Skip to main content
  • ASM Journals
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Latest Articles
    • Special Issues
    • COVID-19 Special Collection
    • Editor's Picks
    • Special Series: Sponsored Minireviews and Video Abstracts
    • Archive
  • Topics
    • Applied and Environmental Science
    • Ecological and Evolutionary Science
    • Host-Microbe Biology
    • Molecular Biology and Physiology
    • Novel Systems Biology Techniques
    • Early-Career Systems Microbiology Perspectives
  • For Authors
    • Getting Started
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics
  • About the Journal
    • About mSystems
    • Editor in Chief
    • Board of Editors
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • ASM Journals
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
mSystems
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Latest Articles
    • Special Issues
    • COVID-19 Special Collection
    • Editor's Picks
    • Special Series: Sponsored Minireviews and Video Abstracts
    • Archive
  • Topics
    • Applied and Environmental Science
    • Ecological and Evolutionary Science
    • Host-Microbe Biology
    • Molecular Biology and Physiology
    • Novel Systems Biology Techniques
    • Early-Career Systems Microbiology Perspectives
  • For Authors
    • Getting Started
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics
  • About the Journal
    • About mSystems
    • Editor in Chief
    • Board of Editors
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
Research Article | Host-Microbe Biology

Heritable Gut Microbiome Associated with Salmonella enterica Serovar Pullorum Infection in Chickens

Jinmei Ding, Hao Zhou, Lingxiao Luo, Lu Xiao, Kaixuan Yang, Lingyu Yang, Yuming Zheng, Ke Xu, Chuan He, Chengxiao Han, Huaixi Luo, Chao Qin, Fisayo T. Akinyemi, Caiju Gu, Zhenxiang Zhou, Qizhong Huang, He Meng
Holly Bik, Editor
Jinmei Ding
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hao Zhou
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lingxiao Luo
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lu Xiao
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kaixuan Yang
bAnimal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lingyu Yang
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yuming Zheng
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ke Xu
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Chuan He
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Chengxiao Han
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Huaixi Luo
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Chao Qin
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Fisayo T. Akinyemi
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Caiju Gu
bAnimal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zhenxiang Zhou
bAnimal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Qizhong Huang
bAnimal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
He Meng
aShanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for He Meng
Holly Bik
University of Georgia
Roles: Editor
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/mSystems.01192-20
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Pullorum disease is one of the most common diarrhea-related diseases caused by Salmonella enterica subspecies enterica serovar Gallinarum biovar Pullorum (S. Pullorum); it negatively affects the poultry industry. However, limited studies have explored the association between the gut microbiota and S. Pullorum infection in chickens. In the present study, we performed a microbiome comparison and a microbiome genome-wide association study (mGWAS) to investigate the association among the host genetics, the gut microbiota, and pullorum disease in chickens. We found that S. Pullorum infection in chickens could alter the abundance of 39 bacterial genera (P < 0.05). The altered structure and composition of the gut microbiota were also detected in the offspring. mGWAS results revealed host genetic variants to be prominently associated with gut microbial diversity and individual microbes. The pathogens Pelomonas and Brevundimonas, which had a high abundance in positive parent chickens and their offspring, were significantly associated with several genetic mutations in immunity-related genes, such as TGIF1, TTLL12, and CCR7. This finding explained why Pelomonas and Brevundimonas were heritable in S. Pullorum-infected chickens. The heritable gut microbes and identified genetic variants could provide references for the selection of resistant chickens and the elimination of pullorum disease.

IMPORTANCE The present study investigated the association among the host genome, the gut microbiome, and S. Pullorum infection in chickens. The results suggested that the gut microbial structure is altered in S. Pullorum-infected chickens. The diversity and abundance of the gut microbiota remarkably differed between the offspring coming from S. Pullorum-positive and S. Pullorum-negative chickens. Heritable gut microbiota were detected in the offspring. Moreover, host genetic variants were associated with microbial diversity and individual gut microbes. The pathogens Pelomonas and Brevundimonas, which exhibited a high heritability in S. Pullorum-positive parents and their offspring, were associated with several genetic mutations in immunity-related genes.

INTRODUCTION

Pullorum disease is an acute systemic disease specific to poultry. The disease mainly occurs in young chicks, causing white diarrhea with high mortality. It is caused by Salmonella enterica subspecies enterica serovar Gallinarum biovar Pullorum (S. Pullorum) (1, 2). Since the early 20th century, pullorum disease has caused substantial economic losses in the poultry industry (3, 4). This disease has a higher mortality rate in 2- to 3-week-old chicks. It rarely occurs in adult birds, and only some infected adult birds show the symptoms of weight loss, diarrhea, inappetence, lesions, and reproductive tract abnormalities (5). Pullorum disease is widely spread and is difficult to cure because of the vertical and horizontal transmission of S. Pullorum (6). Some infected chickens are asymptomatic carriers and can transmit the bacteria to their offspring and other chickens in the flock (7). In 1927, Runnells et al. developed a rapid slide agglutination test based on the S. Pullorum antigen antibody reaction to eliminate infected individuals (8). Although a strict eradication program has been implemented for the S. Pullorum-infected chicken population and some success has been achieved (9, 10), the outbreaks of pullorum disease in chickens indicate that S. Pullorum infection is still frequent and results in considerable economic losses in the poultry industry (11–14). This is mainly because the eradication program results are erratic, including false-negative reactions and a lack of sensitivity (15). Therefore, a new insight is needed to prevent S. Pullorum infection in chickens.

The variation of gut microbial composition and function could be linked to various diseases in mammals and birds, including obesity (16, 17), diarrhea (18, 19), cancer (20), and inflammatory bowel disease (21). In the case of acute inflammation triggered by enteric pathogens, such as Salmonella, the pathogens compete with the gut microbiota and overcome the host immune defenses (22). For example, S. Typhimurium can overcome colonization resistance by abusing the host’s inflammatory immune response to gain an edge over the normal gut microbial community (23, 24). Moreover, S. Pullorum challenge has been found to induce ileal inflammation mediated by proinflammatory cytokines and influence the abundance and diversity of ileal microbes in laying hens (25). Chickens with various genetic backgrounds exhibit various levels of resistance to Salmonella (26). The susceptibility or resistance to Salmonella is related to the host genetics (27, 28). Several studies have discovered candidate genes associated with the death and carrier state of chickens after Salmonella infection (29, 30). Our previous study revealed 43 host genetic markers associated with S. Pullorum infection in chickens (31). These findings indicate that the resistance to Salmonella is closely associated with the gut microbiota and host genetic variants. The gut microbiome can be treated as phenotypes in microbiome genome-wide association studies (mGWAS) to explore the interaction between the microbiota and host genetic variants (32–34). However, few studies have examined chicken pullorum disease, a complex and vertically transmitted bacterial disease, from the perspective of host genetic variants and the gut microbiota.

In the present study, we performed a microbiome comparison and mGWAS to investigate the association among the host genetics, the gut microbiota, and pullorum disease in chickens (Fig. 1a). Microbiome comparison between S. Pullorum-negative and S. Pullorum-positive chickens (groups N and P, respectively) and their respective offspring (groups ON and OP, respectively) was carried out to assess the association between the gut microbial composition and S. Pullorum infection. mGWAS was used to evaluate the contribution of host genomic loci to microbial beta diversity and the abundance of individual microbes. Our discovery provides more information to identify heritable gut microbiota and potential genetic loci associated with S. Pullorum infection and could help in the elimination of infected chickens and the selection of resistant chickens.

FIG 1
  • Open in new tab
  • Download powerpoint
FIG 1

Gut microbial characteristics of S. Pullorum-negative (group N) and S. Pullorum-positive (group P) chickens. (a) Diagram of genetic material and gut microbial transmission from parents to offspring. (b) Comparison of the gut microbiota between groups P and N at the phylum level. (c) Gut microbial composition of group N at the genus level. (d) Gut microbial composition of group P at the genus level. Only the major taxonomic groups are shown. (e) Comparison of the microbial functional pathways between groups N and P at KEGG level two. (f) Comparison of the microbial functional pathways between groups N and P at KEGG level three.

RESULTS

S. Pullorum infection altered the gut microbial characteristics of chickens.To examine the effect of S. Pullorum infection on the gut microbiome of chickens, the microbial composition in groups N and P was compared. Nineteen phyla were detected in the two groups. The dominant phyla were Firmicutes (65.5% in group N and 62.1% in group P), Fusobacteria (16.3% in group N and 18.7% in group P), and Proteobacteria (9.37% in group N and 9.95% in group P) (Fig. 1b). The preponderant genera were Lactobacillus (Firmicutes), Fusobacterium (Fusobacteria), Peptoclostridium (Firmicutes), and Gallibacterium (Proteobacteria) (Fig. 1c and d). Microbiota comparisons at the genus level revealed that the abundance of 39 genera differed between the two groups (P < 0.05), with the difference being significant in the case of 33 out of 39 genera (P < 0.01) (Table 1). Klebsiella (Proteobacteria), Neisseria (Proteobacteria), Enhydrobacter (Proteobacteria), Leuconostoc (Firmicutes), Faecalibaculum (Firmicutes), Enterococcus (Firmicutes), and Mobilitalea (Firmicutes) were enriched in group N, while Anaerobiospirillum (Proteobacteria), Deinococcus (Deinococcus-Thermus), Phascolarctobacterium (Firmicutes), Brevundimonas (Proteobacteria), Pelomonas (Proteobacteria), Oscillibacter (Firmicutes), and Serratia (Proteobacteria) were more abundant in group P than in group N. The metabolic pathways of Staphylococcus aureus infection, beta-lactam resistance, and penicillin and cephalosporin biosynthesis (related to infectious diseases and the biosynthesis of other secondary metabolites) were more enriched in group N than in group P (P < 0.05) (Fig. 1e and f).

View this table:
  • View inline
  • View popup
  • Download powerpoint
TABLE 1

Gut microbiota with significant differences between groups P and N at the genus level

S. Pullorum infection in chickens altered the offspring’s gut microbial composition.To investigate the influence of the host genetics and S. Pullorum infection on the gut microbiota of the offspring, we compared the gut microbial composition between the offspring in groups OP and ON. The alpha diversity indices abundance-based coverage estimator (ACE) and Chao1 suggested that the community richness of group OP was remarkably lower than that of group ON (P < 0.05) (Fig. 2a). By applying principal component analysis (PCA) to microbial beta diversity, the offspring could be classified into their respective groups (Fig. 2b). Of the 13 phyla detected, Firmicutes, Proteobacteria, and Bacteroidetes were the dominant phyla (see Fig. S1a in the supplemental material). The abundance of Parcubacteria was significantly enriched in group OP (P < 0.01) (Table S1). At the genus level, Lactobacillus was more abundant in group ON (62%) than in group OP (53%), while the percentage of Enterococcus, Fusobacterium, and Helicobacter (Proteobacteria) was greater in group OP than in group ON (Fig. S1b). Forty-one genera were prominently different between the two groups (P < 0.05) (Table S2). The abundance of potentially harmful bacteria, namely, Corynebacterium (Actinobacteria), Novosphingobium (Proteobacteria), Vibrionimonas (Bacteroidetes), Aeribacillus (Firmicutes), and Enterococcus, was higher in group OP than in group ON (P < 0.05) (Fig. 2c). On the other hand, beneficial bacteria, such as Kurthia (Firmicutes), Acidovorax (Proteobacteria), and Comamonas (Proteobacteria), were abundant in group ON (Table S2). Pelomonas and Brevundimonas, which were enriched in group P, were more abundant in group OP than in group ON (Table S2).

FIG 2
  • Open in new tab
  • Download powerpoint
FIG 2

Gut microbial composition in offspring from S. Pullorum-positive parents and S. Pullorum-negative parents (groups OP and ON, respectively). (a) Microbial alpha diversity indices ACE and Chao1 between groups OP and ON (*, P < 0.05). (b) Microbial beta diversity in groups OP and ON with a principal component analysis (PCA) plot. (c) Comparison of the abundance of potentially harmful bacteria between groups OP and ON (*, P < 0.05).

FIG S1

Gut microbial characteristics of groups OP and ON. Download FIG S1, TIF file, 0.9 MB.
Copyright © 2021 Ding et al.

This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.

TABLE S1

Gut microbiota with significant difference between groups OP and ON at the phylum level. Download Table S1, DOCX file, 0.01 MB.
Copyright © 2021 Ding et al.

This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.

TABLE S2

Gut microbiota with significant difference between groups OP and ON at the genus level. Download Table S2, DOCX file, 0.02 MB.
Copyright © 2021 Ding et al.

This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.

Host genetic loci associated with gut microbial beta diversity.mGWAS was used to study the association between host genetics and gut microbial diversity. In total, 109 significant single-nucleotide polymorphisms (SNPs) (P < 3.1 × 10−7) were identified in group P (Fig. 3a and Table S3). The most significant SNP is at 7,092,243 bp on Gallus gallus chromosome (GGA) 27, which is located at the intergenic region of IKZF3 and ZPBP2 (Table S3). Seventeen SNPs were clustered on GGA2 in group P (Fig. 3a). Of these, 4 SNPs were located at the intronic region of MAPKKK3L (Fig. 3a). In group P, 162 genes located in the 500 kb upstream and downstream regions around genome-wide significant SNPs were considered candidate genes and were annotated to the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. These genes were enriched in 41 GO functions and 9 pathways. Several enriched pathways related to the host immune system were detected, such as the Wnt signaling pathway and the inflammatory mediator regulation of TRP channels pathway. PRKCA, ADCY9, CAMK2B, and PRKCD were involved in these pathways in group P (Table S4). Meanwhile, 131 significant SNPs were discovered in group N (P < 3.1 × 10−7) (Table S3). In this group, 16 and 13 SNPs were enriched on GGA1 and GGA8, respectively (Fig. 3b and Table S3). Accordingly, the genes located 500 kb upstream and downstream of genome-wide significant SNPs in group N were screened. They were involved in GO functions, such as skeletal muscle tissue regeneration, ATP hydrolysis-coupled proton transport, and cellular response to retinoic acid (Table S4).

FIG 3
  • Open in new tab
  • Download powerpoint
FIG 3

Host genetic variants associated with gut microbiota. A Manhattan plot of genome-wide associations between host genetic variants and gut microbial beta diversity in group P (a) and group N (b). SNPs above the red line were considered significant SNPs (P < 3.1 × 10−7).

TABLE S3

Significant SNPs (P < 3.1 × 10−7) in groups P and N. Download Table S3, XLSX file, 0.03 MB.
Copyright © 2021 Ding et al.

This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.

TABLE S4

Candidate genes in groups P and N mapped to the GO and KEGG databases. Download Table S4, XLSX file, 0.02 MB.
Copyright © 2021 Ding et al.

This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.

Host genetic loci interacted with individual gut microbes.To identify host genes influencing the gut microbiota, mGWAS was performed using 159,272 SNPs identified from groups P and N. Detailed information on the experimental chickens, SNPs, operational taxonomic units (OTUs), and genera used for mGWAS is provided in Table S5. In group P, 1,219 SNPs were associated (P < 6.28 × 10−6) with 177 bacterial genera (Table S6). The most significant association was noted between the SNP at 52,541,080 bp on GGA5 and Facklamia (Firmicutes). This SNP was close to NUDT14 (Table S6). Ten of the 177 genera were more abundant in group P than in group N (P < 0.05) (Table 1 and Fig. 4a). Of these, Bilophila (Proteobacteria) was associated with 25 genes, such as SRPRB, ZP3, and ACAA1 (Table S6). Victivallis (Lentisphaerae) and Phascolarctobacterium were correlated with 16 and 11 genes, respectively (Fig. 4a). Pelomonas, which had a high abundance in group P, was found to be associated with 13 SNPs, one of which was located at the intronic region of NCOA7. Genes such as TGIF1 and TTLL12 were found to be close to the locus of this SNP. Moreover, Anaerobiospirillum, which was more enriched in group P, was associated with 13 SNPs. Of these, one SNP was located at the exonic region of ANPEP on GGA10. Another SNP was located at the intronic region of PSMD14 (Fig. 4a). Furthermore, 12 of the 1,219 significant SNPs were distributed in the exonic region. Of them, 3 nonsynonymous SNPs located at RARRES2, PIP5K1A, and TAP2 were associated with Facklamia and Weissella, which belonged to the order Lactobacillales (Table 2).

FIG 4
  • Open in new tab
  • Download powerpoint
FIG 4

Association between host genetic variants and significantly different bacteria between groups P and N. (a) Significantly different microbes associated with the identified SNPs in group P. (b) Significantly different microbes associated with the identified SNPs in group N.

View this table:
  • View inline
  • View popup
  • Download powerpoint
TABLE 2

The genus significantly associated with exonic single-nucleotide variants (SNVs) (P < 6.28e−06) in groups P and N

TABLE S5

Numbers of experimental animals, SNPs, OTUs, and genera and the significant thresholds used for mGWAS. Download Table S5, DOCX file, 0.01 MB.
Copyright © 2021 Ding et al.

This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.

TABLE S6

Identification of host genetic variants associated with the gut microbiota. Download Table S6, XLSX file, 0.2 MB.
Copyright © 2021 Ding et al.

This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.

In group N, 1,118 SNPs were prominently linked to 181 genera (Table S6). The most significant association was noted between the SNP located at 175,426,469 bp on GGA1 and Brachybacterium (Actinobacteria) (P = 1.29 × 10−11). This SNP was located at the intergenic region between RFC3 and PDS5B (Table S6). Moreover, the SNP located at the intronic region of KY was found to be associated with Mobilitalea (Fig. 4b). The abundance of Mobilitalea was higher in group N. Brevundimonas was associated with 14 SNPs, two of which were located at 10,120,154 bp and 10,120,159 bp on GGA17 at the intronic region of PBX3 (Fig. 4b). The remaining SNPs associated with Brevundimonas were located at the intergenic regions of several immune-related genes, including CCR7 and PLEKHJ1 (Table S6). Furthermore, 19 synonymous SNPs were detected at the exonic region of 15 genes that were associated with 15 genera (Table 2). Among them, 4 SNPs in CENPC were associated with the abundance of Gastranaerophilales_unidentified (Cyanobacteria).

DISCUSSION

In this study, we explored the relationship among host genetics, gut microbiome, and S. Pullorum infection in chickens. Microbiome comparison revealed that S. Pullorum infection in chickens altered the gut microbial composition, resulting in variation of the microbial metabolic function. The abundance of 39 bacterial genera differed between groups P and N. Moreover, compared to group ON, group OP showed a remarkable difference in microbial composition and a high abundance of potentially harmful bacteria. Pelomonas and Brevundimonas exhibited heritability in the offspring coming from S. Pullorum-infected chickens. Pelomonas has been reported to be the dominant bacterium in patients with serious inflammatory bowel disease (35). Similarly, an increased abundance of Brevundimonas has been found in the intestinal mucosa of patients with ulcerative colitis (36). These findings suggest that S. Pullorum infection disturbs the structure of the gut microbiota and the abundance of related microbes in infected individuals and their offspring.

In addition, the heritable gut microbiota was found to be influenced by host genetic variants. In group P, Pelomonas was associated with SNPs that were close to genes such as TGIF1 and TTLL12. TGIF1 promotes the endothelial cell inflammatory response in the gut of mice (37). TTLL12 specifically inhibits the expression of the downstream genes of innate immunity pathways (38). These findings suggest that TGIF1 and TTLL12 interact with Pelomonas to affect intestinal homeostasis in S. Pullorum-infected chickens and cause symptoms such as diarrhea. In group N, the abundance of Brevundimonas was associated with 14 SNPs. These SNPs were close to several immune-related genes in the chicken genome. Among them, CCR7 plays a critical role in controlling T-cell retention/egress to maintain intestinal homeostasis in mice (39). The interaction between CCR7 and Brevundimonas may play a role in the maintenance of gut homeostasis in chickens.

In group P, 4 SNPs associated with gut microbial beta diversity were located at the intronic region of MAPKKK3L, which belongs to the MAPKKK family. MAPKKK can regulate several signal transduction pathways, including c-Jun NH2-terminal kinase, ERK, and nuclear factor-κB (NF-κB), by stimulating the Toll-like receptor (40–42). Moreover, it can stimulate immune cells, such as macrophages, dendritic cells, and neutrophils, to produce various chemokines, including gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF) (43). MYD88, located downstream of MAPKKK3L (less than 26 kb), is associated with susceptibility to S. Pullorum infection (44). Thus, MAPKKK3L may be a vital candidate gene associated with the gut microbiota in S. Pullorum-infected chickens. Therefore, the heritable bacteria Pelomonas and Brevundimonas and significant markers located on related genes, such as TGIF1, TTLL12, CCR7, and MAPKKK3L, could be used for the selection of resistant chickens and the elimination of pullorum disease.

MATERIALS AND METHODS

Animals and sampling.The chickens (Xin Pudong chickens) used in the present study were obtained from the Animal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, Shanghai, China. None of the chickens had been treated with antibiotics. The wing venous blood and feces of 275 hens (52 weeks old; 140 in group N and 135 in group P) were collected. Ten positive roosters were mated with 10 positive hens, and 10 negative roosters were mated with 10 negative hens to obtain their respective offspring. All the chickens were maintained at the same location and fed the same diets. Eighty fecal samples of the offspring were collected at the age of 10 days (40 in group ON and 40 in group OP). In total, 355 fecal samples were collected and stored at −80°C. The protocols in the present study were approved by the Laboratory Animal Research (ILAR) guide for the care and use of laboratory animals at Shanghai Jiao Tong University, China.

Rapid slide agglutination test.S. Pullorum infections in chickens were diagnosed by S. Pullorum and S. Gallinarum polyvalent antigen rapid slide agglutination test reagents (product code 03.01.001.001; Beijing Zhonghai Biotech Co., Ltd., China). In brief, 50 μl of polyvalent antigen and 50 μl of venous blood were placed on a clean glass slide. The antigen and blood were thoroughly mixed and smeared into a circle of 2 cm on the glass slide. The samples were considered positive if 50% or more agglutination occurs in the mixture within 2 min. Samples without agglutination were deemed negative.

16S rRNA gene sequencing.Microbiome DNA was isolated from the fecal samples using the Tiangen DNA stool minikit (number DP328; Tiangen, China) by following the manufacturer’s instructions. The extracted DNA was quantified on a NanoDrop spectrophotometer (Thermo Scientific). The DNA samples were stored at −20°C for further analysis. The V3-V4 regions of the 16S rRNA of all the fecal samples were amplified by PCR using barcoded fusion primers (forward primer 338F, ACTCCTACGGGAGGCAGCA; reverse primer 806R, GGACTACHVGGGTWTCTAAT). The PCR conditions were 98°C for 2 min; 98°C for 15 s; 55°C for 30 s and 72°C for 30 s, repeated for 30 cycles; and 72°C for 5 min. PCR amplicons were excised from a 1.5% agarose gel and purified using the QIAquick gel extraction kit (number 28706; Qiagen, Germany). Purified PCR products were combined at equal concentrations and used to construct a metagenomic library using the Illumina TruSeq sample preparation kit (Illumina) according to the manufacturer's protocol. Sequencing was performed by Shanghai Personal Biotechnology Limited Company (Shanghai, China) using the Illumina MiSeq sequencing platform (Illumina).

Sequence quality control.The Quantitative Insights Into Microbial Ecology (QIIME, v1.8.0) pipeline was employed to process the sequencing data, as described previously (45). In brief, raw sequencing reads with exact matches to the barcodes were assigned to the respective samples and identified as valid sequences. Low-quality sequences were filtered out according to the following criteria (46, 47): sequences with a length of <150 bp, average Phred scores of <20, contained ambiguous bases, and contained mononucleotide repeats of >8 bp. Chimeric sequences were removed using USEARCH (v5.2.236) in QIIME. Paired-end reads with an overlap longer than 10 bp between read 1 and read 2 and without any mismatch were assembled using FLASH (48).

Microbial taxonomic annotation.The filtered high-quality sequences were clustered into operational taxonomic units (OTUs) at 97% sequence identity using UCLUST (49). A representative sequence was selected from each OTU using default parameters. OTU taxonomic annotation was performed by BLAST searching the representative sequences set against the Silva database (50) using the best hit (51). An OTU table was further generated to record the abundance of each OTU in each sample and the taxonomy of OTUs. OTUs containing less than 0.001% of the total sequences across all the samples were discarded. To minimize the difference of the sequencing depth across samples, an averaged, rounded, rarefied OTU table was generated by averaging 100 evenly resampled OTU subsets under 90% of the minimum sequencing depth for further analysis.

Microbiome comparison analysis.Microbiome comparisons were performed between group N versus group P and group ON versus group OP. OTU-level alpha diversity indices, Chao1 richness estimator (52), and abundance-based coverage estimator (ACE) (53) were calculated using the OTU table in QIIME. Principal component analysis (PCA) was conducted based on the genus level compositional profiles and the plot drawn by R (54). Box plots and bar charts were created using SigmaPlot (55). Two-sided Welch’s t test and Benjamini-Hochberg false discovery rate (FDR) (P < 0.05) (56) correction were used in two-group analysis. Microbial functions were predicted using PICRUSt (57). OTUs were mapped to the gg13.5 database at 97% similarity using the QIIME’s command “pick_closed_otus.” OTU abundance was automatically normalized using the 16S rRNA gene copy numbers from known bacterial genomes in Integrated Microbial Genomes (IMG). The predicted genes and their functions were aligned to the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, and the differences among groups were compared using the STAMP software (58).

Genotyping of populations from groups P and N.The genomic DNA of 135 chickens in group P and 140 chickens in group N was extracted from their venous blood using the TIANamp blood DNA kit (number DP348; Tiangen, China). The DNA was used to construct double-digest genotyping-by-sequencing (dd-GBS) libraries (31), which were sequenced at Shanghai Personal Biotechnology Co., Ltd. The dd-GBS data were analyzed using the chicken genome (GRCg6a). The approach of single-nucleotide polymorphism (SNP) calling was consistent with that in our previous study (31). Next, quality control was performed for the genotyping data with call rate thresholds of ≥50%, minor allele frequency (MAF) of ≥5%, and Hardy-Weinberg equilibrium (HWE) P value of >1 × 10−6 using PLINK. A final set of 159,272 SNPs that passed the quality control assessment were used for further analyses.

mGWAS for assessing the association between host genetic loci and gut microbial beta diversity.The association between the gut microbial community and the host genetics was analyzed by performing mGWAS (59). In total, 159,272 SNPs were used as genotyping data, and a pairwise microbiome distance matrix of weighted UniFrac was used as the microbiome data. Mutations with adjusted P values that passed the genome-wide significance threshold (0.05/SNP number) were considered significant. SNPs were annotated using ANNOVAR (60), and the genes that contained significant SNPs were annotated by Gene Ontology (GO) and KEGG analysis using DAVID (61).

mGWAS for assessing the effect of host genetic variants on gut microbial abundance.To identify the genetic variants in groups P and N that were associated with the abundance of individual gut bacteria, a statistical test was performed for each association between SNPs and the taxa. The analysis was performed using the MiBioGen miQTL pipeline (62). In brief, the taxa that were detected in at least 10% of the samples were included; their relative abundance was log transformed and controlled for the effects of the first three genetic principal components. The taxa were treated as quantitative traits, and a linear regression model of their log-transformed relative abundance was adopted with Fisher’s test-based P value estimation. In total, 197 taxa were defined as a binary trait (absence/presence) using logistic regression with chi-square-based P value estimation. The binary and quantitative models were used for groups N and P, respectively. The genome-wide significance threshold for the association was set at 0.05/SNP number. The suggestive significance level was determined by 1/SNP number.

Data availability.Raw read sequences are publicly available in the Sequence Read Archive at National Center for Biotechnology Information (NCBI) under the BioProject accession number PRJNA679403.

ACKNOWLEDGMENTS

This work was supported by the National Natural Science Foundation of China (grant no. 32072701).

We thank Yan Zhang (Carilion Clinic, Roanoke, VA, United States) for editing the manuscript.

FOOTNOTES

    • Received November 16, 2020.
    • Accepted December 11, 2020.
  • Copyright © 2021 Ding et al.

This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

REFERENCES

  1. 1.↵
    1. Rettger LF
    . 1909. Further studies on fatal septicemia in young chickens, or “white diarrhea.” J Med Res 21:115–123.
    OpenUrlPubMed
  2. 2.↵
    1. Persson U,
    2. Jendteg S
    . 1992. The economic impact of poultry-borne salmonellosis: how much should be spent on prophylaxis? Int J Food Microbiol 15:207–213. doi:10.1016/0168-1605(92)90050-d.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Rettger LF,
    2. Harvey SC
    . 1908. Fatal septicemia in young chickens, or “white diarrhea.” J Med Res 18:277–290.
    OpenUrlPubMed
  4. 4.↵
    1. Koutsoumanis K,
    2. Allende A,
    3. Alvarez-Ordóñez A,
    4. Bolton D,
    5. Bover-Cid S,
    6. Chemaly M,
    7. De Cesare A,
    8. Herman L,
    9. Hilbert F,
    10. Lindqvist R,
    11. Nauta M,
    12. Peixe L,
    13. Ru G,
    14. Simmons M,
    15. Skandamis P,
    16. Suffredini E,
    17. Dewulf J,
    18. Hald T,
    19. Michel V,
    20. Niskanen T,
    21. Ricci A,
    22. Snary E,
    23. Boelaert F,
    24. Messens W,
    25. Davies R, EFSA Panel on Biological Hazards (EFSA BIOHAZ Panel)
    . 2019. Salmonella control in poultry flocks and its public health impact. EFSA J 17:e05596. doi:10.2903/j.efsa.2019.5596.
    OpenUrlCrossRef
  5. 5.↵
    1. Shivaprasad HL
    . 2000. Fowl typhoid and pullorum disease. Rev Sci Tech 19:405–424. doi:10.20506/rst.19.2.1222.
    OpenUrlCrossRefPubMedWeb of Science
  6. 6.↵
    1. Berchieri AJ,
    2. Murphy CK,
    3. Marston K,
    4. Barrow PA
    . 2001. Observations on the persistence and vertical transmission of Salmonella enterica serovars Pullorum and Gallinarum in chickens: effect of bacterial and host genetic background. Avian Pathol 30:221–231. doi:10.1080/03079450120054631.
    OpenUrlCrossRefPubMedWeb of Science
  7. 7.↵
    1. Wigley P,
    2. Berchieri AJ,
    3. Page KL,
    4. Smith AL,
    5. Barrow PA
    . 2001. Salmonella enterica Serovar Pullorum persists in splenic macrophages and in the reproductive tract during persistent, disease-free carriage in chickens. Infect Immun 69:7873–7879. doi:10.1128/IAI.69.12.7873-7879.2001.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    1. Runnells RA,
    2. Coon CJ,
    3. Farley H,
    4. Thorp F
    . 1927. An application of the rapid method agglutination test to the diagnosis of bacillary white diarrhoea infection. J Am Vet Med Assoc 70:660–667.
    OpenUrl
  9. 9.↵
    1. Barrow PA,
    2. Freitas Neto OC
    . 2011. Pullorum disease and fowl typhoid–new thoughts on old diseases: a review. Avian Pathol 40:1–13. doi:10.1080/03079457.2010.542575.
    OpenUrlCrossRefPubMedWeb of Science
  10. 10.↵
    World Organisation for Animal Health. 2020. Pullorum disease timelines (1996–2020). World Organisation for Animal Health (OIE), Paris, France. https://www.oie.int/wahis_2/public/wahid.php/Diseaseinformation/Diseasetimelines. Retrieved 10 September 2020.
  11. 11.↵
    1. Salem M,
    2. Odor EM,
    3. Pope C
    . 1992. Pullorum disease in Delaware roasters. Avian Dis 36:1076–1080. doi:10.2307/1591579.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Sato Y,
    2. Sato G,
    3. Tuchili L,
    4. Pandey GS,
    5. Nakajima A,
    6. Chimana H,
    7. Sinsungwe H
    . 1997. Status of Salmonella gallinarum-pullorum infections in poultry in Zambia. Avian Dis 41:490–495. doi:10.2307/1592212.
    OpenUrlCrossRefPubMedWeb of Science
  13. 13.↵
    1. Pan Z,
    2. Wang X,
    3. Zhang X,
    4. Geng S,
    5. Chen X,
    6. Pan W,
    7. Cong Q,
    8. Liu X,
    9. Jiao X,
    10. Liu X
    . 2009. Changes in antimicrobial resistance among Salmonella enterica subspecies enterica serovar Pullorum isolates in China from 1962 to 2007. Vet Microbiol 136:387–392. doi:10.1016/j.vetmic.2008.11.015.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Barrow PA,
    2. Jones MA,
    3. Smith AL,
    4. Wigley P
    . 2012. The long view: salmonella–the last forty years. Avian Pathol 41:413–420. doi:10.1080/03079457.2012.718071.
    OpenUrlCrossRefPubMedWeb of Science
  15. 15.↵
    1. Proux K,
    2. Humbert F,
    3. Jouy E,
    4. Houdayer C,
    5. Lalande F,
    6. Oger A,
    7. Salvat G
    . 2002. Improvements required for the detection of Salmonella Pullorum and Gallinarum. Can J Vet Res 66:151–157.
    OpenUrlPubMed
  16. 16.↵
    1. Turnbaugh PJ,
    2. Hamady M,
    3. Yatsunenko T,
    4. Cantarel BL,
    5. Duncan A,
    6. Ley RE,
    7. Sogin ML,
    8. Jones WJ,
    9. Roe BA,
    10. Affourtit JP,
    11. Egholm M,
    12. Henrissat B,
    13. Heath AC,
    14. Knight R,
    15. Gordon JI
    . 2009. A core gut microbiome in obese and lean twins. Nature 457:480–484. doi:10.1038/nature07540.
    OpenUrlCrossRefPubMedWeb of Science
  17. 17.↵
    1. Ding J,
    2. Zhao L,
    3. Wang L,
    4. Zhao W,
    5. Zhai Z,
    6. Leng L,
    7. Wang Y,
    8. He C,
    9. Zhang Y,
    10. Zhang H,
    11. Li H,
    12. Meng H
    . 2016. Divergent selection-induced obesity alters the composition and functional pathways of chicken gut microbiota. Genet Sel Evol 48:93. doi:10.1186/s12711-016-0270-5.
    OpenUrlCrossRef
  18. 18.↵
    1. Liu S,
    2. Zhao L,
    3. Zhai Z,
    4. Zhao W,
    5. Ding J,
    6. Dai R,
    7. Sun T,
    8. Meng H
    . 2015. Porcine epidemic diarrhea virus infection induced the unbalance of gut microbiota in piglets. Curr Microbiol 71:643–649. doi:10.1007/s00284-015-0895-6.
    OpenUrlCrossRef
  19. 19.↵
    1. Huang A,
    2. Cai R,
    3. Wang Q,
    4. Shi L,
    5. Li C,
    6. Yan H
    . 2019. Dynamic change of gut microbiota during porcine epidemic diarrhea virus infection in suckling piglets. Front Microbiol 10:322. doi:10.3389/fmicb.2019.00322.
    OpenUrlCrossRef
  20. 20.↵
    1. McQuade JL,
    2. Daniel CR,
    3. Helmink BA,
    4. Wargo JA
    . 2019. Modulating the microbiome to improve therapeutic response in cancer. Lancet Oncol 20:e77–e91. doi:10.1016/S1470-2045(18)30952-5.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Lavelle A,
    2. Sokol H
    . 2020. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 17:223–237. doi:10.1038/s41575-019-0258-z.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Thiennimitr P,
    2. Winter SE,
    3. Bäumler AJ
    . 2012. Salmonella, the host and its microbiota. Curr Opin Microbiol 15:108–114. doi:10.1016/j.mib.2011.10.002.
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Stecher B,
    2. Robbiani R,
    3. Walker AW,
    4. Westendorf AM,
    5. Barthel M,
    6. Kremer M,
    7. Chaffron S,
    8. Macpherson AJ,
    9. Buer J,
    10. Parkhill J,
    11. Dougan G,
    12. von Mering C,
    13. Hardt WD
    . 2007. Salmonella enterica serovar typhimurium exploits inflammation to compete with the intestinal microbiota. PLoS Biol 5:2177–2189. doi:10.1371/journal.pbio.0050244.
    OpenUrlCrossRefPubMedWeb of Science
  24. 24.↵
    1. Lawley TD,
    2. Bouley DM,
    3. Hoy YE,
    4. Gerke C,
    5. Relman DA,
    6. Monack DM
    . 2008. Host transmission of Salmonella enterica serovar Typhimurium is controlled by virulence factors and indigenous intestinal microbiota. Infect Immun 76:403–416. doi:10.1128/IAI.01189-07.
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    1. Wang W,
    2. Jia H,
    3. Zhang H,
    4. Wang J,
    5. Lv H,
    6. Wu S,
    7. Qi G
    . 2019. Supplemental plant extracts from flos lonicerae in combination with baikal skullcap attenuate intestinal disruption and modulate gut microbiota in laying hens challenged by Salmonella pullorum. Front Microbiol 10:1681. doi:10.3389/fmicb.2019.01681.
    OpenUrlCrossRef
  26. 26.↵
    1. Li X,
    2. Nie C,
    3. Zhang Z,
    4. Wang Q,
    5. Shao P,
    6. Zhao Q,
    7. Chen Y,
    8. Wang D,
    9. Li Y,
    10. Jiao W,
    11. Li L,
    12. Qin S,
    13. He L,
    14. Jia Y,
    15. Ning Z,
    16. Qu L
    . 2018. Evaluation of genetic resistance to Salmonella Pullorum in three chicken lines. Poult Sci 97:764–769. doi:10.3382/ps/pex354.
    OpenUrlCrossRef
  27. 27.↵
    1. Lamont SJ,
    2. Kaiser MG,
    3. Liu W
    . 2002. Candidate genes for resistance to Salmonella enteritidis colonization in chickens as detected in a novel genetic cross. Vet Immunol Immunopathol 87:423–428. doi:10.1016/S0165-2427(02)00064-8.
    OpenUrlCrossRefPubMedWeb of Science
  28. 28.↵
    1. Barrow PA,
    2. Bumstead N,
    3. Marston K,
    4. Lovell MA,
    5. Wigley P
    . 2004. Faecal shedding and intestinal colonization of Salmonella enterica in in-bred chickens: the effect of host-genetic background. Epidemiol Infect 132:117–126. doi:10.1017/s0950268803001274.
    OpenUrlCrossRefPubMedWeb of Science
  29. 29.↵
    1. Calenge F,
    2. Lecerf F,
    3. Demars J,
    4. Feve K,
    5. Vignoles F,
    6. Pitel F,
    7. Vignal A,
    8. Velge P,
    9. Sellier N,
    10. Beaumont C
    . 2009. QTL for resistance to Salmonella carrier state confirmed in both experimental and commercial chicken lines. Anim Genet 40:590–597. doi:10.1111/j.1365-2052.2009.01884.x.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    1. Li X,
    2. Nie C,
    3. Liu Y,
    4. Chen Y,
    5. Lv X,
    6. Wang L,
    7. Zhang J,
    8. Li K,
    9. Jia Y,
    10. Ban L,
    11. Ning Z,
    12. Qu L
    . 2019. A genome-wide association study explores the genetic determinism of host resistance to Salmonella pullorum infection in chickens. Genet Sel Evol 51:51. doi:10.1186/s12711-019-0492-4.
    OpenUrlCrossRef
  31. 31.↵
    1. Xiao L,
    2. He C,
    3. Luo L,
    4. Yang K,
    5. Yang L,
    6. Xu K,
    7. Zheng Y,
    8. Gu C,
    9. Huang Q,
    10. Meng H
    . 2019. Genome-wide association study identified genes in the response to Salmonella pullorum infection in chickens. Anim Genet 50:403–406. doi:10.1111/age.12787.
    OpenUrlCrossRef
  32. 32.↵
    1. Hua X,
    2. Goedert JJ,
    3. Landi MT,
    4. Shi J
    . 2016. Identifying host genetic variants associated with microbiome composition by testing multiple beta diversity matrices. Hum Hered 81:117–126. doi:10.1159/000448733.
    OpenUrlCrossRef
  33. 33.↵
    1. Goodrich JK,
    2. Davenport ER,
    3. Beaumont M,
    4. Jackson MA,
    5. Knight R,
    6. Ober C,
    7. Spector TD,
    8. Bell JT,
    9. Clark AG,
    10. Ley RE
    . 2016. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe 19:731–743. doi:10.1016/j.chom.2016.04.017.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Goodrich JK,
    2. Davenport ER,
    3. Waters JL,
    4. Clark AG,
    5. Ley RE
    . 2016. Cross-species comparisons of host genetic associations with the microbiome. Science 352:532–535. doi:10.1126/science.aad9379.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    1. Schäffler H,
    2. Herlemann DPR,
    3. Alberts C,
    4. Kaschitzki A,
    5. Bodammer P,
    6. Bannert K,
    7. Köller T,
    8. Warnke P,
    9. Kreikemeyer B,
    10. Lamprecht G
    . 2016. Mucosa-attached bacterial community in Crohn's disease coheres with the clinical disease activity index. Environ Microbiol Rep 8:614–621. doi:10.1111/1758-2229.12411.
    OpenUrlCrossRef
  36. 36.↵
    1. Dheer R,
    2. Davies JM,
    3. Quintero MA,
    4. Damas OM,
    5. Deshpande AR,
    6. Kerman DH,
    7. Sawyer WP,
    8. Pignac-Kobinger J,
    9. Ban Y,
    10. Fernandez I,
    11. Burgueno JF,
    12. Phillips MC,
    13. Abreu MT
    . 2020. Microbial signatures and innate immune gene expression in lamina propria phagocytes of inflammatory bowel disease patients. Cell Mol Gastroenterol Hepatol 9:387–402. doi:10.1016/j.jcmgh.2019.10.013.
    OpenUrlCrossRef
  37. 37.↵
    1. Hneino M,
    2. Blirando K,
    3. Buard V,
    4. Tarlet G,
    5. Benderitter M,
    6. Hoodless P,
    7. François A,
    8. Milliat F
    . 2012. The TG-interacting factor TGIF1 regulates stress-induced proinflammatory phenotype of endothelial cells. J Biol Chem 287:38913–38921. doi:10.1074/jbc.M112.388389.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Ju L-G,
    2. Zhu Y,
    3. Lei P-J,
    4. Yan D,
    5. Zhu K,
    6. Wang X,
    7. Li Q-L,
    8. Li X-J,
    9. Chen J-W,
    10. Li L-Y,
    11. Wu M
    . 2017. TTLL12 Inhibits the activation of cellular antiviral signaling through interaction with VISA/MAVS. J Immunol 198:1274–1284. doi:10.4049/jimmunol.1601194.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. McNamee EN,
    2. Masterson JC,
    3. Veny M,
    4. Collins CB,
    5. Jedlicka P,
    6. Byrne FR,
    7. Ng GY,
    8. Rivera-Nieves J
    . 2015. Chemokine receptor CCR7 regulates the intestinal TH1/TH17/Treg balance during Crohn's-like murine ileitis. J Leukoc Biol 97:1011–1022. doi:10.1189/jlb.3HI0614-303R.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Medzhitov R,
    2. Horng T
    . 2009. Transcriptional control of the inflammatory response. Nat Rev Immunol 9:692–703. doi:10.1038/nri2634.
    OpenUrlCrossRefPubMedWeb of Science
  41. 41.↵
    1. Gantke T,
    2. Sriskantharajah S,
    3. Sadowski M,
    4. Ley SC
    . 2012. IκB kinase regulation of the TPL-2/ERK MAPK pathway. Immunol Rev 246:168–182. doi:10.1111/j.1600-065X.2012.01104.x.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Gantke T,
    2. Sriskantharajah S,
    3. Ley SC
    . 2011. Regulation and function of TPL-2, an IκB kinase-regulated MAP kinase kinase kinase. Cell Res 21:131–145. doi:10.1038/cr.2010.173.
    OpenUrlCrossRefPubMedWeb of Science
  43. 43.↵
    1. Malinin NL,
    2. Boldin MP,
    3. Kovalenko AV,
    4. Wallach D
    . 1997. MAP3K-related kinase involved in NF-kappaB induction by TNF, CD95 and IL-1. Nature 385:540–544. doi:10.1038/385540a0.
    OpenUrlCrossRefPubMedWeb of Science
  44. 44.↵
    1. Liu XQ,
    2. Wang F,
    3. Jin J,
    4. Zhou YG,
    5. Ran JS,
    6. Feng ZQ,
    7. Wang Y,
    8. Liu YP
    . 2015. MyD88 polymorphisms and association with susceptibility to Salmonella Pullorum. Biomed Res Int 2015:1–7. doi:10.1155/2015/692973.
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. Caporaso J,
    2. Kuczynski J,
    3. Stombaugh J,
    4. Bittinger K,
    5. Bushman F,
    6. Costello E,
    7. Fierer N,
    8. Pena A,
    9. Goodrich J,
    10. Gordon J,
    11. Ga H,
    12. Kelley S,
    13. Knights D,
    14. Koenig J,
    15. Ley R,
    16. Lozupone C,
    17. McDonald D,
    18. Muegge B,
    19. Pirrung M,
    20. Reeder J,
    21. Sevinsky J,
    22. Turnbaugh P,
    23. Walters W,
    24. Widmann J,
    25. Yatsunenko T,
    26. Zaneveld J,
    27. Knight R
    . 2010. Qiime allows analysis of high-throughput community sequencing data. Nat Methods 7:335–336. doi:10.1038/nmeth.f.303.
    OpenUrlCrossRefPubMedWeb of Science
  46. 46.↵
    1. Gill SR,
    2. Pop M,
    3. Deboy RT,
    4. Eckburg PB,
    5. Turnbaugh PJ,
    6. Samuel BS,
    7. Gordon JI,
    8. Relman DA,
    9. Fraser-Liggett CM,
    10. Nelson KE
    . 2006. Metagenomic analysis of the human distal gut microbiome. Science 312:1355–1359. doi:10.1126/science.1124234.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. Chen H,
    2. Jiang W
    . 2014. Application of high-throughput sequencing in understanding human oral microbiome related with health and disease. Front Microbiol 5:508. doi:10.3389/fmicb.2014.00508.
    OpenUrlCrossRef
  48. 48.↵
    1. Magoc T,
    2. Salzberg S
    . 2011. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics 27:2957–2963. doi:10.1093/bioinformatics/btr507.
    OpenUrlCrossRefPubMedWeb of Science
  49. 49.↵
    1. Edgar RC
    . 2010. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26:2460–2461. doi:10.1093/bioinformatics/btq461.
    OpenUrlCrossRefPubMedWeb of Science
  50. 50.↵
    1. Quast C,
    2. Pruesse E,
    3. Yilmaz P,
    4. Gerken J,
    5. Schweer T,
    6. Yarza P,
    7. Peplies J,
    8. Glöckner FO
    . 2013. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res 41:D590–D596. doi:10.1093/nar/gks1219.
    OpenUrlCrossRefPubMedWeb of Science
  51. 51.↵
    1. Altschul SF,
    2. Madden TL,
    3. Schäffer AA,
    4. Zhang J,
    5. Zhang Z,
    6. Miller W,
    7. Lipman DJ
    . 1997. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res 25:3389–3402. doi:10.1093/nar/25.17.3389.
    OpenUrlCrossRefPubMedWeb of Science
  52. 52.↵
    1. Chao A
    . 1984. Nonparametric estimation of the number of classes in a population. Scand J Stat 11:265–270.
    OpenUrlWeb of Science
  53. 53.↵
    1. Chao A,
    2. Ma MC,
    3. Yang MCK
    . 1993. Stopping rules and estimation for recapture debugging with unequal failure rates. Biometrika 80:193–201. doi:10.1093/biomet/80.1.193.
    OpenUrlCrossRefWeb of Science
  54. 54.↵
    1. Dessau RB,
    2. Pipper CB
    . 2008. R–project for statistical computing. Ugeskr Laeger 170:328–330.
    OpenUrlCrossRefPubMed
  55. 55.↵
    1. Kornbrot D
    . 2000. Statistical software for microcomputers: sigmaPlot 2000 and SigmaStat2. Br J Math Stat Psychol 53:335–337.
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. Benjamini Y,
    2. Hochberg Y
    . 1995. Controlling the false discovery rate: apractical and powerful approach to multiple testing. J R Statist Soc B 57:289–300. doi:10.1111/j.2517-6161.1995.tb02031.x.
    OpenUrlCrossRefPubMed
  57. 57.↵
    1. Langille MGI,
    2. Zaneveld J,
    3. Caporaso JG,
    4. McDonald D,
    5. Knights D,
    6. Reyes JA,
    7. Clemente JC,
    8. Burkepile DE,
    9. Vega Thurber RL,
    10. Knight R,
    11. Beiko RG,
    12. Huttenhower C
    . 2013. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol 31:814–821. doi:10.1038/nbt.2676.
    OpenUrlCrossRefPubMed
  58. 58.↵
    1. Parks DH,
    2. Beiko RG
    . 2010. Identifying biologically relevant differences between metagenomic communities. Bioinformatics 26:715–721. doi:10.1093/bioinformatics/btq041.
    OpenUrlCrossRefPubMedWeb of Science
  59. 59.↵
    1. Hua X,
    2. Song L,
    3. Yu G,
    4. Goedert J,
    5. Abnet C,
    6. Landi M,
    7. Shi J
    . 2015. MicrobiomeGWAS: a tool for identifying host genetic variants associated with microbiome composition. bioRxiv doi:10.1101/031187.
    OpenUrlAbstract/FREE Full Text
  60. 60.↵
    1. Wang K,
    2. Li M,
    3. Hakonarson H
    . 2010. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res 38:e164. doi:10.1093/nar/gkq603.
    OpenUrlCrossRefPubMed
  61. 61.↵
    1. Jiao X,
    2. Sherman BT,
    3. Huang DW,
    4. Stephens R,
    5. Baseler MW,
    6. Lane HC,
    7. Lempicki RA
    . 2012. DAVID-WS: a stateful web service to facilitate gene/protein list analysis. Bioinformatics 28:1805–1806. doi:10.1093/bioinformatics/bts251.
    OpenUrlCrossRefPubMedWeb of Science
  62. 62.↵
    1. Wang J,
    2. Kurilshikov A,
    3. Radjabzadeh D,
    4. Turpin W,
    5. Croitoru K,
    6. Bonder MJ,
    7. Jackson MA,
    8. Medina-Gomez C,
    9. Frost F,
    10. Homuth G,
    11. Rühlemann M,
    12. Hughes D,
    13. Kim H-N,
    14. Spector TD,
    15. Bell JT,
    16. Steves CJ,
    17. Timpson N,
    18. Franke A,
    19. Wijmenga C,
    20. Meyer K,
    21. Kacprowski T,
    22. Franke L,
    23. Paterson AD,
    24. Raes J,
    25. Kraaij R,
    26. Zhernakova A, MiBioGen Consortium Initiative
    . 2018. Meta-analysis of human genome-microbiome association studies: the MiBioGen consortium initiative. Microbiome 6:101. doi:10.1186/s40168-018-0479-3.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
Download PDF
Citation Tools
Heritable Gut Microbiome Associated with Salmonella enterica Serovar Pullorum Infection in Chickens
Jinmei Ding, Hao Zhou, Lingxiao Luo, Lu Xiao, Kaixuan Yang, Lingyu Yang, Yuming Zheng, Ke Xu, Chuan He, Chengxiao Han, Huaixi Luo, Chao Qin, Fisayo T. Akinyemi, Caiju Gu, Zhenxiang Zhou, Qizhong Huang, He Meng
mSystems Jan 2021, 6 (1) e01192-20; DOI: 10.1128/mSystems.01192-20

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print
Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this mSystems article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Heritable Gut Microbiome Associated with Salmonella enterica Serovar Pullorum Infection in Chickens
(Your Name) has forwarded a page to you from mSystems
(Your Name) thought you would be interested in this article in mSystems.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Heritable Gut Microbiome Associated with Salmonella enterica Serovar Pullorum Infection in Chickens
Jinmei Ding, Hao Zhou, Lingxiao Luo, Lu Xiao, Kaixuan Yang, Lingyu Yang, Yuming Zheng, Ke Xu, Chuan He, Chengxiao Han, Huaixi Luo, Chao Qin, Fisayo T. Akinyemi, Caiju Gu, Zhenxiang Zhou, Qizhong Huang, He Meng
mSystems Jan 2021, 6 (1) e01192-20; DOI: 10.1128/mSystems.01192-20
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • INTRODUCTION
    • RESULTS
    • DISCUSSION
    • MATERIALS AND METHODS
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

heritable
host genetic variants
gut microbiota
mGWAS
Salmonella Pullorum
chicken

Related Articles

Cited By...

About

  • About mSystems
  • Author Videos
  • Board of Editors
  • Policies
  • Overleaf Pilot
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Author Warranty
  • Types of Articles
  • Getting Started
  • Ethics
  • Contact Us

Follow #mSystemsJ

@ASMicrobiology

       

 

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Online ISSN: 2379-5077