Inflammatory pathways amongst people living with HIV in Malawi differ according to socioeconomic status

Christine Kelly, Willard Tinago, Dagmar Alber, Patricia Hunter, Natasha Luckhurst, Jake Connolly, Francesca Arrigoni, Alejandro Garcia Abner, Raphael Kamn'gona, Irene Sheha, Mishek Chammudzi, Kondwani Jambo, Jane Mallewa, Alicja Rapala, Patrick W G Mallon, Henry Mwandumba, Nigel Klein, Saye Khoo, Christine Kelly, Willard Tinago, Dagmar Alber, Patricia Hunter, Natasha Luckhurst, Jake Connolly, Francesca Arrigoni, Alejandro Garcia Abner, Raphael Kamn'gona, Irene Sheha, Mishek Chammudzi, Kondwani Jambo, Jane Mallewa, Alicja Rapala, Patrick W G Mallon, Henry Mwandumba, Nigel Klein, Saye Khoo

Abstract

Background: Non-communicable diseases (NCDs) are increased amongst people living with HIV (PLWH) and are driven by persistent immune activation. The role of socioeconomic status (SES) in immune activation amongst PLWH is unknown, especially in low-income sub-Saharan Africa (SSA), where such impacts may be particularly severe.

Methods: We recruited Malawian adults with CD4<100 cells/ul two weeks after starting ART in the REALITY trial (NCT01825031), as well as volunteers without HIV infection. Clinical assessment, socioeconomic evaluation, blood draw for immune activation markers and carotid femoral pulse wave velocity (cfPWV) were carried out at 2- and 42-weeks post-ART initiation. Socioeconomic risk factors for immune activation and arterial stiffness were assessed using linear regression models.

Results: Of 279 PLWH, the median (IQR) age was 36 (31-43) years and 122 (44%) were female. Activated CD8 T-cells increased from 70% amongst those with no education to 88% amongst those with a tertiary education (p = 0.002); and from 71% amongst those earning less than 10 USD/month to 87% amongst those earning between 100-150 USD/month (p = 0.0001). Arterial stiffness was also associated with higher SES (car ownership p = 0.003, television ownership p = 0.012 and electricity access p = 0.029). Conversely, intermediate monocytes were higher amongst those with no education compared to a tertiary education (12.6% versus 7.3%; p = 0.01) and trended towards being higher amongst those earning less than 10 USD/month compared to 100-150 USD/month (10.5% versus 8.0%; p = 0.08). Water kiosk use showed a protective association against T cell activation (p = 0.007), as well as endothelial damage (MIP1β, sICAM1 and sVCAM1 p = 0.047, 0.026 and 0.031 respectively).

Conclusions: Socioeconomic risk factors for persistent inflammation amongst PLWH in SSA differ depending on the type of inflammatory pathway. Understanding these pathways and their socioeconomic drivers will help identify those at risk and target interventions for NCDs. Future studies assessing drivers of inflammation in HIV should include an SES assessment.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Cell surface immune activation markers…
Fig 1. Cell surface immune activation markers according to education and income category.
*p0.001 ***p0.01.
Fig 2. Adjusted fold change in cell…
Fig 2. Adjusted fold change in cell surface immune activation markers according to socio-economic risk factors.
Models for the effect of socio-economic variables on CD8 T cell and monocyte phenotypes, adjusted for age and sex, are shown. The x axis shows fold change with 95% confidence intervals for the following socio-economic comparisons: i) Income >25 USD per month compared to income = 25USD/month ii) some education compared to no education iii) water kiosk as source of water compared to all other water sources iv) grows household crops compared to doesn’t grow household crops.
Fig 3. Adjusted fold change in plasma…
Fig 3. Adjusted fold change in plasma inflammatory markers according to socio-economic risk factors.
All inflammatory biomarkers measured in μg/mL apart from those marked * which are in pg/mL.
Fig 4. Hypothesis for the role of…
Fig 4. Hypothesis for the role of socio-economic determinants in chronic inflammation and endothelial damage amongst PLWH in low income SSA.
Hypothesis for the impact of socio-economic factors on inflammation mediated non-communicable diseases in people living with HIV in low-income settings. This builds on previously documented relationships between drivers of inflammation, and its effect on endothelial damage in this setting [4, 5]. Further research will be required to evaluate and improve our understanding of the factors driving immune activation and non-communicable disease in low-income settings.

References

    1. Gouda H.N., et al.., Burden of non-communicable diseases in sub-Saharan Africa, 1990–2017: results from the Global Burden of Disease Study 2017. Lancet Glob Health, 2019. 7(10): p. e1375–e1387. doi: 10.1016/S2214-109X(19)30374-2
    1. Peterson T.E. and Baker J.V., Assessing inflammation and its role in comorbidities among persons living with HIV. Curr Opin Infect Dis, 2019. 32(1): p. 8–15. doi: 10.1097/QCO.0000000000000510
    1. Kuller L.H., et al.., Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med, 2008. 5(10): p. e203. doi: 10.1371/journal.pmed.0050203
    1. Kelly C., et al.., HIV-related arterial stiffness in Malawian adults is associated with proportion of PD-1 expressing CD8 T-cells and reverses with anti-retroviral therapy. J Infect Dis, 2019.
    1. Kelly C., et al.., Inflammatory Phenotypes Predict Changes in Arterial Stiffness following ART Initiation. Clin Infect Dis, 2020.
    1. Estimates of global, regional, and national incidence, prevalence, and mortality of HIV, 1980–2015: the Global Burden of Disease Study 2015. Lancet HIV, 2016. 3(8): p. e361–e387. doi: 10.1016/S2352-3018(16)30087-X
    1. Brenchley J.M., et al.., Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med, 2006. 12(12): p. 1365–71. doi: 10.1038/nm1511
    1. Christensen-Quick A., et al.., Subclinical Cytomegalovirus DNA Is Associated with CD4 T Cell Activation and Impaired CD8 T Cell CD107a Expression in People Living with HIV despite Early Antiretroviral Therapy. J Virol, 2019. doi: 10.1128/JVI.00179-19
    1. Peterson I., et al.., Unstructured treatment interruption: an important risk factor for arterial stiffness in adult Malawian patients with antiretroviral treatment. AIDS, 2016. 30(15): p. 2373–8. doi: 10.1097/QAD.0000000000001198
    1. Siika A., et al.., Late Presentation With HIV in Africa: Phenotypes, Risk, and Risk Stratification in the REALITY Trial. Clin Infect Dis, 2018. 66(suppl_2): p. S140–S146. doi: 10.1093/cid/cix1142
    1. Laurent S., et al.., Expert consensus document on arterial stiffness: methodological issues and clinical applications. Eur Heart J, 2006. 27(21): p. 2588–605. doi: 10.1093/eurheartj/ehl254
    1. Vlachopoulos C., Aznaouridis K., and Stefanadis C., Prediction of cardiovascular events and all-cause mortality with arterial stiffness: a systematic review and meta-analysis. J Am Coll Cardiol, 2010. 55(13): p. 1318–27. doi: 10.1016/j.jacc.2009.10.061
    1. Mattace-Raso F.U., et al.., Arterial stiffness and risk of coronary heart disease and stroke: the Rotterdam Study. Circulation, 2006. 113(5): p. 657–63. doi: 10.1161/CIRCULATIONAHA.105.555235
    1. Van Bortel L.M., et al.., Expert consensus document on the measurement of aortic stiffness in daily practice using carotid-femoral pulse wave velocity. J Hypertens, 2012. 30(3): p. 445–8. doi: 10.1097/HJH.0b013e32834fa8b0
    1. Glennie S.J., et al.., Do multiple concurrent infections in African children cause irreversible immunological damage? Immunology, 2012. 135(2): p. 125–32. doi: 10.1111/j.1365-2567.2011.03523.x
    1. Elliott A.M., et al.., Associations between helminth infection and CD4+ T cell count, viral load and cytokine responses in HIV-1-infected Ugandan adults. Trans R Soc Trop Med Hyg, 2003. 97(1): p. 103–8. doi: 10.1016/s0035-9203(03)90040-x
    1. Tusting L.S., et al.., Housing Improvements and Malaria Risk in Sub-Saharan Africa: A Multi-Country Analysis of Survey Data. PLoS Med, 2017. 14(2): p. e1002234. doi: 10.1371/journal.pmed.1002234
    1. Wingfield T., et al.., Addressing social determinants to end tuberculosis. Lancet, 2018. 391(10126): p. 1129–1132. doi: 10.1016/S0140-6736(18)30484-7
    1. Bourke C.D., et al.., Cotrimoxazole reduces systemic inflammation in HIV infection by altering the gut microbiome and immune activation. Sci Transl Med, 2019. 11(486). doi: 10.1126/scitranslmed.aav0537
    1. Leddy A.M., et al.., Food Insecurity Is Associated With Inflammation Among Women Living With HIV. J Infect Dis, 2019. 219(3): p. 429–436. doi: 10.1093/infdis/jiy511
    1. Hunt P.W., et al.., Valganciclovir reduces T cell activation in HIV-infected individuals with incomplete CD4+ T cell recovery on antiretroviral therapy. J Infect Dis, 2011. 203(10): p. 1474–83. doi: 10.1093/infdis/jir060
    1. Hsue P.Y., et al.., Safety and Impact of Low-Dose Methotrexate on Endothelial Function and Inflammation in Individuals with Treated Human Immunodeficiency Virus: AIDS Clinical Trials Group Study A5314. Clin Infect Dis, 2018.
    1. Dube M.P., et al.., A Randomized, Double-Blinded, Placebo-Controlled Trial of Sitagliptin for Reducing Inflammation and Immune Activation in Treated and Suppressed HIV Infection. Clin Infect Dis, 2018.
    1. Tann C.J., et al.., Prevalence of bloodstream pathogens is higher in neonatal encephalopathy cases vs. controls using a novel panel of real-time PCR assays. PLoS One, 2014. 9(5): p. e97259. doi: 10.1371/journal.pone.0097259
    1. Reid M.J.A., et al.., Higher Body Mass Index Is Associated With Greater Proportions of Effector CD8+ T Cells Expressing CD57 in Women Living With HIV. J Acquir Immune Defic Syndr, 2017. 75(5): p. e132–e141. doi: 10.1097/QAI.0000000000001376
    1. Simpson R.J. and Guy K., Coupling aging immunity with a sedentary lifestyle: has the damage already been done?—a mini-review. Gerontology, 2010. 56(5): p. 449–58. doi: 10.1159/000270905
    1. Allen L., et al.., Socioeconomic status and non-communicable disease behavioural risk factors in low-income and lower-middle-income countries: a systematic review. Lancet Glob Health, 2017. 5(3): p. e277–e289. doi: 10.1016/S2214-109X(17)30058-X
    1. Chandna J., et al.., Effects of improved complementary feeding and improved water, sanitation and hygiene on early child development among HIV-exposed children: substudy of a cluster randomised trial in rural Zimbabwe. BMJ Glob Health, 2020. 5(1): p. e001718. doi: 10.1136/bmjgh-2019-001718
    1. Fraga S., et al.., Childhood socioeconomic conditions are associated with increased chronic low-grade inflammation over adolescence: findings from the EPITeen cohort study. Arch Dis Child, 2020. doi: 10.1136/archdischild-2019-317525
    1. Loucks E.B., et al.., Life course socioeconomic position is associated with inflammatory markers: the Framingham Offspring Study. Soc Sci Med, 2010. 71(1): p. 187–95. doi: 10.1016/j.socscimed.2010.03.012
    1. Loucks E.B., et al.., Association of educational level with inflammatory markers in the Framingham Offspring Study. Am J Epidemiol, 2006. 163(7): p. 622–8. doi: 10.1093/aje/kwj076
    1. Dhabhar F.S., Effects of stress on immune function: the good, the bad, and the beautiful. Immunol Res, 2014. 58(2–3): p. 193–210. doi: 10.1007/s12026-014-8517-0
    1. Sima L.C., et al.., Relationship between use of water from community-scale water treatment refill kiosks and childhood diarrhea in Jakarta. Am J Trop Med Hyg, 2012. 87(6): p. 979–84. doi: 10.4269/ajtmh.2012.12-0224
    1. Rogawski E.T., et al.., Use of quantitative molecular diagnostic methods to investigate the effect of enteropathogen infections on linear growth in children in low-resource settings: longitudinal analysis of results from the MAL-ED cohort study. Lancet Glob Health, 2018. 6(12): p. e1319–e1328. doi: 10.1016/S2214-109X(18)30351-6

Source: PubMed

Подписаться