Skip to main content
Top
Published in: Gut Pathogens 1/2023

Open Access 01-12-2023 | Clostridium | Research

Molecular characterization of Clostridium perfringens isolates from a tertiary children’s hospital in Guangzhou, China, establishing an association between bacterial colonization and food allergies in infants

Authors: Kun-yi Huang, Bing-shao Liang, Xiao-yan Zhang, Huan Chen, Ni Ma, Jiao-li Lan, Ding-You Li, Zhen-wen Zhou, Min Yang

Published in: Gut Pathogens | Issue 1/2023

Login to get access

Abstract

Background

Cow’s milk protein allergy (CMPA) is one of the most common types of food allergy in infants. Faecal pathogen cultures showed that the positive rate of Clostridium perfringens was more than 30%, which was significantly higher than that for other bacteria. Therefore, it is speculated that Clostridium perfringens colonization may be one of the pathogenetic factors for CMPA in infants. We conducted a real-world evidence study. Infants aged 0–6 months with diarrhoea and mucoid and/or bloody stools were recruited from a large tertiary hospital in China. Faecal pathogen cultures for the detection of Clostridium perfringens were confirmed by flight mass spectrometry, and potential toxin genes were identified using PCR. After 12 months of follow-up, the diagnoses of CMPA and food allergy were recorded. The correlation was assessed by Pearson correlation analysis.

Results

In this study, 358 infants aged 0–6 months with gastrointestinal symptoms and faecal pathogen cultures were recruited. A total of 270 (44.07% girls; mean age, 2.78 ± 2.84 months) infants were followed up for 12 months. Overall, the rate of positivity for Clostridium perfringens in faecal pathogen cultures was 35.75% (128/358) in infants aged ≤ 6 months. The earliest Clostridium perfringens colonization was detected within 2 days after birth. The majority of Clostridium perfringens isolates were classified as type C in 85 stool samples. In the Clostridium perfringens-positive group, 48.21% (54/112) of infants were clinically diagnosed with food allergies after 12 months, including 37.5% (42/112) with CMPA, which was significantly higher than that of the negative group, with 7.59% (12/158) exhibiting food allergies and 5.06% (8/158) presenting CMPA (P < 0.0001). Faecal Clostridium perfringens positivity was significantly correlated with CMPA, food allergy, faecal occult blood, faecal white blood cells, antibiotic use, increased peripheral blood platelet counts, and decreased haemoglobin levels (P < 0.0001).

Conclusions

This study demonstrates that intestinal colonization by Clostridium perfringens is common in infants. The majority of Clostridium perfringens isolates are classified as type C. Colonization of the intestine by Clostridium perfringens is associated with the development of CMPA and food allergy in infants.
Appendix
Available only for authorised users
Literature
1.
go back to reference Zhou B, Yuan Y, Zhang S, Guo C, Li X, Li G, et al. Intestinal flora and disease mutually shape the regional immune system in the intestinal tract. Front Immunol. 2020;11:575.CrossRefPubMedPubMedCentral Zhou B, Yuan Y, Zhang S, Guo C, Li X, Li G, et al. Intestinal flora and disease mutually shape the regional immune system in the intestinal tract. Front Immunol. 2020;11:575.CrossRefPubMedPubMedCentral
2.
go back to reference Lee MJ, Park YM, Kim B, Tae IH, Kim NE, Pranata M, et al. Disordered development of gut microbiome interferes with the establishment of the gut ecosystem during early childhood with atopic dermatitis. Gut Microbes. 2022;14:2068366.CrossRefPubMedPubMedCentral Lee MJ, Park YM, Kim B, Tae IH, Kim NE, Pranata M, et al. Disordered development of gut microbiome interferes with the establishment of the gut ecosystem during early childhood with atopic dermatitis. Gut Microbes. 2022;14:2068366.CrossRefPubMedPubMedCentral
3.
go back to reference Boutin RCT, Sbihi H, McLaughlin RJ, Hahn AS, Konwar KM, Loo RS, et al. Composition and associations of the infant gut fungal microbiota with environmental factors and childhood allergic outcomes. mBio. 2021;12:e0339620.CrossRefPubMed Boutin RCT, Sbihi H, McLaughlin RJ, Hahn AS, Konwar KM, Loo RS, et al. Composition and associations of the infant gut fungal microbiota with environmental factors and childhood allergic outcomes. mBio. 2021;12:e0339620.CrossRefPubMed
4.
go back to reference Aziz M, Prince JM, Wang P. Gut microbiome and necrotizing enterocolitis: understanding the connection to find a cure. Cell Host Microbe. 2022;30:612–6.CrossRefPubMed Aziz M, Prince JM, Wang P. Gut microbiome and necrotizing enterocolitis: understanding the connection to find a cure. Cell Host Microbe. 2022;30:612–6.CrossRefPubMed
5.
go back to reference Al Radaideh AJ, Badran EF, Shehabi AA. Diversity of toxin genotypes and antimicrobial susceptibility of Clostridium perfringens isolates from feces of infants. Germs. 2019;9:28–34.CrossRefPubMedPubMedCentral Al Radaideh AJ, Badran EF, Shehabi AA. Diversity of toxin genotypes and antimicrobial susceptibility of Clostridium perfringens isolates from feces of infants. Germs. 2019;9:28–34.CrossRefPubMedPubMedCentral
6.
go back to reference Tonooka T, Sakata S, Kitahara M, Hanai M, Ishizeki S, Takada M, et al. Detection and quantification of four species of the genus Clostridium in infant feces. Microbiol Immunol. 2005;49:987–92.CrossRefPubMed Tonooka T, Sakata S, Kitahara M, Hanai M, Ishizeki S, Takada M, et al. Detection and quantification of four species of the genus Clostridium in infant feces. Microbiol Immunol. 2005;49:987–92.CrossRefPubMed
7.
go back to reference Nagpal R, Tsuji H, Takahashi T, Nomoto K, Kawashima K, Nagata S, et al. Gut dysbiosis following C-section instigates higher colonisation of toxigenic Clostridium perfringens in infants. Benef Microbes. 2017;8:353–65.CrossRefPubMed Nagpal R, Tsuji H, Takahashi T, Nomoto K, Kawashima K, Nagata S, et al. Gut dysbiosis following C-section instigates higher colonisation of toxigenic Clostridium perfringens in infants. Benef Microbes. 2017;8:353–65.CrossRefPubMed
8.
go back to reference Fallani M, Rigottier-Gois L, Aguilera M, Bridonneau C, Collignon A, Edwards CA, et al. Clostridium difficile and Clostridium perfringens species detected in infant faecal microbiota using 16S rRNA targeted probes. J Microbiol Methods. 2006;67:150–61.CrossRefPubMed Fallani M, Rigottier-Gois L, Aguilera M, Bridonneau C, Collignon A, Edwards CA, et al. Clostridium difficile and Clostridium perfringens species detected in infant faecal microbiota using 16S rRNA targeted probes. J Microbiol Methods. 2006;67:150–61.CrossRefPubMed
9.
go back to reference Dittmar E, Beyer P, Fischer D, Schäfer V, Schoepe H, Bauer K, et al. Necrotizing enterocolitis of the neonate with Clostridium perfringens: diagnosis, clinical course, and role of alpha toxin. Eur J Pediatr. 2008;167:891–5.CrossRefPubMed Dittmar E, Beyer P, Fischer D, Schäfer V, Schoepe H, Bauer K, et al. Necrotizing enterocolitis of the neonate with Clostridium perfringens: diagnosis, clinical course, and role of alpha toxin. Eur J Pediatr. 2008;167:891–5.CrossRefPubMed
10.
go back to reference Ma Z, Chen L, Xian R, Fang H, Wang J, Hu Y. Time trends of childhood food allergy in China: three cross-sectional surveys in 1999, 2009, and 2019. Pediatr Allergy Immunol. 2021;32:1073–9.CrossRefPubMed Ma Z, Chen L, Xian R, Fang H, Wang J, Hu Y. Time trends of childhood food allergy in China: three cross-sectional surveys in 1999, 2009, and 2019. Pediatr Allergy Immunol. 2021;32:1073–9.CrossRefPubMed
11.
go back to reference Yang M, Tan M, Wu J, Chen Z, Long X, Zeng Y, et al. Prevalence, characteristics, and outcome of cow’s milk protein allergy in chinese infants: a population-based survey. JPEN J Parenter Enteral Nutr. 2019;43:803–8.CrossRefPubMed Yang M, Tan M, Wu J, Chen Z, Long X, Zeng Y, et al. Prevalence, characteristics, and outcome of cow’s milk protein allergy in chinese infants: a population-based survey. JPEN J Parenter Enteral Nutr. 2019;43:803–8.CrossRefPubMed
12.
go back to reference Nakano V, Ignacio A, Llanco L, Bueris V, Sircili MP, Avila-Campos MJ. Multilocus sequence typing analyses of Clostridium perfringens type a strains harboring tpeL and netB genes. Anaerobe. 2017;44:99–105.CrossRefPubMed Nakano V, Ignacio A, Llanco L, Bueris V, Sircili MP, Avila-Campos MJ. Multilocus sequence typing analyses of Clostridium perfringens type a strains harboring tpeL and netB genes. Anaerobe. 2017;44:99–105.CrossRefPubMed
13.
go back to reference Shaw AG, Cornwell E, Sim K, Thrower H, Scott H, Brown JCS, et al. Dynamics of toxigenic Clostridium perfringens colonisation in a cohort of prematurely born neonatal infants. BMC Pediatr. 2020;20:75.CrossRefPubMedPubMedCentral Shaw AG, Cornwell E, Sim K, Thrower H, Scott H, Brown JCS, et al. Dynamics of toxigenic Clostridium perfringens colonisation in a cohort of prematurely born neonatal infants. BMC Pediatr. 2020;20:75.CrossRefPubMedPubMedCentral
14.
go back to reference Parish WE. Evaluation of in vitro predictive tests for irritation and allergic sensitization. Food Chem Toxicol. 1986;24:481–94.CrossRefPubMed Parish WE. Evaluation of in vitro predictive tests for irritation and allergic sensitization. Food Chem Toxicol. 1986;24:481–94.CrossRefPubMed
15.
go back to reference Gaboriau-Routhiau V, Moreau MC. Oral tolerance to ovalbumin in mice: induction and long-term persistence unaffected by Staphylococcus aureus enterotoxin B and Clostridium perfringens type a enterotoxin. Pediatr Res. 1997;42:503–8.CrossRefPubMed Gaboriau-Routhiau V, Moreau MC. Oral tolerance to ovalbumin in mice: induction and long-term persistence unaffected by Staphylococcus aureus enterotoxin B and Clostridium perfringens type a enterotoxin. Pediatr Res. 1997;42:503–8.CrossRefPubMed
16.
go back to reference Morris WE, Fernández-Miyakawa ME. Toxins of Clostridium perfringens. Rev Argent Microbiol. 2009;41:251–60.PubMed Morris WE, Fernández-Miyakawa ME. Toxins of Clostridium perfringens. Rev Argent Microbiol. 2009;41:251–60.PubMed
17.
go back to reference Azimirad M, Gholami F, Yadegar A, Knight DR, Shamloei S, Aghdaei HA, et al. Prevalence and characterization of Clostridium perfringens toxinotypes among patients with antibiotic-associated diarrhea in Iran. Sci Rep. 2019;9:7792.CrossRefPubMedPubMedCentral Azimirad M, Gholami F, Yadegar A, Knight DR, Shamloei S, Aghdaei HA, et al. Prevalence and characterization of Clostridium perfringens toxinotypes among patients with antibiotic-associated diarrhea in Iran. Sci Rep. 2019;9:7792.CrossRefPubMedPubMedCentral
18.
go back to reference Vaishnavi C, Kaur S. Clostridium perfringens enterotoxin in antibiotic-associated diarrhea. Indian J Pathol Microbiol. 2008;51:198–9.CrossRefPubMed Vaishnavi C, Kaur S. Clostridium perfringens enterotoxin in antibiotic-associated diarrhea. Indian J Pathol Microbiol. 2008;51:198–9.CrossRefPubMed
19.
go back to reference Woo PC, Lau SK, Chan KM, Fung AM, Tang BS, Yuen KY. Clostridium bacteraemia characterised by 16S ribosomal RNA gene sequencing. J Clin Pathol. 2005;58:301–7.CrossRefPubMedPubMedCentral Woo PC, Lau SK, Chan KM, Fung AM, Tang BS, Yuen KY. Clostridium bacteraemia characterised by 16S ribosomal RNA gene sequencing. J Clin Pathol. 2005;58:301–7.CrossRefPubMedPubMedCentral
20.
go back to reference Gohari IM, Navarro MA, Li J, Shrestha A, Uzal F, McClane BA. Pathogenicity and virulence of Clostridium perfringens. Virulence. 2021;12:723–53.CrossRef Gohari IM, Navarro MA, Li J, Shrestha A, Uzal F, McClane BA. Pathogenicity and virulence of Clostridium perfringens. Virulence. 2021;12:723–53.CrossRef
21.
go back to reference Li Z, Yan C, Gong X, Wang J. Severe intravascular hemolysis from Clostridium perfringens septicemia in a neonate with necrotizing enterocolitis in China: a case report. Infect Drug Resist. 2022;15:1461–5.CrossRefPubMedPubMedCentral Li Z, Yan C, Gong X, Wang J. Severe intravascular hemolysis from Clostridium perfringens septicemia in a neonate with necrotizing enterocolitis in China: a case report. Infect Drug Resist. 2022;15:1461–5.CrossRefPubMedPubMedCentral
22.
go back to reference Vernacchio L, Vezina RM, Mitchell AA, Lesko SM, Plaut AG, Acheson DW. Diarrhea in american infants and young children in the community setting: incidence, clinical presentation and microbiology. Pediatr Infect Dis J. 2006;25:2–7.CrossRefPubMed Vernacchio L, Vezina RM, Mitchell AA, Lesko SM, Plaut AG, Acheson DW. Diarrhea in american infants and young children in the community setting: incidence, clinical presentation and microbiology. Pediatr Infect Dis J. 2006;25:2–7.CrossRefPubMed
23.
go back to reference Sung K, Kim JY, Lee YJ, Hwang EH, Park JH. High incidence of staphylococcus aureus and norovirus gastroenteritis in infancy: a single-center, 1-year experience. Pediatr Gastroenterol Hepatol Nutr. 2014;17:140–6.CrossRefPubMedPubMedCentral Sung K, Kim JY, Lee YJ, Hwang EH, Park JH. High incidence of staphylococcus aureus and norovirus gastroenteritis in infancy: a single-center, 1-year experience. Pediatr Gastroenterol Hepatol Nutr. 2014;17:140–6.CrossRefPubMedPubMedCentral
24.
go back to reference Mitchell LA, Koval M. Specificity of interaction between Clostridium perfringens enterotoxin and claudin-family tight junction proteins. Toxins (Basel). 2010;2:1595–611.CrossRefPubMed Mitchell LA, Koval M. Specificity of interaction between Clostridium perfringens enterotoxin and claudin-family tight junction proteins. Toxins (Basel). 2010;2:1595–611.CrossRefPubMed
25.
go back to reference Daneshmand A, Kermanshahi H, Mohammed J, Sekhavati MH, Javadmanesh A, Ahmadian M, et al. Intestinal changes and immune responses during Clostridium perfringens-induced necrotic enteritis in broiler chickens. Poult Sci. 2022;101:101652.CrossRefPubMed Daneshmand A, Kermanshahi H, Mohammed J, Sekhavati MH, Javadmanesh A, Ahmadian M, et al. Intestinal changes and immune responses during Clostridium perfringens-induced necrotic enteritis in broiler chickens. Poult Sci. 2022;101:101652.CrossRefPubMed
26.
go back to reference Lin JD, Devlin JC, Yeung F, McCauley C, Leung JM, Chen YH, et al. Rewilding Nod2 and Atg16l1 mutant mice uncovers genetic and environmental contributions to microbial responses and immune cell composition. Cell Host Microbe. 2020;27:830–40e4.CrossRefPubMedPubMedCentral Lin JD, Devlin JC, Yeung F, McCauley C, Leung JM, Chen YH, et al. Rewilding Nod2 and Atg16l1 mutant mice uncovers genetic and environmental contributions to microbial responses and immune cell composition. Cell Host Microbe. 2020;27:830–40e4.CrossRefPubMedPubMedCentral
27.
go back to reference Nakayama J, Kobayashi T, Tanaka S, Korenori Y, Tateyama A, Sakamoto N, et al. Aberrant structures of fecal bacterial community in allergic infants profiled by 16S rRNA gene pyrosequencing. FEMS Immunol Med Microbiol. 2011;63:397–406.CrossRefPubMed Nakayama J, Kobayashi T, Tanaka S, Korenori Y, Tateyama A, Sakamoto N, et al. Aberrant structures of fecal bacterial community in allergic infants profiled by 16S rRNA gene pyrosequencing. FEMS Immunol Med Microbiol. 2011;63:397–406.CrossRefPubMed
28.
go back to reference Kalliomäki M, Kirjavainen P, Eerola E, Kero P, Salminen S, Isolauri E. Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin Immunol. 2001;107:129–34.CrossRefPubMed Kalliomäki M, Kirjavainen P, Eerola E, Kero P, Salminen S, Isolauri E. Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin Immunol. 2001;107:129–34.CrossRefPubMed
29.
go back to reference Smehilová M, Vlková E, Nevoral J, Flajsmanová K, Killer J, Rada V. Comparison of intestinal microflora in healthy infants and infants with allergic colitis. Folia Microbiol (Praha). 2008;53:255–8.CrossRefPubMed Smehilová M, Vlková E, Nevoral J, Flajsmanová K, Killer J, Rada V. Comparison of intestinal microflora in healthy infants and infants with allergic colitis. Folia Microbiol (Praha). 2008;53:255–8.CrossRefPubMed
30.
go back to reference Roessler A, Forssten SD, Glei M, Ouwehand AC, Jahreis G. The effect of probiotics on faecal microbiota and genotoxic activity of faecal water in patients with atopic dermatitis: a randomized, placebo-controlled study. Clin Nutr. 2012;31:22–9.CrossRefPubMed Roessler A, Forssten SD, Glei M, Ouwehand AC, Jahreis G. The effect of probiotics on faecal microbiota and genotoxic activity of faecal water in patients with atopic dermatitis: a randomized, placebo-controlled study. Clin Nutr. 2012;31:22–9.CrossRefPubMed
31.
go back to reference Rood JI, Adams V, Lacey J, Lyras D, McClane BA, Melville SB, et al. Expansion of the Clostridium perfringens toxin-based typing scheme. Anaerobe. 2018;53:5–10.CrossRefPubMedPubMedCentral Rood JI, Adams V, Lacey J, Lyras D, McClane BA, Melville SB, et al. Expansion of the Clostridium perfringens toxin-based typing scheme. Anaerobe. 2018;53:5–10.CrossRefPubMedPubMedCentral
32.
go back to reference Harrison B, Raju D, Garmory HS, Brett MM, Titball RW, Sarker MR. Molecular characterization of Clostridium perfringens isolates from humans with sporadic diarrhea: evidence for transcriptional regulation of the beta2-toxin-encoding gene. Appl Environ Microbiol. 2005;71:8362–70.CrossRefPubMedPubMedCentral Harrison B, Raju D, Garmory HS, Brett MM, Titball RW, Sarker MR. Molecular characterization of Clostridium perfringens isolates from humans with sporadic diarrhea: evidence for transcriptional regulation of the beta2-toxin-encoding gene. Appl Environ Microbiol. 2005;71:8362–70.CrossRefPubMedPubMedCentral
Metadata
Title
Molecular characterization of Clostridium perfringens isolates from a tertiary children’s hospital in Guangzhou, China, establishing an association between bacterial colonization and food allergies in infants
Authors
Kun-yi Huang
Bing-shao Liang
Xiao-yan Zhang
Huan Chen
Ni Ma
Jiao-li Lan
Ding-You Li
Zhen-wen Zhou
Min Yang
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Gut Pathogens / Issue 1/2023
Electronic ISSN: 1757-4749
DOI
https://doi.org/10.1186/s13099-023-00572-x

Other articles of this Issue 1/2023

Gut Pathogens 1/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine