A Mycobacterium tuberculosis Specific IgG3 Signature of Recurrent Tuberculosis

Stephanie Fischinger, Deniz Cizmeci, Sally Shin, Leela Davies, Patricia S Grace, Aida Sivro, Nonhlanhla Yende-Zuma, Hendrik Streeck, Sarah M Fortune, Douglas A Lauffenburger, Kogieleum Naidoo, Galit Alter, Stephanie Fischinger, Deniz Cizmeci, Sally Shin, Leela Davies, Patricia S Grace, Aida Sivro, Nonhlanhla Yende-Zuma, Hendrik Streeck, Sarah M Fortune, Douglas A Lauffenburger, Kogieleum Naidoo, Galit Alter

Abstract

South Africa has the highest prevalence of HIV and tuberculosis (TB) co-infection globally. Recurrent TB, caused by relapse or reinfection, makes up the majority of TB cases in South Africa, and HIV infected individuals have a greater likelihood of developing recurrent TB. Given that TB remains a leading cause of death for HIV infected individuals, and correlates of TB recurrence protection/risk have yet to be defined, here we sought to understand the antibody associated mechanisms of recurrent TB by investigating the humoral response in a longitudinal cohort of HIV co-infected individuals previously treated for TB with and without recurrent disease during follow-up, in order to identify antibody correlates of protection between individuals who do not have recurrent TB and individuals who do. We used a high-throughput, "systems serology" approach to profile biophysical and functional characteristics of antibodies targeting antigens from Mycobacterium tuberculosis (Mtb). Differences in antibody profiles were noted between individuals with and without recurrent TB, albeit these differences were largely observed close to the time of re-diagnosis. Individuals with recurrent TB had decreased Mtb-antigen specific IgG3 titers, but not other IgG subclasses or IgA, compared to control individuals. These data point to a potential role for Mtb-specific IgG3 responses as biomarkers or direct mediators of protective immunity against Mtb recurrence.

Trial registration: ClinicalTrials.gov NCT01539005.

Keywords: IgG3; Mycobacterium tuberculosis; antibodies; recurrence; recurrent tuberculosis.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Fischinger, Cizmeci, Shin, Davies, Grace, Sivro, Yende-Zuma, Streeck, Fortune, Lauffenburger, Naidoo and Alter.

Figures

Figure 1
Figure 1
Mtb-specific antibody profile across recurrent/no recurrent TB patient cohort. (A) The annotated heatmap shows antibody measurements across time, groups and displays demographic data. The first row of the heatmap delivers information of grouping into individuals who go on to get recurrent TB (orange) versus who do not get recurrent TB (grey). Days to TB recurrence for the recurrence group is indicated in shades of orange, days from cure are colored in grey for all individuals. The row below in blue shades reveals if participants had (1) or did not (2) have previous TB before enrollment into the parent study. Age, gender, CD4 levels and viral load are indicated. Antibody effector functions, titer and Fc-receptor binding are displayed, antigens for the functions are PPD, EDAT6/CFP10 and LAM, additional antigens for titer and Fc-receptor binding include Hspx, Ag85, RV0826 and RV1363, in this order. (B) IgG1 and IgG3 titers are shown over time for the recurrent TB group (top) and the no recurrence group (bottom) for PPD, ESAT6/CFP10, LAM and Hspx. X axis depicts time since TB cure1 at enrollment into the study and color in the recurrence group represents time to TB recurrence in shades of orange.
Figure 2
Figure 2
Mtb-specific antibody IgG1 titers correlate across different antigens. Graphs show the most distant time point from time since cure (and closest time point to recurrence for the recurrent TB group) for each individual in both groups. (A) Nightingale rose charts show comparisons between IgG1 titers against different TB-antigens between individuals with recurrent and no recurrent TB. Petals are scaled based on the median of the percent rank of each feature per group. (B) The Spearman correlation heatmap depicts correlation r values for the no recurrence group (grey) and the recurrence group (orange) for IgG1 titers across antigens. Color depth indicates r values, asterisk represent significance, adjusted for multiple comparison using Bonferroni correction method, adjusted p value *< 0.05, **< 0.01, ***<0.001, ****<0.0001.
Figure 3
Figure 3
Individuals who do not get recurrent TB have higher titers of IgG3. All graphs show data for the most distant time point from time since cure. (A) Nightingale rose charts allow for comparison between groups for antibody titer (IgG1-IgG4, IgA, IgM), Fc-receptor binding levels (FCR2A, 2B, 3A, 3B) and the ability to induce effector functions (ADCP, ADNP) at the most distant time point from TB cure1. The left column shows flowers for the recurrent TB group, the right side for the no recurrence group. Relative responses for PPD (top), ESAT6/CFP10 (middle) and LAM (bottom) are shown. (B) Dot plots show univariate IgG3 levels (MFI) across different Mtb antigens, median is indicated by line. For Hspx, IgG3:IgG2 ratios are depicted additionally. Grey depicts no recurrence, orange recurrent TB. Each dot represents the average within each matching bin. P-values were calculated using Wilcoxon signed-rank test and adjusted for multiple comparison using Benjamini-Hochberg method.
Figure 4
Figure 4
Mixed effects model supports the indication of higher IgG3 levels in no recurrent TB individuals. Dot plots and the model depict the most distant time point from time since cure for every individual. The mixed effects models depict the overall differences in measured antibody features between individuals with (left side) and without (right side) recurrent TB. The models include recurrent and non-recurrent individuals taking into account age, gender, as well as days since TB cure1. The X-axis depicts the effect size between the groups and y-axis shows a measure of statistical significance.

References

    1. WHO . WHO Global Tuberculosis Report 2018. (2018). p. 6–8. doi: 10.1016/j.pharep.2017.02.021
    1. Mabunda TE, Ramalivhana NJ, Dambisya YM. Mortality Associated With Tuberculosis/HIV Co-Infection Among Patients on TB Treatment in the Limpopo Province, South Africa. Afr Health Sci (2014) 14:849–54. doi: 10.4314/ahs.v14i4.12
    1. Bruchfeld J, Correia-Neves M, Källenius G. Tuberculosis and HIV Coinfection. Cold Spring Harb Perspect Med (2015) 5:a017871. doi: 10.1101/cshperspect.a017871
    1. Crampin AC, Mwaungulu JN, Mwaungulu FD, Mwafulirwa DT, Munthali K, Floyd S, et al. . Recurrent TB: Relapse or Reinfection? The Effect of HIV in a General Population Cohort in Malawi. AIDS (2010) 24:417–26. doi: 10.1097/QAD.0b013e32832f51cf
    1. Pawlowski A, Jansson M, Sköld M, Rottenberg ME, Källenius G. Tuberculosis and HIV Co-Infection. PloS Pathog (2012) 8:e1002464–e1002464. doi: 10.1371/journal.ppat.1002464
    1. Abdool Karim SS, Churchyard GJ, Karim QA, Lawn SD. HIV Infection and Tuberculosis in South Africa: An Urgent Need to Escalate the Public Health Response. Lancet (London England) (2009) 374:921–33. doi: 10.1016/S0140-6736(09)60916-8
    1. Chaisson RE, Churchyard GJ. Recurrent Tuberculosis: Relapse, Reinfection, and HIV. J Infect Dis (2010) 201:653–5. doi: 10.1086/650531
    1. Millet J-P, Shaw E, Orcau A, Casals M, Miró JM, Caylà JA. Barcelona Tuberculosis Recurrence Working Group. Tuberculosis Recurrence After Completion Treatment in a European City: Reinfection or Relapse? PloS One (2013) 8:e64898. doi: 10.1371/journal.pone.0064898
    1. Narayanan S, Swaminathan S, Supply P, Shanmugam S, Narendran G, Hari L, et al. . Impact of HIV Infection on the Recurrence of Tuberculosis in South India. J Infect Dis (2010) 201:691–703. doi: 10.1086/650528
    1. Sivro A, McKinnon LR, Yende-Zuma N, Gengiah S, Samsunder N, Abdool Karim SS, et al. . Plasma Cytokine Predictors of Tuberculosis Recurrence in Antiretroviral-Treated Human Immunodeficiency Virus-Infected Individuals From Durban, South Africa. Clin Infect Dis (2017) 65:819–26. doi: 10.1093/cid/cix357
    1. Welch RJ, Lawless KM, Litwin CM. Antituberculosis IgG Antibodies as a Marker of Active Mycobacterium Tuberculosis Disease. Clin Vaccine Immunol (2012) 19:522–6. doi: 10.1128/CVI.05573-11
    1. Abebe F, Bjune G. The Protective Role of Antibody Responses During Mycobacterium Tuberculosis Infection. Clin Exp Immunol (2009) 157:235–43. doi: 10.1111/j.1365-2249.2009.03967.x
    1. Suliman S, Thompson EG, Sutherland J, Weiner J, Ota MOC, Shankar S, et al. . Four-Gene Pan-African Blood Signature Predicts Progression to Tuberculosis. Am J Respir Crit Care Med (2018) 197:1198–208. doi: 10.1164/rccm.201711-2340OC
    1. Lu LL, Smith MT, Yu KKQ, Luedemann C, Suscovich TJ, Grace PS, et al. . IFN-γ-Independent Immune Markers of Mycobacterium Tuberculosis Exposure. Nat Med (2019) 25:977–87. doi: 10.1038/s41591-019-0441-3
    1. Lu LL, Chung AW, Rosebrock TR, Ghebremichael M, Yu WH, Grace PS, et al. . A Functional Role for Antibodies in Tuberculosis. Cell (2016) 167:433–43.e14. doi: 10.1016/j.cell.2016.08.072
    1. Lu LL, Das J, Grace PS, Fortune SM, Restrepo BI, Alter G. Antibody Fc Glycosylation Discriminates Between Latent and Active Tuberculosis. J Infect Dis (2020) 222:2093–102. doi: 10.1093/infdis/jiz643
    1. Balu S, Reljic R, Lewis MJ, Pleass RJ, McIntosh R, van Kooten C, et al. . A Novel Human IgA Monoclonal Antibody Protects Against Tuberculosis. J Immunol (2011) 186:3113–9. doi: 10.4049/jimmunol.1003189
    1. Hamasur B, Haile M, Pawlowski A, Schroder U, Kallenius G, Svenson SB. A Mycobacterial Lipoarabinomannan Specific Monoclonal Antibody and Its F(Ab’) Fragment Prolong Survival of Mice Infected With Mycobacterium Tuberculosis. Clin Exp Immunol (2004) 138:30–8. doi: 10.1111/j.1365-2249.2004.02593.x
    1. Guirado E, Amat I, Gil O, Díaz J, Arcos V, Caceres N, et al. . Passive Serum Therapy With Polyclonal Antibodies Against Mycobacterium tuberculosis Protects Against Post-Chemotherapy Relapse of Tuberculosis Infection in SCID Mice. Microb Inf (2006) 8(5):1252–9. doi: 10.1016/j.micinf.2005.12.004
    1. Darboe F, Mbandi SK, Naidoo K, Yende-Zuma N, Lewis L, Thompson EG, et al. . Detection of Tuberculosis Recurrence, Diagnosis and Treatment Response by a Blood Transcriptomic Risk Signature in HIV-Infected Persons on Antiretroviral Therapy. Front Microbiol (2019) 10:1–16. doi: 10.3389/fmicb.2019.01441
    1. Naidoo K, Dookie N, Naidoo K, Yende-Zuma N, Chimukangara B, Bhushan A, et al. . Recurrent Tuberculosis Among HIV-Coinfected Patients: a Case Series From KwaZulu-Natal. Infect Drug Resist (2018) 11:1413–21. doi: 10.2147/IDR.S150644
    1. Maharaj B, Gengiah TN, Yende-Zuma N, Gengiah S, Naidoo A, Naidoo K. Implementing Isoniazid Preventive Therapy in a Tuberculosis Treatment-Experienced Cohort on ART. Int J Tuberc Lung Dis (2017) 21:537–43. doi: 10.5588/ijtld.16.0775
    1. Abdool Karim SS, Naidoo K, Grobler A, Padayatchi N, Baxter C, Gray A, et al. . Timing of Initiation of Antiretroviral Drugs During Tuberculosis Therapy. N Engl J Med (2010) 362:697–706. doi: 10.1056/NEJMoa0905848
    1. Brown EP, Licht AF, Dugast AS, Choi I, Bailey-Kellogg C, Alter G, et al. . High-Throughput, Multiplexed IgG Subclassing of Antigen-Specific Antibodies From Clinical Samples. J Immunol Methods (2012) 386:117–23. doi: 10.1016/j.jim.2012.09.007
    1. Ackerman ME, Moldt B, Wyatt RT, Dugast A-S, McAndrew E, Tsoukas S, et al. . A Robust, High-Throughput Assay to Determine the Phagocytic Activity of Clinical Antibody Samples. J Immunol Methods (2011) 366:8–19. doi: 10.1016/j.jim.2010.12.016
    1. Karsten CB, Mehta N, Shin SA, Diefenbach TJ, Slein MD, Karpinski W, et al. . A Versatile High-Throughput Assay to Characterize Antibody-Mediated Neutrophil Phagocytosis. J Immunol Methods (2019) 471:46–56. doi: 10.1016/j.jim.2019.05.006
    1. Bates D, Mächler M, Bolker B, Walker S. Fitting Linear Mixed-Effects Models Using Lme4. J Stat Softw (2015) 67:1–46. doi: 10.18637/jss.v067.i01
    1. Serra-Vidal MM, Latorre I, Franken KLCM, Díaz J, de Souza-Galvão ML, Casas I, et al. . Immunogenicity of 60 Novel Latency-Related Antigens of Mycobacterium Tuberculosis. Front Microbiol (2014) 5:1–13. doi: 10.3389/fmicb.2014.00517
    1. Gomez MP, Donkor S, Adetifa IM, Ota MOC, Sutherland JS. Analysis of LAM and 38 kDa Antibody Levels for Diagnosis of TB in a Case-Control Study in West Africa. ISRN Immunol (2012) 2012:1–6. doi: 10.5402/2012/237823
    1. Taylor JL, Wieczorek A, Keyser AR, Grover A, Flinkstrom R, Karls RK, et al. . HspX-Mediated Protection Against Tuberculosis Depends on Its Chaperoning of a Mycobacterial Molecule. Immunol Cell Biol (2012) 90:945–54. doi: 10.1038/icb.2012.34
    1. Huygen K. The Immunodominant T-Cell Epitopes of the Mycolyl-Transferases of the Antigen 85 Complex of M. Tuberculosis. Front Immunol (2014) 5:1–11. doi: 10.3389/fimmu.2014.00321
    1. Vidarsson G, Dekkers G, Rispens T. IgG Subclasses and Allotypes: From Structure to Effector Functions. Front Immunol (2014) 5:520. doi: 10.3389/fimmu.2014.00520
    1. Banerjee K, Klasse PJ, Sanders RW, Pereyra F, Michael E, Lu M, et al. . IgG Subclass Profiles in Infected HIV Type 1 Controllers and Chronic Progressors and in Uninfected Recipients of Env Vaccines. AIDS Res Hum Retroviruses (2010) 26:445–58. doi: 10.1089/aid.2009.0223
    1. Friedland G, Churchyard GJ, Nardell E. Tuberculosis and HIV Coinfection: Current State of Knowledge and Research Priorities. J Infect Dis (2007) 196:S1–3. doi: 10.1086/518667
    1. Parvaresh L, Crighton T, Martinez E, Bustamante A, Chen S, Sintchenko V. Recurrence of Tuberculosis in a Low-Incidence Setting: A Retrospective Cross-Sectional Study Augmented by Whole Genome Sequencing. BMC Infect Dis (2018) 18:265. doi: 10.1186/s12879-018-3164-z
    1. Kawahara JY, Irvine EB. Alter G. A Case for Antibodies as Mechanistic Correlates of Immunity in Tuberculosis. Front Immunol (2019) 10:1–9. doi: 10.3389/fimmu.2019.00996
    1. Abebe F, Belay M, Legesse M, Ottenhoff THM, K L M C F . IgA and IgG Against Mycobacterium Tuberculosis Rv2031 Discriminate Between Pulmonary Tuberculosis Patients, Mycobacterium Tuberculosis-Infected and non-Infected Individuals. PloS One (2018) 13:e0190989. doi: 10.1371/journal.pone.0190989
    1. Akpogheneta OJ, Duah NO, Tetteh KKA, Dunyo S, Lanar DE, Pinder M, et al. . Duration of Naturally Acquired Antibody Responses to Blood-Stage Plasmodium Falciparum Is Age Dependent and Antigen Specific. Infect Immun (2008) 76:1748–55. doi: 10.1128/IAI.01333-07
    1. Keating SM, Pilcher CD, Jain V, Lebedeva M, Hampton D, Abdel-Mohsen M, et al. . HIV Antibody Level as a Marker of HIV Persistence and Low-Level Viral Replication. J Infect Dis (2017) 216:72–81. doi: 10.1093/infdis/jix225
    1. Wang M, Yuan J, Li T, Liu Y, Wu J, Di B, et al. . Antibody Dynamics of 2009 Influenza A (H1N1) Virus in Infected Patients and Vaccinated People in China. PloS One (2011) 6:e16809. doi: 10.1371/journal.pone.0016809
    1. Azzurri A, Kanaujia GV, Sow OY, Bah B, Diallo A, Del PG, et al. . Serological Markers of Pulmonary Tuberculosis and of Response to Anti-Tuberculosis Treatment in a Patient Population in Guinea. Int J Immunopathol Pharmacol (2006) 19:205873920601900. doi: 10.1177/205873920601900120
    1. Walzl G, Ronacher K, Hanekom W, Scriba TJ, Zumla A. Immunological Biomarkers of Tuberculosis. Nat Rev Immunol (2011) 11:343–54. doi: 10.1038/nri2960
    1. Silva VMC, Kanaujia G, Gennaro ML, Menzies D. Factors Associated With Humoral Response to ESAT-6, 38 kDa and 14 kDa in Patients With a Spectrum of Tuberculosis. Int J Tuberc Lung Dis (2003) 7:478–84. doi: 10.1183/09031936.06.00042706
    1. Stavnezer J, Schrader CE. IgH Chain Class Switch Recombination: Mechanism and Regulation. J Immunol (2014) 193:5370–8. doi: 10.4049/jimmunol.1401849
    1. Damelang T, Rogerson SJ, Kent SJ, Chung AW. Role of IgG3 in Infectious Diseases. Trends Immunol (2019) 40:197–211. doi: 10.1016/j.it.2019.01.005
    1. Roussilhon C, Oeuvray C, Müller-Graf C, Tall A, Rogier C, Trape J-F, et al. . Long-Term Clinical Protection From Falciparum Malaria Is Strongly Associated With IgG3 Antibodies to Merozoite Surface Protein 3. PloS Med (2007) 4:e320. doi: 10.1371/journal.pmed.0040320
    1. Fischinger S, Dolatshahi S, Jennewein MF, Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al. . IgG3 Collaborates With IgG1 and IgA to Recruit Effector Function in RV144 Vaccinees. JCI Insight (2020) 5:1–16. doi: 10.1172/jci.insight.140925
    1. Roy Chowdhury R, Vallania F, Yang Q, Lopez Angel CJ, Darboe F, Penn-Nicholson A, et al. . A Multi-Cohort Study of the Immune Factors Associated With M. Tuberculosis Infection Outcomes. Nature (2018) 560:644–8. doi: 10.1038/s41586-018-0439-x
    1. Bruhns P, Iannascoli B, England P, Mancardi DA, Fernandez N, Jorieux S, et al. . Specificity and Affinity of Human Fcγ Receptors and Their Polymorphic Variants for Human IgG Subclasses. Blood (2009) 113:3716–25. doi: 10.1182/blood-2008-09-179754
    1. Gupta S, Shende N, Bhatia AS, Kumar S, Harinath BC. IgG Subclass Antibody Response to Mycobacterial Serine Protease at Different Stages of Pulmonary Tuberculosis. Med Sci Monit (2005) 11(12):CR585–8.
    1. Dobaño C, Santano R, Vidal M, Jiménez A, Jairoce C, Ubillos I, et al. . Differential Patterns of IgG Subclass Responses to Plasmodium Falciparum Antigens in Relation to Malaria Protection and RTS,S Vaccination. Front Immunol (2019) 10:439. doi: 10.3389/fimmu.2019.00439

Source: PubMed

3
Abonneren