HIV status alters disease severity and immune cell responses in beta variant SARS-CoV-2 infection wave

  1. Farina Karim
  2. Inbal Gazy
  3. Sandile Cele
  4. Yenzekile Zungu
  5. Robert Krause
  6. Mallory Bernstein
  7. Khadija Khan
  8. Yashica Ganga
  9. Hylton Errol Rodel
  10. Ntombifuthi Mthabela
  11. Matilda Mazibuko
  12. Daniel Muema
  13. Dirhona Ramjit
  14. Thumbi Ndung'u
  15. Willem Hanekom
  16. Bernadett Gosnell
  17. Richard J Lessells
  18. Emily B Wong
  19. Tulio de Oliveira
  20. Yunus Moosa
  21. Gil Lustig
  22. Alasdair Leslie  Is a corresponding author
  23. Henrik Kløverpris  Is a corresponding author
  24. Alex Sigal  Is a corresponding author
  1. Africa Health Research Institute, South Africa
  2. University of KwaZulu-Natal, South Africa
  3. Africa Health Research Institute; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, South Africa
  4. Africa Health Research Institute; Division of Infection and Immunity, University College London, South Africa
  5. Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, South Africa
  6. KwaZulu-Natal Research Institute for TB-HIV, South Africa
  7. University of KwaZulu-Natal,SA, South Africa
  8. Centre for the AIDS Programme of Research in South Africa, South Africa
  9. African Health Research Institute, South Africa
  10. Africa Health Research Institute, University of KwaZulu-Natal, South Africa

Abstract

There are conflicting reports on the effects of HIV on COVID-19. Here we analyzed disease severity and immune cell changes during and after SARS-CoV-2 infection in 236 participants from South Africa, of which 39% were people living with HIV (PLWH), during the first and second (beta dominated) infection waves. The second wave had more PLWH requiring supplemental oxygen relative to HIV negative participants. Higher disease severity was associated with low CD4 T cell counts and higher neutrophil to lymphocyte ratios (NLR). Yet, CD4 counts recovered and NLR stabilized after SARS-CoV-2 clearance in wave 2 infected PLWH, arguing for an interaction between SARS-CoV-2 and HIV infection leading to low CD4 and high NLR. The first infection wave, where severity in HIV negative and PLWH was similar, still showed some HIV modulation of SARS-CoV-2 immune responses. Therefore, HIV infection can synergize with the SARS-CoV-2 variant to change COVID-19 outcomes.

Data availability

All data generated or analysed during this study are included in the manuscript and supporting files

Article and author information

Author details

  1. Farina Karim

    Division of Clinical Studies, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  2. Inbal Gazy

    University of KwaZulu-Natal, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  3. Sandile Cele

    Systems Infection Biology, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  4. Yenzekile Zungu

    Africa Health Research Institute, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  5. Robert Krause

    Africa Health Research Institute, Africa Health Research Institute; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  6. Mallory Bernstein

    Africa Health Research Institute, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  7. Khadija Khan

    Division of Clinical Studies, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  8. Yashica Ganga

    Africa Health Research Institute, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  9. Hylton Errol Rodel

    Systems Infection Biology, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  10. Ntombifuthi Mthabela

    Africa Health Research Institute, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  11. Matilda Mazibuko

    Africa Health Research Institute, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  12. Daniel Muema

    Africa Health Research Institute; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Africa Health Research Institute; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  13. Dirhona Ramjit

    Africa Health Research Institute, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  14. Thumbi Ndung'u

    Africa Health Research Institute, Africa Health Research Institute, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-2962-3992
  15. Willem Hanekom

    Africa Health Research Institute; Division of Infection and Immunity, University College London, Africa Health Research Institute; Division of Infection and Immunity, University College London, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  16. Bernadett Gosnell

    Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durbans, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  17. Richard J Lessells

    University of KwaZulu-Natal, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-0926-710X
  18. Emily B Wong

    KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  19. Tulio de Oliveira

    School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal,SA, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  20. Yunus Moosa

    Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  21. Gil Lustig

    Centre for the AIDS Programme of Research in South Africa, Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
    Competing interests
    The authors declare that no competing interests exist.
  22. Alasdair Leslie

    African Health Research Institute, Durban, South Africa
    For correspondence
    Al.Leslie@ahri.org
    Competing interests
    The authors declare that no competing interests exist.
  23. Henrik Kløverpris

    Africa Health Research Institute, Africa Health Research Institute, Durban, South Africa
    For correspondence
    Henrik.Kloverpris@ahri.org
    Competing interests
    The authors declare that no competing interests exist.
  24. Alex Sigal

    School of Laboratory Medicine and Medical Sciences, Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
    For correspondence
    alex.sigal@ahri.org
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-8571-2004

Funding

Bill and Melinda Gates Foundation (INV-018944)

  • Alex Sigal

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Lishomwa Ndhlovu

Ethics

Human subjects: The study protocol was approved by the University of KwaZulu-Natal Institutional Review Board (approval BREC/00001275/2020). Adult patients ($>$18 years old) presenting either at King Edward VIII or Clairwood Hospitals in Durban, South Africa, between 8 June to 25 September 2020, diagnosed to be SARS-CoV-2 positive as part of their clinical workup and able to provide informed consent were eligible for the study. Written informed consent was obtained for all enrolled participants.

Version history

  1. Received: February 9, 2021
  2. Accepted: September 7, 2021
  3. Accepted Manuscript published: October 5, 2021 (version 1)
  4. Accepted Manuscript updated: October 6, 2021 (version 2)
  5. Version of Record published: December 16, 2021 (version 3)

Copyright

© 2021, Karim et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 1,292
    views
  • 244
    downloads
  • 29
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Farina Karim
  2. Inbal Gazy
  3. Sandile Cele
  4. Yenzekile Zungu
  5. Robert Krause
  6. Mallory Bernstein
  7. Khadija Khan
  8. Yashica Ganga
  9. Hylton Errol Rodel
  10. Ntombifuthi Mthabela
  11. Matilda Mazibuko
  12. Daniel Muema
  13. Dirhona Ramjit
  14. Thumbi Ndung'u
  15. Willem Hanekom
  16. Bernadett Gosnell
  17. Richard J Lessells
  18. Emily B Wong
  19. Tulio de Oliveira
  20. Yunus Moosa
  21. Gil Lustig
  22. Alasdair Leslie
  23. Henrik Kløverpris
  24. Alex Sigal
(2021)
HIV status alters disease severity and immune cell responses in beta variant SARS-CoV-2 infection wave
eLife 10:e67397.
https://doi.org/10.7554/eLife.67397

Share this article

https://doi.org/10.7554/eLife.67397

Further reading

    1. Microbiology and Infectious Disease
    Hina Khan, Partha Paul ... Dibyendu Sarkar
    Research Article

    Survival of Mycobacterium tuberculosis within the host macrophages requires the bacterial virulence regulator PhoP, but the underlying reason remains unknown. 3′,5′-Cyclic adenosine monophosphate (cAMP) is one of the most widely used second messengers, which impacts a wide range of cellular responses in microbial pathogens including M. tuberculosis. Herein, we hypothesized that intra-bacterial cAMP level could be controlled by PhoP since this major regulator plays a key role in bacterial responses against numerous stress conditions. A transcriptomic analysis reveals that PhoP functions as a repressor of cAMP-specific phosphodiesterase (PDE) Rv0805, which hydrolyzes cAMP. In keeping with these results, we find specific recruitment of the regulator within the promoter region of rv0805 PDE, and absence of phoP or ectopic expression of rv0805 independently accounts for elevated PDE synthesis, leading to the depletion of intra-bacterial cAMP level. Thus, genetic manipulation to inactivate PhoP-rv0805-cAMP pathway decreases cAMP level, stress tolerance, and intracellular survival of the bacillus.

    1. Biochemistry and Chemical Biology
    2. Microbiology and Infectious Disease
    Natalia E Ketaren, Fred D Mast ... John D Aitchison
    Research Advance

    To date, all major modes of monoclonal antibody therapy targeting SARS-CoV-2 have lost significant efficacy against the latest circulating variants. As SARS-CoV-2 omicron sublineages account for over 90% of COVID-19 infections, evasion of immune responses generated by vaccination or exposure to previous variants poses a significant challenge. A compelling new therapeutic strategy against SARS-CoV-2 is that of single-domain antibodies, termed nanobodies, which address certain limitations of monoclonal antibodies. Here, we demonstrate that our high-affinity nanobody repertoire, generated against wild-type SARS-CoV-2 spike protein (Mast et al., 2021), remains effective against variants of concern, including omicron BA.4/BA.5; a subset is predicted to counter resistance in emerging XBB and BQ.1.1 sublineages. Furthermore, we reveal the synergistic potential of nanobody cocktails in neutralizing emerging variants. Our study highlights the power of nanobody technology as a versatile therapeutic and diagnostic tool to combat rapidly evolving infectious diseases such as SARS-CoV-2.