Biomarkers of post-discharge mortality among children with complicated severe acute malnutrition

James M Njunge, Agnes Gwela, Nelson K Kibinge, Moses Ngari, Lydia Nyamako, Emily Nyatichi, Johnstone Thitiri, Gerard Bryan Gonzales, Robert H J Bandsma, Judd L Walson, Evelyn N Gitau, James A Berkley, James M Njunge, Agnes Gwela, Nelson K Kibinge, Moses Ngari, Lydia Nyamako, Emily Nyatichi, Johnstone Thitiri, Gerard Bryan Gonzales, Robert H J Bandsma, Judd L Walson, Evelyn N Gitau, James A Berkley

Abstract

High mortality after discharge from hospital following acute illness has been observed among children with Severe Acute Malnutrition (SAM). However, mechanisms that may be amenable to intervention to reduce risk are unknown. We performed a nested case-control study among HIV-uninfected children aged 2-59 months treated for complicated SAM according to WHO recommendations at four Kenyan hospitals. Blood was drawn from 1778 children when clinically judged stable before discharge from hospital. Cases were children who died within 60 days. Controls were randomly selected children who survived for one year without readmission to hospital. Untargeted proteomics, total protein, cytokines and chemokines, and leptin were assayed in plasma and corresponding biological processes determined. Among 121 cases and 120 controls, increased levels of calprotectin, von Willebrand factor, angiotensinogen, IL8, IL15, IP10, TNFα, and decreased levels of leptin, heparin cofactor 2, and serum paraoxonase were associated with mortality after adjusting for possible confounders. Acute phase responses, cellular responses to lipopolysaccharide, neutrophil responses to bacteria, and endothelial responses were enriched among cases. Among apparently clinically stable children with SAM, a sepsis-like profile is associated with subsequent death. This may be due to ongoing bacterial infection, translocated bacterial products or deranged immune response during nutritional recovery.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Plasma total protein concentration is significantly different between cases (n = 119) and controls (n = 119). Box plot summarize the median and interquartile ranges of plasma total protein and P values < 0.05 are significant.
Figure 2
Figure 2
Systemic inflammatory proteins are increased among cases. (A) Bar plot of differentially expressed proteins from the mass spectrometry-based proteome analysis between cases (n = 121) and controls (n = 120). The analysis was performed using proteome measurements in plasma among cases and controls. The bar graph depicts the log10 of the fold change of differentially expressed proteins. Red and green bars indicate significantly up- and downregulated proteins among cases. (B) Box plot summaries of the median and interquartile ranges of natural logarithm concentrations of 5 cytokines that were significantly different between cases (n = 112) and controls (n = 113). Overlaid dots represent individual data points. GCSF, granulocyte-colony stimulating factor; IL8, interleukin 8; IL15, interleukin 15; IP10, Interferon gamma-induced protein 10 (IP-10)/chemokine (C-X-C motif) ligand 10; TNFA, tumour necrosis factor; P values represent the adjusted P value using the Benjamini and Hochberg (BH) method for multiple testing.
Figure 3
Figure 3
Pairwise correlation heatmap among differentially expressed proteins FDR P < 0.05 and those with bold and underlined are P < 0.001.
Figure 4
Figure 4
Leptin concentrations are significantly different between cases (n = 119) and controls (n = 119). (A) Box plot summary of the median and IQRs of natural logarithms of leptin concentrations. Overlaid dots represent individual data points. (B) Leptin concentration, showing only those <100 pg/ml among cases and controls. The dot plot depicts the 35 pg/ml leptin cut-off and dots represent the individual leptin data points <100 pg/ml. P < 0.05 are significant.
Figure 5
Figure 5
Enrichment analysis of upregulated proteins among cases. Biological processes associated with early post-discharge mortality in children with SAM based on Gene Ontology (GO) analysis. The analysis for pathways enriched was performed using the DAVID Bioinformatics Resources version 6.8. The Bar graphs depict the enriched GO process category and the −log10 of the P value. The P value depicts the probability that a particular biological process is enriched in a group of proteins relative to other biological processes.

References

    1. Black RE, et al. Maternal and child undernutrition and overweight in low-income and middle-income countries. Lancet. 2013;382(9890):427–451. doi: 10.1016/S0140-6736(13)60937-X.
    1. Liu L, et al. Global, regional, and national causes of under-5 mortality in 2000–15: an updated systematic analysis with implications for the Sustainable Development Goals. Lancet. 2016;388(10063):3027–3035. doi: 10.1016/S0140-6736(16)31593-8.
    1. Pelletier DL, et al. The effects of malnutrition on child mortality in developing countries. Bull World Health Organ. 1995;73(4):443–8.
    1. Caulfield LE, et al. Undernutrition as an underlying cause of child deaths associated with diarrhea, pneumonia, malaria, and measles. Am J Clin Nutr. 2004;80(1):193–198. doi: 10.1093/ajcn/80.1.193.
    1. Nair H, et al. Global and regional burden of hospital admissions for severe acute lower respiratory infections in young children in 2010: a systematic analysis. Lancet. 2013;381(9875):1380–1390. doi: 10.1016/S0140-6736(12)61901-1.
    1. Man WDC, et al. Nutritional status of children admitted to hospital with different diseases and its relationship to outcome in The Gambia, West Africa. Trop Med Int Health. 1998;3(8):678–686. doi: 10.1046/j.1365-3156.1998.00283.x.
    1. Walson JL, Berkley JA. The impact of malnutrition on childhood infections. Curr Opin Infect Dis. 2018;31(3):231–236.
    1. WHO. Guideline: Updates on the Management of Severe Acute Malnutrition in Infants and Children (World Health Organization, Geneva, 2013).
    1. Bachou H, et al. Reduction of unnecessary transfusion and intravenous fluids in severely malnourished children is not enough to reduce mortality. Ann Trop Paediatr. 2008;28(1):23–33. doi: 10.1179/146532808X270644.
    1. Brewster DR. Inpatient management of severe malnutrition: time for a change in protocol and practice. Ann Trop Paediatr. 2011;31(2):97–107. doi: 10.1179/146532811X12925735813887.
    1. Page A-L, et al. Infections in children admitted with complicated severe acute malnutrition in Niger. PLoS One. 2013;8(7):e68699. doi: 10.1371/journal.pone.0068699.
    1. Maitland K, et al. Children with severe malnutrition: can those at highest risk of death be identified with the WHO Protocol. PLOS Medicine. 2006;3(12):e500. doi: 10.1371/journal.pmed.0030500.
    1. Moïsi Jennifer, Gatakaa Hellen, Berkley James, Maitland Kathryn, Mturi Neema, Newton Charles, Njuguna Patricia, Nokes James, Ojal John, Bauni Evasius, Tsofa Benjamin, Peshu Norbert, Marsh Kevin, Williams Thomas, Scott J Anthony. Excess child mortality after discharge from hospital in Kilifi, Kenya: a retrospective cohort analysis. Bulletin of the World Health Organization. 2011;89(10):725–732. doi: 10.2471/BLT.11.089235.
    1. Ngari MM, et al. Mortality after inpatient treatment for severe pneumonia in children: a cohort study. Paediatr Perinat Epidemiol. 2017;31(3):233–242. doi: 10.1111/ppe.12348.
    1. Wiens M O, Kumbakumba E, Larson C P, Ansermino J M, Singer J, Kissoon N, Wong H, Ndamira A, Kabakyenga J, Kiwanuka J, Zhou G. Postdischarge mortality in children with acute infectious diseases: derivation of postdischarge mortality prediction models. BMJ Open. 2015;5(11):e009449. doi: 10.1136/bmjopen-2015-009449.
    1. Chhibber AV, et al. child mortality after discharge from a health facility following suspected pneumonia, meningitis or septicaemia in rural Gambia: a cohort study. PLoS One. 2015;10(9):e0137095. doi: 10.1371/journal.pone.0137095.
    1. Veirum JE, et al. Increased mortality in the year following discharge from a paediatric ward in Bissau, Guinea-Bissau. Acta Pædiatrica. 2007;96(12):1832–1838. doi: 10.1111/j.1651-2227.2007.00562.x.
    1. Wiens MO, et al. pediatric post-discharge mortality in resource poor countries: a systematic review. PLoS One. 2013;8(6):e66698. doi: 10.1371/journal.pone.0066698.
    1. Berkley JA, et al. Daily co-trimoxazole prophylaxis to prevent mortality in children with complicated severe acute malnutrition: a multicentre, double-blind, randomised placebo-controlled trial. Lancet Glob Health. 2016;4(7):e464–e473. doi: 10.1016/S2214-109X(16)30096-1.
    1. Dubray C, et al. Treatment of severe malnutrition with 2-day intramuscular ceftriaxone vs 5-day amoxicillin. Ann Trop Paediatr. 2008;28(1):13–22. doi: 10.1179/146532808X270635.
    1. Kerac M, et al. Probiotics and prebiotics for severe acute malnutrition (PRONUT study): a double-blind efficacy randomised controlled trial in Malawi. Lancet. 2009;374(9684):136–44. doi: 10.1016/S0140-6736(09)60884-9.
    1. Trehan I, et al. Antibiotics as part of the management of severe acute malnutrition. N Engl J Med. 2013;368(5):425–35. doi: 10.1056/NEJMoa1202851.
    1. Nemetchek B, et al. Paediatric postdischarge mortality in developing countries: a systematic review. BMJ Open. 2018;8(12):e023445. doi: 10.1136/bmjopen-2018-023445.
    1. Bourke CD, Berkley JA, Prendergast AJ. Immune dysfunction as a cause and consequence of malnutrition. Trends Immunol. 2016;37(6):386–398. doi: 10.1016/j.it.2016.04.003.
    1. Tate JR, et al. Harmonising adult and paediatric reference intervals in australia and new zealand: an evidence-based approach for establishing a first panel of chemistry analytes. Clin Biochem Rev. 2014;35(4):213–35.
    1. Karita E, et al. CLSI-derived hematology and biochemistry reference intervals for healthy adults in Eastern and Southern Africa. PLoS One. 2009;4(2):e4401. doi: 10.1371/journal.pone.0004401.
    1. Hickmans EM, Finch E, Tonks E. Plasma protein values in infants. Arch Dis Child. 1943;18(94):96–97. doi: 10.1136/adc.18.94.96.
    1. Elias JE, Gygi SP. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat Methods. 2007;4:207. doi: 10.1038/nmeth1019.
    1. Bartz S, et al. Severe acute malnutrition in childhood: hormonal and metabolic status at presentation, response to treatment, and predictors of mortality. J Clin Endocrinol Metab. 2014;99(6):2128–37. doi: 10.1210/jc.2013-4018.
    1. Rytter MJH, et al. The immune system in children with malnutrition—a systematic review. PLoS One. 2014;9(8):e105017. doi: 10.1371/journal.pone.0105017.
    1. Jones KDJ, Berkley JA. Severe acute malnutrition and infection. Paediatr Int Child Health. 2014;34(Suppl 1):1–29. doi: 10.1179/2046904714Z.000000000218.
    1. Vandal K, et al. Blockade of S100A8 and S100A9 suppresses neutrophil migration in response to lipopolysaccharide. J Immunol. 2003;171(5):2602–2609. doi: 10.4049/jimmunol.171.5.2602.
    1. Huang H, Tu L. Expression of S100 family proteins in neonatal rats with sepsis and its significance. Int J Clin Exp Pathol. 2015;8(2):1631–9.
    1. Simm M, et al. Performance of plasma calprotectin as a biomarker of early sepsis: a pilot study. Biomark Med. 2016;10(8):811–818. doi: 10.2217/bmm-2016-0032.
    1. Jonsson N, et al. Calprotectin as an early biomarker of bacterial infections in critically ill patients: an exploratory cohort assessment. Crit Care Resusc. 2017;19(3):205–213.
    1. Kitabayashi A, et al. Granulocyte colony-stimulating factor downregulates allogeneic immune responses by posttranscriptional inhibition of tumor necrosis factor- alpha production. Blood. 1995;86(6):2220–2227.
    1. Hartung T, et al. Effect of granulocyte colony-stimulating factor treatment on ex vivo blood cytokine response in human volunteers. Blood. 1995;85(9):2482–2489.
    1. Opal SM, et al. Relationship between plasma levels of lipopolysaccharide (LPS) and LPS-binding protein in patients with severe sepsis and septic shock. J Infect Dis. 1999;180(5):1584–1589. doi: 10.1086/315093.
    1. Zweigner J, et al. High concentrations of lipopolysaccharide-binding protein in serum of patients with severe sepsis or septic shock inhibit the lipopolysaccharide response in human monocytes. Blood. 2001;98(13):3800–3808. doi: 10.1182/blood.V98.13.3800.
    1. Rogers CL, Rozanski EA. Von Willebrand Factor antigen concentration in dogs with sepsis. J Vet Intern Med. 2010;24(1):229–230. doi: 10.1111/j.1939-1676.2009.0436.x.
    1. Kasten KR, et al. T cells are potent early mediators of the host response to sepsis. Shock. 2010;34(4):327–336. doi: 10.1097/SHK.0b013e3181e14c2e.
    1. Dufour JH, et al. IFN-γ-Inducible Protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. J Immunol. 2002;168(7):3195–3204. doi: 10.4049/jimmunol.168.7.3195.
    1. Attia S, et al. Mortality in children with complicated severe acute malnutrition is related to intestinal and systemic inflammation: an observational cohort study. Am J Clin Nutr. 2016;104(5):1441–1449. doi: 10.3945/ajcn.116.130518.
    1. Reynolds JV, et al. Impaired gut barrier function in malnourished patients. Br J Surg. 1996;83(9):1288–1291. doi: 10.1002/bjs.1800830934.
    1. Amadi B, et al. Impaired barrier function and autoantibody generation in malnutrition enteropathy in Zambia. EBioMedicine. 2017;22:191–199. doi: 10.1016/j.ebiom.2017.07.017.
    1. Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395:763. doi: 10.1038/27376.
    1. Klok MD, Jakobsdottir S, Drent ML. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: a review. Obes Rev. 2007;8(1):21–34. doi: 10.1111/j.1467-789X.2006.00270.x.
    1. Lord GM, et al. Leptin modulates the T-cell immune response and reverses starvation-induced immunosuppression. Nature. 1998;394:897. doi: 10.1038/29795.
    1. SÁNchez-Margalet V, et al. Role of leptin as an immunomodulator of blood mononuclear cells: mechanisms of action. Clin Exp Immunol. 2003;133(1):11–19. doi: 10.1046/j.1365-2249.2003.02190.x.
    1. Santos-Alvarez J, Goberna R, Sánchez-Margalet V. Human leptin stimulates proliferation and activation of human circulating monocytes. Cell Immunol. 1999;194(1):6–11. doi: 10.1006/cimm.1999.1490.
    1. Mody A, et al. Effects of HIV infection on the metabolic and hormonal status of children with severe acute malnutrition. PLoS One. 2014;9(7):e102233. doi: 10.1371/journal.pone.0102233.
    1. Popa C, et al. Markers of inflammation are negatively correlated with serum leptin in rheumatoid arthritis. Ann Rheum Dis. 2005;64(8):1195–8. doi: 10.1136/ard.2004.032243.
    1. Pérez-Pérez A, et al. Role of leptin as a link between metabolism and the immune system. Cytokine Growth Factor Rev. 2017;35:71–84. doi: 10.1016/j.cytogfr.2017.03.001.
    1. Abella V, et al. Leptin in the interplay of inflammation, metabolism and immune system disorders. Nat Rev Rheumatol. 2017;13:100. doi: 10.1038/nrrheum.2016.209.
    1. Ahima RS, Prabakaran D, Flier JS. Postnatal leptin surge and regulation of circadian rhythm of leptin by feeding. Implications for energy homeostasis and neuroendocrine function. J Clin Invest. 1998;101(5):1020–1027. doi: 10.1172/JCI1176.
    1. Ruggeri ZM. The role of von Willebrand Factor in thrombus formation. Thromb Res. 2007;120(Suppl 1):S5–S9. doi: 10.1016/j.thromres.2007.03.011.
    1. van Mourik JA, et al. von Willebrand Factor propeptide in vascular disorders: a tool to distinguish between acute and chronic endothelial cell perturbation. Blood. 1999;94(1):179–185.
    1. Gragnano F, et al. The role of von Willebrand Factor in vascular inflammation: from pathogenesis to targeted therapy. Mediators Inflamm. 2017;2017:5620314. doi: 10.1155/2017/5620314.
    1. Kremer Hovinga JA, et al. ADAMTS-13, von Willebrand factor and related parameters in severe sepsis and septic shock. J Thromb Haemost. 2007;5(11):2284–2290. doi: 10.1111/j.1538-7836.2007.02743.x.
    1. Cleland SJ, et al. Endothelial dysfunction as a possible link between C-reactive protein levels and cardiovascular disease. Clin Sci. 2000;98(5):531–535. doi: 10.1042/cs0980531.
    1. Hingorani AD, et al. Acute systemic inflammation impairs endothelium-dependent dilatation in humans. Circulation. 2000;102(9):994–9. doi: 10.1161/01.CIR.102.9.994.
    1. Stenvinkel P. Endothelial dysfunction and inflammation—is there a link? Nephrol Dial Transplant. 2001;16(10):1968–1971. doi: 10.1093/ndt/16.10.1968.
    1. Lu H, et al. Structure and functions of angiotensinogen. Hypertens Res. 2016;39:492. doi: 10.1038/hr.2016.17.
    1. Lee JW. Fluid and electrolyte disturbances in critically ill patients. Electrolyte Blood Press. 2010;8(2):72–81. doi: 10.5049/EBP.2010.8.2.72.
    1. Chistiakov DA, et al. Paraoxonase and atherosclerosis-related cardiovascular diseases. Biochimie. 2017;132:19–27. doi: 10.1016/j.biochi.2016.10.010.
    1. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protocols. 2008;4(1):44–57. doi: 10.1038/nprot.2008.211.
    1. WHO. Pocket book for hospital care of children: guidelines for the management of common illness with limited resources (2005).
    1. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotech. 2008;26(12):1367–1372. doi: 10.1038/nbt.1511.
    1. Cox J, et al. Andromeda: A peptide search engine integrated into the MaxQuant environment. J Proteome Res. 2011;10(4):1794–1805. doi: 10.1021/pr101065j.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol. 1995;57(1):289–300.
    1. Team, R. C. R: A language and environment for statistical computing. R Foundation for Statistical Computing (2017).
    1. Stata Corp. STATA survival analysis reference manual release 15, S. Press, Editor. Stata Press, p. 475 (2017).
    1. Vizcaino JA, et al. ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat Biotech. 2014;32(3):223–226. doi: 10.1038/nbt.2839.

Source: PubMed

3
Předplatit