Summary

Epithelial Cell Infection Analyses with Shigella

Published: February 09, 2024
doi:

Summary

The present protocol describes infection assays to interrogate Shigella adherence, invasion, and intracellular replication using in vitro epithelial cell lines.

Abstract

The human-adapted enteric bacterial pathogen Shigella causes millions of infections each year, creates long-term growth effects among pediatric patients, and is a leading cause of diarrheal deaths worldwide. Infection induces watery or bloody diarrhea as a result of the pathogen transiting the gastrointestinal tract and infecting the epithelial cells lining the colon. With staggering increases in antibiotic resistance and the current lack of approved vaccines, standardized research protocols are critical to studying this formidable pathogen. Here, methodologies are presented to examine the molecular pathogenesis of Shigella using in vitro analyses of bacterial adherence, invasion, and intracellular replication in colonic epithelial cells. Prior to infection analyses, the virulence phenotype of Shigella colonies was verified by the uptake of the Congo red dye on agar plates. Supplemented laboratory media can also be considered during bacterial culturing to mimic in vivo conditions. Bacterial cells are then used in a standardized protocol to infect colonic epithelial cells in tissue culture plates at an established multiplicity of infection with adaptations to analyze each stage of infection. For adherence assays, Shigella cells are incubated with reduced media levels to promote bacterial contact with epithelial cells. For both invasion and intracellular replication assays, gentamicin is applied for various time intervals to eliminate extracellular bacteria and enable assessment of invasion and/or the quantification of intracellular replication rates. All infection protocols enumerate adherent, invaded, and/or intracellular bacteria by serially diluting infected epithelial cell lysates and plating bacterial colony forming units relative to infecting titers on Congo red agar plates. Together, these protocols enable independent characterization and comparisons for each stage of Shigella infection of epithelial cells to study this pathogen successfully.

Introduction

Diarrheal diseases caused by enteric bacterial pathogens are a significant global health burden. In 2016, diarrheal diseases were responsible for 1.3 million deaths worldwide and were the fourth leading cause of death in children younger than five years of age1,2. The Gram-negative, enteric bacterial pathogen Shigella is the causative agent of shigellosis, a major cause of diarrheal deaths worldwide3. Shigellosis causes significant morbidity and mortality each year in children from lower- and middle-income countries4,5, while infections in high-income countries are linked to daycare center, foodborne, and waterborne outbreaks6,7,8,9. Ineffective vaccine development10 and rising rates of antimicrobial resistance (AMR)11,12 have complicated the management of large-scale Shigella outbreaks. Recent Centers for Disease Control and Prevention data show that nearly 46% of Shigella infections in the United States displayed drug resistance in 202013,14, while the World Health Organization has declared Shigella as an AMR priority pathogen for which new therapies are urgently needed15.

Shigella infections are easily transmitted via the fecal-oral route upon ingestion of contaminated food or water, or through direct human contact. Shigella has evolved to be an efficient, human-adapted pathogen, with an infectious dose of 10-100 bacteria sufficient to cause disease16. During small intestinal transit, Shigella is exposed to environmental signals, such as elevated temperature and bile17. Detection of these signals induces transcriptional changes to express virulence factors that enhance the ability of the bacteria to infect the human colon17,18,19. Shigella does not invade the colonic epithelium from the apical surface, but rather transits across the epithelial layer following uptake into specialized antigen-presenting microfold cells (M cells) within the follicle-associated epithelium20,21,22. Following transcytosis, Shigella cells are phagocytosed by resident macrophages. Shigella rapidly escapes the phagosome and triggers macrophage cell death, resulting in the release of pro-inflammatory cytokines5,23,24. Shigella then invades colonic epithelial cells from the basolateral side, lyses the macropinocytic vacuole, and establishes a replicative niche in the cytoplasm5,25. Pro-inflammatory cytokines, particularly interleukin-8 (IL-8), recruit polymorphonuclear neutrophil leukocytes (PMNs) to the site of infection, which weakens epithelial tight junctions, and enables bacterial infiltration of the epithelial lining to exacerbate basolateral infection5. The PMNs destroy the infected epithelial lining to contain the infection, which results in the characteristic symptoms of bacillary (bloody) dysentery5. Although invasion and intracellular replication mechanisms have been thoroughly characterized, new research is demonstrating important new concepts in Shigella infection, including virulence regulation during gastrointestinal (GI) transit17, adherence19, improved basolateral access through barrier permeability26, and asymptomatic carriage in malnourished children27.

The ability of Shigella spp. to cause diarrheal disease is restricted to humans and non-human primates (NHP)28. Shigella intestinal infection models have been developed for zebrafish29, mice30, guinea pigs31, rabbits21,32,33, and pigs34,35. However, none of these model systems can accurately replicate the disease characteristics observed during human infection36. Although NHP models of shigellosis have been established to study Shigella pathogenesis, these model systems are expensive to implement and require artificially high infectious doses, up to nine orders of magnitude higher than the infectious dose of humans37,38,39,40,41,42. Thus, the remarkable adaptation of Shigella for infection of human hosts necessitates the use of human-derived cell cultures to recreate physiologically relevant models for accurate interrogation of Shigella pathogenesis.

Here, detailed procedures are described to measure the rates of Shigella adherence to, invasion of, and replication within HT-29 colonic epithelial cells. Using these standardized protocols, the molecular mechanisms by which bacterial virulence genes and environmental signals impact each step of Shigella infection can be interrogated to better understand the dynamic host-pathogen interaction relationship.

Protocol

1. Preparation of reagents and materials NOTE: All volumes are consistent with an assay using two 6-well plates. TSB medium: Add 0.5 L of deionized (DI) water to 15 g of Tryptic Soy Broth (TSB, see Table of Materials) medium and autoclave. Store at room temperature. Bile salts medium (TSB + BS): To prepare TSB containing 0.4% (w/v) bile salts, resuspend 0.06 g of bile salts (BS, see Table of Materials) in 15 mL of autocla…

Representative Results

Adherence, invasion, and intracellular replication assays were performed comparing S. flexneri 2457T wild type (WT) to S. flexneri ΔVF (ΔVF), a mutant hypothesized to negatively regulate Shigella virulence. Since Shigella uses bile salts as a signal to regulate virulence17,18,47, experiments were performed after bacterial subculture in TSB media as well as TSB supplemented w…

Discussion

This protocol describes a set of three standardized assays to study Shigella adherence, invasion, and intracellular replication of intestinal epithelial cells. Although these methods are merely modified versions of classical gentamicin assays used to study the invasion and intracellular replication of various bacterial pathogens within host cells49,50,51, special considerations must be applied when studying Shigella…

Disclosures

The authors have nothing to disclose.

Acknowledgements

Support for the authors includes Massachusetts General Hospital's Department of Pediatrics, the Executive Committee on Research Interim Support Funding (ISF) award 2022A009041, the National Institute of Allergy and Infectious Diseases grant R21AI146405, and the National Institute of Diabetes and Digestive and Kidney Diseases grant Nutrition Obesity Research Center at Harvard (NORCH) 2P30DK040561-26. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Materials

0.22 μm PES filter Millipore-Sigma SCGP00525 Sterile, polyethersulfone filter for sterilizing up to 50 mL media
14 mL culture tubes Corning 352059 17 mm x 100 mm polypropylene test tubes with cap
50 mL conical tubes Corning 430829 50 mL clear polypropylene conical bottom centrifuge tubes with leak-proof cap
6-well tissue culture plates Corning 3516 Plates are treated for optimal cell attachment
Bile salts Sigma-Aldrich B8756 1:1 ratio of cholate to deoxycholate
Congo red dye Sigma-Aldrich C6277 A benzidine-based anionic diazo dye, >85% purity
Countess cell counting chamber slide Invitrogen C10283 To be used with the Countess Automated Cell Counter
Dimethyl sulfoxide (DMSO) Sigma-Aldrich D8418 A a highly polar organic reagent
Dulbecco’s Modified Eagle Medium (DMEM) Gibco 10569-010 DMEM is supplemented with high glucose, sodium pyruvate, GlutaMAX, and Phenol Red
Fetal Bovine Serum (FBS) Sigma-Aldrich F4135 Heat-inactivated, sterile
Gentamicin Sigma-Aldrich G3632 Stock concentration is 50 mg/mL
HT-29 cell line ATCC HTB-38 Adenocarcinoma cell line; colorectal in origin
Paraffin film Bemis PM999 Laboratory sealing film
Petri dishes Thermo Fisher Scientific FB0875713 100 mm x 15 mm Petri dishes for solid media
Phosphate-buffered saline (PBS) Thermo Fisher Scientific 10010049 1x concentration; pH 7.4
Select agar Invitrogen 30391023 A mixture of polysaccharides extracted from red seaweed cell walls to make bacterial plating media
T75 flasks Corning 430641U Tissue culture flasks
Triton X-100 Sigma-Aldrich T8787 A common non-ionic surfactant and emulsifier 
Trypan blue stain Invitrogen T10282 A dye to detect dead tissue culture cells; only live cells can exclude the dye
Trypsin-EDTA Gibco 25200-056 Reagent for cell dissociation for cell line maintenance and passaging
Tryptic Soy Broth (TSB) Sigma-Aldrich T8907 Bacterial growth media

References

  1. Karambizi, N. U., McMahan, C. S., Blue, C. N., Temesvari, L. A. Global estimated Disability-Adjusted Life-Years (DALYs) of diarrheal diseases: A systematic analysis of data from 28 years of the global burden of disease study. PloS one. 16 (10), e0259077 (2021).
  2. WHO. WHO methods and data sources for country-level causes of death 2000-2016. World Health Organization. , (2018).
  3. Kotloff, K. L. Shigella infection in children and adults: a formidable foe. Lancet Glob Health. 5 (12), e1166-e1167 (2017).
  4. Kotloff, K. L., 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. 382 (9888), 209-222 (2013).
  5. Schroeder, G. N., Hilbi, H. Molecular pathogenesis of Shigella spp.: Controlling host cell signaling, invasion, and death by type III secretion. Clin Microbiol Rev. 21 (1), 134-156 (2008).
  6. Arvelo, W., et al. Transmission risk factors and treatment of pediatric shigellosis during a large daycare center-associated outbreak of multidrug resistant shigella sonnei: Implications for the management of shigellosis outbreaks among children. Pediatr Infect Dis J. 28 (11), 976-980 (2009).
  7. Kozyreva, V. K., et al. Recent outbreaks of Shigellosis in California caused by two distinct populations of Shigella sonnei with either increased virulence or fluoroquinolone resistance. mSphere. 1 (6), 1-18 (2016).
  8. Bowen, A., et al. Importation and domestic transmission of Shigella sonnei resistant to ciprofloxacin – United States, May 2014-February 2015. MMWR Morb Mortal Wkly Rep. 64 (12), 318-320 (2015).
  9. Tansarli, G. S., et al. Genomic reconstruction and directed interventions in a multidrug-resistant Shigellosis outbreak in Seattle, WA, USA: a genomic surveillance study. Lancet. 3099 (22), 1-11 (2023).
  10. Barry, E. M., et al. Progress and pitfalls in Shigella vaccine research. Nat Rev Gastroenterol Hepatol. 10 (4), 245-255 (2013).
  11. Increase in Extensively Drug-Resistant Shigellosis in the United States. CDC Health Alert Network. Centers for Disease Control and Prevention Available from: https://emergency.cdc.gov/han/2023/han00486.asp?ACSTrackingID=USCDC_511-DM100260&ACSTrackingLabel=HAN%20486%20-%20General%20Public&deliveryName=USCDC_511-DM100260 (2023)
  12. Shiferaw, B., et al. Antimicrobial susceptibility patterns of Shigella isolates in Foodborne Diseases Active Surveillance Network (FoodNet) sites, 2000-2010. Clin Infect Dis. 54, S458-S463 (2012).
  13. Centers for Disease Control and Prevention. COVID-19: U.S. Impact on Antimicrobial Resistance, Special Report 2022. Atlanta, GA: U.S. Department of Health and Human Services. CDC. , (2022).
  14. Centers for Disease Control and Prevention. Antibiotic resistance threats in the United States, 2019. CDC. 10 (1), (2019).
  15. WHO. Prioritization of pathogens to guide discovery, research and development of new antibiotics for drug-resistant bacterial infections, including tuberculosis. WHO. , (2017).
  16. DuPont, H. L., Levine, M. M., Hornick, R. B., Formal, S. B. Inoculum size in shigellosis and implications for expected mode of transmission. J Infect Dis. 159 (6), 1126-1128 (1989).
  17. Nickerson, K. P., et al. Analysis of Shigella flexneri resistance, biofilm formation, and transcriptional profile in response to bile salts. Infect Immun. 85 (6), 1-18 (2017).
  18. Faherty, C. S., Redman, J. C., Rasko, D. A. Shigella flexneri effectors OspE1 and OspE2 mediate induced adherence to the colonic epithelium following bile salts exposure. Mol Microbiol. 85 (1), 107-121 (2012).
  19. Chanin, R. B., et al. Shigella flexneri adherence factor expression in in vivo-like conditions. mSphere. 4 (6), e00751 (2019).
  20. Baranov, V., Hammarström, S. Carcinoembryonic antigen (CEA) and CEA-related cell adhesion molecule 1 (CEACAM1), apically expressed on human colonic M cells, are potential receptors for microbial adhesion. Histochem Cell Biol. 121 (2), 83-89 (2004).
  21. Wassef, J. S., Keren, D. F., Mailloux, J. L. Role of M cells in initial antigen uptake and in ulcer formation in the rabbit intestinal loop model of shigellosis. Infect Immun. 57 (3), 858-863 (1989).
  22. Sansonetti, P. J., Arondel, J., Cantey, J. R., Prévost, M. C., Huerre, M. Infection of rabbit Peyer’s patches by Shigella flexneri: Effect of adhesive or invasive bacterial phenotypes on follicle-associated epithelium. Infect Immun. 64 (7), 2752-2764 (1996).
  23. Sansonetti, P. J., et al. Caspase-1 activation of IL-1beta and IL-18 are essential for Shigella flexneri-induced inflammation. Immunity. 12 (5), 581-590 (2000).
  24. Zychlinsky, A., Fitting, C., Cavaillon, J. M., Sansonetti, P. J. Interleukin 1 is released by murine macrophages during apoptosis induced by Shigella flexneri. J Clin Invest. 94 (3), 1328-1332 (1994).
  25. Sansonetti, P. J., Ryter, A., Clerc, P., Maurelli, A. T., Mounier, J. Multiplication of Shigella flexneri within HeLa cells: lysis of the phagocytic vacuole and plasmid-mediated contact hemolysis. Infect Immun. 51 (2), 461-469 (1986).
  26. Maldonado-Contreras, A., et al. Shigella depends on SepA to destabilize the intestinal epithelial integrity via cofilin activation. Gut Microbes. 8 (6), 544-560 (2017).
  27. Collard, J. -. M., et al. High prevalence of small intestine bacteria overgrowth and asymptomatic carriage of enteric pathogens in stunted children in Antananarivo, Madagascar. PLoS Negl Trop Dis. 16 (5), e0009849 (2022).
  28. Mattock, E., Blocker, A. J. How do the virulence factors of shigella work together to cause disease. Front Cell Infect Microbiol. 7, 1-24 (2017).
  29. Mostowy, S., et al. The zebrafish as a new model for the in vivo study of Shigella flexneri interaction with phagocytes and bacterial autophagy. PLoS Pathog. 9 (9), e1003588 (2013).
  30. Martinez-Becerra, F. J., et al. Parenteral immunization with IpaB/IpaD protects mice against lethal pulmonary infection by Shigella. Vaccine. 31 (24), 2667-2672 (2013).
  31. Shim, D. -. H., et al. New animal model of shigellosis in the Guinea pig: its usefulness for protective efficacy studies. J Immunol. 178 (4), 2476-2482 (2007).
  32. Marteyn, B., et al. Modulation of Shigella virulence in response to available oxygen in vivo. Nature. 465 (7296), 355-358 (2010).
  33. West, N. P., et al. Optimization of virulence functions through glucosylation of Shigella LPS. Science. 307 (5713), 1313-1317 (2005).
  34. Maurelli, A. T., et al. Shigella infection as observed in the experimentally inoculated domestic pig, Sus scrofa domestica. Microbial Pathog. 25 (4), 189-196 (1998).
  35. Jeong, K. -. I., Zhang, Q., Nunnari, J., Tzipori, S. A piglet model of acute gastroenteritis induced by Shigella dysenteriae Type 1. J Infect Dis. 201 (6), 903-911 (2010).
  36. Kim, Y. -. J., Yeo, S. -. G., Park, J. -. H., Ko, H. -. J. Shigella vaccine development: prospective animal models and current status. Curr Pharm Biotechnol. 14 (10), 903-912 (2013).
  37. Kent, T. H., Formal, S. B., LaBrec, E. H., Sprinz, H., Maenza, R. M. Gastric shigellosis in rhesus monkeys. Am J Pathol. 51 (2), 259-267 (1967).
  38. Shipley, S. T., et al. A challenge model for Shigella dysenteriae 1 in cynomolgus monkeys (Macaca fascicularis). Comp Med. 60 (1), 54-61 (2010).
  39. Higgins, R., Sauvageau, R., Bonin, P. Shigella flexneri Type 2 Infection in captive nonhuman primates. Can Vet J. 26 (12), 402-403 (1985).
  40. Oaks, E. V., Hale, T. L., Formal, S. B. Serum immune response to Shigella protein antigens in rhesus monkeys and humans infected with Shigella spp. Infect Immun. 53 (1), 57-63 (1986).
  41. Formal, S. B., et al. Protection of monkeys against experimental shigellosis with a living attenuated oral polyvalent dysentery vaccine. J Bacteriol. 92 (1), 17-22 (1966).
  42. Levine, M. M., Kotloff, K. L., Barry, E. M., Pasetti, M. F., Sztein, M. B. Clinical trials of Shigella vaccines: two steps forward and one step back on a long, hard road. Nat Rev Microbiol. 5 (7), 540-553 (2007).
  43. Payne, S. M. Laboratory cultivation and storage of Shigella. Curr Protoc Microbiol. 55 (1), 93 (2019).
  44. NIH Guidelines. NIH guidelines for research involving recombinant or synthetic nucleic acid molecules. NIH Guidelines. 2, 142 (2019).
  45. Maurelli, A. T., Blackmon, B., Curtiss, R. Loss of pigmentation in Shigella flexneri 2a is correlated with loss of virulence and virulence-associated plasmid. Infect Immun. 43 (1), 397-401 (1984).
  46. HT-29 cell line product sheet. ATCC Available from: https://www.atcc.org/products/htb-38 (2023)
  47. Sistrunk, J. R., Nickerson, K. P., Chanin, R. B., Rasko, D. A., Faherty, C. S. Survival of the fittest: How bacterial pathogens utilize bile to enhance infection. Clin Microbiol Rev. 29 (4), 819-836 (2016).
  48. Stensrud, K. F., et al. Deoxycholate interacts with IpaD of Shigella flexneri in inducing the recruitment of IpaB to the type III secretion apparatus needle tip. J Biol Chem. 283 (27), 18646-18654 (2008).
  49. Mandell, G. L. Interaction of intraleukocytic bacteria and antibiotics. J Clin Invest. 52 (7), 1673-1679 (1973).
  50. Elsinghorst, E. A. Measurement of invasion by gentamicin resistance. Methods Enzymo. 236 (1979), 405-420 (1994).
  51. Elsinghorst, E. A., Weitz, J. A. Epithelial cell invasion and adherence directed by the enterotoxigenic Escherichia coli tib locus is associated with a 104-kilodalton outer membrane protein. Infect Immun. 62 (8), 3463-3471 (1994).
  52. Dorman, C. J., McKenna, S., Beloin, C. Regulation of virulence gene expression in Shigella flexneri, a facultative intracellular pathogen. Int J Med Microbiol. 291 (2), 89-96 (2001).
  53. Porter, M. E., Dorman, C. J. Positive regulation of Shigella flexneri virulence genes by integration host factor. J Bacteriol. 179 (21), 6537-6550 (1997).
  54. Maurelli, A. T., Blackmon, B., Curtiss, R. Temperature-dependent expression of virulence genes in Shigella species. Infect Immun. 43 (1), 195-201 (1984).
  55. Schuch, R., Maurelli, A. T. Virulence plasmid instability in Shigella flexneri 2a is induced by virulence gene expression. Infect Immun. 65 (9), 3686-3692 (1997).
  56. Formal, S. B., Hale, T. L., Sansonetti, P. J. Invasive enteric pathogens. Rev Infect Dis. 5, S702-S707 (1983).
  57. Pál, T., Hale, T. L. Plasmid-associated adherence of Shigella flexneri in a HeLa cell model. Infect Immun. 57 (8), 2580-2582 (1989).
  58. Noben, M., et al. Human intestinal epithelium in a dish: Current models for research into gastrointestinal pathophysiology. United European Gastroenterol J. 5 (8), 1073-1081 (2017).
  59. Liévin-Le Moal, V., Servin, A. L. Pathogenesis of human enterovirulent bacteria: lessons from cultured, fully differentiated human colon cancer cell lines. Microbiol Mol Biol Rev R. 77 (3), 380-439 (2013).
  60. Mitchell, D. M., Ball, J. M. Characterization of a spontaneously polarizing HT-29 cell line, HT-29/cl.f8. In Vitro Cell Dev Biol – Anim. 40 (10), 297-302 (2004).
  61. Gagnon, M., Zihler Berner, A., Chervet, N., Chassard, C., Lacroix, C. Comparison of the Caco-2, HT-29 and the mucus-secreting HT29-MTX intestinal cell models to investigate Salmonella adhesion and invasion. J Microbiol Methods. 94 (3), 274-279 (2013).
  62. Koestler, B. J., et al. Human intestinal enteroids as a model system of Shigella pathogenesis. Infect Immun. 87 (4), 00733 (2019).
  63. Ranganathan, S., et al. Evaluating Shigella flexneri pathogenesis in the human enteroid model. Infect Immun. 87 (4), (2019).
  64. Nickerson, K. P., et al. A versatile human intestinal organoid-derived epithelial monolayer model for the study of enteric pathogens. Microbiol Spectr. 9 (1), 1-17 (2021).
  65. Perlman, M., Senger, S., Verma, S., Carey, J., Faherty, C. S. A foundational approach to culture and analyze malnourished organoids. Gut Microbes. 15 (2), 2248713 (2023).
  66. Pope, L. M., Reed, K. E., Payne, S. M. Increased protein secretion and adherence to HeLa cells by Shigella spp. following growth in the presence of bile salts. Infect Immun. 63 (9), 3642-3648 (1995).
  67. Faherty, C. S., et al. The synthesis of OspD3 (ShET2) in Shigella flexneri is independent of OspC1. Gut Microbes. 7 (6), 486-502 (2016).
  68. Ridlon, J. M., Kang, D. -. J., Hylemon, P. B. Bile salt biotransformations by human intestinal bacteria. J Lipid Res. 47 (2), 241-259 (2006).
  69. Köseoğlu, V. K., Hall, C. P., Rodríguez-López, E. M., Agaisse, H. The Autotransporter IcsA promotes Shigella flexneri biofilm formation in the presence of bile salts. Infect Immun. 87 (7), 1-14 (2019).
check_url/66426?article_type=t

Play Video

Cite This Article
Poore, K., Lenneman, B. R., Faherty, C. S. Epithelial Cell Infection Analyses with Shigella. J. Vis. Exp. (204), e66426, doi:10.3791/66426 (2024).

View Video