Skip to main content
Advertisement
  • Loading metrics

Enteric infection and dysfunction—A new target for PLOS Neglected Tropical Diseases

  • Michael B. Arndt ,

    MArndt@uw.edu

    Affiliations PATH, Seattle, Washington, United States of America, Department of Global Health, University of Washington, Seattle, Washington, United States of America

  • Judd L. Walson

    Affiliations Department of Global Health, University of Washington, Seattle, Washington, United States of America, Department of Epidemiology, University of Washington, Seattle, Washington, United States of America, Department of Medicine, University of Washington, Seattle, Washington, United States of America, Department of Pediatrics, University of Washington, Seattle, Washington, United States of America, Childhood Acute Illness and Nutrition Network, Nairobi, Kenya

The past decade has seen a dramatic increase in global attention to neglected tropical diseases, with remarkable momentum in policy, programs, funding, and research. As we look to the future, it is critical for PLOS Neglected Tropical Diseases to continue to reevaluate the scope of the journal and to define which syndromes and diseases continue to disproportionately impact neglected populations in low-resource settings.

Many pathogens highlighted in PLOS Neglected Tropical Diseases have profound impacts on the gastrointestinal system. There is increasing recognition of the role of the intestine as the critical site where the human host encounters pathogens, initiates the immune response, protects from pathogen invasion, controls nutrient and calorie absorption, and modulates hormonal response. Enteric dysfunction—as a result of diarrheal disease, parasitic infections, alterations in the gut microbial environment, and other causes—has been linked to malnutrition, delayed cognitive development, increased susceptibility to other infectious diseases, poor oral vaccine response, and increased mortality risk. The collective long-term impacts of these conditions on the most marginalized communities are tremendous. Given the importance of these diseases and syndromes, PLOS Neglected Tropical Diseases has decided to include a new section focused on enteric infections and enteric dysfunction. Of course, many diseases and syndromes—including the soil-transmitted helminths, schistosomiasis, giardiasis, cholera, and other enteric pathogens that impact the physiology of the gut—are already included within the scope of PLOS Neglected Tropical Diseases and have been highlighted previously in this journal [1]. It is our hope that by highlighting the shared impacts of enteric infection and dysfunction as a specific target for the journal, we can serve as a major platform to bring together valuable discoveries and innovations that will drive progress in this important field.

Diarrhea remains a leading cause of child mortality globally. The Global Enteric Multicenter Study (GEMS) reported Shigella spp., rotavirus, adenovirus, heat-stable toxin enterotoxigenic Escherichia coli (ST-ETEC), Cryptosporidium spp., and Campylobacter spp. as the most common pathogens associated with moderate-to-severe diarrhea in infants and young children in low-resource settings [2, 3]. However, mounting evidence also suggests that infection with enteric pathogens need not cause diarrhea to pose threats to child health. Even in the absence of diarrhea, many pathogens can increase gut inflammation and permeability and may result in systemic inflammation [4]. For example, high Campylobacter pathogen burden was associated with a significant decrease in height for age z-score among children enrolled in a multisite birth cohort study (MAL-ED), independent of diarrhea symptoms [5]. In addition to negative growth impacts, there is mounting evidence that frequent enteric infections and poor gut health in infants and children may also impair cognitive development and increase risk of metabolic and cardiovascular diseases later in life [68]. Although innovations such as low-osmolarity oral rehydration solution and zinc have greatly reduced the acute mortality from diarrhea in low-resource settings, interventions to reduce these long-term sequelae associated with repeated symptomatic and asymptomatic exposure to enteric pathogens are limited.

In many low-resource environments, environmental enteric dysfunction (EED)—a small bowel intestinal disorder characterized by mucosal inflammation, reduced barrier integrity, and nutrient malabsorption (e.g., villus blunting)—is widespread among infants and children, including children with no other identified comorbidity [911]. Noninvasive biomarkers reflective of intestinal inflammation, intestinal permeability, and microbial translocation appear to be elevated among more than half of all children in many settings. In addition to direct effects within the gut, EED can lead to translocation of bacterial products across the compromised intestinal barrier, with resulting systemic inflammation. Such inflammation appears to interfere with the growth-hormone (GH) axis and leads to a state of GH resistance that ultimately suppresses growth. GH resistance, defined by elevated systemic GH levels and low levels of insulin-like growth factor 1 (IGF-1; the hormone principally responsible for stimulating local bone and tissue growth), is exhibited in states of caloric and protein undernutrition, isolated micronutrient deficiencies (zinc, vitamin A, magnesium), and in response to chronic systemic inflammation [1216]. Evidence for the relationships between EED, systemic inflammation, and GH resistance in children comes from studies of young children in Brazil and Zimbabwe in which blood-based biomarkers of systemic inflammation (e.g., C-reactive protein and Alpha-1-acid glycoprotein) were negatively associated with systemic IGF-1 (and IGF binding protein 3) and linear growth, and in Brazil, positively associated with stool myeloperoxidase, a marker of intestinal inflammation [13, 16].

However, despite the highly prevalent nature of EED and the associated morbidity and mortality risk, there are major gaps in our understanding of EED and enteric dysfunction across the entire spectrum of translational research. PLOS Neglected Tropical Diseases is uniquely positioned to highlight research addressing these gaps from basic research to the implementation of interventions at scale.

Because the etiology and physiology of enteric dysfunction is incompletely defined, it is essential that model systems (e.g., animal models, organoids, in vitro cell models) be developed to elucidate some of the underlying features of the condition. Few published studies have evaluated the histopathologic features of EED. One such study produced a mouse model of EED using a combination of protein-deficient diet and exposure to a specific microbial environment [17]. Ongoing work using this model is exploring the impact of EED histopathology on brain development, both through direct (dissection) and indirect (behavioral) methods. Further work must be done to determine whether this model is sufficiently robust to serve as a preclinical model for EED interventions and/or oral vaccine efficacy in low-resource settings and to screen clinical candidates prior to use in phase 1 studies. It also remains unclear to what extent changes in the gut are protective and appropriate rather than destructive and harmful to the health of the host. To date, limited research has explored the potential role of host genetics in inflammatory, metabolic, and immune pathways involved in enteric dysfunction. In addition, it is not clear whether there is interplay between the composition and activity of the gut microbiome and the severity of morbidity experienced by children with EED. Despite evidence suggesting the importance of the microbiome in the normal development of endocrine, digestive, and immune systems, the optimal microbial composition of the gut remains unclear [18]. Identification of therapeutic or preventative targets for EED could draw heavily upon the insights derived from research into these areas.

There remain major challenges in establishing formal case definition(s) for enteric dysfunction that can be used in clinical trials and surveillance studies. The lack of well-validated biomarkers suitable for use in low-resource settings and predictive of morbid sequelae limits the ability of clinicians to diagnose and manage EED. The optimal method to assess gut abnormalities is endoscopy with biopsy to determine villus blunting and other histopathologic features. However, these invasive procedures may not be practical or ethical to utilize among children in low-resource settings. Because several forms of enteric dysfunction are often coendemic in low-resource settings, it is critical to harmonize case definitions to ensure proper classification of similar clinical entities from differing etiologies, because the etiology is likely relevant for elicitation of response to a given treatment approach. A combination of anthropometric, intestinal biomarker, and systemic biomarker information could potentially be used as the foundation for a case definition for EED [19]; however, more data are needed to form consensus around the ideal criteria. A case definition may also be helpful in focusing field and laboratory research on outcomes of enteric infections other than acute diarrhea, such as linear growth faltering, impairment in cognitive development, and later-life cardiovascular and metabolic disease [6]. Outside of clinical research, a case definition would provide information on the geographic distribution of EED and could enable the identification of potential risk factors that cluster geospatially. Such a case definition would also be instrumental in quantifying the long-term societal and economic impacts of EED (and chronic enteropathogen infections) in low-resource settings. An improved understanding of the epidemiology of EED will be useful in establishing whether EED is a syndrome or a disease and will be useful in guiding the approach to evaluating treatment and/or prevention interventions.

Many candidate biomarkers that can be analyzed noninvasively from stool, blood, and urine samples have been evaluated, and several have been associated with linear growth and other health outcomes [20]. Noninvasive markers indicative of intestinal inflammation (fecal neopterin, myeloperoxidase, and calprotectin), barrier disruption (fecal alpha-1-antitrypsin), and intestinal damage and repair (serum intestinal fatty acid binding protein, serum glucagon-like peptide 2, and fecal regenerating family member 1- beta [Reg1β]) [2124] have been associated with reduced growth and adverse health outcomes. The dual sugar permeability test, which measures the ratio of urinary lactulose excretion to rhamnose (or mannitol) excretion, following fasting and saccharide administration, is a measure of intestinal barrier function that has long been utilized as a biomarker for EED [25, 26]. Numerous blood-based biomarkers of systemic inflammation have been correlated with linear growth faltering and/or oral vaccine failure, including acute phase proteins (i.e., alpha-1-glycoprotein, ferritin, and C-reactive protein), innate immune response (soluble cluster of differentiation [CD]14), indoleamine 2, 3-dioxygenase 1 (IDO1) activity (kynurenine and tryptophan), and cytokines (e.g., interleukin [IL]-1b and IL-10) [4, 2729]. However, relationships between these markers and health outcomes have not been observed consistently across studies conducted in different geographies and age groups, and this heterogeneity is not yet well understood [30]. It is unclear whether there are important differences in etiology underlying enteric dysfunction in each setting, and although some studies have sought to combine information from different markers into composite scores [22, 31, 32], methods and markers have differed. These fecal, urinary, and systemic biomarkers nonetheless represent potential tools for diagnosing EED and stratifying child populations with increased morbidity risk into clinical trials with therapeutic candidates.

A number of interventional approaches for enteric dysfunction have been tested in children over the past 10 to 15 years, including water, sanitation, and hygiene (WASH) interventions; zinc; antibiotics; probiotics; and the targeted administration of locally active anti-inflammatory drugs. Most of these studies have used the dual sugar permeability test as an outcome measure, with many focusing on lactulose excretion alone (rather than the ratio with mannitol or rhamnose) due to its reduced complexity. Neither a probiotic (Lactobacillus rhamnosus GG) nor antibiotic (rifaximin) reduced lactulose excretion in children living in Malawi [33, 34]. Although an initial study observed a significant reduction in the change in lactulose:mannitol (L:M) ratio in Malawian children receiving albendazole or zinc tablets, a follow-up study did not observe reduced L:M ratio or lactulose excretion after 12 to 24 weeks in children given a combination of zinc, albendazole, and multiple micronutrients in comparison with children given a placebo [35, 36]. A study in 3-to-9-month-old Gambian infants also did not observe a reduction in lactulose excretion or intestinal inflammation (fecal calprotectin levels) in children supplemented with long-chain polyunsaturated fatty acids (fish oil) [37]. A small pilot study in young Kenyan children with severe acute malnutrition and stunting concluded that mesalamine was safe to administer in the population and observed modest reductions in markers of intestinal inflammation (fecal calprotectin) and systemic inflammation (plasma Immunoglobulin G to endotoxin core antibody) in the treatment arm but reported better nutritional recovery in the placebo arm [38]. An upcoming publication will report on the results of a clinical trial in Malawi that tested the effect of lactoferrin and lysozyme administration on intestinal integrity and growth in infants [39]. Results from two major cluster-randomized studies of WASH interventions, the WASH Benefits trial (Bangladesh and Kenya) and SHINE (Zimbabwe), examined whether such interventions impact growth, gut integrity, or inflammation. Neither trial has reported improvements in child linear growth, diarrhea, or cognitive scores in individual water, sanitation, hygiene, or combined WASH arms compared with their control groups [40, 41], and secondary analyses of other outcomes are currently being analyzed or are in press. Plausible explanations for the lack of health improvements in these WASH trials come from analyses of environmental samples collected in Kenya that suggest the great importance of child contact with surface water, soil, and public surfaces as sources of enteric pathogen exposure [42, 43]. Additional innovation(s) may be necessary to effectively alter the pertinent behavioral, environmental, and spatial conditions to disrupt enteric pathogen exposure and prevent associated health outcomes.

PLOS Neglected Tropical Diseases looks forward to publishing the results of studies and trials targeting the definition, classification, mechanisms, treatment, and/or prevention of enteric infection and dysfunction. PLOS Neglected Tropical Diseases is prepared to serve as a platform to bring together and disseminate important discoveries and innovations that will drive solutions to these enteric conditions that disproportionately impact populations in low-resource settings.

References

  1. 1. Bartelt LA, Lima AA, Kosek M, Peñataro Yori P, Lee G, Guerrant RL. "Barriers" to child development and human potential: the case for including the "neglected enteric protozoa" (NEP) and other enteropathy-associated pathogens in the NTDs. PLoS Negl Trop Dis. 2013;7(4):e2125. pmid:23593514
  2. 2. Kotloff KL, Nataro JP, Blackwelder WC, Nasrin D, Farag TH, Panchalingam S, et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case-control study. Lancet. 2013;382(9888):209–22. pmid:23680352
  3. 3. Liu J, Platts-Mills JA, Juma J, Kabir F, Nkeze J, Okoi C, et al. Use of quantitative molecular diagnostic methods to identify causes of diarrhoea in children: a reanalysis of the GEMS case-control study. Lancet. 2016;388(10051):1291–301. pmid:27673470
  4. 4. Kosek MN, Investigators M-EN. Causal Pathways from Enteropathogens to Environmental Enteropathy: Findings from the MAL-ED Birth Cohort Study. EBioMedicine. 2017;18:109–17. pmid:28396264
  5. 5. Amour C, Gratz J, Mduma E, Svensen E, Rogawski ET, McGrath M, et al. Epidemiology and Impact of Campylobacter Infection in Children in 8 Low-Resource Settings: Results From the MAL-ED Study. Clin Infect Dis. 2016.
  6. 6. Nataro JP, Guerrant RL. Chronic consequences on human health induced by microbial pathogens: Growth faltering among children in developing countries. Vaccine. 2017;35(49 Pt A):6807–12.
  7. 7. Lee GO, Olortegui MP, Salas MS, Yori PP, Trigoso DR, Kosek P, et al. Environmental enteropathy is associated with cardiometabolic risk factors in Peruvian children. J Dev Orig Health Dis. 2017;8(3):337–48. pmid:28264759
  8. 8. Guerrant RL, DeBoer MD, Moore SR. The impoverished gut—a triple burden of diarrhoea, stunting and chronic disease. Nature Reviews. 2013.
  9. 9. Keusch GT, Denno DM, Black RE, Duggan C, Guerrant RL, Lavery JV, et al. Environmental Enteric Dysfunction: Pathogenesis, Diagnosis, and Clinical Consequences. Clinical Infectious Diseases. 2014;59(suppl 4):S207–S12.
  10. 10. Korpe PS, Petri WA. Environmental enteropathy: critical implications of a poorly understood condition. Trends Mol Med. 2012;18(6):328–36. pmid:22633998
  11. 11. Prendergast A, Kelly P. Enteropathies in the developing world: neglected effects on global health. Am J Trop Med Hyg. 2012;86(5):756–63. pmid:22556071
  12. 12. Jones AD, Rukobo S, Chasekwa B, Mutasa K, Ntozini R, Mbuya MN, et al. Acute illness is associated with suppression of the growth hormone axis in Zimbabwean infants. Am J Trop Med Hyg. 2015;92(2):463–70. pmid:25535308
  13. 13. Prendergast A, S, Rukobo S, Rukobo r, Chasekwa B, Mutasa K, et al. Stunting is characterized by chronic inflammation in Zimbabwean infants. PLoS ONE. 2014;9(2):e86928. pmid:24558364
  14. 14. Fazeli P, Klibanski A. Determinants of GH resistance in malnutrition. The Journal of Endocrinology. 2014;220(3):R57–65. pmid:24363451
  15. 15. Fazeli P, Misra M, Goldstein M, Miller K, Klibanski A. Fibroblast growth factor-21 may mediate growth hormone resistance in anorexia nervosa. The Journal of Clinical Endocrinology and Metabolism. 2010;95(1):369–74. pmid:19926712
  16. 16. DeBoer MD, Scharf RJ, Leite AM, Férrer A, Havt A, Pinkerton R, et al. Systemic inflammation, growth factors, and linear growth in the setting of infection and malnutrition. Nutrition. 2017;33:248–53. pmid:27712965
  17. 17. Brown EM, Wlodarska M, Willing BP, Vonaesch P, Han J, Reynolds LA, et al. Diet and specific microbial exposure trigger features of environmental enteropathy in a novel murine model. Nat Commun. 2015;6:7806. pmid:26241678
  18. 18. Schwarzer M, Makki K, Storelli G, Machuca-Gayet I, Srutkova D, Hermanova P, et al. Lactobacillus plantarum strain maintains growth of infant mice during chronic undernutrition. Science. 2016;351(6275):854–7. pmid:26912894
  19. 19. Denno DM, Tarr PI, Nataro JP. Environmental Enteric Dysfunction: A Case Definition for Intervention Trials. Am J Trop Med Hyg. 2017.
  20. 20. McGrath CJ, Arndt MB, Walson JL. Biomarkers to Stratify Risk Groups among Children with Malnutrition in Resource-Limited Settings and to Monitor Response to Intervention. Horm Res Paediatr. 2017.
  21. 21. Peterson KM, Buss J, Easley R, Yang Z, Korpe PS, Niu F, et al. REG1B as a predictor of childhood stunting in Bangladesh and Peru. Am J Clin Nutr. 2013;97(5):1129–33. pmid:23553156
  22. 22. Kosek M, Haque R, Lima A, Babji S, Shrestha S, Qureshi S, et al. Fecal markers of intestinal inflammation and permeability associated with the subsequent acquisition of linear growth deficits in infants. Am J Trop Med Hyg. 2013;88(2):390–6. pmid:23185075
  23. 23. Guerrant RL, Leite AM, Pinkerton R, Medeiros PH, Cavalcante PA, DeBoer M, et al. Biomarkers of Environmental Enteropathy, Inflammation, Stunting, and Impaired Growth in Children in Northeast Brazil. PLoS ONE. 2016;11(9):e0158772. pmid:27690129
  24. 24. Arndt MB, Richardson BA, Ahmed T, Mahfuz M, Haque R, John-Stewart GC, et al. Fecal Markers of Environmental Enteropathy and Subsequent Growth in Bangladeshi Children. Am J Trop Med Hyg. 2016;95(3):694–701. pmid:27352872
  25. 25. Denno DM, VanBuskirk K, Nelson ZC, Musser CA, Hay Burgess DC, Tarr PI. Use of the Lactulose to Mannitol Ratio to Evaluate Childhood Environmental Enteric Dysfunction: A Systematic Review. Clinical Infectious Diseases. 2014;59(suppl 4):S213–S9.
  26. 26. Faubion W, Camilleri M, Murray J, Kelly P, Amadi B, Kosek M, et al. Improving the detection of environmental enteric dysfunction: a lactulose, rhamnose assay of intestinal permeability in children aged under 5 years exposed to poor sanitation and hygiene. BMJ Global Health. 2016;1(1):e000066. pmid:28588929
  27. 27. Kosek MN, Mduma E, Kosek PS, Lee GO, Svensen E, Pan WK, et al. Plasma Tryptophan and the Kynurenine-Tryptophan Ratio are Associated with the Acquisition of Statural Growth Deficits and Oral Vaccine Underperformance in Populations with Environmental Enteropathy. Am J Trop Med Hyg. 2016;95(4):928–37. pmid:27503512
  28. 28. Naylor C, Lu M, Haque R, Mondal D, Buonomo E, Nayak U, et al. Environmental Enteropathy, Oral Vaccine Failure and Growth Faltering in Infants in Bangladesh. EBioMedicine. 2015;2(11):1759–66. pmid:26870801
  29. 29. Prendergast A, Rukobo S, Chasekwa B, Mutasa K, Ntozini R, Mbuya M, et al. Stunting Is Characterized by Chronic Inflammation in Zimbabwean Infants. PLoS ONE. 2014;9(2):e86928. pmid:24558364
  30. 30. Harper KM, Mutasa M, Prendergast AJ, Humphrey J, Manges AR. Environmental enteric dysfunction pathways and child stunting: A systematic review. PLoS Negl Trop Dis. 2018;12(1):e0006205. pmid:29351288
  31. 31. Campbell RK, Schulze KJ, Shaikh S, Raqib R, Wu LSF, Ali H, et al. Environmental enteric dysfunction and systemic inflammation predict reduced weight but not length gain in rural Bangladeshi children. Br J Nutr. 2018;119(4):407–14. pmid:29498344
  32. 32. Campbell RK, Schulze KJ, Shaikh S, Mehra S, Ali H, Wu L, et al. Biomarkers of Environmental Enteric Dysfunction Among Children in Rural Bangladesh. J Pediatr Gastroenterol Nutr. 2017;65(1):40–6. pmid:28644348
  33. 33. Galpin L, Manary M, Fleming K, Ou C-N, Ashorn P, Shulman R. Effect of Lactobacillus GG on intestinal integrity in Malawian children at risk of tropical enteropathy. The American journal of clinical nutrition. 2005;82(5):1040–5. pmid:16280436
  34. 34. Trehan I, Shulman R, Ou C-N, Maleta K, Manary M. A Randomized, Double-Blind, Placebo-Controlled Trial of Rifaximin, a Nonabsorbable Antibiotic, in the Treatment of Tropical Enteropathy. The American Journal of Gastroenterology. 2009;104(9):2326–33. pmid:19491826
  35. 35. Wang AZ, Shulman RJ, Crocker AH, Thakwalakwa C, Maleta KM, Devaraj S, et al. A Combined Intervention of Zinc, Multiple Micronutrients, and Albendazole Does Not Ameliorate Environmental Enteric Dysfunction or Stunting in Rural Malawian Children in a Double-Blind Randomized Controlled Trial. J Nutr. 2016.
  36. 36. Ryan KN, Stephenson KB, Trehan I, Shulman RJ, Thakwalakwa C, Murray E, et al. Zinc or Albendazole Attenuates the Progression of Environmental Enteropathy: A Randomized Controlled Trial. Clinical Gastroenterology and Hepatology. 2014.
  37. 37. van der Merwe LF, Moore SE, Fulford AJ, Halliday KE, Drammeh S, Young S, et al. Long-chain PUFA supplementation in rural African infants: a randomized controlled trial of effects on gut integrity, growth, and cognitive development. Am J Clin Nutr. 2013;97(1):45–57. pmid:23221579
  38. 38. Jones KD, Hünten-Kirsch B, Laving AM, Munyi CW, Ngari M, Mikusa J, et al. Mesalazine in the initial management of severely acutely malnourished children with environmental enteric dysfunction: a pilot randomized controlled trial. BMC Med. 2014;12:133. pmid:25189855
  39. 39. Cheng WD, Wold KJ, Benzoni NS, Thakwalakwa C, Maleta KM, Manary MJ, et al. Lactoferrin and lysozyme to reduce environmental enteric dysfunction and stunting in Malawian children: study protocol for a randomized controlled trial. Trials. 2017;18(1):523. pmid:29110675
  40. 40. Null C, Stewart CP, Pickering AJ, Dentz HN, Arnold BF, Arnold CD, et al. Effects of water quality, sanitation, handwashing, and nutritional interventions on diarrhoea and child growth in rural Kenya: a cluster-randomised controlled trial. Lancet Glob Health. 2018;6(3):e316–e29. pmid:29396219
  41. 41. Luby SP, Rahman M, Arnold BF, Unicomb L, Ashraf S, Winch PJ, et al. Effects of water quality, sanitation, handwashing, and nutritional interventions on diarrhoea and child growth in rural Bangladesh: a cluster randomised controlled trial. Lancet Glob Health. 2018;6(3):e302–e15. pmid:29396217
  42. 42. N Medgyesi D, Sewell D, Senesac R, Cumming O, Mumma J, Baker K. The Landscape of Enteric Pathogen Exposure of Young Children in Public Domains of Low-Income, Urban Kenya: The Influence of Exposure Pathway and Spatial Range of Play on Multi-Pathogen Exposure Risks2018.
  43. 43. Baker KK, Senesac R, Sewell D, Sen Gupta A, Cumming O, Mumma J. Fecal Fingerprints of Enteric Pathogen Contamination in Public Environments of Kisumu, Kenya, Associated with Human Sanitation Conditions and Domestic Animals. Environ Sci Technol. 2018;52(18):10263–74. pmid:30106283