3
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Low pre-existing endemic human coronavirus (HCoV-NL63)-specific T cell frequencies are associated with impaired SARS-CoV-2-specific T cell responses in people living with HIV

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Understanding how HIV affects SARS-CoV-2 immunity is crucial for managing COVID-19 in sub-Saharan populations due to frequent coinfections. Our previous research showed that unsuppressed HIV is associated with weaker immune responses to SARS-CoV-2, but the underlying mechanisms are unclear. We investigated how pre-existing T cell immunity against an endemic human coronavirus HCoV-NL63 impacts SARS-CoV-2 T cell responses in people living with HIV (PLWH) compared to uninfected individuals, and how HIV-related T cell dysfunction influences responses to SARS-CoV-2 variants.

          Methods

          We used flow cytometry to measure T cell responses following PBMC stimulation with peptide pools representing beta, delta, wild-type, and HCoV-NL63 spike proteins. Luminex bead assay was used to measure circulating plasma chemokine and cytokine levels. ELISA and MSD V-PLEX COVID-19 Serology and ACE2 Neutralization assays were used to measure humoral responses.

          Results

          Regardless of HIV status, we found a strong positive correlation between responses to HCoV-NL63 and SARS-CoV-2. However, PLWH exhibited weaker CD4 + T cell responses to both HCoV-NL63 and SARS-CoV-2 than HIV-uninfected individuals. PLWH also had higher proportions of functionally exhausted (PD-1high) CD4 + T cells producing fewer proinflammatory cytokines (IFNγ and TNFα) and had elevated plasma IL-2 and IL-12(p70) levels compared to HIV-uninfected individuals. HIV status didn’t significantly affect IgG antibody levels against SARS-CoV-2 antigens or ACE2 binding inhibition activity.

          Conclusion

          Our results indicate that the decrease in SARS-CoV-2 specific T cell responses in PLWH may be attributable to reduced frequencies of pre-existing cross-reactive responses. However, HIV infection minimally affected the quality and magnitude of humoral responses, and this could explain why the risk of severe COVID-19 in PLWH is highly heterogeneous.

          Related collections

          Most cited references46

          • Record: found
          • Abstract: found
          • Article: not found

          Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals

          Summary Understanding adaptive immunity to SARS-CoV-2 is important for vaccine development, interpreting coronavirus disease 2019 (COVID-19) pathogenesis, and calibration of pandemic control measures. Using HLA class I and II predicted peptide ‘megapools’, circulating SARS-CoV-2−specific CD8+ and CD4+ T cells were identified in ∼70% and 100% of COVID-19 convalescent patients, respectively. CD4+ T cell responses to spike, the main target of most vaccine efforts, were robust and correlated with the magnitude of the anti-SARS-CoV-2 IgG and IgA titers. The M, spike and N proteins each accounted for 11-27% of the total CD4+ response, with additional responses commonly targeting nsp3, nsp4, ORF3a and ORF8, among others. For CD8+ T cells, spike and M were recognized, with at least eight SARS-CoV-2 ORFs targeted. Importantly, we detected SARS-CoV-2−reactive CD4+ T cells in ∼40-60% of unexposed individuals, suggesting cross-reactive T cell recognition between circulating ‘common cold’ coronaviruses and SARS-CoV-2.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Longitudinal analyses reveal immunological misfiring in severe COVID-19

            Recent studies have provided insights into the pathogenesis of coronavirus disease 2019 (COVID-19) 1–4 . However, the longitudinal immunological correlates of disease outcome remain unclear. Here we serially analysed immune responses in 113 patients with moderate or severe COVID-19. Immune profiling revealed an overall increase in innate cell lineages, with a concomitant reduction in T cell number. An early elevation in cytokine levels was associated with worse disease outcomes. Following an early increase in cytokines, patients with moderate COVID-19 displayed a progressive reduction in type 1 (antiviral) and type 3 (antifungal) responses. By contrast, patients with severe COVID-19 maintained these elevated responses throughout the course of the disease. Moreover, severe COVID-19 was accompanied by an increase in multiple type 2 (anti-helminths) effectors, including interleukin-5 (IL-5), IL-13, immunoglobulin E and eosinophils. Unsupervised clustering analysis identified four immune signatures, representing growth factors (A), type-2/3 cytokines (B), mixed type-1/2/3 cytokines (C), and chemokines (D) that correlated with three distinct disease trajectories. The immune profiles of patients who recovered from moderate COVID-19 were enriched in tissue reparative growth factor signature A, whereas the profiles of those with who developed severe disease had elevated levels of all four signatures. Thus, we have identified a maladapted immune response profile associated with severe COVID-19 and poor clinical outcome, as well as early immune signatures that correlate with divergent disease trajectories.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls

              Memory T cells induced by previous pathogens can shape susceptibility to, and the clinical severity of, subsequent infections1. Little is known about the presence in humans of pre-existing memory T cells that have the potential to recognize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we studied T cell responses against the structural (nucleocapsid (N) protein) and non-structural (NSP7 and NSP13 of ORF1) regions of SARS-CoV-2 in individuals convalescing from coronavirus disease 2019 (COVID-19) (n = 36). In all of these individuals, we found CD4 and CD8 T cells that recognized multiple regions of the N protein. Next, we showed that patients (n = 23) who recovered from SARS (the disease associated with SARS-CoV infection) possess long-lasting memory T cells that are reactive to the N protein of SARS-CoV 17 years after the outbreak of SARS in 2003; these T cells displayed robust cross-reactivity to the N protein of SARS-CoV-2. We also detected SARS-CoV-2-specific T cells in individuals with no history of SARS, COVID-19 or contact with individuals who had SARS and/or COVID-19 (n = 37). SARS-CoV-2-specific T cells in uninfected donors exhibited a different pattern of immunodominance, and frequently targeted NSP7 and NSP13 as well as the N protein. Epitope characterization of NSP7-specific T cells showed the recognition of protein fragments that are conserved among animal betacoronaviruses but have low homology to 'common cold' human-associated coronaviruses. Thus, infection with betacoronaviruses induces multi-specific and long-lasting T cell immunity against the structural N protein. Understanding how pre-existing N- and ORF1-specific T cells that are present in the general population affect the susceptibility to and pathogenesis of SARS-CoV-2 infection is important for the management of the current COVID-19 pandemic.
                Bookmark

                Author and article information

                Contributors
                URI : https://loop.frontiersin.org/people/1115013Role: Role: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/2590156Role: Role:
                URI : https://loop.frontiersin.org/people/2603044Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1885724Role: Role: Role:
                Role: Role: Role:
                Role: Role:
                Role: Role: Role:
                Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1042727Role: Role: Role:
                URI : https://loop.frontiersin.org/people/476355Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1722391Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1428444Role: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/341207Role: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/332108Role: Role: Role: Role: Role: Role: Role: Role: Role: Role: Role:
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                26 January 2024
                2023
                : 14
                : 1291048
                Affiliations
                [1] 1 Africa Health Research Institute (AHRI), Nelson R. Mandela School of Medicine , Durban, South Africa
                [2] 2 Emory-University of Georgia, Center of Excellence of Influenza Research and Surveillance (CEIRS) , Lusaka, Zambia
                [3] 3 Center for Family Health Research in Zambia (CFHRZ), formerly Zambia Emory HIV Research Project (ZEHRP) , Lusaka, Zambia
                [4] 4 Infection and Immunity Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme , Blantyre, Malawi
                [5] 5 Human Immunodeficiency Virus (HIV) Pathogenesis Program, School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal , Durban, South Africa
                [6] 6 Department of Clinical Sciences, Liverpool School of Tropical Medicine , Liverpool, United Kingdom
                [7] 7 Division of Infection and Immunity, University College London , London, United Kingdom
                [8] 8 Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard University , Cambridge, MA, United States
                Author notes

                Edited by: Alberto Beretta, Independent Researcher, Milano, Italy

                Reviewed by: Alireza Haghparast, Ferdowsi University of Mashhad, Iran

                Laura Fantuzzi, National Institute of Health (ISS), Italy

                *Correspondence: Zaza M. Ndhlovu, zaza.ndhlovu@ 123456ahri.org
                Article
                10.3389/fimmu.2023.1291048
                10853422
                38343437
                0ff2a0fe-b709-49b5-925e-b8648a667f97
                Copyright © 2024 Ng’uni, Musale, Nkosi, Mandolo, Mvula, Michelo, Karim, Moosa, Khan, Jambo, Hanekom, Sigal, Kilembe and Ndhlovu

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 08 September 2023
                : 18 December 2023
                Page count
                Figures: 5, Tables: 3, Equations: 1, References: 47, Pages: 15, Words: 7497
                Funding
                The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was funded by; HHMI International research scholar award (Grant #55008743 to ZN), the Wellcome Trust (Grant# 226137/Z/22/Z Awarded to AS and ZN), the BMGF [Awarded to AS and ZN (INV-018944), awarded to KJ (INV-039481)], and SAMRC (Awarded to WH, # 96838). This work was also partially funded by the Sub-Saharan African Network for TB/HIV Research Excellence (SANTHE) collaborative award (to ZN # SANTHE-COL016), a DELTAS Africa Initiative (grant # DEL-15-006). The DELTAS Africa Initiative is an independent funding scheme of the African Academy of Sciences (AAS)’s Alliance for Accelerating Excellence in Science in Africa (AESA) and supported by the New Partnership for Africa’s Development Planning and Coordinating Agency (NEPAD Agency) with funding from the Wellcome Trust (grant # 107752/Z/15/Z) and the UK government. This project was also funded in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under Contract No. HHSN272201400004C (NIAID Centers of Excellence for Influenza Research and Surveillance, CEIRS).
                Categories
                Immunology
                Original Research
                Custom metadata
                Viral Immunology

                Immunology
                hiv,sars-cov-2,hcov-nl63,covid-19,t-cell response,antibody response
                Immunology
                hiv, sars-cov-2, hcov-nl63, covid-19, t-cell response, antibody response

                Comments

                Comment on this article