50 nm. Intranasal immunization of mice with these particles without additional adjuvants induced strong humoral immune response against M2e and the whole virus. Particles carrying both M2e and HA2 induced antigen-specific multifunctional CD4+ effector memory T cells. Immunization provided high protection of mice against the lethal challenge with different subtypes of influenza A virus. The obtained self-assembling nanoparticles can be used to develop a universal influenza vaccine.
Collapse
Affiliation(s)
- Anna A Zykova
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Elena A Blokhina
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Liudmila A Stepanova
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, St. Petersburg, Russia
| | - Marina A Shuklina
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, St. Petersburg, Russia
| | - Liudmila M Tsybalova
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, St. Petersburg, Russia
| | - Victor V Kuprianov
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Nikolai V Ravin
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
20
|
Carre C, Wong G, Narang V, Tan C, Chong J, Chin HX, Xu W, Lu Y, Chua M, Poidinger M, Tambyah P, Nyunt M, Ng TP, Larocque D, Hessler C, Bosco N, Quemeneur L, Larbi A. Endoplasmic reticulum stress response and bile acid signatures associate with multi-strain seroresponsiveness during elderly influenza vaccination. iScience 2021; 24:102970. [PMID: 34471863 PMCID: PMC8387917 DOI: 10.1016/j.isci.2021.102970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 03/25/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022] Open
Abstract
The elderly are an important target for influenza vaccination, and the determination of factors that underlie immune responsiveness is clinically valuable. We evaluated the immune and metabolic profiles of 205 elderly Singaporeans administered with Vaxigrip. Despite high seroprotection rates, we observed heterogeneity in the response. We stratified the cohort into complete (CR) or incomplete responders (IR), where IR exhibited signs of accelerated T cell aging. We found a higher upregulation of genes associated with the B-cell endoplasmic-reticulum stress response in CR, where XBP-1 acts as a key upstream regulator. B-cells from IR were incapable of matching the level of XBP-1 upregulation observed in CR after inducing ER stress with tunicamycin in vitro. Metabolic signatures also distinguished CR and IR - as CR presented with a greater diversity of bile acids. Our findings suggest that the ER-stress pathway activation could improve influenza vaccination in the elderly.
Collapse
Affiliation(s)
| | - Glenn Wong
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Vipin Narang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Crystal Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Joni Chong
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Hui Xian Chin
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Weili Xu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Yanxia Lu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Michelle Chua
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Paul Tambyah
- Division of Infectious Diseases, National University Hospital, Singapore
| | - Ma Nyunt
- Division of Infectious Diseases, National University Hospital, Singapore
| | - Tze Pin Ng
- Division of Infectious Diseases, National University Hospital, Singapore
| | | | | | - Nabil Bosco
- Nestlé Research, Nestlé Institute of Health Sciences, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | | | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| |
Collapse
|
21
|
Gonzalez NM, Zou D, Gu A, Chen W. Schrödinger's T Cells: Molecular Insights Into Stemness and Exhaustion. Front Immunol 2021; 12:725618. [PMID: 34512656 PMCID: PMC8427607 DOI: 10.3389/fimmu.2021.725618] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/11/2021] [Indexed: 01/16/2023] Open
Abstract
T cell stemness and exhaustion coexist as two key contrasting phenomena during chronic antigen stimulation, such as infection, transplant, cancer, and autoimmunity. T cell exhaustion refers to the progressive loss of effector function caused by chronic antigen exposure. Exhausted T (TEX) cells highly express multiple inhibitory receptors and exhibit severe defects in cell proliferation and cytokine production. The term T cell stemness describes the stem cell-like behaviors of T cells, including self-renewal, multipotency, and functional persistence. It is well accepted that naïve and some memory T cell subsets have stem cell-like properties. When investigating the exhaustive differentiation of T cells in chronic infection and cancer, recent studies highlighted the stemness of "precursors of exhausted" T (TPEX) cells prior to their terminal differentiation to TEX cells. Clinically successful checkpoint blockades for cancer treatment appear to invigorate antitumor TPEX cells but not TEX cells. Here we discuss the transcriptional and epigenetic regulations of T cell stemness and exhaustion, with a focus on how systems immunology was and will be utilized to define the molecular basis underlying the transition of TPEX to TEX cells. We suggest a "stepwise model" of T cell stemness and exhaustion, in which loss of stemness and exhaustion progression are gradual multi-step processes. We provide perspectives on the research needed to define T cell stemness and exhaustion in the transplantation setting, in which allogenic T cells are also chronically exposed to alloantigens. A better understanding of T cell stemness and exhaustion will shed light on developing novel strategies for immunotherapies.
Collapse
Affiliation(s)
- Nancy M Gonzalez
- Immunobiology & Transplant Science Center, Department of Surgery, Houston Methodist Research Institute & Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States.,College of Medicine, Texas A&M Health Science Center, College Station, TX, United States
| | - Dawei Zou
- Immunobiology & Transplant Science Center, Department of Surgery, Houston Methodist Research Institute & Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Andy Gu
- Immunobiology & Transplant Science Center, Department of Surgery, Houston Methodist Research Institute & Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Wenhao Chen
- Immunobiology & Transplant Science Center, Department of Surgery, Houston Methodist Research Institute & Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States.,Department of Surgery, Weill Cornell Medicine, Cornell University, New York, NY, United States
| |
Collapse
|
22
|
Chu CF, Sabath F, Fibi-Smetana S, Sun S, Öllinger R, Noeßner E, Chao YY, Rinke L, Winheim E, Rad R, Krug AB, Taher L, Zielinski CE. Convalescent COVID-19 Patients Without Comorbidities Display Similar Immunophenotypes Over Time Despite Divergent Disease Severities. Front Immunol 2021; 12:601080. [PMID: 34867933 PMCID: PMC8634761 DOI: 10.3389/fimmu.2021.601080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 07/23/2021] [Indexed: 01/12/2023] Open
Abstract
COVID-19, the disease caused by SARS-CoV-2 infection, can assume a highly variable disease course, ranging from asymptomatic infection, which constitutes the majority of cases, to severe respiratory failure. This implies a diverse host immune response to SARS-CoV-2. However, the immunological underpinnings underlying these divergent disease courses remain elusive. We therefore set out to longitudinally characterize immune signatures of convalescent COVID-19 patients stratified according to their disease severity. Our unique convalescent COVID-19 cohort consists of 74 patients not confounded by comorbidities. This is the first study of which we are aware that excludes immune abrogations associated with non-SARS-CoV-2 related risk factors of disease severity. Patients were followed up and analyzed longitudinally (2, 4 and 6 weeks after infection) by high-dimensional flow cytometric profiling of peripheral blood mononuclear cells (PBMCs), in-depth serum analytics, and transcriptomics. Immune phenotypes were correlated to disease severity. Convalescence was overall associated with uniform immune signatures, but distinct immune signatures for mildly versus severely affected patients were detectable within a 2-week time window after infection.
Collapse
Affiliation(s)
- Chang-Feng Chu
- Institute of Virology, Technical University of Munich, Munich, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
- Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Friedrich Schiller University, Jena, Germany
| | - Florian Sabath
- Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Silvia Fibi-Smetana
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria
| | - Shan Sun
- Institute of Virology, Technical University of Munich, Munich, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Rupert Öllinger
- Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Elfriede Noeßner
- Immunoanalytics-Tissue Control of Immunocytes, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Ying-Yin Chao
- Institute of Virology, Technical University of Munich, Munich, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Linus Rinke
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Elena Winheim
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Roland Rad
- Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Anne B. Krug
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Leila Taher
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria
| | - Christina E. Zielinski
- Institute of Virology, Technical University of Munich, Munich, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
- Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
23
|
Qin S, Chen R, Jiang Y, Zhu H, Chen L, Chen Y, Shen M, Lin X. Multifunctional T cell response in active pulmonary tuberculosis patients. Int Immunopharmacol 2021; 99:107898. [PMID: 34333359 DOI: 10.1016/j.intimp.2021.107898] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Tuberculosis still threatens human health. We aimed to investigate the T cell immune status and the role of multifunctional T cells in pulmonary tuberculosis patients. METHODS Thirty active pulmonary tuberculosis (APTB) patients, 30 latent tuberculosis infection (LTBI) patients, 25 cured pulmonary tuberculosis (CPTB) patients and 25 healthy controls (HCs) enrolled in this study. Flow cytometer for detecting T cell phenotype and function. CBA Flex Set was used to measure chemokine. RESULTS Compared with HCs and LTBI patients, APTB patients had fewer CD4+ T and CD8+ T cells, but the expression of granzyme A, granzyme B and perforin on CD8+ T cells increased. Compared to LTBI and CPTB patients, Mycobacterium tuberculosis-specific CD8+ T cells in APTB patients appeared to be more differentiated CD45RA-CCR7- cells, and there were more multifunctional CD4+ T and CD8+ T cells. Importantly, the frequency of multifunctional CD4+ T cells in the pleural fluid of APTB patients was higher than that of peripheral blood. And the proportion of multifunctional CD4+ T cells expressing the migration receptor CXCR3 in the peripheral blood of APTB patients decreased, while the concentrations of its ligands, chemokine MIG, IP-10 and I-TAC increased significantly in plasma, especially in pleural fluid. CONCLUSIONS Decreased T lymphocytes in APTB patients may cause compensatory activation of CD8+ T cells. Multifunctional CD4+ T cells in peripheral blood could migrate to the lungs under the action of CXCR3 and associated chemokine. Multifunctional CD4+ T cells and Multifunctional CD8+ T cells were of great significance in monitoring disease treatment.
Collapse
Affiliation(s)
- Shuang Qin
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ruiqi Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yujie Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hengyue Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lijiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yanfan Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Mo Shen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Xiangyang Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
24
|
Rapaka RR, Cross AS, McArthur MA. Using Adjuvants to Drive T Cell Responses for Next-Generation Infectious Disease Vaccines. Vaccines (Basel) 2021; 9:vaccines9080820. [PMID: 34451945 PMCID: PMC8402546 DOI: 10.3390/vaccines9080820] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Using adjuvants to drive features of T cell responses to vaccine antigens is an important technological challenge in the design of new and improved vaccines against infections. Properties such as T helper cell function, T cell memory, and CD8+ T cell cytotoxicity may play critical roles in optimal and long-lived immunity through vaccination. Directly manipulating specific immune activation or antigen delivery pathways with adjuvants may selectively augment desired T cell responses in vaccination and may improve the effectiveness and durability of vaccine responses in humans. In this review we outline recently studied adjuvants in their potential for antigen presenting cell and T cell programming during vaccination, with an emphasis on what has been observed in studies in humans as available.
Collapse
|
25
|
Smith-Norowitz TA, Shidid S, Norowitz YM, Kohlhoff S. Chlamydia pneumoniae-Induced IFN-Gamma Responses in Peripheral Blood Mononuclear Cells Increase Numbers of CD4+ but Not CD8+ T Effector Memory Cells. J Blood Med 2021; 12:385-394. [PMID: 34104025 PMCID: PMC8178698 DOI: 10.2147/jbm.s303275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/20/2021] [Indexed: 11/23/2022] Open
Abstract
Background Chlamydia pneumoniae causes respiratory infection in adults and children. Previous studies in our laboratory identified significantly higher in vitro T lymphocyte responses to C. pneumoniae in children with asthma compared to healthy controls which may indicate the presence of T effector memory (TEM) lymphocytes. Aim In the present study, healthy subjects were screened for the presence of TEM cells and their cytokines. CCR7 negative effector TEMs may indicate persistent infection with C. pneumoniae. Methods Peripheral blood mononuclear cells (PBMC) (1×106/mL) from adult non-asthmatic subjects were infected for 1h ± C. pneumoniae TW-183 at a multiplicity of infection (MOI) = 0.1 and cultured (48 hrs). Distributions of lymphocytes (CD4+, CD8+) and TEM cells (CD4+CCR7+CD45RA+CD154+, CD8+CCR7+CD45RA+CD154+) were determined. Levels of intracellular interleukin (IL)-2, IL-4, and interferon (IFN)-gamma were measured (flow microfluorimetry); IFN-gamma was measured in supernatants (ELISA). Results C. pneumoniae infection led to a decrease in numbers of CD8+ TEM and CD8+CD154+ cells; CD4+TEM and CD4+CD154+ cells did not change. Numbers of TEM cells (CD4+IL-2+, CD8+ IL-2+) also decreased. However, number of TEM cells (CD4+IL4-+, CD8+ IL-4+) and (CD4+ IFN-gamma+, CD8+IFN-gamma+) did not change. When stratified according to IFN-gamma+ status, numbers of CD4+ IL-2+ and CD4+IL-4+ TEMs increased; CD8+IL-2+ and CD8+ IL-4+ TEMs decreased. Conclusion C. pneumoniae-induced PBMC IFN-gamma+ responses increased numbers of CD4+ IL-2+ and CD4+IL-4+ TEM cells, while CD8+IL-2+ and CD8+IL-4+ TEMs decreased. Production of IFN-gamma by C. pneumoniae infected PBMC should be further studied as a biomarker of persistent infection in humans.
Collapse
Affiliation(s)
- Tamar A Smith-Norowitz
- Department of Pediatrics, Division of Infectious Diseases, State University of New York Downstate Medical Center, Brooklyn, NY, 11203, USA
| | - Sarah Shidid
- Department of Pediatrics, Division of Infectious Diseases, State University of New York Downstate Medical Center, Brooklyn, NY, 11203, USA
| | - Yitzchok M Norowitz
- Department of Pediatrics, Division of Infectious Diseases, State University of New York Downstate Medical Center, Brooklyn, NY, 11203, USA
| | - Stephan Kohlhoff
- Department of Pediatrics, Division of Infectious Diseases, State University of New York Downstate Medical Center, Brooklyn, NY, 11203, USA
| |
Collapse
|
26
|
Mazzoni A, Maggi L, Capone M, Vanni A, Spinicci M, Salvati L, Tekle Kiros S, Semeraro R, Pengue L, Colao MG, Magi A, Rossolini GM, Liotta F, Cosmi L, Bartoloni A, Annunziato F. Heterogeneous magnitude of immunological memory to SARS-CoV-2 in recovered individuals. Clin Transl Immunology 2021; 10:e1281. [PMID: 33976879 PMCID: PMC8101693 DOI: 10.1002/cti2.1281] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/02/2021] [Accepted: 04/09/2021] [Indexed: 12/26/2022] Open
Abstract
Objective Although the adaptive immune response to SARS‐CoV‐2 has been characterised in the acute and early convalescent phase of the disease, few studies explore whether natural infection elicits long‐lasting immunological memory in recovered individuals. In this work, we aimed to assess the maintenance of immunological memory to SARS‐CoV‐2. Methods We evaluated the long‐term virus‐specific cellular and humoral immune response in the members of an Italian Serie A football team, who experienced a cluster of COVID‐19 in March 2020, which was strictly evaluated in the following months. Results Our results highlight a heterogeneous magnitude of immunological memory at 5 months after infection. Indeed, 20% of the subjects displayed a weak cellular and humoral memory to SARS‐CoV‐2, suggesting that they may be at higher risk of reinfection. In addition, a history of symptomatic COVID‐19 was associated with higher levels of SARS‐CoV‐2‐reactive CD4+ T cells and specific antibody levels than in asymptomatic individuals. Conclusion Collectively, these data demonstrate that immunity to SARS‐CoV‐2 is maintained five months postinfection even if the magnitude of response is heterogeneous among individuals. This finding suggests that some COVID‐19‐recovered subjects may benefit from vaccination.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Anna Vanni
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine University of Florence Florence Italy.,Infectious and Tropical Diseases Unit Careggi University Hospital Florence Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Seble Tekle Kiros
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Luca Pengue
- Sports Medicine Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Maria Grazia Colao
- Microbiology and Virology Unit Careggi University Hospital Florence Italy
| | - Alberto Magi
- Department of Information Engineering University of Florence Florence Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine University of Florence Florence Italy.,Microbiology and Virology Unit Careggi University Hospital Florence Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine University of Florence Florence Italy.,Immunology and Cell Therapy Unit Careggi University Hospital Florence Italy.,Flow Cytometry Diagnostic Center and Immunotherapy (CDCI) Careggi University Hospital Florence Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine University of Florence Florence Italy.,Immunology and Cell Therapy Unit Careggi University Hospital Florence Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine University of Florence Florence Italy.,Infectious and Tropical Diseases Unit Careggi University Hospital Florence Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine University of Florence Florence Italy.,Flow Cytometry Diagnostic Center and Immunotherapy (CDCI) Careggi University Hospital Florence Italy
| |
Collapse
|
27
|
Functional reconstitution of HBV-specific CD8 T cells by in vitro polyphenol treatment in chronic hepatitis B. J Hepatol 2021; 74:783-793. [PMID: 33188902 DOI: 10.1016/j.jhep.2020.10.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS In chronic HBV infection, mitochondrial functions and proteostasis are dysregulated in exhausted HBV-specific CD8 T cells. To better characterise the potential involvement of deregulated protein degradation mechanisms in T cell exhaustion, we analysed lysosome-mediated autophagy in HBV-specific CD8 T cells. Bioactive compounds able to simultaneously target both mitochondrial functions and proteostasis were tested to identify optimal combination strategies to reconstitute efficient antiviral CD8 T cell responses in patients with chronic HBV infection. METHODS Lysosome-mediated degradation pathways were analysed by flow cytometry in virus-specific CD8 T cells from patients with chronic HBV infection. Mitochondrial function, intracellular proteostasis, and cytokine production were evaluated in HBV-peptide-stimulated T cell cultures, in the presence or absence of the polyphenols resveratrol (RSV) and oleuropein (OLE) and their metabolites, either alone or in combination with other bioactive compounds. RESULTS HBV-specific CD8 T cells from patients with CHB showed impaired autophagic flux. RSV and OLE elicited a significant improvement in mitochondrial, proteostasis and antiviral functions in CD8 T cells. Cytokine production was also enhanced by synthetic metabolites, which correspond to those generated by RSV and OLE metabolism in vivo, suggesting that these polyphenols may also display an effect after transformation in vivo. Moreover, polyphenolic compounds improved the T cell revitalising effect of mitochondria-targeted antioxidants and of programmed cell death protein 1/programmed cell death ligand 1 blockade. CONCLUSIONS Simultaneously targeting multiple altered intracellular pathways with the combination of mitochondria-targeted antioxidants and natural polyphenols may represent a promising immune reconstitution strategy for the treatment of chronic HBV infection. LAY SUMMARY In chronic hepatitis B, antiviral T lymphocytes are deeply impaired, with many altered intracellular functions. In vitro exposure to polyphenols, such as resveratrol and oleuropein, can correct some of the deregulated intracellular pathways and improve antiviral T cell function. This effect can be further strengthened by the association of polyphenols with antioxidant compounds in a significant proportion of patients. Thus, the combination of antioxidants and natural polyphenols represents a promising strategy for chronic hepatitis B therapy.
Collapse
|
28
|
Lilleri D, Fornara C. Detection of the potentiality before the actuality: Measurement of T-cell proliferation before cell division occurs. Cytometry A 2021; 99:769-771. [PMID: 33538118 DOI: 10.1002/cyto.a.24312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 11/07/2022]
Affiliation(s)
- Daniele Lilleri
- Virologia Molecolare, Microbiologia e Virologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Fornara
- Virologia Molecolare, Microbiologia e Virologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
29
|
Early T cell reconstitution and cytokine profile may help to guide a personalized management of human cytomegalovirus infection after allogeneic hematopoietic stem cell transplantation. J Clin Virol 2021; 135:104734. [PMID: 33476929 DOI: 10.1016/j.jcv.2021.104734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 12/04/2020] [Accepted: 01/06/2021] [Indexed: 11/20/2022]
Abstract
Human cytomegalovirus (HCMV) infection is one of the major causes of mortality and morbidity after allo-hematopoietic stem cell transplantation (HSCT). Antiviral therapies are associated with toxicity and high economic burden. The aim of this retrospective study was to identify allo-HSCT HCMV-seropositive recipients at low risk of clinically significant HCMV infection who could avoid antiviral therapies. Sixty adult patients who underwent allo-HSCT were clustered in two groups: i) 22 (37%) spontaneously controlling HCMV reactivation (Controllers); ii) 38 (63%) developing clinically significant HCMV infection and receiving pre-emptive therapy (Non-Controllers). We analyzed several patient baseline characteristics, total/HCMV-specific CD4+ and CD8+ T-cell counts and their cytokine production (IFNγ, TNFα, IL2). Controllers presented a higher number of total/HCMV-specific CD4+ and CD8+ T-cells (P=0.001 and P=0.017 for total CD4+ and CD8+ T-cells respectively; P<0.001 for HCMV-specific T-cells) and a lower percentage of mono-functional IFNγ-producing HCMV-specific CD8+ T-cells (P=0.002). In bi-variable models, the prognostic impact of the percentage of mono-functional HCMV-specific CD8+ T-cells on treatment-free survival, adjusted for total/HCMVspecific CD4+ and CD8+ T-cells, was confirmed. An HCMV-seronegative donor was the only baseline characteristic associated with a clinically significant infection. These data, when confirmed by a larger prospective study, may provide information for guiding the personalized management of HCMV infection in allo-HSCT recipients.
Collapse
|
30
|
Mazzoni A, Maggi L, Capone M, Spinicci M, Salvati L, Colao MG, Vanni A, Kiros ST, Mencarini J, Zammarchi L, Mantengoli E, Menicacci L, Caldini E, Romagnani S, Liotta F, Morettini A, Rossolini GM, Bartoloni A, Cosmi L, Annunziato F. Cell-mediated and humoral adaptive immune responses to SARS-CoV-2 are lower in asymptomatic than symptomatic COVID-19 patients. Eur J Immunol 2020; 50:2013-2024. [PMID: 33080068 DOI: 10.1002/eji.202048915] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/23/2020] [Indexed: 11/11/2022]
Abstract
The characterization of cell-mediated and humoral adaptive immune responses to SARS-CoV-2 is fundamental to understand COVID-19 progression and the development of immunological memory to the virus. In this study, we detected T-cells reactive to SARS-CoV-2 proteins M, S, and N, as well as serum virus-specific IgM, IgA, IgG, in nearly all SARS-CoV-2 infected individuals, but not in healthy donors. Virus-reactive T cells exhibited signs of in vivo activation, as suggested by the surface expression of immune-checkpoint molecules PD1 and TIGIT. Of note, we detected antigen-specific adaptive immune response both in asymptomatic and symptomatic SARS-CoV-2 infected subjects. More importantly, symptomatic patients displayed a significantly higher magnitude of both cell-mediated and humoral adaptive immune response to the virus, as compared to asymptomatic individuals. These findings suggest that an uncontrolled adaptive immune response contribute to the development of the life-threatening inflammatory phase of the disease. Finally, this study might open the way to develop effective vaccination strategies.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Grazia Colao
- Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Anna Vanni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Seble Tekle Kiros
- Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Jessica Mencarini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Zammarchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | | | - Lorenzo Menicacci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eleonora Caldini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | | | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Flow Cytometry Diagnostic Center and Immunotherapy (CDCI), Careggi University Hospital, Florence, Italy
| |
Collapse
|
31
|
Tincati C, Cannizzo ES, Giacomelli M, Badolato R, d'Arminio Monforte A, Marchetti G. Heightened Circulating Interferon-Inducible Chemokines, and Activated Pro-Cytolytic Th1-Cell Phenotype Features Covid-19 Aggravation in the Second Week of Illness. Front Immunol 2020; 11:580987. [PMID: 33193384 PMCID: PMC7606391 DOI: 10.3389/fimmu.2020.580987] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/24/2020] [Indexed: 01/08/2023] Open
Abstract
Covid-19 features a delayed onset of critical illness occurring approximately one week from the beginning of symptoms, which corresponds to the bridging of innate and adaptive immunity. We reasoned that the immune events occurring at the turning point of disease might mark the direction toward pathogenic versus protective inflammatory responses. Subjects with either severe (s; PaO2/FiO2 ratio <200) or mild (m; PaO2/FiO2 ratio>300) Covid-19 were enrolled. A range of chemokines and cytokines as well as reactive oxygen species (ROS) were measured in plasma. Dendritic and NK cell frequency, monocyte and B-/T-cell phenotype and SARS-CoV-2-specific T-cell responses were assessed in PBMC. Twenty mCovid-19 and 20 sCovid-19 individuals were studied. sCovid-19 patients displayed higher non-classical monocytes, plasma chemokines (CXCL8, CXCL9, CXCL10), cytokines (IL-6, IL-10), and ROS versus mCovid-19. sCovid-19 also showed significantly increased activated CD38+HLA-DR+ T-lymphocyte, and granzyme-B+/perforin+ pro-cytolytic T-cells. All Covid-19 patients showed SARS-CoV-2 specific-T-cell response with a predominance of Th1 bi- or trifunctional IFN-γ/IL-2/TNF-α-expressing CD4+, while no difference according to disease severity was observed. Severe Covid-19 features heightened circulating IFN-inducible chemokines and activated pro-cytolytic Th1 cell phenotype in the second week of illness, yet SARS-CoV-2-specific responses are similar to that of mild illness. Altogether, our observations suggest Th1 polarization coupled to higher cytolytic profile in sCovid-19 as correlate of disease pathogenesis and as potential targets to be investigated in the roadmap to therapy and vaccine development.
Collapse
Affiliation(s)
- Camilla Tincati
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - E Stefania Cannizzo
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Mauro Giacomelli
- Dipartimento di Scienze Cliniche e Sperimentali, Università degli Studi di Brescia, ASST Spedali Civili and A. Nocivelli Institute of Molecular Medicine, c/o Spedali Civili, Brescia, Italy
| | - Raffaele Badolato
- Dipartimento di Scienze Cliniche e Sperimentali, Università degli Studi di Brescia, ASST Spedali Civili and A. Nocivelli Institute of Molecular Medicine, c/o Spedali Civili, Brescia, Italy
| | - Antonella d'Arminio Monforte
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Giulia Marchetti
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| |
Collapse
|
32
|
Rapaka RR, Wahid R, Fresnay S, Booth JS, Darton TC, Jones C, Waddington CS, Levine MM, Pollard AJ, Sztein MB. Human Salmonella Typhi exposure generates differential multifunctional cross-reactive T-cell memory responses against Salmonella Paratyphi and invasive nontyphoidal Salmonella. Clin Transl Immunology 2020; 9:e1178. [PMID: 33005416 PMCID: PMC7512505 DOI: 10.1002/cti2.1178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
Objective There are no vaccines for most of the major invasive Salmonella strains causing severe infection in humans. We evaluated the specificity of adaptive T memory cell responses generated after Salmonella Typhi exposure in humans against other major invasive Salmonella strains sharing capacity for dissemination. Methods T memory cells from eleven volunteers who underwent controlled oral challenge with wtS. Typhi were characterised by flow cytometry for cross‐reactive cellular cytokine/chemokine effector responses or evidence of degranulation upon stimulation with autologous B‐lymphoblastoid cells infected with either S. Typhi, Salmonella Paratyphi A (PA), S. Paratyphi B (PB) or an invasive nontyphoidal Salmonella strain of the S. Typhimurium serovar (iNTSTy). Results Blood T‐cell effector memory (TEM) responses after exposure to S. Typhi in humans evolve late, peaking weeks after infection in most volunteers. Induced multifunctional CD4+ Th1 and CD8+ TEM cells elicited after S. Typhi challenge were cross‐reactive with PA, PB and iNTSTy. The magnitude of multifunctional CD4+ TEM cell responses to S. Typhi correlated with induction of cross‐reactive multifunctional CD8+ TEM cells against PA, PB and iNTSTy. Highly multifunctional subsets and T central memory and T effector memory cells that re‐express CD45 (TEMRA) demonstrated less heterologous T‐cell cross‐reactivity, and multifunctional Th17 elicited after S. Typhi challenge was not cross‐reactive against other invasive Salmonella. Conclusion Gaps in cross‐reactive immune effector functions in human T‐cell memory compartments were highly dependent on invasive Salmonella strain, underscoring the importance of strain‐dependent vaccination in the design of T‐cell‐based vaccines for invasive Salmonella.
Collapse
Affiliation(s)
- Rekha R Rapaka
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Rezwanul Wahid
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Stephanie Fresnay
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA.,Present address: Stephanie Fresnay GlaxoSmithKline Rockville MD USA
| | - Jayaum S Booth
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Thomas C Darton
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK.,Present address: Thomas C Darton University of Sheffield Medical School Sheffield UK
| | - Claire Jones
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK
| | - Claire S Waddington
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK.,Present address: University of Cambridge Cambridge UK
| | - Myron M Levine
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Andrew J Pollard
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| |
Collapse
|
33
|
Mazzoni A, Salvati L, Maggi L, Capone M, Vanni A, Spinicci M, Mencarini J, Caporale R, Peruzzi B, Antonelli A, Trotta M, Zammarchi L, Ciani L, Gori L, Lazzeri C, Matucci A, Vultaggio A, Rossi O, Almerigogna F, Parronchi P, Fontanari P, Lavorini F, Peris A, Rossolini GM, Bartoloni A, Romagnani S, Liotta F, Annunziato F, Cosmi L. Impaired immune cell cytotoxicity in severe COVID-19 is IL-6 dependent. J Clin Invest 2020; 130:4694-4703. [PMID: 32463803 DOI: 10.1172/jci138554] [Citation(s) in RCA: 375] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/20/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUNDCoronavirus disease 19 (COVID-19) is an emerging infectious disease caused by SARS-CoV-2. Antiviral immune response is crucial to achieve pathogen clearance; however, in some patients an excessive and aberrant host immune response can lead to an acute respiratory distress syndrome. The comprehension of the mechanisms that regulate pathogen elimination, immunity, and pathology is essential to better characterize disease progression and widen the spectrum of therapeutic options.METHODSWe performed a flow cytometric characterization of immune cell subsets from 30 patients with COVID-19 and correlated these data with clinical outcomes.RESULTSPatients with COVID-19 showed decreased numbers of circulating T, B, and NK cells and exhibited a skewing of CD8+ T cells toward a terminally differentiated/senescent phenotype. In agreement, CD4+ T and CD8+ T, but also NK cells, displayed reduced antiviral cytokine production capability. Moreover, a reduced cytotoxic potential was identified in patients with COVID-19, particularly in those who required intensive care. The latter group of patients also showed increased serum IL-6 levels that inversely correlated to the frequency of granzyme A-expressing NK cells. Off-label treatment with tocilizumab restored the cytotoxic potential of NK cells.CONCLUSIONThe association between IL-6 serum levels and the impairment of cytotoxic activity suggests the possibility that targeting this cytokine may restore antiviral mechanisms.FUNDINGThis study was supported by funds from the Department of Experimental and Clinical Medicine of University of Florence (the ex-60% fund and the "Excellence Departments 2018-2022 Project") derived from Ministero dell'Istruzione, dell'Università e della Ricerca (Italy).
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Anna Vanni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Infectious and Tropical Diseases Unit
| | - Jessica Mencarini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Infectious and Tropical Diseases Unit
| | | | | | - Alberto Antonelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Lorenzo Zammarchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Infectious and Tropical Diseases Unit
| | - Luca Ciani
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Leonardo Gori
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Chiara Lazzeri
- Intensive Care Unit and Regional Extracorporeal Membrane Oxygenation (ECMO) Referral Centre
| | | | | | | | - Fabio Almerigogna
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Immunoallergology Unit
| | - Paola Parronchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Immunology and Cell Therapy Unit
| | | | - Federico Lavorini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Pneumology and Intensive Care Unit, and
| | - Adriano Peris
- Intensive Care Unit and Regional Extracorporeal Membrane Oxygenation (ECMO) Referral Centre
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Clinical Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Infectious and Tropical Diseases Unit
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Immunology and Cell Therapy Unit
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Flow Cytometry Diagnostic Center and Immunotherapy (CDCI)
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Immunology and Cell Therapy Unit
| |
Collapse
|
34
|
Healy ZR, Weinhold KJ, Murdoch DM. Transcriptional Profiling of CD8+ CMV-Specific T Cell Functional Subsets Obtained Using a Modified Method for Isolating High-Quality RNA From Fixed and Permeabilized Cells. Front Immunol 2020; 11:1859. [PMID: 32983102 PMCID: PMC7492549 DOI: 10.3389/fimmu.2020.01859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/10/2020] [Indexed: 01/04/2023] Open
Abstract
Previous studies suggest that the presence of antigen-specific polyfunctional T cells is correlated with improved pathogen clearance, disease control, and clinical outcomes; however, the molecular mechanisms responsible for the generation, function, and survival of polyfunctional T cells remain unknown. The study of polyfunctional T cells has been, in part, limited by the need for intracellular cytokine staining (ICS), necessitating fixation and cell membrane permeabilization that leads to unacceptable degradation of RNA. Adopting elements from prior research efforts, we developed and optimized a modified protocol for the isolation of high-quality RNA (i.e., RIN > 7) from primary human T cells following aldehyde-fixation, detergent-based permeabilization, intracellular cytokines staining, and sorting. Additionally, this method also demonstrated utility preserving RNA when staining for transcription factors. This modified protocol utilizes an optimized combination of an RNase inhibitor and high-salt buffer that is cost-effective while maintaining the ability to identify and resolve cell populations for sorting. Overall, this protocol resulted in minimal loss of RNA integrity, quality, and quantity during cytoplasmic staining of cytokines and subsequent flourescence-activated cell sorting. Using this technique, we obtained the transcriptional profiles of functional subsets (i.e., non-functional, monofunctional, bifunctional, polyfunctional) of CMV-specific CD8+T cells. Our analyses demonstrated that these functional subsets are molecularly distinct, and that polyfunctional T cells are uniquely enriched for transcripts involved in viral response, inflammation, cell survival, proliferation, and metabolism when compared to monofunctional cells. Polyfunctional T cells demonstrate reduced activation-induced cell death and increased proliferation after antigen re-challenge. Further in silico analysis of transcriptional data suggested a critical role for STAT5 transcriptional activity in polyfunctional cell activation. Pharmacologic inhibition of STAT5 was associated with a significant reduction in polyfunctional cell cytokine expression and proliferation, demonstrating the requirement of STAT5 activity not only for proliferation and cell survival, but also cytokine expression. Finally, we confirmed this association between CMV-specific CD8+ polyfunctionality with STAT5 signaling also exists in immunosuppressed transplant recipients using single cell transcriptomics, indicating that results from this study may translate to this vulnerable patient population. Collectively, these results shed light on the mechanisms governing polyfunctional T cell function and survival and may ultimately inform multiple areas of immunology, including but not limited to the development of new vaccines, CAR-T cell therapies, and adoptive T cell transfer.
Collapse
Affiliation(s)
- Zachary R Healy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Duke University Hospital, Durham, NC, United States
| | - Kent J Weinhold
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - David M Murdoch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Duke University Hospital, Durham, NC, United States
| |
Collapse
|
35
|
Barham MS, Abrahams DA, Khayumbi J, Ongalo J, Tonui J, Campbell A, de Kock M, Ouma SG, Odhiambo FH, Hanekom WA, Gandhi NR, Day CL. HIV Infection Is Associated With Downregulation of BTLA Expression on Mycobacterium tuberculosis-Specific CD4 T Cells in Active Tuberculosis Disease. Front Immunol 2019; 10:1983. [PMID: 31497018 PMCID: PMC6712065 DOI: 10.3389/fimmu.2019.01983] [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: 05/21/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
Nearly a quarter of the global population is infected with Mycobacterium tuberculosis (Mtb), with 10 million people developing active tuberculosis (TB) annually. Co-infection with human immunodeficiency virus (HIV) has long been recognized as a significant risk factor for progression to TB disease, yet the mechanisms whereby HIV impairs T cell-mediated control of Mtb infection remain poorly defined. We hypothesized that HIV infection may promote upregulation of inhibitory receptors on Mtb-specific CD4 T cells, a mechanism that has been associated with antigen-specific T cell dysfunction in chronic infections. Using cohorts of HIV-infected and HIV-uninfected individuals with latent Mtb infection (LTBI) and with active TB disease, we stimulated peripheral blood mononuclear cells (PBMC) for 6 hours with Mtb peptide pools and evaluated co-expression profiles of the inhibitory receptors BTLA, CTLA-4, and PD-1 on IFN-γ+/TNF-α+ Mtb-specific CD4 T cells. Mtb-specific CD4 T cells in all participant groups expressed predominately either one or no inhibitory receptors, unlike cytomegalovirus- and HIV-specific CD4 T cells circulating in the same individuals, which were predominately CTLA-4+PD-1+. There were no significant differences in inhibitory receptor expression profiles of Mtb-specific CD4 T cells between HIV-uninfected and HIV-infected individuals with LTBI. Surprisingly, BTLA expression, both alone and in combination with CTLA-4 and PD-1, was markedly downregulated on Mtb-specific CD4 T cells in HIV-infected individuals with active TB. Together, these data provide novel evidence that the majority of Mtb-specific CD4 T cells do not co-express multiple inhibitory receptors, regardless of HIV infection status; moreover, they highlight a previously unrecognized role of BTLA expression on Mtb-specific CD4 T cells that could be further explored as a potential biomarker of Mtb infection status, particularly in people living with HIV, the population at greatest risk for development of active TB disease.
Collapse
Affiliation(s)
- Morgan S Barham
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Deborah A Abrahams
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jeremiah Khayumbi
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Joshua Ongalo
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Joan Tonui
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Angela Campbell
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Marwou de Kock
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Samuel Gurrion Ouma
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | | | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Neel R Gandhi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Cheryl L Day
- Emory Vaccine Center, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
36
|
Chigbu DI, Loonawat R, Sehgal M, Patel D, Jain P. Hepatitis C Virus Infection: Host⁻Virus Interaction and Mechanisms of Viral Persistence. Cells 2019; 8:cells8040376. [PMID: 31027278 PMCID: PMC6523734 DOI: 10.3390/cells8040376] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/25/2019] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatitis C (HCV) is a major cause of liver disease, in which a third of individuals with chronic HCV infections may develop liver cirrhosis. In a chronic HCV infection, host immune factors along with the actions of HCV proteins that promote viral persistence and dysregulation of the immune system have an impact on immunopathogenesis of HCV-induced hepatitis. The genome of HCV encodes a single polyprotein, which is translated and processed into structural and nonstructural proteins. These HCV proteins are the target of the innate and adaptive immune system of the host. Retinoic acid-inducible gene-I (RIG-I)-like receptors and Toll-like receptors are the main pattern recognition receptors that recognize HCV pathogen-associated molecular patterns. This interaction results in a downstream cascade that generates antiviral cytokines including interferons. The cytolysis of HCV-infected hepatocytes is mediated by perforin and granzyme B secreted by cytotoxic T lymphocyte (CTL) and natural killer (NK) cells, whereas noncytolytic HCV clearance is mediated by interferon gamma (IFN-γ) secreted by CTL and NK cells. A host-HCV interaction determines whether the acute phase of an HCV infection will undergo complete resolution or progress to the development of viral persistence with a consequential progression to chronic HCV infection. Furthermore, these host-HCV interactions could pose a challenge to developing an HCV vaccine. This review will focus on the role of the innate and adaptive immunity in HCV infection, the failure of the immune response to clear an HCV infection, and the factors that promote viral persistence.
Collapse
Affiliation(s)
- DeGaulle I Chigbu
- Department of Microbiology and Immunology, and the Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA 19129, USA.
- Pennsylvania College of Optometry at Salus University, Elkins Park, PA 19027, USA.
| | - Ronak Loonawat
- Department of Microbiology and Immunology, and the Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA 19129, USA.
| | - Mohit Sehgal
- Immunology, Microenvironment & Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| | - Dip Patel
- Department of Microbiology and Immunology, and the Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA 19129, USA.
| | - Pooja Jain
- Department of Microbiology and Immunology, and the Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA 19129, USA.
| |
Collapse
|
37
|
Potent Anti-hepatitis C Virus (HCV) T Cell Immune Responses Induced in Mice Vaccinated with DNA-Launched RNA Replicons and Modified Vaccinia Virus Ankara-HCV. J Virol 2019; 93:JVI.00055-19. [PMID: 30674625 DOI: 10.1128/jvi.00055-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C is a liver disease caused by the hepatitis C virus (HCV) affecting 71 million people worldwide with no licensed vaccines that prevent infection. Here, we have generated four novel alphavirus-based DNA-launched self-amplifying RNA replicon (DREP) vaccines expressing either structural core-E1-E2 or nonstructural p7-NS2-NS3 HCV proteins of genotype 1a placed under the control of an alphavirus promoter, with or without an alphaviral translational enhancer (grouped as DREP-HCV or DREP-e-HCV, respectively). DREP vectors are known to induce cross-priming and further stimulation of immune responses through apoptosis, and here we demonstrate that they efficiently trigger apoptosis-related proteins in transfected cells. Immunization of mice with the DREP vaccines as the priming immunization followed by a heterologous boost with a recombinant modified vaccinia virus Ankara (MVA) vector expressing the nearly full-length genome of HCV (MVA-HCV) induced potent and long-lasting HCV-specific CD4+ and CD8+ T cell immune responses that were significantly stronger than those of a homologous MVA-HCV prime/boost immunization, with the DREP-e-HCV/MVA-HCV combination the most immunogenic regimen. HCV-specific CD4+ and CD8+ T cell responses were highly polyfunctional, had an effector memory phenotype, and were mainly directed against E1-E2 and NS2-NS3, respectively. Additionally, DREP/MVA-HCV immunization regimens induced higher antibody levels against HCV E2 protein than homologous MVA-HCV immunization. Collectively, these results provided an immunization protocol against HCV by inducing high levels of HCV-specific T cell responses as well as humoral responses. These findings reinforce the combined use of DREP-based vectors and MVA-HCV as promising prophylactic and therapeutic vaccines against HCV.IMPORTANCE HCV represents a global health problem as more than 71 million people are chronically infected worldwide. Direct-acting antiviral agents can cure HCV infection in most patients, but due to the high cost of these agents and the emergence of resistant mutants, they do not represent a feasible and affordable strategy to eradicate the virus. Therefore, a vaccine is an urgent goal that requires efforts to understand the correlates of protection for HCV clearance. Here, we describe for the first time the generation of novel vaccines against HCV based on alphavirus DNA replicons expressing HCV antigens. We demonstrate that potent T cell immune responses, as well as humoral immune responses, against HCV can be achieved in mice by using a combined heterologous prime/boost immunization protocol consisting of the administration of alphavirus replicon DNA vectors as the priming immunization followed by a boost with a recombinant modified vaccinia virus Ankara vector expressing HCV antigens.
Collapse
|
38
|
Booth JS, Patil SA, Goldberg E, Barnes RS, Greenwald BD, Sztein MB. Attenuated Oral Typhoid Vaccine Ty21a Elicits Lamina Propria and Intra-Epithelial Lymphocyte Tissue-Resident Effector Memory CD8 T Responses in the Human Terminal Ileum. Front Immunol 2019; 10:424. [PMID: 30923521 PMCID: PMC6426796 DOI: 10.3389/fimmu.2019.00424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 02/18/2019] [Indexed: 11/16/2022] Open
Abstract
Tissue-resident memory T cells (TRM) are newly defined memory T cells (TM) distinct from circulating TM subsets which have the potential to mount rapid protective immune responses at the site of infection. However, very limited information is available regarding the role and contribution of TRM in vaccine-mediated immune responses in humans at the site of infection. Here, we studied the role and contribution of tissue resident memory T cells (TRM) located in the terminal ileum (TI) (favored site of infection for S. Typhi) following oral Ty21a immunization in humans. We examined TI-lamina propria mononuclear cells (LPMC) and intra-epithelial lymphocytes (IEL) CD8+ TRM subsets obtained from healthy volunteers undergoing medically-indicated colonoscopies who were either immunized with Ty21a or unvaccinated. No significant differences in the frequencies of LPMC CD8+ TRM and CD8+CD69+CD103– T cells subsets were observed following Ty21a-immunization. However, LPMC CD8+ TRM exhibited significantly higher levels of cytokines (IFN-γ, IL-17A, and TNF-α) ex-vivo in Ty21a-vaccinated than in unvaccinated volunteers. LPMC CD8+ TRMS. Typhi-specific responses were evaluated using S. Typhi-infected targets and found to produce significantly higher levels of S. Typhi-specific IL-17A. In contrast, LPMC CD8+CD69+CD103- T cells produced significantly increased S. Typhi-specific levels of IFN-γ, IL-2, and IL-17A. Finally, we assessed CD8+ TRM in IEL and observed that the frequency of IEL CD8+ TRM is significantly lower following Ty21a immunization. However, ex-vivo IEL CD8+ TRM elicited by Ty21a immunization spontaneously produced significantly higher levels of cytokines (IFN-γ, IL-17A, IL-2, and TNF-α). This study provides the first demonstration of the effect of oral Ty21a vaccination on CD8+ TRM subsets (spontaneous and S. Typhi-specific) responses in the LPMC and IEL compartment of the human terminal ileum mucosa, contributing novel information to our understanding of the generation of mucosal immune responses following oral Ty21a-immunization.
Collapse
Affiliation(s)
- Jayaum S Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Seema A Patil
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Eric Goldberg
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Robin S Barnes
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bruce D Greenwald
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
39
|
Shannon I, White CL, Murphy A, Qiu X, Treanor JJ, Nayak JL. Differences in the influenza-specific CD4 T cell immunodominance hierarchy and functional potential between children and young adults. Sci Rep 2019; 9:791. [PMID: 30692574 PMCID: PMC6349841 DOI: 10.1038/s41598-018-37167-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/04/2018] [Indexed: 01/31/2023] Open
Abstract
Studies of the B cell repertoire suggest that early childhood influenza infections profoundly shape later reactivity by creating an “imprint” that impacts subsequent vaccine responses and may provide lasting protection against influenza strains within the same viral group. However, there is little known about how these early childhood influenza exposures shape CD4 T cell reactivity later in life. To investigate the effect of age on influenza-specific CD4 T cell specificity and functionality, reactivity in cohorts of 2 year old children and young adult subjects was compared. Intracellular cytokine staining was used to determine the viral antigen specificity and expression levels of various cytokines following stimulation of peripheral blood mononuclear cells with complete peptide pools representing the entire translated sequences of the pH1, H3, HA-B, NP, and M1 proteins. We found that the influenza protein-specific immunodominance pattern in children differs from that in young adults, with much lower reactivity to the NP internal virion protein in young children. Alterations in CD4 T cell functionality were also noted, as responding CD4 T cells from children produced less IFNγ and were less likely to express multiple cytokines. These differences in the repertoire of influenza-specific CD4 T cells available for recall on influenza challenge in early childhood could possibly contribute to early imprinting of influenza-specific immunity as well as the increased susceptibility of children to this viral infection.
Collapse
Affiliation(s)
- Ian Shannon
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, 601 Elmwood Ave, Box 690, Rochester, NY, 14642, USA
| | - Chantelle L White
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, 601 Elmwood Ave, Box 690, Rochester, NY, 14642, USA
| | - Amy Murphy
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, 601 Elmwood Ave, Box 690, Rochester, NY, 14642, USA
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 265 Crittenden Blvd, Box 630, Rochester, NY, 14642, USA
| | - John J Treanor
- Biomedical Advanced Research and Development Authority (BARDA)/HHS/ASPR, Influenza and Emerging Diseases Division 21J14, 200 C St SW, Washington, DC, 20515, USA
| | - Jennifer L Nayak
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, 601 Elmwood Ave, Box 690, Rochester, NY, 14642, USA.
| |
Collapse
|
40
|
Natale MA, César GA, Alvarez MG, Castro Eiro MD, Lococo B, Bertocchi G, Albareda MC, Laucella SA. Trypanosoma cruzi-specific IFN-γ-producing cells in chronic Chagas disease associate with a functional IL-7/IL-7R axis. PLoS Negl Trop Dis 2018; 12:e0006998. [PMID: 30517089 PMCID: PMC6281225 DOI: 10.1371/journal.pntd.0006998] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/14/2018] [Indexed: 11/19/2022] Open
Abstract
Background The severity of cardiac disease in chronic Chagas disease patients is associated with different features of T-cell exhaustion. Here, we assessed whether the ability of T cells to secrete IFN-γ in response to T. cruzi was linked to disruption in immune homeostasis and inflammation in patients with chronic Chagas disease. Methodology/Principal findings PBMCs from chronic Chagas disease patients and uninfected controls were examined for frequencies of T. cruzi-responsive IFN-γ-producing cells by ELISPOT and cellular expression and function of IL-7R using flow cytometry. Serum levels of IL-7, IL-21, IL-27, soluble IL-7R, and inflammatory cytokines were also evaluated by ELISA or CBA techniques. Patients possessing T. cruzi-specific IFN-γ-producing cells (i.e. IFN-γ producers) had higher levels of memory T cells capable of modulating the alpha chain of IL-7R and an efficient response to IL-7 compared to that in patients lacking (i.e. IFN-γ nonproducers) parasite-specific T-cell responses. IFN-γ producers also showed low levels of soluble IL-7R, high basal expression of Bcl-2 in T cells and low basal frequencies of activated CD25+ T cells. Modulation of IL-7R was inversely associated with serum IL-6 levels and positively associated with serum IL-8 levels. Circulating IL-21 and IL-27 levels were not associated with the frequency of IFN-γ producing cells but were reduced in less severe clinical forms of the disease. In vitro stimulation of PBMCs with IL-7 or IL-27 enhanced IFN-γ production in IFN-γ producers but not in IFN-γ nonproducers. Conclusions/Significance Alterations of the IL-7/IL-7R axis and in the levels of inflammatory cytokines were linked to impaired T. cruzi-specific IFN-γ production. These alterations might be responsible of the process of immune exhaustion observed in chronic Chagas disease. Mechanisms of acquired immune response against Trypanosoma cruzi antigens include both humoral and cellular components that might be critical in a chronic infection. Through a vast number of studies, several groups have postulated that, similar to other chronic infections, T-cell responses in chronic Trypanosoma cruzi infection are driven to exhaustion. Alterations in T-cell signaling pathways have emerged as one of the mechanisms of immune exhaustion. Here, we investigated whether the ability of T cells to secrete IFN-γ in response to T. cruzi was linked to the expression and function of the IL-7 receptor and the cytokines involved in regulating this axis in patients with different clinical forms of chronic Chagas disease. This study showed that the ability of T cells to secrete IFN-γ in response to T. cruzi is linked to an efficient modulation and function of IL-7R and low levels of inflammatory cytokines. Low IFN-γ-ELISPOT responses could not be reverted by in vitro treatment with IL-7. These findings contribute to our understanding of the long-term consequences of T. cruzi-infection and might be useful to delineate novel therapeutic strategies.
Collapse
Affiliation(s)
- María A. Natale
- Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Gonzalo A. César
- Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - María G. Alvarez
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | | | - Bruno Lococo
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - Graciela Bertocchi
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - María C. Albareda
- Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Susana A. Laucella
- Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
41
|
Xiao M, Chen X, He R, Ye L. Differentiation and Function of Follicular CD8 T Cells During Human Immunodeficiency Virus Infection. Front Immunol 2018; 9:1095. [PMID: 29872434 PMCID: PMC5972284 DOI: 10.3389/fimmu.2018.01095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/02/2018] [Indexed: 11/13/2022] Open
Abstract
The combination antiretroviral therapeutic (cART) regime effectively suppresses human immunodeficiency virus (HIV) replication and prevents progression to acquired immunodeficiency diseases. However, cART is not a cure, and viral rebound will occur immediately after treatment is interrupted largely due to the long-term presence of an HIV reservoir that is composed of latently infected target cells that maintain a quiescent state or persistently produce infectious viruses. CD4 T cells that reside in B-cell follicles within lymphoid tissues, called follicular helper T cells (TFH), have been identified as a major HIV reservoir. Due to their specialized anatomical structure, HIV-specific CD8 T cells are largely insulated from this TFH reservoir. It is increasingly clear that the elimination of TFH reservoirs is a key step toward a functional cure for HIV infection. Recently, several studies have suggested that a fraction of HIV-specific CD8 T cells can differentiate into a CXCR5-expressing subset, which are able to migrate into B-cell follicles and inhibit viral replication. In this review, we discuss the differentiation and functions of this newly identified CD8 T-cell subset and propose potential strategies for purging TFH HIV reservoirs by utilizing this unique population.
Collapse
Affiliation(s)
- Minglu Xiao
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Xiangyu Chen
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Ran He
- Department of Immunology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, China
| |
Collapse
|
42
|
Immunological and physical evaluation of the multistage tuberculosis subunit vaccine candidate H56/CAF01 formulated as a spray-dried powder. Vaccine 2018; 36:3331-3339. [PMID: 29699790 DOI: 10.1016/j.vaccine.2018.04.055] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/16/2018] [Accepted: 04/19/2018] [Indexed: 01/11/2023]
Abstract
Liquid vaccine dosage forms have limited stability and require refrigeration during their manufacture, distribution and storage. In contrast, solid vaccine dosage forms, produced by for example spray drying, offer improved storage stability and reduced dependence on cold-chain facilities. This is advantageous for mass immunization campaigns for global public health threats, e.g., tuberculosis (TB), and offers cheaper vaccine distribution. The multistage subunit vaccine antigen H56, which is a fusion protein of the Mycobacterium tuberculosis (Mtb) antigens Ag85B, ESAT-6, and Rv2660, has been shown to confer protective efficacy against active TB before and after Mtb exposure in preclinical models, and it is currently undergoing clinical phase 2a testing. In several studies, including a recent study comparing multiple clinically relevant vaccine adjuvants, the T helper type 1 (Th1)/Th17-inducing adjuvant CAF01 was the most efficacious adjuvant for H56 to stimulate protective immunity against Mtb. With the long-term goal of designing a thermostable and self-administrable dry powder vaccine based on H56 and CAF01 for inhalation, we compared H56 spray-dried with CAF01 with the non-spray-dried H56/CAF01 vaccine with respect to their ability to induce systemic Th1, Th17 and humoral responses after subcutaneous immunization. Here we show that spray drying of the H56/CAF01 vaccine results in preserved antigenic epitope recognition and adjuvant activity of CAF01, and the spray-dried, reconstituted vaccine induces antigen-specific Th1, Th17 and humoral immune responses, which are comparable to those stimulated by the non-spray-dried H56/CAF01 vaccine. In addition, the spray-dried and reconstituted H56/CAF01 vaccine promotes similar polyfunctional CD4+ T-cell responses as the non-spray-dried vaccine. Thus, our study provides proof-of-concept that spray drying of the subunit vaccine H56/CAF01 preserves vaccine-induced humoral and cell-mediated immune responses. These results support our ongoing efforts to develop a thermostable, dry powder-based TB vaccine.
Collapse
|
43
|
Stepanova LA, Mardanova ES, Shuklina MA, Blokhina EA, Kotlyarov RY, Potapchuk MV, Kovaleva AA, Vidyaeva IG, Korotkov AV, Eletskaya EI, Ravin NV, Tsybalova LM. Flagellin-fused protein targeting M2e and HA2 induces potent humoral and T-cell responses and protects mice against various influenza viruses a subtypes. J Biomed Sci 2018; 25:33. [PMID: 29631629 PMCID: PMC5891888 DOI: 10.1186/s12929-018-0433-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/27/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Current influenza vaccines are mainly strain-specific and have limited efficacy in preventing new, potentially pandemic, influenza strains. Efficient control of influenza A infection can potentially be achieved through the development of broad-spectrum vaccines based on conserved antigens. A current trend in the design of universal flu vaccines is the construction of recombinant proteins based on combinations of various conserved epitopes of viral proteins (M1, M2, HA2, NP). In this study, we compared the immunogenicity and protective action of two recombinant proteins which feature different designs and which target different antigens. RESULTS Balb/c mice were immunized subcutaneously with Flg-HA2-2-4M2ehs or FlgSh-HA2-2-4M2ehs; these constructs differ in the location of hemagglutinin's HA2-2(76-130) insertion into flagellin (FliC). The humoral and T-cell immune responses to these constructs were evaluated. The simultaneous expression of different M2e and HA2-2(76-130) in recombinant protein form induces a strong M2e-specific IgG response and CD4+/ CD8+ T-cell response. The insertion of HA2-2(76-130) into the hypervariable domain of flagellin greatly increases antigen-specific T-cell response, as evidenced by the formation of multi-cytokine-secreting CD4+, CD8+ T-cells, Tem, and Tcm. Both proteins provide full protection from lethal challenge with A/H3N2 and A/H7N9. CONCLUSION Our results show that highly conserved M2e and HA2-2(76-130) can be used as important targets for the development of universal flu vaccines. The location of the HA2-2(76-130) peptide's insertion into the hypervariable domain of flagellin had a significant effect on the T-cell response to influenza antigens, as seen by forming of multi-cytokine-secreting CD4+ and CD8+ T-cells.
Collapse
Affiliation(s)
- Liudmila A Stepanova
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia.
| | - Eugenia S Mardanova
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Leninsky Ave. 33, building 2, 119071, Moscow, Russia
| | - Marina A Shuklina
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Elena A Blokhina
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Leninsky Ave. 33, building 2, 119071, Moscow, Russia
| | - Roman Y Kotlyarov
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Leninsky Ave. 33, building 2, 119071, Moscow, Russia
| | - Marina V Potapchuk
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Anna A Kovaleva
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Inna G Vidyaeva
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Alexandr V Korotkov
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Elizaveta I Eletskaya
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Nikolai V Ravin
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Leninsky Ave. 33, building 2, 119071, Moscow, Russia
| | - Liudmila M Tsybalova
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| |
Collapse
|
44
|
Chiacchio T, Petruccioli E, Vanini V, Cuzzi G, La Manna MP, Orlando V, Pinnetti C, Sampaolesi A, Antinori A, Caccamo N, Goletti D. Impact of antiretroviral and tuberculosis therapies on CD4 + and CD8 + HIV/M. tuberculosis-specific T-cell in co-infected subjects. Immunol Lett 2018; 198:33-43. [PMID: 29635002 DOI: 10.1016/j.imlet.2018.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/17/2018] [Accepted: 04/04/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Human Immunodeficiency Virus (HIV) infection is a risk factor for tuberculosis (TB). Antiretroviral therapy (ART) changed HIV clinical management but it is still unclear how pre-existing HIV/Mycobacterium tuberculosis (Mtb)-specific CD4+ and CD8+ T-cells are restored. AIM to evaluate the impact of ART and TB therapies on the functional and phenotypic profile of Mtb-specific antigen-response of CD4+ and CD8+ T-cells in prospectively enrolled HIV-TB co-infected patients. METHODS ART-naïve HIV-infected patients, with or without active TB or latent TB infection (LTBI), were enrolled before and after starting ART and TB therapies. Peripheral blood mononuclear cells (PBMC) were stimulated overnight with Mtb and HIV antigens (GAG). Cytokine expression and phenotype profile were evaluated by flow cytometry. Cytomegalovirus (CMV) and staphylococcal enterotoxin B (SEB) were also used. RESULTS The median of absolute number of CD4+ T-cells increased after ART and TB therapies in all groups analyzed, while the median of absolute number of CD8+ T-cells decreases in HIV and HIV-LTBI groups. Treatments significantly increased the frequency of Mtb-specific CD4+ T-cells in the HIV-LTBI (p = 0.015) with a rise of the central memory compartment. The magnitude of the CD4+ T-cell response to HIV-GAG significantly increased in active TB (p = 0.03), whereas the magnitude of CMV-specific CD4+ T-cell response decreased in all the groups. Similarly, the treatments increased the number of Mtb-specific CD8+ responders in both HIV-LTBI and HIV-TB groups, whereas the phenotype distribution was dependent on the antigens used and on the stage of infection/disease. CONCLUSIONS After therapies the median of absolute number and the proportion of CD4+ T-cells increased in all groups whereas the median of absolute count and proportion of CD8+ T-cells decreased in the HIV and HIV-LTBI subjects. Interestingly, an increased frequency of CD4+ T-cell response to RD1 proteins in HIV-LTBI subjects was found. These results contribute to a better understanding of the effect of ART and TB therapies on the modulation of Mtb-specific CD4+ and CD8+ T-cells subsets.
Collapse
Affiliation(s)
- Teresa Chiacchio
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Via Portuense 292, 00149 Rome, Italy
| | - Elisa Petruccioli
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Via Portuense 292, 00149 Rome, Italy
| | - Valentina Vanini
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Via Portuense 292, 00149 Rome, Italy
| | - Gilda Cuzzi
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Via Portuense 292, 00149 Rome, Italy
| | - Marco Pio La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Azienda Ospedaliera Universitaria Policlinico P. Giaccone, Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Valentina Orlando
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Azienda Ospedaliera Universitaria Policlinico P. Giaccone, Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Carmela Pinnetti
- HIV/AIDS Department, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Roma, Italy
| | - Alessandro Sampaolesi
- HIV/AIDS Department, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Roma, Italy
| | - Andrea Antinori
- HIV/AIDS Department, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Roma, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Azienda Ospedaliera Universitaria Policlinico P. Giaccone, Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Via Portuense 292, 00149 Rome, Italy.
| |
Collapse
|
45
|
Lee YJ, Yu JE, Kim P, Lee JY, Cheong YC, Lee YJ, Chang J, Seong BL. Eliciting unnatural immune responses by activating cryptic epitopes in viral antigens. FASEB J 2018; 32:4658-4669. [PMID: 29570395 PMCID: PMC6103170 DOI: 10.1096/fj.201701024rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Antigenic variation in viral surface antigens is a strategy for escaping the host's adaptive immunity, whereas regions with pivotal functions for infection are less subject to antigenic variability. We hypothesized that genetically invariable and immunologically dormant regions of a viral surface antigen could be exposed to the host immune system and activated by rendering them susceptible to antigen-processing machinery in professional antigen-presenting cells (APCs). Considering the frequent antigen drift and shift in influenza viruses, we identified and used structural modeling to evaluate the conserved regions on the influenza hemagglutinin (HA) surface as potential epitopes. Mutant viruses containing the cleavage motifs of cathepsin S within HA were generated. Immunization of mice showed that the mutant, but not the wild-type virus, elicited specific antibodies against the cryptic epitope. Those antibodies were purified, and specific binding to HA was confirmed. These results suggest that an unnatural immune response can be elicited through the processing of target antigens in APCs, followed by presentation via the major histocompatibility complex, if not subjected to regulatory pathways. By harnessing the antigen-processing machinery, our study shows a proof-of-principle for designer vaccines with increased efficacy and safety by either activating cryptic, or inactivating naturally occurring, epitopes of viral antigens.-Lee, Y. J., Yu, J. E., Kim, P., Lee, J.-Y., Cheong, Y. C., Lee, Y. J., Chang, J., Seong, B. L. Eliciting unnatural immune responses by activating cryptic epitopes in viral antigens.
Collapse
Affiliation(s)
- Young Jae Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Ji Eun Yu
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Paul Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Jeong-Yoon Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Yu Cheol Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Yoon Jae Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea.,Vaccine Translational Research Center (VTRC), Yonsei University, Seoul, South Korea
| | - Baik Lin Seong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.,Vaccine Translational Research Center (VTRC), Yonsei University, Seoul, South Korea
| |
Collapse
|
46
|
Lam JKP, Hui KF, Ning RJ, Xu XQ, Chan KH, Chiang AKS. Emergence of CD4+ and CD8+ Polyfunctional T Cell Responses Against Immunodominant Lytic and Latent EBV Antigens in Children With Primary EBV Infection. Front Microbiol 2018; 9:416. [PMID: 29599759 PMCID: PMC5863510 DOI: 10.3389/fmicb.2018.00416] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/21/2018] [Indexed: 11/13/2022] Open
Abstract
Long term carriers were shown to generate robust polyfunctional T cell (PFC) responses against lytic and latent antigens of Epstein-Barr virus (EBV). However, the time of emergence of PFC responses against EBV antigens, pattern of immunodominance and difference between CD4+ and CD8+ T cell responses during various stages of EBV infection are not clearly understood. A longitudinal study was performed to assess the development of antigen-specific PFC responses in children diagnosed to have primary symptomatic (infectious mononucleosis [IM]) and asymptomatic (AS) EBV infection. Evaluation of IFN-γ secreting CD8+ T cell responses upon stimulation by HLA class I-specific peptides of EBV lytic and latent proteins by ELISPOT assay followed by assessment of CD4+ and CD8+ PFC responses upon stimulation by a panel of overlapping EBV peptides for co-expression of IFN-γ, TNF-α, IL-2, perforin and CD107a by flow cytometry were performed. Cytotoxicity of T cells against autologous lymphoblastoid cell lines (LCLs) as well as EBV loads in PBMC and plasma were also determined. Both IM and AS patients had elevated PBMC and plasma viral loads which declined steadily during a 12-month period from the time of diagnosis whilst decrease in the magnitude of CD8+ T cell responses toward EBV lytic peptides in contrast to increase toward latent peptides was shown with no significant difference between those of IM and AS patients. Both lytic and latent antigen-specific CD4+ and CD8+ T cells demonstrated polyfunctionality (defined as greater or equal to three functions) concurrent with enhanced cytotoxicity against autologous LCLs and steady decrease in plasma and PBMC viral loads over time. Immunodominant peptides derived from BZLF1, BRLF1, BMLF1 and EBNA3A-C proteins induced the highest proportion of CD8+ as well as CD4+ PFC responses. Diverse functional subtypes of both CD4+ and CD8+ PFCs were shown to emerge at 6–12 months. In conclusion, EBV antigen-specific CD4+ and CD8+ PFC responses emerge during the first year of primary EBV infection, with greatest responses toward immunodominant epitopes in both lytic and latent proteins, correlating to steady decline in PBMC and plasma viral loads.
Collapse
Affiliation(s)
- Janice K P Lam
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - K F Hui
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Raymond J Ning
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - X Q Xu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - K H Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Alan K S Chiang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
47
|
Falconer O, Newell ML, Jones CE. The Effect of Human Immunodeficiency Virus and Cytomegalovirus Infection on Infant Responses to Vaccines: A Review. Front Immunol 2018; 9:328. [PMID: 29552009 PMCID: PMC5840164 DOI: 10.3389/fimmu.2018.00328] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/06/2018] [Indexed: 12/11/2022] Open
Abstract
The success of prevention of mother to child transmission programs over the last two decades has led to an increasing number of infants who are exposed to human immunodeficiency virus (HIV), but who are not themselves infected (HIV-exposed, uninfected infants). Although the morbidity and mortality among HIV-exposed, uninfected infants is considerably lower than that among HIV-infected infants, they may remain at increased risk of infections in the first 2 years of life compared with their HIV-unexposed peers, especially in the absence of breastfeeding. There is some evidence of immunological differences in HIV-exposed, uninfected infants, which could play a role in susceptibility to infection. Cytomegalovirus (CMV) may contribute to the increased immune activation observed in HIV-exposed, uninfected infants. Infants born to HIV-infected women are at increased risk of congenital CMV infection, as well as early acquisition of postnatal CMV infection. In infants with HIV infection, CMV co-infection in early life is associated with higher morbidity and mortality. This review considers how HIV infection, HIV exposure, and CMV infection affect infant responses to vaccination, and explores possible immunological and other explanations for these findings. HIV-infected infants have lower vaccine-induced antibody concentrations following tetanus, diphtheria, pertussis, hepatitis B, and pneumococcal vaccination, although the clinical relevance of this difference is not known. Despite lower concentrations of maternal-specific antibody at birth, HIV-exposed, uninfected infants respond to vaccination at least as well as their HIV-unexposed uninfected peers. CMV infection leads to an increase in activation and differentiation of the whole T-cell population, but there is limited data on the effects of CMV infection on infant vaccine responses. In light of growing evidence of poor clinical outcomes associated with CMV infection in HIV-exposed, uninfected infants, further studies are particularly important in this group. A clearer understanding of the mechanisms by which maternal viral infections influence the developing infant immune system is critical to the success of maternal and infant vaccination strategies.
Collapse
Affiliation(s)
- Olivia Falconer
- Institute for Life Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Marie-Louise Newell
- Institute of Developmental Science, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Christine E Jones
- Institute for Life Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| |
Collapse
|
48
|
Pathakumari B, Devasundaram S, Maddineni P, Raja A. Rv2204c, Rv0753c and Rv0009 antigens specific T cell responses in latent and active TB – a flow cytometry-based analysis. Int J Med Microbiol 2018; 308:297-305. [DOI: 10.1016/j.ijmm.2017.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 10/17/2017] [Accepted: 12/04/2017] [Indexed: 12/17/2022] Open
|
49
|
Ngu LN, Nji NN, Ambada G, Ngoh AA, Njambe Priso GD, Tchadji JC, Lissom A, Magagoum SH, Sake CN, Tchouangueu TF, Chukwuma GO, Okoli AS, Sagnia B, Chukwuanukwu R, Tebit DM, Esimone CO, Waffo AB, Park CG, Überla K, Nchinda GW. Dendritic cell targeted HIV-1 gag protein vaccine provides help to a recombinant Newcastle disease virus vectored vaccine including mobilization of protective CD8 + T cells. IMMUNITY INFLAMMATION AND DISEASE 2017; 6:163-175. [PMID: 29205929 PMCID: PMC5818444 DOI: 10.1002/iid3.209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022]
Abstract
Introduction Recombinant Newcastle Disease virus (rNDV) vectored vaccines are safe mucosal applicable vaccines with intrinsic immune‐modulatory properties for the induction of efficient immunity. Like all viral vectored vaccines repeated inoculation via mucosal routes invariably results to immunity against viral vaccine vectors. To obviate immunity against viral vaccine vectors and improve the ability of rNDV vectored vaccines in inducing T cell immunity in murine air way we have directed dendritic cell targeted HIV‐1 gag protein (DEC‐Gag) vaccine; for the induction of helper CD4+ T cells to a Recombinant Newcastle disease virus expressing codon optimized HIV‐1 Gag P55 (rNDV‐L‐Gag) vaccine. Methods We do so through successive administration of anti‐DEC205‐gagP24 protein plus polyICLC (DEC‐Gag) vaccine and rNDV‐L‐Gag. First strong gag specific helper CD4+ T cells are induced in mice by selected targeting of anti‐DEC205‐gagP24 protein vaccine to dendritic cells (DC) in situ together with polyICLC as adjuvant. This targeting helped T cell immunity develop to a subsequent rNDV‐L‐Gag vaccine and improved both systemic and mucosal gag specific immunity. Results This sequential DEC‐Gag vaccine prime followed by an rNDV‐L‐gag boost results to improved viral vectored immunization in murine airway, including mobilization of protective CD8+ T cells to a pathogenic virus infection site. Conclusion Thus, complementary prime boost vaccination, in which prime and boost favor distinct types of T cell immunity, improves viral vectored immunization, including mobilization of protective CD8+T cells to a pathogenic virus infection site such as the murine airway.
Collapse
Affiliation(s)
- Loveline N Ngu
- Department of Biochemistry, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon.,Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon
| | - Nadesh N Nji
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon
| | - Georgia Ambada
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Apeh A Ngoh
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of biomedical sciences, University of Dschang, Dschang, Cameroon
| | - Ghislain D Njambe Priso
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Jules C Tchadji
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Abel Lissom
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Suzanne H Magagoum
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Carol N Sake
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Microbiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Thibau F Tchouangueu
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of biochemistry, University of Dschang, Dschang, Cameroon
| | - George O Chukwuma
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Medical Laboratory Science College of Medicine, Nnewi Campus, Nnamdi Azikiwe University, Awka, Anambra
| | | | - Bertrand Sagnia
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon
| | - Rebecca Chukwuanukwu
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Medical Laboratory Science College of Medicine, Nnewi Campus, Nnamdi Azikiwe University, Awka, Anambra
| | - Denis M Tebit
- Myles Thaler Center for AIDS and Human Retrovirus Research, Department of Microbiology, Immunology and Cancer Biology, Jordan Hall 7088, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22903, USA
| | - Charles O Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| | - Alain B Waffo
- Department of Biological Sciences # 223, Alabama State University, 1627, Hall Street, Montgomery, Alabama 36104, USA
| | - Chae G Park
- Laboratory of Immunology, Brain Korea 21 PLUS Project for Medical Science, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.,Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York 10065, USA
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Erlangen, Germany
| | - Godwin W Nchinda
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
50
|
Comte D, Karampetsou MP, Yoshida N, Kis-Toth K, Kyttaris VC, Tsokos GC. Signaling Lymphocytic Activation Molecule Family Member 7 Engagement Restores Defective Effector CD8+ T Cell Function in Systemic Lupus Erythematosus. Arthritis Rheumatol 2017; 69:1035-1044. [PMID: 28076903 DOI: 10.1002/art.40038] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/05/2017] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Effector CD8+ T cell function is impaired in systemic lupus erythematosus (SLE) and is associated with a compromised ability to fight infections. Signaling lymphocytic activation molecule family member 7 (SLAMF7) engagement has been shown to enhance natural killer cell degranulation. This study was undertaken to characterize the expression and function of SLAMF7 on CD8+ T cell subsets isolated from the peripheral blood of SLE patients and healthy subjects. METHODS CD8+ T cell subset distribution, SLAMF7 expression, and expression of cytolytic enzymes (perforin, granzyme A [GzmA], and GzmB) on cells isolated from SLE patients and healthy controls were analyzed by flow cytometry. CD107a expression and interferon-γ (IFNγ) production in response to viral antigenic stimulation in the presence or absence of an anti-SLAMF7 antibody were assessed by flow cytometry. Antiviral cytotoxic activity in response to SLAMF7 engagement was determined using a flow cytometry-based assay. RESULTS The distribution of CD8+ T cell subsets was altered in the peripheral blood of SLE patients, with a decreased effector cell subpopulation. Memory CD8+ T cells from SLE patients displayed decreased amounts of SLAMF7, a surface receptor that characterizes effector CD8+ T cells. Ligation of SLAMF7 increased CD8+ T cell degranulation capacity and the percentage of IFNγ-producing cells in response to antigen challenge in SLE patients and healthy controls. Moreover, SLAMF7 engagement promoted cytotoxic lysis of target cells in response to stimulation with viral antigens. CONCLUSION CD8+ T cell activation in response to viral antigens is defective in SLE patients. Activation of SLAMF7 through a specific monoclonal antibody restores CD8+ T cell antiviral effector function to normal levels and thus represents a potential therapeutic option in SLE.
Collapse
Affiliation(s)
- Denis Comte
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, and Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Maria P Karampetsou
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Nobuya Yoshida
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Katalin Kis-Toth
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Vasileios C Kyttaris
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|