Next Article in Journal
Decidua Parietalis Mesenchymal Stem/Stromal Cells and Their Secretome Diminish the Oncogenic Properties of MDA231 Cells In Vitro
Next Article in Special Issue
IL-19 Contributes to the Development of Nonalcoholic Steatohepatitis by Altering Lipid Metabolism
Previous Article in Journal
Orai3 Calcium Channel Regulates Breast Cancer Cell Migration through Calcium-Dependent and -Independent Mechanisms
Previous Article in Special Issue
Significance of Interleukin (IL)-4 and IL-13 in Inflammatory Arthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Interleukins Orchestrate Mucosal Immune Responses to Salmonella Infection in the Intestine

Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
Submission received: 14 November 2021 / Revised: 3 December 2021 / Accepted: 8 December 2021 / Published: 10 December 2021
(This article belongs to the Collection The Increasingly Fascinating World of Interleukins)

Abstract

:
Salmonella infection remains one of the major public health problems in the world, with increasing resistance to antibiotics. The resolution is to explore the pathogenesis of the infection and search for alternative therapy other than antibiotics. Immune responses to Salmonella infection include innate and adaptive immunity. Flagellin or muramyl dipeptide from Salmonella, recognized by extracellular Toll-like receptors and intracellular nucleotide-binding oligomerization domain2, respectively, induce innate immunity involving intestinal epithelial cells, neutrophils, macrophages, dendric cells and lymphocytes, including natural killer (NK) and natural killer T (NKT) cells. The cytokines, mostly interleukins, produced by the cells involved in innate immunity, stimulate adaptive immunity involving T and B cells. The mucosal epithelium responds to intestinal pathogens through its secretion of inflammatory cytokines, chemokines, and antimicrobial peptides. Chemokines, such as IL-8 and IL-17, recruit neutrophils into the cecal mucosa to defend against the invasion of Salmonella, but induce excessive inflammation contributing to colitis. Some of the interleukins have anti-inflammatory effects, such as IL-10, while others have pro-inflammatory effects, such as IL-1β, IL-12/IL-23, IL-15, IL-18, and IL-22. Furthermore, some interleukins, such as IL-6 and IL-27, exhibit both pro- and anti-inflammatory functions and anti-microbial defenses. The majority of interleukins secreted by macrophages and lymphocytes contributes antimicrobial defense or protective effects, but IL-8 and IL-10 may promote systemic Salmonella infection. In this article, we review the interleukins involved in Salmonella infection in the literature.

1. Introduction

Salmonella infection remains one of the major public health problems in the world, with increasing resistance to antibiotics. Non-typhoidal Salmonella (NTS) usually causes self-limiting diarrhea in immunocompetent hosts, but may develop into sepsis or complications in immunocompromised hosts.
Immune responses to Salmonella infection include innate and adaptive immunity. The different stages of Salmonella infection are reflected in the innate and acquired immunity, orchestrated by a variety of immune cells to defend against this bacterium, having a different importance during distinct infection stages. The innate immune system can restrict the replication of Salmonella to a certain degree, but acquired immunity is essential for the effective control and eradication of bacteria.
Besides intestinal epithelial cells which form a physical barrier and produce inflammatory cytokines, chemokines, and antimicrobial peptides [1,2], a variety of immune cells accomplish the innate immunity against Salmonella infection, including dendritic cells, neutrophils, macrophages, natural killer (NK), and γδ T cells. Phagocytes, central to the control of Salmonella infection during the initial stages of Salmonella infection, are recruited and activated by the inflammation of the infected tissues and large amounts of IFN-γ produced by a variety of cells, with NK cells being an important source [3]. Interleukin (IL)-8 recruits neutrophils from the circulation system into the infected tissue to defend against the invasion of Salmonella [4]. However, the accumulation of neutrophils gives rise to characteristic pathological changes of colitis. Macrophages appear to be crucial for protective immunity against intracellular Salmonella by phagocytosis of the bacteria and, along with dendritic cells, are major sources of many interleukins [5,6]. Moreover, dendritic cells play an important bridge between innate and acquired immunity via interleukins [7]. T cells are important for the control of S. typhimurium infection and CD4+ T cells in particular, but also CD8+ T cells and perhaps γδ T cells are involved [5]. Humoral immunity plays critical role(s) in the response to Salmonella infection, especially at the late phase [8]. Among intracellular bacteria, B cells play a notable role in resistance to Salmonella.
Infection leads to Toll-like receptors or intracellular nucleotide-binding oligomerization domain activation, the production of inflammatory interleukins such as IL-1α/β, IL-6, IL-8, IL-10, IL12/23, IL-15, IL-17A, IL-18, IL-25, IL-27, TNF-α, chemokine (C-C motif) ligand 2 (CCL2), IFN-γ, and neutrophil and macrophage recruitment. The plasma pro-inflammatory versus anti-inflammatory cytokine profile in patients with severe sepsis has been demonstrated to predict mortality [9,10,11,12,13,14]. In this article, we review the interleukins orchestrate intestinal mucosa responses to Salmonella infection in the literature (Table 1 and Table 2).

2. IL-1

Interleukin (IL)-1α and IL-1β are equally potent inflammatory cytokines but reveal highly dissimilar functions and biogenesis. IL-1α is constitutively expressed in many cell types at a steady state, and can be induced in response to cell stress, injury, infection, or pro-inflammatory stimuli [64]. IL-1α can function as a plasma membrane-bound cytokine. The exposure of cells to bacterial infection stimulates the intracellular expression of IL-1α as well as resulting in membrane IL-1α expression [65,66,67]. Although the biogenesis of IL-1α and its distinctive role in the inflammatory process remain poorly defined, recombinant murine TNF-α and IL-1α can protect mice from lethal bacterial infections [68]. The resistance of mice to a lethal dose of S. typhimurium could be enhanced if the mice were pretreated with IL-1α [15]. IL-1β is a key mediator of the inflammatory response and plays a major role on the pathogenesis of inflammatory bowel disease (IBD), consistent with the finding that IL-1β is up-regulated in IBD patients [48] and IBD colonic macrophages release mature IL-1β on exposure to lipopolysaccharide (LPS) [47]. The IL-1β-induced increase in the intestinal epithelial tight junction permeability may contribute to intestinal inflammation. Autophagy protein Atg16L1 polymorphisms in Crohn disease exhibit an excessive production of IL-1β and overwhelming inflammation in the colon [69,70]. Active vitamin D might enhance autophagy and decrease the IL-1β response to defend against Salmonella invasiveness and prevent the host from detrimental inflammation [2]. In vivo, active vitamin D3 attenuates the severity of Salmonella colitis in mice by decreasing IL-1β expression [50]. Furthermore, specific blockade of IL-1 receptors reduces the inflammatory responses in rabbit immune complex colitis [49]

3. IL-6

Interleukin (IL)-6 produced by enterocytes has anti-inflammatory and cell-protective effects in intestinal mucosa and enterocytes. It may counteract some of the injurious effects of sepsis and endotoxemia [17,71]. IL-6 has been reported to be a mediator of the epithelial barrier protection [72] and endogenous IL-6 plays an essential, non-redundant role in limiting intestinal injury [16]. Salmonella-induced intense inflammation causes the breakdown of the intestinal epithelial barrier, translocation of bacteria, and absorption of endotoxins into the circulation [73] and, consequently, bacteremia as well as endotoxemia. The probiotic Lactobacillus paracasei may exert some of their beneficial effects by enhancing IL-6 production in enterocytes subjected to an inflammatory stimulus [18]. PJ-34, a potent poly (ADP-ribose) polymerase-1 (PARP-1) inhibitor, may exert defense on intestinal epithelial cells (IECs) against invasive Salmonella infection by up-regulating IL-6 production through the ERK and NF-κB signal pathway [19].

4. IL-8

Current data indicate that IECs orchestrate mucosal innate immunity through their production of inflammatory cytokines, chemokines, and antimicrobial peptides [74,75]. Chemokines, such as IL-8, recruits neutrophils into cecal mucosa to defend against the invasion of Salmonella [20,21,22]. However, the accumulation of neutrophils gives rise to colitis [76] and sepsis [51]. Toll-like receptor 5 (TLR5) and intracellular nucleotide-binding oligomerization domain 2 (NOD2) are two important pattern recognition receptors involved in innate immunity to invading pathogens. Flagellin, a ligand for TLR5, is a dominant pro-inflammatory determinant in IECs infected by Salmonella. The cooperation of flagellin and muramyl dipeptide, a NOD2 agonist, in IECs synergistically upregulates inflammatory IL-8 response to Salmonella infection [23]. Intracellular Salmonella infection in both macrophages and IECs induces cholesterol accumulation in the Salmonella-containing vesicles (SCVs) [77] within which the virulent bacteria survive and replicate [78,79]. Plasma membrane cholesterol supports the survival of Salmonella in IECs through the PI3K-dependent anti-IL-8 pathway [24]. Contrasting to membrane cholesterol, sphingolipids act on epithelial defense against the invasive pathogen by enhancing the NOD2-mediated human beta-defensin 2 (hBD-2) response [80]. The probiotic Lactobacillus plantarum Lp62 inhibited IL-8 production by Salmonella Typhi-stimulated IECs and prevented the adhesion of pathogens to the cells [57]. The treatment of probiotics before and after infection having different effects on the Salmonella-induced IL-8 response in IECs suggests the critical timing of probiotic supplementation [25]. Active vitamin D modulates the pro-inflammatory IL-8 response in Salmonella-infected IECs to prevent the host from detrimental extreme inflammation [1].

5. IL-10

IL-10 is a powerful anti-inflammatory Th2 cytokine with a broad range of target cell types, primarily of the innate cells, such as dendric cells (DCs), macrophages, neutrophils, B cells, and T cells. Mice deficient in IL-10 spontaneously develop colitis resembling IBD. During Clostridium difficile infection, IL-10 is required to avoid an excessive immune reaction and prevent the host from more severe acute colitis [81]. IL-10 can dampen the secretion of pro-inflammatory cytokines, such as IL-12 [26] and prevents tissue damage [82] by acting on antigen-presenting cells. Furthermore, IL-10 facilities the regulatory T (Treg) cell-mediated suppression of T helper 17 (Th17)-driven colitis in mice [27]. On the other hand, S. Typhimurium can infect and persist in B cells [83], which provide additional signals to transform T cells to Treg cells. IL-10 produced by T and B cells promotes systemic Salmonella enterica serovar Typhimurium infection in mice [52]. The in vivo administration of the anti-IL-10 monoclonal antibody significantly enhanced host resistance at the early stage of Salmonella infection by accelerating macrophage functions and, consequently, the activation of γδT cells and enhanced levels of monokine mRNA, including IL-lα, tumor necrosis factor-α (TNF-α), and IL-12 [53].

6. IL-12

IL-12, similar to IL-23, is a pro-inflammatory cytokine produced by activated DCs, macrophages in response to microbial pathogens [84,85], and stimulates natural killer (NK), T, and natural killer T (NKT) cells to produce IFN-γ, which, in turn, aid in the elimination of intramacrophage pathogens, including S. Typhimurium [30,86]. These two cytokines must be considered together, because both share a common p40 subunit and their biology is closely interlocked [7]. IL-12 and IL-23 can also induce TNF-α and GM-CSF production in T cells via IFN-γ-independent signal transduction and bactericidal mechanisms [28]. In a human study, a high incidence of invasive Salmonella diseases in patients with IL-12/IL-23-INF-γ-axis deficiency highlights the importance of IL-12/IL-23- INF-γ-axis for immunity against Salmonella in humans [29]. Clinicians should consider an underlying IL-12/IL-23-INF-γ-axis deficiency in patients with recurrent, extraintestinal, and invasive Salmonella diseases, which usually require extensive treatment. Recombinant gamma interferon enhances the internalization and early intracellular killing of Salmonella enterica serovar Typhimurium by human macrophages [30], which release more IL-12 and less IL-10. Ustekinumab, the monoclonal antibody targeting the shared p40 subunit, has been approved for Crohn’s disease (CD) and has demonstrated promising results in the treatment of ulcerative colitis [54].

7. IL-15

Interleukin-15 (IL-15) is a cytokine that resembles IL-2 in its biological activities [31], stimulating macrophages, NK cells, T cells, and B cells to secrete cytokines, and has pro-inflammatory properties by itself [55]. It is upregulated under the conditions of tissue destruction and during infection [87,88]. IL-15 was reported to be involved in protection against bacterial infection mediated by NK and γδ T cells [31]. IL-15 overexpression promotes intestinal dysbiosis with a decrease in luminal butyrate-producing bacteria, lowers butyrate levels, and is associated with an increased susceptibility to colitis [56] and impacts on the pathogenesis of intestinal inflammatory diseases, such as celiac disease and IBD. Endogenous IL-15 had an important function in early protection against infection with an avirulent strain of S. choleraesuis through the activation of NK cells and IFN- γ production [32].

8. IL-17

Interleukin-17 (IL-17), a cytokine produced by Th17 cells, recruits neutrophils and plays a crucial role in host defense against extracellular bacteria [33]. Additionally, IL-17 helps to orchestrate mucosal inflammation by inducing the production of neutrophil chemoattractants (e.g., IL-8, CCL20, Lipocalin-2, and iNOS) in the intestines [41]. In IBD patients, the increased expression of IL-17 was observed in the intestinal mucosa [89]. In the mouse colitis model of S. Typhimurium infection, IL-23 orchestrates mucosal responses to release IL-17 [41] that contribute specifically to neutrophil recruitment into the cecal mucosa to prevent Salmonella dissemination [34,35]. iNKT cells play a protective role against Salmonella enterocolitis by downregulating IL-17-producing γδT cells [58]. The probiotic Lactobacillus plantarum ZS2058 significantly reduced the pathogenicity of Salmonella colitis by promoting the colon IL23/IL-22 axis in the mouse [36]. Lactobacillus plantarum Lp62 was able to suppress IL-17, IL1-β, and TNF-α production in LPS-stimulated J774 macrophages [57].

9. IL-18

IL-18 is a cytokine that binds to a specific receptor expressed on various types of cells and has pleiotropic functions [37,59]. Its capability to promote INF-γ production by T cells and NK cells leads to pro-inflammatory activity. Colon epithelial cells constitutively produce IL-18, which increases upon NLRP6 inflammasome and in turn activates the epithelial cells to produce antimicrobial peptides to maintain microbiome homeostasis [38]. Moreover, the binding of TLR5 to its ligand flagellin not only results in a NF-κB-mediated pro-inflammatory cytokine responses, including IL-8, TNFα, and the matrix metalloprotease (MMP)-9, but induces IL-18 secretion and Th1-like cytokine responses in human peripheral blood mononuclear cells (PBMC) [37]. Salmonella pathogenicity islands (SPI)-1 effector secretion leads to NF-κB signaling and caspase-1-mediated IL-1β/IL-18 activation [60].

10. IL-22

The major functions of IL-22 in the intestine are the inflammatory response, enhanced antimicrobial activity, the maintenance of the mucosal barrier, resistance to colonization of opportunistic pathogens, enhancement of epithelial regeneration, and wound healing [40,90]. IL-22 can restrict the growth of M. tuberculosis intracellularly in macrophages by enhancing phagosomal fusion. IL-22 can also orchestrate mucosal inflammation by inducing the production of neutrophil chemoattractants (e.g., IL-8, CCL20, Lipocalin-2, and iNOS) in the intestines [41]. IL-22 promotes the intracellular fusion of SCVs with lysosomes, leading to the phagolysosomal killing of S. Typhimurium in human epithelial cells [39]. Salmonella-induced IL-22 production can suppress the growth of commensal Enterobacteriaceae, the closest competitors for Salmonella, in the inflamed gut, thereby enhancing the Salmonella colonization of mucosal layers [91]. Furthermore, the ability of IL-22 to heal intestinal inflammation and promote epithelial repair from acute injury [40] highlights IL-22 as a promising target for future IBD therapy.

11. IL-23

Interleukin-23 (IL-23) is a member of the IL-12 family of cytokines with pro-inflammatory properties. Its ability to potently enhance the expansion of Th17 cells indicates the responsibility for many of the inflammatory autoimmune responses. IL-23 is a key participant in the central regulation of the cellular mechanisms involved in inflammation [61]. IL-23 is produced by various immune cells such as dendritic cells, monocytes, as well as type 1 macrophages (Mϕ1) upon Toll-like receptor signaling [92] in response to the binding of pathogens. In addition, neutrophils have been identified as a potential source of IL-23 production [93]. Recent studies have identified an increased production of IL-23 in various mouse models of colitis and IBD patients (review in [63]). IL-23 is able to accelerate the proliferation of both murine and human memory T cells producing Th17 cytokines such as IL-17A, IL-17F, and IL-22 [62]. IL-23 is known to induce IFN-γ production and is associated with host innate immunity against Salmonella. The IL-23/IL-22 axis during innate immunity against Salmonella may contribute to protection against Salmonella infection by several ways, such as IL-22-regulated expression of anti-microbial peptides and acute phase proteins and IL-17A-dependent neutrophil recruitment [42]. IL-23 deficient mice were unable to express IL-17A during S. Typhimurium-induced colitis [41]. Infection with Salmonella can induce IL-23, IL-18, and IL-1β, but not IL-12, production in monocytes and type 1 pro-inflammatory macrophages [94].

12. IL-27

Interleukin (IL)-27, one of heterodimeric cytokines that belongs to the IL-12 family, has fundamental roles in innate and adaptive immune regulation [95]. It is produced in myeloid cells in response to bacterial infection and has both anti- and pro-inflammatory functions [95,96]. Accordingly, the role of IL-27 in human and experimental mouse colitis is controversial. IL-27 enhanced TLR4 or TLR5 expression in human monocytes and macrophages, induced greater LPS/flagellin-mediated signaling, and significantly enhanced pro-inflammatory cytokines IL-12p40, TNF-α, and IL-6 production in S. typhimurium-infected cells [43,44,46]. These findings support the role for IL-27 in anti-microbial defenses by altering the expression of innate immune sensors such as TLR4 or TLR5 [45].

13. Conclusions

Salmonella infection remains one of the major public health problems in the world, with increasing resistance to antibiotics. The resolution is to look for alternative therapy other than antibiotics based on the host immune reaction to infection. Interleukins play a crucial role in orchestrating mucosal immune responses to Salmonella infection in the intestine. How to take advantage of is knowledge and exploit effective reagents to treat Salmonella infection would be the essential next-generation issue.
In recent years, our group has contributed much effort to exploit the biotherapy that could prevent or treat Salmonellosis. PJ-34, a PARP-1 inhibitor, may exert its protective effect on intestinal epithelial cells against invasive Salmonella infection by up-regulating IL-6 production, which has anti-inflammatory and cell-protective effects [19], and counteracts some of the injurious effects of sepsis and endotoxemia.
Salmonella-induced plasma membrane cholesterol accumulation in SCVs [24] may, subsequently, protect intestinal epithelial cells from apoptosis and produce an anti-inflammatory signal, both of which may contribute to the establishment of a Salmonella infection in cells. Contrasting to the utilization of membrane cholesterol on the maintenance of Salmonella-containing vacuoles and anti-inflammatory responses, sphingolipids act on the epithelial defense against the invasive pathogen [80]. Simvastatin or Fluvastatin, cholesterol-lowering statins, can suppress the pro-inflammatory IL-8 response in Salmonella-infected IECs to prevent the detrimental effects of overwhelming inflammation in the host [97].
Our recent in vitro and in vivo studies observed that active vitamin D prevented the host from the detrimental effects of overwhelming inflammation by downregulating pro-inflammatory responses (IL-6, TNF-α, IL-8, and IL-1β) [1,2,25] in Salmonella-infected IECs and decreased the severity of colitis in mice. The different regulation of probiotics on Salmonella-induced IL-8 responses in Caco-2 cells according to the administered timing supports a rationale for the therapeutic use of probiotics in the treatment of Salmonella colitis and inflammatory bowel disease [25]. Furthermore, we observed the synergistic effects of probiotics or postbiotics and active vitamin D on anti-inflammatory responses (IL-6, TNF-α, IL-8, and IL-1β) in Salmonella colitis mice [98,99].
It is mandatory to elucidate the pathogenesis of Salmonella infection for the design of intervention strategies that might reduce the use of antimicrobial agents and decrease the incidence of multidrug-resistant Salmonellosis. All these novel findings and thoughtful explorations of health knowledge could be applied to perform clinical trials and preventive medicine for the better lives of future generations.

Funding

This research was funded by the Ministry of Science and Technology, grant number MOST 107-2314-B-182-046-MY2, and the Chang Gung Memorial Hospital, grant number CMRPG8H0681 and CMRPG8K0421.

Acknowledgments

We thank Yi-Lee and the Chang Gung Medical Foundation Kaohsiung Chang Gung Memorial Hospital Tissue Bank Core Lab and Biobank (CLRPG8I0032) for excellent technical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Huang, F.C. The differential effects of 1,25-dihydroxyvitamin D3 on Salmonella-induced interleukin-8 and human beta-defensin-2 in intestinal epithelial cells. Clin. Exp. Immunol. 2016, 185, 98–106. [Google Scholar] [CrossRef] [Green Version]
  2. Huang, F.C. Vitamin D differentially regulates Salmonella-induced intestine epithelial autophagy and interleukin-1beta expression. World J. Gastroenterol. 2016, 22, 10353–10363. [Google Scholar] [CrossRef] [PubMed]
  3. Ramarathinam, L.; Niesel, D.W.; Klimpel, G.R. Salmonella typhimurium induces IFN-gamma production in murine splenocytes. Role of natural killer cells and macrophages. J. Immunol. 1993, 150, 3973–3981. [Google Scholar] [PubMed]
  4. Fleckenstein, J.M.; Kopecko, D.J. Breaching the mucosal barrier by stealth: An emerging pathogenic mechanism for enteroadherent bacterial pathogens. J. Clin. Investig. 2001, 107, 27–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Mittrucker, H.W.; Kaufmann, S.H. Immune response to infection with Salmonella typhimurium in mice. J. Leukoc. Biol. 2000, 67, 457–463. [Google Scholar] [CrossRef] [PubMed]
  6. Tam, M.A.; Rydstrom, A.; Sundquist, M.; Wick, M.J. Early cellular responses to Salmonella infection: Dendritic cells, monocytes, and more. Immunol. Rev. 2008, 225, 140–162. [Google Scholar] [CrossRef] [PubMed]
  7. Trinchieri, G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev. Immunol. 2003, 3, 133–146. [Google Scholar] [CrossRef] [PubMed]
  8. Takaya, A.; Yamamoto, T.; Tokoyoda, K. Humoral Immunity vs. Salmonella. Front. Immunol. 2019, 10, 3155. [Google Scholar] [CrossRef] [PubMed]
  9. Sullivan, J.S.; Kilpatrick, L.; Costarino, A.T., Jr.; Lee, S.C.; Harris, M.C. Correlation of plasma cytokine elevations with mortality rate in children with sepsis. J. Pediatr. 1992, 120, 510–515. [Google Scholar] [CrossRef]
  10. Casey, L.C.; Balk, R.A.; Bone, R.C. Plasma cytokine and endotoxin levels correlate with survival in patients with the sepsis syndrome. Ann. Intern. Med. 1993, 119, 771–778. [Google Scholar] [CrossRef] [PubMed]
  11. Wong, H.R.; Cvijanovich, N.; Wheeler, D.S.; Bigham, M.T.; Monaco, M.; Odoms, K.; Macias, W.L.; Williams, M.D. Interleukin-8 as a stratification tool for interventional trials involving pediatric septic shock. Am. J. Respir. Crit. Care Med. 2008, 178, 276–282. [Google Scholar] [CrossRef]
  12. Carrol, E.D.; Payton, A.; Payne, D.; Miyajima, F.; Chaponda, M.; Mankhambo, L.A.; Banda, D.L.; Molyneux, E.M.; Cox, H.; Jacobson, G.; et al. The IL1RN promoter rs4251961 correlates with IL-1 receptor antagonist concentrations in human infection and is differentially regulated by GATA-1. J. Immunol. 2011, 186, 2329–2335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. van Dissel, J.T.; van Langevelde, P.; Westendorp, R.G.; Kwappenberg, K.; Frolich, M. Anti-inflammatory cytokine profile and mortality in febrile patients. Lancet 1998, 351, 950–953. [Google Scholar] [CrossRef]
  14. Gogos, C.A.; Drosou, E.; Bassaris, H.P.; Skoutelis, A. Pro-versus anti-inflammatory cytokine profile in patients with severe sepsis: A marker for prognosis and future therapeutic options. J. Infect. Dis. 2000, 181, 176–180. [Google Scholar] [CrossRef]
  15. Morrissey, P.J.; Charrier, K. Treatment of mice with IL-1 before infection increases resistance to a lethal challenge with Salmonella typhimurium. The effect correlates with the resistance allele at the Ity locus. J. Immunol. 1994, 153, 212–219. [Google Scholar] [PubMed]
  16. Jin, X.; Zimmers, T.A.; Zhang, Z.; Pierce, R.H.; Koniaris, L.G. Interleukin-6 is an important in vivo inhibitor of intestinal epithelial cell death in mice. Gut 2010, 59, 186–196. [Google Scholar] [CrossRef]
  17. Xing, Z.; Gauldie, J.; Cox, G.; Baumann, H.; Jordana, M.; Lei, X.F.; Achong, M.K. IL-6 is an antiinflammatory cytokine required for controlling local or systemic acute inflammatory responses. J. Clin. Investig. 1998, 101, 311–320. [Google Scholar] [CrossRef] [PubMed]
  18. Reilly, N.; Poylin, V.; Menconi, M.; Onderdonk, A.; Bengmark, S.; Hasselgren, P.O. Probiotics potentiate IL-6 production in IL-1beta-treated Caco-2 cells through a heat shock-dependent mechanism. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 293, R1169–R1179. [Google Scholar] [CrossRef]
  19. Huang, F.C. Upregulation of Salmonella-induced IL-6 production in Caco-2 cells by PJ-34, PARP-1 inhibitor: Involvement of PI3K, p38 MAPK, ERK, JNK, and NF-kappaB. Mediat. Inflamm. 2009, 2009, 103890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. McCormick, B.A.; Hofman, P.M.; Kim, J.; Carnes, D.K.; Miller, S.I.; Madara, J.L. Surface attachment of Salmonella typhimurium to intestinal epithelia imprints the subepithelial matrix with gradients chemotactic for neutrophils. J. Cell Biol. 1995, 131, 1599–1608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Eckmann, L.; Kagnoff, M.F.; Fierer, J. Epithelial cells secrete the chemokine interleukin-8 in response to bacterial entry. Infect. Immun. 1993, 61, 4569–4574. [Google Scholar] [CrossRef] [Green Version]
  22. Kucharzik, T.; Williams, I.R. Neutrophil migration across the intestinal epithelial barrier--summary of in vitro data and description of a new transgenic mouse model with doxycycline-inducible interleukin-8 expression in intestinal epithelial cells. Pathobiology 2002, 70, 143–149. [Google Scholar] [CrossRef]
  23. Huang, F.C. Regulation of Salmonella flagellin-induced interleukin-8 in intestinal epithelial cells by muramyl dipeptide. Cell Immunol. 2012, 278, 1–9. [Google Scholar] [CrossRef]
  24. Huang, F.C. Plasma membrane cholesterol plays a critical role in the Salmonella-induced anti-inflammatory response in intestinal epithelial cells. Cell Immunol. 2011, 271, 480–487. [Google Scholar] [CrossRef]
  25. Huang, F.C.; Huang, S.C. The different effects of probiotics treatment on Salmonella-induced interleukin-8 response in intestinal epithelia cells via PI3K/Akt and NOD2 expression. Benef. Microbes 2016, 7, 739–748. [Google Scholar] [CrossRef]
  26. Ma, X.; Yan, W.; Zheng, H.; Du, Q.; Zhang, L.; Ban, Y.; Li, N.; Wei, F. Regulation of IL-10 and IL-12 production and function in macrophages and dendritic cells. F1000Research 2015, 4. [Google Scholar] [CrossRef] [Green Version]
  27. Chaudhry, A.; Samstein, R.M.; Treuting, P.; Liang, Y.; Pils, M.C.; Heinrich, J.M.; Jack, R.S.; Wunderlich, F.T.; Bruning, J.C.; Muller, W.; et al. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 2011, 34, 566–578. [Google Scholar] [CrossRef] [Green Version]
  28. van de Vosse, E.; Ottenhoff, T.H. Human host genetic factors in mycobacterial and Salmonella infection: Lessons from single gene disorders in IL-12/IL-23-dependent signaling that affect innate and adaptive immunity. Microbes Infect. 2006, 8, 1167–1173. [Google Scholar] [CrossRef] [PubMed]
  29. MacLennan, C.; Fieschi, C.; Lammas, D.A.; Picard, C.; Dorman, S.E.; Sanal, O.; MacLennan, J.M.; Holland, S.M.; Ottenhoff, T.H.; Casanova, J.L.; et al. Interleukin (IL)-12 and IL-23 are key cytokines for immunity against Salmonella in humans. J. Infect. Dis. 2004, 190, 1755–1757. [Google Scholar] [CrossRef] [Green Version]
  30. Gordon, M.A.; Jack, D.L.; Dockrell, D.H.; Lee, M.E.; Read, R.C. Gamma interferon enhances internalization and early nonoxidative killing of Salmonella enterica serovar Typhimurium by human macrophages and modifies cytokine responses. Infect. Immun. 2005, 73, 3445–3452. [Google Scholar] [CrossRef] [Green Version]
  31. Carson, W.E.; Giri, J.G.; Lindemann, M.J.; Linett, M.L.; Ahdieh, M.; Paxton, R.; Anderson, D.; Eisenmann, J.; Grabstein, K.; Caligiuri, M.A. Interleukin (IL) 15 is a novel cytokine that activates human natural killer cells via components of the IL-2 receptor. J. Exp. Med. 1994, 180, 1395–1403. [Google Scholar] [CrossRef]
  32. Hirose, K.; Nishimura, H.; Matsuguchi, T.; Yoshikai, Y. Endogenous IL-15 might be responsible for early protection by natural killer cells against infection with an avirulent strain of Salmonella choleraesuis in mice. J. Leukoc. Biol. 1999, 66, 382–390. [Google Scholar] [CrossRef]
  33. Santos, R.L.; Raffatellu, M.; Bevins, C.L.; Adams, L.G.; Tukel, C.; Tsolis, R.M.; Baumler, A.J. Life in the inflamed intestine, Salmonella style. Trends Microbiol. 2009, 17, 498–506. [Google Scholar] [CrossRef] [Green Version]
  34. Raffatellu, M.; Santos, R.L.; Verhoeven, D.E.; George, M.D.; Wilson, R.P.; Winter, S.E.; Godinez, I.; Sankaran, S.; Paixao, T.A.; Gordon, M.A.; et al. Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency promotes Salmonella dissemination from the gut. Nat. Med. 2008, 14, 421–428. [Google Scholar] [CrossRef] [Green Version]
  35. Keestra, A.M.; Godinez, I.; Xavier, M.N.; Winter, M.G.; Winter, S.E.; Tsolis, R.M.; Baumler, A.J. Early MyD88-dependent induction of interleukin-17A expression during Salmonella colitis. Infect. Immun. 2011, 79, 3131–3140. [Google Scholar] [CrossRef] [Green Version]
  36. Liu, J.; Gu, Z.; Song, F.; Zhang, H.; Zhao, J.; Chen, W. Lactobacillus plantarum ZS2058 and Lactobacillus rhamnosus GG Use Different Mechanisms to Prevent Salmonella Infection in vivo. Front. Microbiol. 2019, 10, 299. [Google Scholar] [CrossRef]
  37. Bachmann, M.; Horn, K.; Poleganov, M.A.; Paulukat, J.; Nold, M.; Pfeilschifter, J.; Muhl, H. Interleukin-18 secretion and Th1-like cytokine responses in human peripheral blood mononuclear cells under the influence of the toll-like receptor-5 ligand flagellin. Cell Microbiol. 2006, 8, 289–300. [Google Scholar] [CrossRef]
  38. Levy, M.; Shapiro, H.; Thaiss, C.A.; Elinav, E. NLRP6: A Multifaceted Innate Immune Sensor. Trends Immunol. 2017, 38, 248–260. [Google Scholar] [CrossRef]
  39. Forbester, J.L.; Lees, E.A.; Goulding, D.; Forrest, S.; Yeung, A.; Speak, A.; Clare, S.; Coomber, E.L.; Mukhopadhyay, S.; Kraiczy, J.; et al. Interleukin-22 promotes phagolysosomal fusion to induce protection against Salmonella enterica Typhimurium in human epithelial cells. Proc. Natl. Acad. Sci. USA 2018, 115, 10118–10123. [Google Scholar] [CrossRef] [Green Version]
  40. Mizoguchi, A. Healing of intestinal inflammation by IL-22. Inflamm. Bowel Dis. 2012, 18, 1777–1784. [Google Scholar] [CrossRef]
  41. Godinez, I.; Raffatellu, M.; Chu, H.; Paixao, T.A.; Haneda, T.; Santos, R.L.; Bevins, C.L.; Tsolis, R.M.; Baumler, A.J. Interleukin-23 orchestrates mucosal responses to Salmonella enterica serotype Typhimurium in the intestine. Infect. Immun. 2009, 77, 387–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Siegemund, S.; Schutze, N.; Schulz, S.; Wolk, K.; Nasilowska, K.; Straubinger, R.K.; Sabat, R.; Alber, G. Differential IL-23 requirement for IL-22 and IL-17A production during innate immunity against Salmonella enterica serovar Enteritidis. Int. Immunol. 2009, 21, 555–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Guzzo, C.; Ayer, A.; Basta, S.; Banfield, B.W.; Gee, K. IL-27 enhances LPS-induced proinflammatory cytokine production via upregulation of TLR4 expression and signaling in human monocytes. J. Immunol. 2012, 188, 864–873. [Google Scholar] [CrossRef] [Green Version]
  44. Petes, C.; Wynick, C.; Guzzo, C.; Mehta, D.; Logan, S.; Banfield, B.W.; Basta, S.; Cooper, A.; Gee, K. IL-27 enhances LPS-induced IL-1beta in human monocytes and murine macrophages. J. Leukoc. Biol. 2017, 102, 83–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Petes, C.; Odoardi, N.; Plater, S.M.; Martin, N.L.; Gee, K. IL-27 amplifies cytokine responses to Gram-negative bacterial products and Salmonella typhimurium infection. Sci. Rep. 2018, 8, 13704. [Google Scholar] [CrossRef]
  46. Royle, M.C.; Totemeyer, S.; Alldridge, L.C.; Maskell, D.J.; Bryant, C.E. Stimulation of Toll-like receptor 4 by lipopolysaccharide during cellular invasion by live Salmonella typhimurium is a critical but not exclusive event leading to macrophage responses. J. Immunol. 2003, 170, 5445–5454. [Google Scholar] [CrossRef] [Green Version]
  47. McAlindon, M.E.; Hawkey, C.J.; Mahida, Y.R. Expression of interleukin 1 beta and interleukin 1 beta converting enzyme by intestinal macrophages in health and inflammatory bowel disease. Gut 1998, 42, 214–219. [Google Scholar] [CrossRef]
  48. Ludwiczek, O.; Vannier, E.; Borggraefe, I.; Kaser, A.; Siegmund, B.; Dinarello, C.A.; Tilg, H. Imbalance between interleukin-1 agonists and antagonists: Relationship to severity of inflammatory bowel disease. Clin. Exp. Immunol. 2004, 138, 323–329. [Google Scholar] [CrossRef] [PubMed]
  49. Cominelli, F.; Nast, C.C.; Clark, B.D.; Schindler, R.; Lierena, R.; Eysselein, V.E.; Thompson, R.C.; Dinarello, C.A. Interleukin 1 (IL-1) gene expression, synthesis, and effect of specific IL-1 receptor blockade in rabbit immune complex colitis. J. Clin. Investig. 1990, 86, 972–980. [Google Scholar] [CrossRef] [Green Version]
  50. Huang, F.C.; Huang, S.C. Active vitamin D3 attenuates the severity of Salmonella colitis in mice by orchestrating innate immunity. Immun. Inflamm. Dis. 2021, 9, 481–491. [Google Scholar] [CrossRef] [PubMed]
  51. Gilchrist, J.J.; Heath, J.N.; Msefula, C.L.; Gondwe, E.N.; Naranbhai, V.; Mandala, W.; MacLennan, J.M.; Molyneux, E.M.; Graham, S.M.; Drayson, M.T.; et al. Cytokine Profiles during Invasive Nontyphoidal Salmonella Disease Predict Outcome in African Children. Clin. Vaccine Immunol. 2016, 23, 601–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Salazar, G.A.; Penaloza, H.F.; Pardo-Roa, C.; Schultz, B.M.; Munoz-Durango, N.; Gomez, R.S.; Salazar, F.J.; Pizarro, D.P.; Riedel, C.A.; Gonzalez, P.A.; et al. Interleukin-10 Production by T and B Cells Is a Key Factor to Promote Systemic Salmonella enterica Serovar Typhimurium Infection in Mice. Front. Immunol. 2017, 8, 889. [Google Scholar] [CrossRef] [Green Version]
  53. Arai, T.; Hiromatsu, K.; Nishimura, H.; Kimura, Y.; Kobayashi, N.; Ishida, H.; Nimura, Y.; Yoshikai, Y. Effects of in vivo administration of anti-IL-10 monoclonal antibody on the host defence mechanism against murine Salmonella infection. Immunology 1995, 85, 381–388. [Google Scholar]
  54. Wong, U.; Cross, R.K. Expert opinion on interleukin-12/23 and interleukin-23 antagonists as potential therapeutic options for the treatment of inflammatory bowel disease. Expert Opin. Investig. Drugs 2019, 28, 473–479. [Google Scholar] [CrossRef] [PubMed]
  55. Waldmann, T.A. The biology of interleukin-2 and interleukin-15: Implications for cancer therapy and vaccine design. Nat. Rev. Immunol. 2006, 6, 595–601. [Google Scholar] [CrossRef]
  56. Meisel, M.; Mayassi, T.; Fehlner-Peach, H.; Koval, J.C.; O’Brien, S.L.; Hinterleitner, R.; Lesko, K.; Kim, S.; Bouziat, R.; Chen, L.; et al. Interleukin-15 promotes intestinal dysbiosis with butyrate deficiency associated with increased susceptibility to colitis. ISME J. 2017, 11, 15–30. [Google Scholar] [CrossRef]
  57. Ferreira Dos Santos, T.; Alves Melo, T.; Almeida, M.E.; Passos Rezende, R.; Romano, C.C. Immunomodulatory Effects of Lactobacillus plantarum Lp62 on Intestinal Epithelial and Mononuclear Cells. Biomed. Res. Int. 2016, 2016, 8404156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Noto Llana, M.; Sarnacki, S.H.; Morales, A.L.; Aya Castaneda, M.D.R.; Giacomodonato, M.N.; Blanco, G.; Cerquetti, M.C. Activation of iNKT Cells Prevents Salmonella-Enterocolitis and Salmonella-Induced Reactive Arthritis by Downregulating IL-17-Producing gammadeltaT Cells. Front. Cell Infect. Microbiol. 2017, 7, 398. [Google Scholar] [CrossRef]
  59. Muhl, H.; Pfeilschifter, J. Interleukin-18 bioactivity: A novel target for immunopharmacological anti-inflammatory intervention. Eur. J. Pharm. 2004, 500, 63–71. [Google Scholar] [CrossRef]
  60. Raupach, B.; Peuschel, S.K.; Monack, D.M.; Zychlinsky, A. Caspase-1-mediated activation of interleukin-1beta (IL-1beta) and IL-18 contributes to innate immune defenses against Salmonella enterica serovar Typhimurium infection. Infect. Immun. 2006, 74, 4922–4926. [Google Scholar] [CrossRef] [Green Version]
  61. Tang, C.; Chen, S.; Qian, H.; Huang, W. Interleukin-23: As a drug target for autoimmune inflammatory diseases. Immunology 2012, 135, 112–124. [Google Scholar] [CrossRef] [PubMed]
  62. Buonocore, S.; Ahern, P.P.; Uhlig, H.H.; Ivanov, I.I.; Littman, D.R.; Maloy, K.J.; Powrie, F. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 2010, 464, 1371–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Neurath, M.F. IL-23 in inflammatory bowel diseases and colon cancer. Cytokine Growth Factor Rev. 2019, 45, 1–8. [Google Scholar] [CrossRef]
  64. Di Paolo, N.C.; Shayakhmetov, D.M. Interleukin 1alpha and the inflammatory process. Nat. Immunol. 2016, 17, 906–913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kurt-Jones, E.A.; Kiely, J.M.; Unanue, E.R. Conditions required for expression of membrane IL 1 on B cells. J. Immunol. 1985, 135, 1548–1550. [Google Scholar]
  66. Di Paolo, N.C.; Shafiani, S.; Day, T.; Papayannopoulou, T.; Russell, D.W.; Iwakura, Y.; Sherman, D.; Urdahl, K.; Shayakhmetov, D.M. Interdependence between Interleukin-1 and Tumor Necrosis Factor Regulates TNF-Dependent Control of Mycobacterium tuberculosis Infection. Immunity 2015, 43, 1125–1136. [Google Scholar] [CrossRef] [Green Version]
  67. Kurt-Jones, E.A.; Fiers, W.; Pober, J.S. Membrane interleukin 1 induction on human endothelial cells and dermal fibroblasts. J. Immunol. 1987, 139, 2317–2324. [Google Scholar] [PubMed]
  68. Cross, A.S.; Sadoff, J.C.; Kelly, N.; Bernton, E.; Gemski, P. Pretreatment with recombinant murine tumor necrosis factor alpha/cachectin and murine interleukin 1 alpha protects mice from lethal bacterial infection. J. Exp. Med. 1989, 169, 2021–2027. [Google Scholar] [CrossRef]
  69. Saitoh, T.; Fujita, N.; Jang, M.H.; Uematsu, S.; Yang, B.G.; Satoh, T.; Omori, H.; Noda, T.; Yamamoto, N.; Komatsu, M.; et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature 2008, 456, 264–268. [Google Scholar] [CrossRef] [PubMed]
  70. Plantinga, T.S.; Crisan, T.O.; Oosting, M.; van de Veerdonk, F.L.; de Jong, D.J.; Philpott, D.J.; van der Meer, J.W.; Girardin, S.E.; Joosten, L.A.; Netea, M.G. Crohn’s disease-associated ATG16L1 polymorphism modulates pro-inflammatory cytokine responses selectively upon activation of NOD2. Gut 2011, 60, 1229–1235. [Google Scholar] [CrossRef]
  71. Barton, B.E.; Jackson, J.V. Protective role of interleukin 6 in the lipopolysaccharide-galactosamine septic shock model. Infect. Immun. 1993, 61, 1496–1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Wang, L.; Srinivasan, S.; Theiss, A.L.; Merlin, D.; Sitaraman, S.V. Interleukin-6 induces keratin expression in intestinal epithelial cells: Potential role of keratin-8 in interleukin-6-induced barrier function alterations. J. Biol. Chem. 2007, 282, 8219–8227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ding, J.W.; Andersson, R.; Soltesz, V.; Willen, R.; Bengmark, S. Obstructive jaundice impairs reticuloendothelial function and promotes bacterial translocation in the rat. J. Surg. Res. 1994, 57, 238–245. [Google Scholar] [CrossRef]
  74. Stadnyk, A.W. Intestinal epithelial cells as a source of inflammatory cytokines and chemokines. Can. J. Gastroenterol. 2002, 16, 241–246. [Google Scholar] [CrossRef] [PubMed]
  75. Neutra, M.R.; Mantis, N.J.; Kraehenbuhl, J.P. Collaboration of epithelial cells with organized mucosal lymphoid tissues. Nat. Immunol. 2001, 2, 1004–1009. [Google Scholar] [CrossRef]
  76. Santos, R.L.; Tsolis, R.M.; Baumler, A.J.; Adams, L.G. Pathogenesis of Salmonella-induced enteritis. Braz. J. Med. Biol. Res. 2003, 36, 3–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Catron, D.M.; Sylvester, M.D.; Lange, Y.; Kadekoppala, M.; Jones, B.D.; Monack, D.M.; Falkow, S.; Haldar, K. The Salmonella-containing vacuole is a major site of intracellular cholesterol accumulation and recruits the GPI-anchored protein CD55. Cell Microbiol. 2002, 4, 315–328. [Google Scholar] [CrossRef]
  78. Garcia-del Portillo, F.; Nunez-Hernandez, C.; Eisman, B.; Ramos-Vivas, J. Growth control in the Salmonella-containing vacuole. Curr. Opin. Microbiol. 2008, 11, 46–52. [Google Scholar] [CrossRef] [PubMed]
  79. Steele-Mortimer, O. The Salmonella-containing vacuole: Moving with the times. Curr. Opin. Microbiol. 2008, 11, 38–45. [Google Scholar] [CrossRef] [Green Version]
  80. Huang, F.C. De Novo sphingolipid synthesis is essential for Salmonella-induced autophagy and human beta-defensin 2 expression in intestinal epithelial cells. Gut Pathog. 2016, 8, 5. [Google Scholar] [CrossRef] [Green Version]
  81. Kim, M.N.; Koh, S.J.; Kim, J.M.; Im, J.P.; Jung, H.C.; Kim, J.S. Clostridium difficile infection aggravates colitis in interleukin 10-deficient mice. World J. Gastroenterol. 2014, 20, 17084–17091. [Google Scholar] [CrossRef]
  82. Iyer, S.S.; Cheng, G. Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit. Rev. Immunol. 2012, 32, 23–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Castro-Eguiluz, D.; Pelayo, R.; Rosales-Garcia, V.; Rosales-Reyes, R.; Alpuche-Aranda, C.; Ortiz-Navarrete, V. B cell precursors are targets for Salmonella infection. Microb. Pathog. 2009, 47, 52–56. [Google Scholar] [CrossRef] [PubMed]
  84. Brigl, M.; Bry, L.; Kent, S.C.; Gumperz, J.E.; Brenner, M.B. Mechanism of CD1d-restricted natural killer T cell activation during microbial infection. Nat. Immunol. 2003, 4, 1230–1237. [Google Scholar] [CrossRef]
  85. O’Shea, J.J.; Paul, W.E. Regulation of T(H)1 differentiation--controlling the controllers. Nat. Immunol. 2002, 3, 506–508. [Google Scholar] [CrossRef]
  86. Ramirez-Alejo, N.; Santos-Argumedo, L. Innate defects of the IL-12/IFN-gamma axis in susceptibility to infections by mycobacteria and salmonella. J. Interferon. Cytokine Res. 2014, 34, 307–317. [Google Scholar] [CrossRef] [Green Version]
  87. Jabri, B.; Abadie, V. IL-15 functions as a danger signal to regulate tissue-resident T cells and tissue destruction. Nat. Rev. Immunol. 2015, 15, 771–783. [Google Scholar] [CrossRef] [PubMed]
  88. Fehniger, T.A.; Caligiuri, M.A. Interleukin 15: Biology and relevance to human disease. Blood 2001, 97, 14–32. [Google Scholar] [CrossRef]
  89. Iacomino, G.; Rotondi Aufiero, V.; Iannaccone, N.; Melina, R.; Giardullo, N.; De Chiara, G.; Venezia, A.; Taccone, F.S.; Iaquinto, G.; Mazzarella, G. IBD: Role of intestinal compartments in the mucosal immune response. Immunobiology 2019, 225, 151849. [Google Scholar] [CrossRef]
  90. Pham, T.A.; Clare, S.; Goulding, D.; Arasteh, J.M.; Stares, M.D.; Browne, H.P.; Keane, J.A.; Page, A.J.; Kumasaka, N.; Kane, L.; et al. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization resistance to an opportunistic pathogen. Cell Host Microbe 2014, 16, 504–516. [Google Scholar] [CrossRef] [Green Version]
  91. Behnsen, J.; Jellbauer, S.; Wong, C.P.; Edwards, R.A.; George, M.D.; Ouyang, W.; Raffatellu, M. The cytokine IL-22 promotes pathogen colonization by suppressing related commensal bacteria. Immunity 2014, 40, 262–273. [Google Scholar] [CrossRef] [Green Version]
  92. Dennehy, K.M.; Willment, J.A.; Williams, D.L.; Brown, G.D. Reciprocal regulation of IL-23 and IL-12 following co-activation of Dectin-1 and TLR signaling pathways. Eur. J. Immunol. 2009, 39, 1379–1386. [Google Scholar] [CrossRef]
  93. Stark, M.A.; Huo, Y.; Burcin, T.L.; Morris, M.A.; Olson, T.S.; Ley, K. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 2005, 22, 285–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. van de Wetering, D.; de Paus, R.A.; van Dissel, J.T.; van de Vosse, E. Salmonella induced IL-23 and IL-1beta allow for IL-12 production by monocytes and Mphi1 through induction of IFN-gamma in CD56 NK/NK-like T cells. PLoS ONE 2009, 4, e8396. [Google Scholar] [CrossRef] [Green Version]
  95. Pflanz, S.; Timans, J.C.; Cheung, J.; Rosales, R.; Kanzler, H.; Gilbert, J.; Hibbert, L.; Churakova, T.; Travis, M.; Vaisberg, E.; et al. IL-27, a heterodimeric cytokine composed of EBI3 and p28 protein, induces proliferation of naive CD4+ T cells. Immunity 2002, 16, 779–790. [Google Scholar] [CrossRef] [Green Version]
  96. Villarino, A.V.; Hunter, C.A. Biology of recently discovered cytokines: Discerning the pro- and anti-inflammatory properties of interleukin-27. Arthritis Res. 2004, 6, 225–233. [Google Scholar] [CrossRef] [Green Version]
  97. Huang, F.C.; Huang, S.C. Differential Effects of Statins on Inflammatory Interleukin-8 and Antimicrobial Peptide Human Beta-Defensin 2 Responses in Salmonella-Infected Intestinal Epithelial Cells. Int. J. Mol. Sci. 2018, 19, 1650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Huang, F.C.; Huang, S.C. The Combined Beneficial Effects of Postbiotic Butyrate on Active Vitamin D3-Orchestrated Innate Immunity to Salmonella Colitis. Biomedicines 2021, 9, 1296. [Google Scholar] [CrossRef] [PubMed]
  99. Huang, F.C.; Huang, S.C. The Cooperation of Bifidobacterium longum and Active Vitamin D3 on Innate Immunity in Salmonella Colitis Mice via Vitamin D Receptor. Microorganisms 2021, 9, 1804. [Google Scholar] [CrossRef] [PubMed]
Table 1. The interleukins orchestrate mucosal immune responses to defense against Salmonella infection in the intestine.
Table 1. The interleukins orchestrate mucosal immune responses to defense against Salmonella infection in the intestine.
InterleukinBiologic FunctionsExperimentsIntervention and EffectsClinical ApplicationsRef.
IL-1αFunction as a plasma membrane cytokine involved in the inflammation and protection from bacterial infections though its role remains poorly definedMiceIL-1α-enhanced resistance of mice to S. Typhimurium infectionSalmonella[15]
IL-6
  • Anti-inflammation
  • Mediator of epithelial barrier protection
  • Protection from sepsis and endotoxemia
IECsPJ-34 up-regulates IL-6 production in Salmonella-infected IECs [16,17,18,19]
Enterocytes
  • Probiotic (L. paracasei) potentiates IL-6 production in IL-1beta-treated Caco-2 cells
IL-8
  • Recruits neutrophils to defense against the invasion of Salmonella
IECs
  • Flagellin and MDP synergistically enhance IL-8
  • Salmonella colitis
  • IBD
[20,21,22,23,24,25]
2.
Plasma membrane cholesterol supports the survival of Salmonella in IECs through anti-IL-8 pathways
3.
Probiotics enhance IL-8 expression
IL-10
  • Anti-inflammation Th2 cytokine
  • Inhibit the development of Th1-type immune responses
  • Reduce NK cell responses
  • Prevent the differentiation of naïve T cells into effector cytotoxic T cells
  • Dampen the secretion of pro-inflammatory cytokines, such as IL-12
  • Induce Treg cell proliferation
  • Suppression of T helper 17 (TH17)-driven colitis
MiceIL-10-deficient mice develop colitisSalmonella sepsis[26,27]
IL-12
  • IL-12/IL-23 component acts on NK and T cells and NKT cells to induce IFN-γ-dependent or IFN-γ-independent immunity against intracellular Salmonella infection
  • A Key cytokine for immunity against invasive Salmonella in humans
HumansIL-12 enhances internalization and early intracellular killing of Salmonella enterica Serovar Typhimurium by human macrophagesRecurrent, extraintestinal and invasive Salmonella diseases[28,29,30]
IL-15
  • Stimulating macrophages, NK cells, T cells, and B cells to proliferate, secrete cytokines, and/or produce antibody
  • Protection against bacterial infection
MiceEndogenous IL-15 functions as early protection against infection with an avirulent strain of S. choleraesuis through activation of NK cells and IFN- γ productionSalmonella infection[31,32]
IL-17A cytokine involved in neutrophil recruitment to defend against extracellular bacteriaMiceProbiotic Lactobacillus plantarum ZS2058 significantly reduced the pathogenicity of Salmonella colitis by promoting the IL23/IL-22 axis in the mouse ileumSalmonella colitis[33,34,35,36]
IL-18
  • Promotes IFN-γ production by T cells and NK cells thereby shaping immunity towards a Th1-like phenotype
  • Activates the colon epithelial cells to produce antimicrobial peptides to maintain microbiome homeostasis
Salmonella pathogenicity islands (SPI)-1 effector secretion leads to NF-kB signaling and caspase-1-mediated IL-1β/IL-18 activation [37,38]
IL-22
  • Inflammatory responses
  • Maintenance of intestine mucosal barrier
  • Enhanced antimicrobial activity, and mucosal healing
  • Resistant to intestinal colonization of opportunistic pathogens
Human epithelial cellsIL-22 promotes intracellular fusion of SCVs with lysosomes leading to phagolysosomal killing of S. Typhimurium in human epithelial cells
  • Salmonella colitis
  • IBD
[39,40]
MiceIL-22 is able to heal intestinal inflammation and promote epithelial repair from acute injury
IL-23
  • A member of the IL-12 family of cytokines with pro-inflammatory properties
  • IL-23 induce IFN-γ and IL-22 production and are associated with host innate immunity against Salmonella
MiceMice deficient for IL-23 is associated with S. Typhimurium colitis
  • IBD
  • Salmonella colitis
[41,42]
IL-27
  • Has fundamental roles in innate and adaptive immune regulation
  • Has both anti- and pro-inflammatory functions
  • Enhance TLR4 or TLR5 expression in human monocytes and macrophages, to cooperate for optimal anti-bacterial responses
  • Monocytes
  • Macrophages
IL-27-enhanced TLR4 or TLR5 expression in human monocytes and macrophages, induced greater LPS/flagellin-mediated signaling, and significantly enhanced pro-inflammatory cytokines IL-12p40, TNF-α, and IL-6 production in S. typhimurium infected cellsSalmonella infection[43,44,45,46]
Abbreviations: IECs, intestine epithelial cells; IBD, inflammatory bowel disease; MDP, muramyl dipeptide; NK, natural killer; Treg: regulatory T cells; NKT, natural killer T; INF, interferon; TNF, tumor necrosis factor; LPS, lipopolysaccharide; Th1, type 1 helper T cells; SCVs, Salmonella-containing vesicles; TLR, Toll-like receptor.
Table 2. The interleukins orchestrate mucosal immune responses to enhance Salmonella colitis.
Table 2. The interleukins orchestrate mucosal immune responses to enhance Salmonella colitis.
InterleukinBiologic FunctionsExperimentsIntervention and EffectsClinical ApplicationsRef.
IL-1β
  • Amplifying intestinal inflammation by increasing intestinal epithelial tight junction permeability
  • Atg16L1 suppresses IL-1β expression in macrophage and IECs
IECsActive vitamin D decrease IL-1β response to Salmonella infection to prevent the host from detrimental inflammationIBD[2,47,48,49,50]
MiceActive vitamin D3 attenuates the severity of Salmonella colitis in mice by decreasing IL-1β response
RabbitBlockade of IL-1 receptors reduces the inflammatory responses in experiment colitis
IL-8Accumulation of neutrophils gives rise to colitis and sepsis IECs
  • Probiotics suppress IL-8 expression
  • Salmonella colitis
  • IBD
[1,25,51]
2.
Active vitamin D3 suppresses IL-8 expression
IL-10Promote systemic S. Typhimurium infection in miceMiceAnti-IL-10 monoclonal antibody block IL-10 to defense against systemic Salmonella infectionSalmonella sepsis[52,53]
IL-12A pro-inflammatory cytokine in response to microbial pathogensHumansUstekinumab, the monoclonal antibody targeting the shared p40 subunit of IL12/IL23, has been approved for treatment of IBDIBD[30,54]
IL-15
  • Pro-inflammatory by itself
  • Promote intestinal dysbiosis that increases susceptibility to colitis
Mice
  • Celiac disease
  • IBD
[32,55,56]
IL-17
  • Orchestrate mucosal inflammation in IBD and Salmonella colitis
  • iNKT cells play a protective role against Salmonella-enterocolitis by downregulating IL-17-producing γδT cells
  • Macrophages
  • iNKT cells
Lactobacillus plantarum Lp62 was able to suppress IL-17, IL1-β and TNF-α production in LPS-stimulated J774 macrophages
  • Salmonella colitis
  • IBD
[36,57,58]
IL-18
  • A member of the IL-1 family of cytokines with pro-inflammatory and tumor-suppressive properties
  • Initiates a pro-inflammatory cytokine cascade in peripheral blood mononuclear cells (PBMC)
  • Salmonella pathogenicity islands (SPI)-1 effector secretion leads to NF-kB signaling and caspase-1-mediated IL-1β/IL-18 activation
  • Animal models suggest suppression of IL-18 bioactivity as a novel therapeutic concept specifically for the treatment of chronic inflammatory diseases
[37,59,60]
IL-23
  • Accelerate proliferation of both murine and human memory T cells producing Th17 cytokines including iIL-17 and IL-22
  • Increased production of IL-23 in various mouse models of colitis and IBD patients
HumanmiceNeutralizing antibodies against IL-12/IL-23 p40 and IL-23 p19 have been successfully used in clinical trials for therapy of IBD
  • IBD
  • Salmonella colitis
[41,61,62,63]
IL-27IL-27 can directly induce expression of IL-1 and TNF-α by primary mast cells and production of IL-1, TNF-α, IL-12p35 and IL-18 by monocytes
  • Monocytes
  • Mast cells
Salmonella infection[44]
Abbreviations: IECs, intestine epithelial cells; IBD, inflammatory bowel disease; MDP, muramyl dipeptide; NK, natural killer; Treg: regulatory T cells; NKT, natural killer T; INF, interferon; TNF, tumor necrosis factor; LPS, lipopolysaccharide; Th1, type 1 helper T cells; SCVs, Salmonella-containing vesicles; TLR, Toll-like receptor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Huang, F.-C. The Interleukins Orchestrate Mucosal Immune Responses to Salmonella Infection in the Intestine. Cells 2021, 10, 3492. https://0-doi-org.brum.beds.ac.uk/10.3390/cells10123492

AMA Style

Huang F-C. The Interleukins Orchestrate Mucosal Immune Responses to Salmonella Infection in the Intestine. Cells. 2021; 10(12):3492. https://0-doi-org.brum.beds.ac.uk/10.3390/cells10123492

Chicago/Turabian Style

Huang, Fu-Chen. 2021. "The Interleukins Orchestrate Mucosal Immune Responses to Salmonella Infection in the Intestine" Cells 10, no. 12: 3492. https://0-doi-org.brum.beds.ac.uk/10.3390/cells10123492

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