Next Article in Journal
Two Novel Variants of WDR26 in Chinese Patients with Intellectual Disability
Previous Article in Journal
Mitochondrial Pseudogenes Suggest Repeated Inter-Species Hybridization among Direct Human Ancestors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transcriptomic Analysis of the Spleen of Different Chicken Breeds Revealed the Differential Resistance of Salmonella Typhimurium

1
State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
2
Animal Production Department, National Research Centre, Dokki, Cairo 12622, Egypt
*
Author to whom correspondence should be addressed.
Genes 2022, 13(5), 811; https://doi.org/10.3390/genes13050811
Submission received: 7 April 2022 / Revised: 28 April 2022 / Accepted: 28 April 2022 / Published: 2 May 2022
(This article belongs to the Section Animal Genetics and Genomics)

Abstract

:
Salmonella Typhimurium (ST) is a foodborne pathogen that adversely affects the health of both animals and humans. Since poultry is a common source and carrier of the disease, controlling ST infection in chickens will have a protective impact on human health. In the current study, Beijing-You (BY) and Cobb chicks (5-day-old specific-pathogen-free) were orally challenged by 2.4 × 1012 CFU ST, spleen transcriptome was conducted 1 day post-infection (DPI) to identify gene markers and pathways related to the immune system. A total of 775 significant differentially expressed genes (DEGs) in comparisons between BY and Cobb were identified, including 498 upregulated and 277 downregulated genes (fold change ≥2.0, p < 0.05). Several immune response pathways against Salmonella were enriched, including natural killer-cell-mediated-cytotoxicity, cytokine–cytokine receptor interaction, antigen processing and presentation, phagosomes, and intestinal immune network for IgA production, for both BY and Cobb chickens. The BY chicks showed a robust response for clearance of bacterial load, immune response, and robust activation of phagosomes, resulting in ST resistance. These results confirmed that BY breed more resistance to ST challenge and will provide a better understanding of BY and Cobb chickens’ susceptibility and resistance to ST infection at the early stages of host immune response, which could expand the known intricacies of molecular mechanisms in chicken immunological responses against ST. Pathways induced by Salmonella infection may provide a novel approach to developing preventive and curative strategies for ST, and increase inherent resistance in animals through genetic selection.

1. Introduction

Salmonella Typhimurium (ST) is a gram-negative intestinal pathogen that doesn’t cause serious morbidity or death; however, chickens can carry this bacterium for several weeks without showing any symptoms, thus posing a serious public health risk [1,2,3]. Recently, ST became the most prevalent serotype in eggs and meat produced in China [4,5,6]. ST can be difficult to treat in humans and animals because it is often very resistant to several antibiotics, such as nalidixic acid, trimethoprim, and sulfamethoxazole [7,8,9]. Salmonella typically infects chickens through the fecal–oral route. The innate immune system in particular is important for Salmonella infections, as adaptive immunity is not yet developed in young chickens. [10]. Therefore, understanding host immune response mechanisms and resistance to ST infection is crucial for reducing economic losses in chicken productions and for protecting animal and human health [11,12]. Beijing-You (BY) is a local Chinese chicken breed that is more resistant to stress and disease than White Leghorn [13]. Candidate SNPs linked to immunological features were identified in BY chicken by a genome-wide association study (antibody responses against SRBC and AIV, serum IgY level, and heterophils and lymphocytes numbers and ratios) [14]. Resistance to Salmonella Pullorum is higher in BY chickens than in Rhode Island Red chickens [15]. Antibody titers raised for Newcastle disease and avian influenza vaccinations were higher in BY chickens than White Leghorn chickens [16]. During Salmonella infection, the spleen plays a vital role in revealing cellular damage and pathogenic mechanisms. In addition, mounting data shows that the spleen in avian species is more responsible for innate response shortly after disease detection by scavenging antigens from the blood than in mammalian species [17,18]. Little is known about the immune system genes and signaling pathways in BY and Cobb chickens during ST infection. This study identifies pathways and genes differentially expressed in BY compared to Cobb chickens post-ST challenging to elucidate effective host resistance mechanisms against ST.

2. Materials and Methods

2.1. Animals and Experimental Diets

The Animal Ethics Committee of the Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (IAS-CAAS, Beijing, China) approved the experimental procedures (IASCAAS2021-31). A total of 100 one-day-old Cobb broiler chicks purchased from Poultry Breeding Co., Ltd. (Beijing, China) and 100 one-day-old BY chicks obtained from the Changping Experimental farm of Institute of Animal Sciences (Beijing, China) were maintained under strict hygienic conditions in two isolated control chamber rooms at the experimental center of China Agricultural University (Beijing, China).
The Salmonella Typhimurium strain was resuscitated overnight in Luria–Bertani (LB) broth (Amresco, Washington, DC, USA) at 37 °C in an orbital shaking incubator at 150 rpm. After recovery, ST was cultured for 12 h and concentrated in a centrifuge. The final number of colony-forming units (CFU) was determined by plating serial dilutions as described in previous studies [19]. Salmonella testing was performed on all the chicks [20], and the positive chicks were eliminated. At 3 d of age, all the birds were challenged orally with 1 mL PBS holding 2.4 × 1012 CFU ST. At 1 DPI, the spleens and livers were collected. The spleens were placed in liquid nitrogen for storage, and the livers were used to measure the bacterial count of ST by calculating the number of colony-forming units on the MacConkey agar. According to the liver bacterial count and symptoms, (i.e., drooping wings, dying, and diarrhea), the spleens of 20 chicks from each group were chosen for RNA sequencing.

2.2. RNA Extraction, cDNA Library Preparation, and RNA-seq

The spleen RNAs were extracted from the 40 chicks using a QIAGEN RNeasy Kit (Qiagen, Hilden, Germany). The quality and quantity of the total RNA were assessed by a 2100 Bioanalyzer and RNA 6000 Nano kit (Agilent, Santa Clara, CA, USA). For the mRNA library construction and deep sequencing, 3 μg total of RNA was prepared using the TruSeq RNA Sample Preparation Kit (Illumina, San Diego, CA, USA) to capture the coding transcriptome. After purification, the RNA was fragmented by using divalent cations at 95 °C. The cleaved RNA fragments were reversely transcribed into first-strand cDNA using TruSeq RNA Library Preparation Kit, followed by second-strand cDNA synthesis. After cDNA fragment purification and adaptor ligation, RNA sequencing was performed on the HiSeq X Ten platform (Illumina, San Diego, CA, USA).
Sequence data can be accessed at the GSA of BIG Data Center, Beijing Institute of Genomics (BIG), Chinese Academy of Sciences and will be publicly accessible at https://ngdc.cncb.ac.cn/gsa/ (access number: CRA006334, accessed on 11 March 2024).

2.3. Differentially Expressed Genes and Function Enrichment Analysis

The quality control of reads were performed using FastQC (v0.11.5) [21]. Sequence adapters and low-quality reads were eliminated. Hisat2 was used mapping the sequencing data to the chicken reference genome (Gallus gallus 6.0) [22]. The expression quantities of the mapped transcripts were calculated using Htseq [23]. The DEGs analyses were conducted by DESeq2 [24]. Genes with fold change ≥2 and padj < 0.5 were considered DEGs. To assess the variation between samples, a cluster map of DEGs was conducted by pheatmap (https://CRAN.R-project.org/package=pheatmap (accessed on 15 April 2021)). To provide an overview of the DEGs, volcano plots were performed by ggplot2 [25]. The transcripts were converted to Entrez Gene IDs, and the gene enrichment analysis based on the DEGs was conducted by Gene Ontology (GO) functional enrichment analysis [26].

3. Results

3.1. Overview of the Bacterial Load between BY and Cobb Chicks

The number of colonies on MacConkey agar was calculated to assess the bacterial burden in the liver. As seen in Figure 1, BY chickens showed significantly lower liver bacterial count than Cobb chickens, indicating that BY chicks were more effective in eliminating ST. These findings explain the severe symptoms that appeared in Cobb more than BY chicks, which might illustrate the higher resistance against Salmonella shown in BY chickens than in Cobb chickens.

3.2. Sequencing of Spleen Transcriptomes

RNA-Seq of the spleen produced >20 Mb clean reads of 40 samples. Approximately 93% of the clean reads had quality scores exceeding the Q30 value. The data proved the reliability of the RNA-seq and could be used for data analysis. After removing the interference reads, the clean reads represented >93%, as shown in Supplementary Table S1.

3.3. DEGs in Response to ST Infection

The DEGs between BY and Cobb chickens were identified by DESeq2. There were 775 significant DEGs in the spleen after ST stimulation (498 up- and 277 down-regulated), as seen in Figure 2A and Supplementary Table S2. Many immune-related genes such as CATH1, AvBD4, BTN3A3, AvBD1, AvBD7, and CNTN3 were significantly upregulated (p < 0.01) between BY and Cobb after challenge with Salmonella (log2 FC 3.5, 3.53, 4.04, 3.6, 4.37, and 3.04, respectively). The Cluster map of DEGs revealed that the up-regulated more than down-regulated genes in spleen between BY and cobb chickens, as shown in Figure 2B.

3.4. Functional Enrichment Analysis of the DEGs

The function analysis of all DEGs was performed using GO and KEGG enrichment. A total of 51 GO terms were enriched and divided into: (i) 24 GO terms under biological process, (ii) 16 GO terms under cellular component, and (iii) 11 GO terms under molecular function, as shown in Supplementary Figure S1. The significantly enriched GO terms related to immune biological processes were mainly involved in immune response (GO:0006955), immune system process (GO:0002376), response to bacterium (GO:0009617), and response to stimulus (GO:0050896), as shown in Table 1.
After KEGG enrichment analysis, we selected the 20 pathways with the most significant (p < 0.05) enrichment. Pathways related to metabolism included Indole alkaloid biosynthesis; pathways related to immune system included toll-like receptor signaling pathways, complement and coagulation cascades, intestinal immune network for IgA production, NOD-like receptor signaling pathways, cytosolic DNA-sensing pathways, and natural-killer-cell-mediated cytotoxicity. In addition, pathways related to cellular processes included focal adhesion, gap junction, and phagosomes; pathways related to signaling molecules and interaction included cell adhesion molecules, ECM-receptor interaction, and neuroactive ligand–receptor interaction, as seen in Figure 3 and Supplementary Table S3.

4. Discussion

Changes in the transcriptomes of chickens after Salmonella infection have been studied extensively, particularly spleen transcriptomes. Although, few investigations on the differences in the transcriptomes of spleen in local and commercial chickens. The expression patterns of the spleen were compared in BY and Cobb chickens to understand functional and transcriptome alterations after Salmonella infection. In addition to clinical signs, the liver bacterial load is considered when evaluating the response of different chicken breeds to ST infection. The spleen expression profiles were determined by transcriptomic analysis.
In the early phases of Salmonella infection in newly hatched chicks, the innate immune system plays a critical role in preventing the spread of the pathogen [27]. The control of bacterial growth is based on host-produced cytokines, but bacterial clearance is dependent on the effective activation of CD4+ T cells, particularly in peripheral immunological organs [28].
Significantly altered signaling pathways associated with bacterial–epithelial interactions and immune responses have been identified in the spleen. The significantly altered pathways after ST infection included neuroactive ligand–receptor interaction and cytokine–cytokine receptor interaction, in line with earlier reports [29,30,31]. High doses of Salmonella can result in the production of excess amounts of proinflammatory cytokines, or a “cytokine storm”, leading to endotoxin shock or sepsis-related deaths [32,33,34]. Thus, the potential influence of over-expression of inflammatory cytokines due to hypersensitivity response to SE in susceptible birds was also considered in this study.
Several pathways related to the immune system were enriched in Cobb chickens, such as NOD-like receptor signaling pathways, neuroactive ligand–receptor interaction, natural-killer-cell-mediated cytotoxicity, and cytokine–cytokine receptor interaction, in agreement with a previous study [19]. Several immune-related pathways were activated in Cobb chickens, such as neuroactive ligand–receptor interaction, cytokine–cytokine receptor interaction, natural-killer-cell-mediated cytotoxicity, and NOD-like receptor signaling pathways. These findings revealed several signaling pathways controlling the invasion and clearance of Salmonella. Moreover, these enriched pathways were shown to be increased by many gene expressions resulting from infection with chicken pathogenic E. coli [35,36,37,38].
Intriguingly, overlapping genes were found in natural-killer-cell-mediated cytotoxicity, and phagosome and antigen processing and presentation pathways, as shown in Figure 4. This indicates that several signaling pathways may be involved in controlling the invasion and clearance of Salmonella. In addition, many genes in these identified pathways are expressed more in response to ST infection. MHC genes in chickens are essential for antigen presentation to T cells, which serve as a vital bridge between adaptive and innate immunity and are associated with resistance to Salmonella [39,40].
The hosts have special strategies to combat the infection. This study has enriched natural-killer-cell-mediated cytotoxicity and phagosomes. NK cells are scavenger cells that play a role in innate defense. Their primary function is to recognize and eliminate virally infected, altered, and neoplastic host cells [41,42]. NK cells play an important role in Salmonellosis in the first days of life [43].
Upon activation, NK cells release lytic granules containing perforin and granzyme, which are used to lyse target cells [44,45]. NK cells also secrete the cytokine IFNγ to attract other cells to initiate phagocytosis [46]. Heterophils ingest pathogens through a process known as phagocytosis. After ingestion, microorganisms are entrapped in a vacuole called the phagosome, which fuse directly with cytoplasmic granules killing trapped pathogens by releasing antimicrobial peptides and proteolytic enzymes from intracellular granules [47].
The intestinal immune network for IgA production also has been enriched. IgA is a major antibody isotype produced on the mucosal surface and plays an essential role in the immune response of the intestine and prevention of tissue damage during inflammation [19,48,49,50]. Once the intestinal immune system recognizes the invasion of pathogenic microorganisms, IgA is stimulated and transferred into the intestinal lumen [51]. Pathogen-binding IgA has been shown to regulate bacterial movement and protect the host from infection [52]. In addition, IgA inhibits bacterial penetration into epithelial cells by blocking the type III secretion system. [53]. As shown in Figure 5, IgA-production-signaling-pathway-related genes were more expressed in BY chickens.
Earlier studies have shown that macrophages initiate glycolytic upregulation while recognizing microbial ligands. Glycolytic energy promotes antibacterial inflammation and cytokine production [54,55]. After Salmonella infection, the spleen is a key immune organ with metabolic variations between BY and Cobb chickens.

5. Conclusions

This study aimed to characterize local chickens (BY) and commercial chickens (Cobb) splenic transcriptomes in response to Salmonella Typhimurium infection. A total of 775 DEGs were identified between BY and Cobb chicks. DEGs are mostly engaged in the innate immune system’s biological processes, and various pathways, including natural-killer-cell-mediated cytotoxicity, phagosomes, and the intestinal immune network for IgA production, are involved in chicken protection from Salmonella. This shows the ability of local chickens to resist Salmonella better than commercial chickens, which can pave a road for breeding strategies to improve the resistance of chickens against harmful bacteria in the food industry.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes13050811/s1, Figure S1: The enriched GO terms based on the DEGs identified in BY and cobb chicken; Table S1: RNA sequencing Summary; Table S2: Differential Gene Expression data for BY and Cobb chicken; Table S3: The top 20 significant KEGG enriched pathways of BY and Cobb chicken.

Author Contributions

Conception, M.S.E., Q.L., G.Z., and J.W.; methodology, M.S.E., H.W., and J.D.; formal analysis and data curation, M.S.E., M.M., Q.W., Q.Z., and N.Z.; funding acquisition, Q.W., Q.L., G.Z., and J.W.; investigation, M.S.E., H.W., M.M., J.D., Q.Z., and N.Z.; writing—original draft, M.S.E. and J.D.; administration, Q.W. and Q.L.; writing—review and editing, M.S.E., Q.W., Q.L., G.Z., and J.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by grants from the National Key R&D Program of China (2018YFE0128000).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of the Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China (IAS2021-31).

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Calenge, F.; Beaumont, C. Toward integrative genomics study of genetic resistance to Salmonella and Campylobacter intestinal colonization in fowl. Front. Genet. 2012, 3, 261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Calenge, F.; Kaiser, P.; Vignal, A.; Beaumont, C. Genetic control of resistance to salmonellosis and to Salmonella carrier-state in fowl: A review. Genet. Sel. Evol. 2010, 42, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Barrow, P.A.; Jones, M.A.; Smith, A.L.; Wigley, P. The long view: Salmonella–the last forty years. Avian Pathol. 2012, 41, 413–420. [Google Scholar] [CrossRef] [PubMed]
  4. Li, Y.; Yang, Q.; Cao, C.; Cui, S.; Wu, Y.; Yang, H.; Xiao, Y.; Yang, B.J.P.s. Prevalence and characteristics of Salmonella isolates recovered from retail raw chickens in Shaanxi Province, China. Poult. Sci. 2020, 99, 6031–6044. [Google Scholar] [CrossRef] [PubMed]
  5. Li, W.; Li, H.; Zheng, S.; Wang, Z.; Sheng, H.; Shi, C.; Shi, X.; Niu, Q.; Yang, B.J.F.i.M. Prevalence, serotype, antibiotic susceptibility, and genotype of Salmonella in eggs from poultry farms and marketplaces in Yangling, Shaanxi province, China. Front. Microbiol. 2020, 11, 1482. [Google Scholar] [CrossRef]
  6. Yang, X.; Wu, Q.; Zhang, J.; Huang, J.; Chen, L.; Wu, S.; Zeng, H.; Wang, J.; Chen, M.; Wu, H.; et al. Prevalence, bacterial load, and antimicrobial resistance of Salmonella serovars isolated from retail meat and meat products China. Front. Microbiol. 2019, 10, 2121. [Google Scholar] [CrossRef]
  7. DuPont, H.L.; Steele, J.H. The human health implication of the use of antimicrobial agents in animal feeds. Vet. Q. 1987, 9, 309–320. [Google Scholar] [CrossRef]
  8. Goldman, E. Antibiotic abuse in animal agriculture: Exacerbating drug resistance in human pathogens. Hum. Ecol. Risk Assess. 2004, 10, 121–134. [Google Scholar] [CrossRef]
  9. Hui, Y.Z. Serotypes and antimicrobial susceptibility of Salmonella spp. isolated from farm animals in China. Front. Microbiol. 2015, 6, 602. [Google Scholar] [CrossRef] [Green Version]
  10. Bar-Shira, E.; Sklan, D.; Friedman, A. Establishment of immune competence in the avian GALT during the immediate post-hatch period. Dev. Comp. Immunol. 2003, 27, 147–157. [Google Scholar] [CrossRef]
  11. Aljumaah, M.R.; Suliman, G.M.; Abdullatif, A.A.; Abudabos, A.M. Effects of phytobiotic feed additives on growth traits, blood biochemistry, and meat characteristics of broiler chickens exposed to Salmonella typhimurium. Poult. Sci. 2020, 99, 5744–5751. [Google Scholar] [CrossRef] [PubMed]
  12. Abudabos, A.M.; Aljumaah, M.R.; Aabdullatif, A.; Suliman, G.M. Feed supplementation with some natural products on Salmonella infected broilers’ performance and intestinal injury during the starter period. Ital. J. Anim. Sci. 2020, 19, 1304–1309. [Google Scholar] [CrossRef]
  13. Wu, C.M.; Zhao, G.P.; Wen, J.; Chen, J.L.; Zheng, M.Q.; Chen, G.H. Diversity of immune traits between Chinese Beijing-You chicken and White Leghorn. Acta Vet. Et Zootech. 2007, 38, 1383–1388. [Google Scholar]
  14. Zhang, L.; Li, P.; Liu, R.; Zheng, M.; Sun, Y.; Wu, D.; Hu, Y.; Wen, J.; Zhao, G. The identification of loci for immune traits in chickens using a genome-wide association study. PLoS ONE 2015, 10, e0117269. [Google Scholar] [CrossRef] [PubMed]
  15. Li, X.; Nie, C.; Zhang, Z.; Wang, Q.; Shao, P.; Zhao, Q.; Chen, Y.; Wang, D.; Li, Y.; Jiao, W. Evaluation of genetic resistance to Salmonella Pullorum in three chicken lines. Poult. Sci. 2018, 97, 764–769. [Google Scholar] [CrossRef]
  16. Liu, L.B.; Wu, C.M.; Wen, J.; Chen, J.L.; Zheng, M.Q.; Zhao, G.P. Association of SNPs in exon 2 of the MHC BF gene with immune traits in two distinct chicken populations: Chinese Beijing-You and White Leghorn. Acta Agric. Scand Sect. A 2009, 59, 4–11. [Google Scholar] [CrossRef]
  17. Smith, K.G.; Hunt, J.L. On the use of spleen mass as a measure of avian immune system strength. Oecologia 2004, 138, 28–31. [Google Scholar] [CrossRef]
  18. Tiron, A.; Vasilescu, C. Spleen and immunity. Immune implications of splenectomy. Chirurgia 2008, 103, 255–263. [Google Scholar]
  19. Wang, F.; Zhang, J.; Zhu, B.; Wang, J.; Wang, Q.; Zheng, M.; Wen, J.; Li, Q.; Zhao, G. Transcriptome analysis of the cecal tonsil of Jingxing yellow chickens revealed the mechanism of differential resistance to Salmonella. Genes 2019, 10, 979. [Google Scholar] [CrossRef] [Green Version]
  20. Li, P.; Xia, P.; Wen, J.; Zheng, M.; Chen, J.; Zhao, J.; Jiang, R.; Liu, R.; Zhao, G. Up-regulation of the MyD88-dependent pathway of TLR signaling in spleen and caecum of young chickens infected with Salmonella serovar Pullorum. Vet. Microbiol. 2010, 143, 346–351. [Google Scholar] [CrossRef]
  21. Chen, S.; Zhou, Y.; Chen, Y.; Gu, J. fastp: An ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 2018, 34, i884–i890. [Google Scholar] [CrossRef] [PubMed]
  22. Kim, D.; Langmead, B.; Salzberg, S.L. HISAT: A fast spliced aligner with low memory requirements. Nat. Methods 2015, 12, 357–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Anders, S.; Pyl, P.T.; Huber, W. HTSeq—A Python framework to work with high-throughput sequencing data. Bioinformatics 2015, 31, 166–169. [Google Scholar] [CrossRef] [PubMed]
  24. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 1–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ginestet, C. ggplot2: Elegant Graphics for Data Analysis; Wiley Online Library: Hoboken, NJ, USA, 2011. [Google Scholar]
  26. Young, M.D.; Wakefield, M.J.; Smyth, G.K.; Oshlack, A. Gene ontology analysis for RNA-seq: Accounting for selection bias. Genome Biol. 2010, 11, R14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Ijaz, A.; Veldhuizen, E.J.; Broere, F.; Rutten, V.P.; Jansen, C.A.J.P. The interplay between salmonella and intestinal innate immune cells in chickens. Pathogens 2021, 10, 1512. [Google Scholar] [CrossRef] [PubMed]
  28. Talbot, S.; Tötemeyer, S.; Yamamoto, M.; Akira, S.; Hughes, K.; Gray, D.; Barr, T.; Mastroeni, P.; Maskell, D.J.; Bryant, C.E. Toll-like receptor 4 signalling through MyD88 is essential to control Salmonella enterica serovar Typhimurium infection, but not for the initiation of bacterial clearance. Immunology 2009, 128, 472–483. [Google Scholar] [CrossRef] [Green Version]
  29. Li, P.; Fan, W.; Li, Q.; Wang, J.; Liu, R.; Everaert, N.; Liu, J.; Zhang, Y.; Zheng, M.; Cui, H. Splenic microRNA expression profiles and integration analyses involved in host responses to Salmonella enteritidis infection in chickens. Front. Cell. Infect. Microbiol. 2017, 7, 377. [Google Scholar] [CrossRef] [Green Version]
  30. Chiang, H.-I.; Swaggerty, C.L.; Kogut, M.H.; Dowd, S.E.; Li, X.; Pevzner, I.Y.; Zhou, H. Gene expression profiling in chicken heterophils with Salmonella enteritidis stimulation using a chicken 44 K Agilent microarray. BMC Genom. 2008, 9, 526. [Google Scholar] [CrossRef] [Green Version]
  31. Matulova, M.; Rajova, J.; Vlasatikova, L.; Volf, J.; Stepanova, H.; Havlickova, H.; Sisak, F.; Rychlik, I. Characterization of chicken spleen transcriptome after infection with Salmonella enterica serovar Enteritidis. PLoS ONE 2012, 7, e48101. [Google Scholar] [CrossRef]
  32. Cohen, J. The immunopathogenesis of sepsis. Nature 2002, 420, 885–891. [Google Scholar] [CrossRef] [PubMed]
  33. Netea, M.G.; Balkwill, F.; Chonchol, M.; Cominelli, F.; Donath, M.Y.; Giamarellos-Bourboulis, E.J.; Golenbock, D.; Gresnigt, M.S.; Heneka, M.T.; Hoffman, H.M. A guiding map for inflammation. Nat. Immunol. 2017, 18, 826–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Clark, I.A.; Vissel, B. The meteorology of cytokine storms, and the clinical usefulness of this knowledge. Semin. Immunopathol. 2017, 39, 505–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Li, G.; Tivendale, K.A.; Liu, P.; Feng, Y.; Wannemuehler, Y.; Cai, W.; Mangiamele, P.; Johnson, T.J.; Constantinidou, C.; Penn, C.W. Transcriptome analysis of avian pathogenic Escherichia coli O1 in chicken serum reveals adaptive responses to systemic infection. Infect. Immun. 2011, 79, 1951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sandford, E.E.; Orr, M.; Shelby, M.; Li, X.; Zhou, H.; Johnson, T.J.; Kariyawasam, S.; Liu, P.; Nolan, L.K.; Lamont, S.J. Leukocyte transcriptome from chickens infected with avian pathogenic Escherichia coli identifies pathways associated with resistance. Results Immunol. 2012, 2, 44–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Sun, H.; Liu, P.; Nolan, L.K.; Lamont, S.J. Novel pathways revealed in bursa of fabricius transcriptome in response to extraintestinal pathogenic Escherichia coli (ExPEC) infection. PLoS ONE 2015, 10, e0142570. [Google Scholar] [CrossRef] [Green Version]
  38. Sun, H.; Liu, P.; Nolan, L.K.; Lamont, S.J. Thymus transcriptome reveals novel pathways in response to avian pathogenic Escherichia coli infection. Poult. Sci. 2016, 95, 2803–2814. [Google Scholar] [CrossRef]
  39. Lamont, S.; Kaiser, M.; Liu, W.J. Candidate genes for resistance to Salmonella enteritidis colonization in chickens as detected in a novel genetic Salmonella. Vet. Immunol. Immunopathol. 2002, 87, 423–428. [Google Scholar] [CrossRef]
  40. Liu, W.; Miller, M.M.; Lamont, S.J.J.I. Association of MHC class I and class II gene polymorphisms with vaccine or challenge response to Salmonella enteritidis in young chicks. Immunogenetics 2002, 54, 582–590. [Google Scholar] [CrossRef]
  41. Morvan, M.G.; Lanier, L.L. NK cells and cancer: You can teach innate cells new tricks. Nat. Cancer 2016, 16, 7–19. [Google Scholar] [CrossRef]
  42. Lodoen, M.B.; Lanier, L.L. Natural killer cells as an initial defense against pathogens. Curr. Opin. Immunol. 2006, 18, 391–398. [Google Scholar] [CrossRef] [PubMed]
  43. Meijerink, N.; van den Biggelaar, R.H.; van Haarlem, D.A.; Stegeman, J.A.; Rutten, V.P.; Jansen, C.A. A detailed analysis of innate and adaptive immune responsiveness upon infection with Salmonella enterica serotype Enteritidis in young broiler chickens. Vet. Res. 2021, 52, 109. [Google Scholar] [CrossRef] [PubMed]
  44. Orange, J.S. Formation and function of the lytic NK-cell immunological synapse. Nat. Rev. Immunol. 2008, 8, 713–725. [Google Scholar] [CrossRef] [Green Version]
  45. Solana, R.; Tarazona, R.; Gayoso, I.; Lesur, O.; Dupuis, G.; Fulop, T. Innate immunosenescence: Effect of aging on cells and receptors of the innate immune system in humans. Semin. Immunol. 2012, 24, 331–341. [Google Scholar] [CrossRef]
  46. Carvajal, B.G.; Methner, U.; Pieper, J.; Berndt, A.J.V. Effects of Salmonella enterica serovar Enteritidis on cellular recruitment and cytokine gene expression in caecum of vaccinated chickens. Vaccine 2008, 26, 5423–5433. [Google Scholar] [CrossRef] [PubMed]
  47. Genovese, K.J.; He, H.; Swaggerty, C.L.; Kogut, M.H. The avian heterophil. Dev. Comp. Immunol. 2013, 41, 334–340. [Google Scholar] [CrossRef]
  48. Pabst, O. New concepts in the generation and functions of IgA. Nat. Rev. Immunol. 2012, 12, 821–832. [Google Scholar] [CrossRef]
  49. Slack, E.; Balmer, M.L.; Fritz, J.H.; Hapfelmeier, S. Functional flexibility of intestinal IgA–broadening the fine line. Front. Immunol. 2012, 3, 100. [Google Scholar] [CrossRef] [Green Version]
  50. Mkaddem, S.B.; Christou, I.; Rossato, E.; Berthelot, L.; Lehuen, A.; Monteiro, R.C. IgA, IgA receptors, and their anti-inflammatory properties. Fc Recept. 2014, 382, 221–235. [Google Scholar]
  51. Palm, N.W.; De Zoete, M.R.; Cullen, T.W.; Barry, N.A.; Stefanowski, J.; Hao, L.; Degnan, P.H.; Hu, J.; Peter, I.; Zhang, W. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 2014, 158, 1000–1010. [Google Scholar] [CrossRef] [Green Version]
  52. Forbes, S.J.; Eschmann, M.; Mantis, N.J. Inhibition of Salmonella enterica serovar typhimurium motility and entry into epithelial cells by a protective antilipopolysaccharide monoclonal immunoglobulin A antibody. Infect. Immun. 2008, 76, 4137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Forbes, S.J.; Bumpus, T.; McCarthy, E.A.; Corthésy, B.; Mantis, N.J. Transient suppression of Shigella flexneri type 3 secretion by a protective O-antigen-specific monoclonal IgA. MBio 2011, 2, e00042-11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Freemerman, A.J.; Johnson, A.R.; Sacks, G.N.; Milner, J.J.; Kirk, E.L.; Troester, M.A.; Macintyre, A.N.; Goraksha-Hicks, P.; Rathmell, J.C.; Makowski, L. Metabolic reprogramming of macrophages: Glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. J. Biol. Chem. 2014, 289, 7884–7896. [Google Scholar] [CrossRef] [Green Version]
  55. Van den Bossche, J.; O’Neill, L.A.; Menon, D. Macrophage immunometabolism: Where are we (going)? Trends Immunol. 2017, 38, 395–406. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Bacterial burden in the liver 1 day after oral infection of chicks with 2.4 × 1012 CFU ST/chick. The results are presented as individual values, mean ± standard deviation, * p < 0.05.
Figure 1. Bacterial burden in the liver 1 day after oral infection of chicks with 2.4 × 1012 CFU ST/chick. The results are presented as individual values, mean ± standard deviation, * p < 0.05.
Genes 13 00811 g001
Figure 2. (A) Volcano plot showing differentially expressed genes (DEGs) in spleen between BY and Cobb chickens after Salmonella infection (log2 FC ≥ 1 and padj < 0.05). (B) The heat map based on DEGs of the RNA-seq data.
Figure 2. (A) Volcano plot showing differentially expressed genes (DEGs) in spleen between BY and Cobb chickens after Salmonella infection (log2 FC ≥ 1 and padj < 0.05). (B) The heat map based on DEGs of the RNA-seq data.
Genes 13 00811 g002
Figure 3. KEGG pathway enrichment analysis. Differentially expressed genes in spleen. y-Axis represents enriched pathways; x-axis represents the rich factor of pathways. Bubble size represents the number of genes, and the color bar represents significance.
Figure 3. KEGG pathway enrichment analysis. Differentially expressed genes in spleen. y-Axis represents enriched pathways; x-axis represents the rich factor of pathways. Bubble size represents the number of genes, and the color bar represents significance.
Genes 13 00811 g003
Figure 4. Interaction between signaling pathways and overlapping genes involved in ST infection.
Figure 4. Interaction between signaling pathways and overlapping genes involved in ST infection.
Genes 13 00811 g004
Figure 5. IgA-production-signaling pathway. Green, significantly downregulated DEGs. Red, significantly upregulated DEGs.
Figure 5. IgA-production-signaling pathway. Green, significantly downregulated DEGs. Red, significantly upregulated DEGs.
Genes 13 00811 g005
Table 1. Immune-related biological processes identified by gene ontology analysis of DEGs in spleen.
Table 1. Immune-related biological processes identified by gene ontology analysis of DEGs in spleen.
TableDescriptionCountp-Value
GO:0006955immune response230.0001
GO:0009617response to bacterium100.001
GO:0098542defense response to other organism110.0013
GO:0002285lymphocyte activation involved in immune response60.0016
GO:0002376immune system process300.0058
GO:0006952defense response180.0067
GO:0051707response to other organism130.0073
GO:0002520immune system development140.0129
GO:0050896response to stimulus940.013
GO:0009607response to biotic stimulus130.0132
GO:0001775cell activation120.0205
GO:0032501multicellular organismal process730.026
GO:0009605response to external stimulus240.031
GO:0007275multicellular organism development560.0324
GO:0030154cell differentiation430.0396
GO:0045087innate immune response80.0462
DEGs between BY and Cobb chickens were used to identify enriched biological functions (p < 0.05).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Elsharkawy, M.S.; Wang, H.; Ding, J.; Madkour, M.; Wang, Q.; Zhang, Q.; Zhang, N.; Li, Q.; Zhao, G.; Wen, J. Transcriptomic Analysis of the Spleen of Different Chicken Breeds Revealed the Differential Resistance of Salmonella Typhimurium. Genes 2022, 13, 811. https://doi.org/10.3390/genes13050811

AMA Style

Elsharkawy MS, Wang H, Ding J, Madkour M, Wang Q, Zhang Q, Zhang N, Li Q, Zhao G, Wen J. Transcriptomic Analysis of the Spleen of Different Chicken Breeds Revealed the Differential Resistance of Salmonella Typhimurium. Genes. 2022; 13(5):811. https://doi.org/10.3390/genes13050811

Chicago/Turabian Style

Elsharkawy, Mohamed Shafey, Hailong Wang, Jiqiang Ding, Mahmoud Madkour, Qiao Wang, Qi Zhang, Na Zhang, Qinghe Li, Guiping Zhao, and Jie Wen. 2022. "Transcriptomic Analysis of the Spleen of Different Chicken Breeds Revealed the Differential Resistance of Salmonella Typhimurium" Genes 13, no. 5: 811. https://doi.org/10.3390/genes13050811

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop