Next Article in Journal
First Molecular Detection and Characterization of Hemotropic Mycoplasma Species in Cattle and Goats from Uganda
Next Article in Special Issue
Genetics of Arthrogryposis and Macroglossia in Piemontese Cattle Breed
Previous Article in Journal
Use of Contrast-Enhanced Ultrasonography for the Characterization of Tumor Thrombi in Seven Dogs
Previous Article in Special Issue
The Role of Innate Immune Response and Microbiome in Resilience of Dairy Cattle to Disease: The Mastitis Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Overview of Research Development on the Role of NF-κB Signaling in Mastitis

1
State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
2
Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
3
Institute of Agricultural Information of CAAS, Beijing 100081, China
*
Author to whom correspondence should be addressed.
Animals 2020, 10(9), 1625; https://doi.org/10.3390/ani10091625
Submission received: 17 August 2020 / Revised: 26 August 2020 / Accepted: 28 August 2020 / Published: 10 September 2020
(This article belongs to the Special Issue Genetics of Animal Health and Disease in Livestock)

Abstract

:

Simple Summary

NF-κB signaling has been widely studied for its role in inflammatory and immunity-related diseases. Mastitis is considered one of the inflammatory and immunity associated diseases which are a serious threat to the global dairy industry. Having such a critical role in immunity and inflammation, NF-κB signaling is currently under target for therapeutic purposes in mastitis control research. The virulent factor, lipopolysaccharides (LPS), of bacteria after attachment with relevant Toll-like receptors (TLRs) on mammary epithelial cells starts its pathogenesis by using NF-κB signaling to cause mastitis. Several studies have proved that the blocking of NF-κB signaling could be a useful strategy for mastitis control.

Abstract

Mastitis is the inflammation of the mammary gland. Escherichia coli and Staphylococcus aureus are the most common bacteria responsible for mastitis. When mammary epithelial cells are infected by microorganisms, this activates an inflammatory response. The bacterial infection is recognized by innate pattern recognition receptors (PRRs) in the mammary epithelial cells, with the help of Toll-like receptors (TLRs). Upon activation by lipopolysaccharides, a virulent agent of bacteria, the TLRs further trigger nuclear factor-κB (NF-κB) signaling to accelerate its pathogenesis. The NF-κB has an essential role in many biological processes, such as cell survival, immune response, inflammation and development. Therefore, the NF-κB signaling triggered by the TLRs then regulates the transcriptional expression of specific inflammatory mediators to initiate inflammation of the mammary epithelial cells. Thus, any aberrant regulation of NF-κB signaling may lead to many inflammatory diseases, including mastitis. Hence, the inhibiting of NF-κB signaling has potential therapeutic applications in mastitis control strategies. In this review, we highlighted the regulation and function of NF-κB signaling in mastitis. Furthermore, the role of NF-κB signaling for therapeutic purposes in mastitis control has been explored in the current review.

1. Introduction

Mastitis is the inflammation of the mammary gland, which is associated with pathological changes in udder tissue and decreases in the quantity and quality of milk [1,2]. Based on its duration and symptoms, mastitis might be acute or chronic [3,4]. Udder swelling, reduced milk yield, clots and increase somatic cell counts in milk are the most common clinical signs of mastitis [5]. All these factors are associated with pathogenic invasion, which is followed by the involvement of neutrophils under a specific stimulus. The inflammatory conditions may lead to chronic inflammation if not properly controlled and treated [6,7]. Different types of etiological invading bacterial pathogens are involved in bovine mastitis, of which Coliforms, Escherichia coli, Streptococci and Staphylococcus aureus are the most common bacteria [8,9,10,11]. Gram-negative bacteria, such as E. coli, can often cause clinical mastitis, and Gram-positive bacteria, such as S. aureus, are involved in subclinical mastitis infection [12,13,14].
Previous reports have documented that mammary epithelial cells work as the first line of defense of the mammary gland by generating multiple inflammatory cytokines against bacteria invading the epithelial cells [15,16]. Toll-like receptors (TLRs) are pattern recognition receptors (PRRs) expressed by many cell types, including mammary epithelial and immune cells [17]. In addition, it has been reported that innate immune systems recognize pathogens through TLRs [18,19,20].
The TLRs are distributed on the host cell surface that regulates the initial sensation of infection [21,22]. Every pathogen uses specific receptors on host cells—for example, S. aureus uses TLR2 and TLR6 [23], while E. coli utilizes TLR2 and TLR4—to transmit their signals inside the cell [21]. This specificity to TLRs depends on the virulent factor of pathogens. The cell wall of S. aureus is composed of lipoteichoic acid and peptidoglycan [24], while Gram-negative bacteria, such as E. coli, have lipopolysaccharides (LPS) in their cell wall [25]. The binding of pathogenic virulent factors to TLRs leads to the activation of several signaling components, including nuclear factor kappa-light-chain-enhancer of activated B (NF-κB) [26], which is considered one of the key players associated with inflammatory action. Besides, NF-κB signaling has been widely studied for its role in regulation of immunity and inflammation. Keeping in view the versatile functions of NF-κB signaling, the current review has specifically concentrated on summarizing possible research development on the role of NF-κB signaling activation and regulation of immunity and inflammation in bovine mastitis.

2. Materials and Methods

All studies which have discussed the role of NF-κB signaling in mammary gland infection, mainly bovine mastitis, were screened through authentic sources, such as PubMed, ScienceDirect, Web of Science, SpringerLink, Scopus and Google Scholar. The major keywords used for the search of literature were milk production, mastitis, NF-κB signaling, TLRs, MYD88, PPRs, cytokines, E. coli- and S. aureus-mastitis. The related data published in the English language in well-reputed peer-reviewed journals have been included for discussion in the current review. Furthermore, we excluded all content available in the form of conference abstracts, books, book chapters and unpublished findings.

3. General Regulatory Pattern of NF-κB Signaling

NF-κB is a common term used for inducible dimeric transcription factors. It is composed of a Rel family DNA binding protein which distinguishes common sequence motifs. Mammals express 5 Rel (NF-κB) proteins which are composed of two classes including Rel A (p65), c-Rel and Rel-B proteins which do not need proteolytic processing as the class is composed of NF-κB1 and NF-κB2 genes, encoded for p105 and p100, respectively, which do not require proteolytic processing to synthesize mature p50 and p52 NF-κB proteins [27]. The NF-κB protein was first found in murine B-lymphocytes, but currently, it has been identified in many cell types, including mammary epithelial cells [28]. Different external stimuli, such as tumor necrosis factor Alpha (TNF-α) [29], interleukin 1-beta (IL-1β) [30], LPS and reactive oxygen species (ROS) [31] after attachment with TLRs, activate NF-κB [32]. NF-κB signaling has an essential role in the regulation of immunity and inflammation [33], cell apoptosis, cell survival and proliferation (Figure 1) [34,35].
In addition, NF-κB signaling plays a vital role in the regulation of inflammatory cytokines, adhesion molecules, chemokines and growth factors involved in mammary gland inflammation [36]. Adhesion molecules are important proteins of tight junctions [37], which are closely related to the link between cell membranes and are required for normal lactation in mammals [38]. Song et al. has shown that LPS disrupt the permeability of the blood–milk barrier by activating the NF-κB signaling pathway. The pro-inflammatory cytokines regulated by the NF-κB signaling pathway promote the process of inflammation and interrupt the integrity of tight junction structures in the mammary epithelial cells [39]. The disruption in the blood–milk barrier has been reported during mastitis, which might be due to damage of the tight junctions responsible for normal lactation [40]. The disruption of tight junctions also may lead to loss of milk which is one of the common signs of mastitis in dairy cattle. Having such a critical role in inflammation and immunity, the NF-κB pathway has been widely targeted in mastitis research [41,42,43,44,45,46].

4. Role of NF-κB Signaling in Normal Physiology of Mammary Gland Development

A regulated pattern of activation of NF-κB during the various stages of the development of mammary glands has been demonstrated [46]. NF-κB activation rises during pregnancy and decreases during lactation, followed by elevation during the mammary gland involution, again [47,48]. This change in pattern suggests that NF-κB plays a significant role during pregnancy and involution. Mammary gland involution is associated with apoptosis of the secretory alveolar epithelium [49], and NF-κB has been explored to mediate the anti-apoptotic proteins [50]. These findings revealed the role of NF-κB in promoting the survival of epithelial cells [51]. It has been demonstrated that NF-κB activates the two essential lactogenic hormones, namely prolactin and oxytocin [52,53]. In addition to playing a role in the developmental process of normal mammary glands, NF-κB activation was found to be associated with mammary gland infections.

5. Role of NF-κB Signaling in Mastitis

The murine model and bovine reports have shown the link of NF-κB regulation with mastitis [43]. Most of the studies investigated the role of NF-κB in mastitis as a regulator of inflammatory cytokines [54,55]. Considerable losses of milk have been observed during mastitis and mammary gland involution which showed the link of both with the up-regulation of NF-κB during a time of milk loss and mammary gland remodeling.

5.1. Mechanism of NF-κB Signaling Activation by Bacteria during Mastitis

LPS, a bacterial virulence factor, interacts with TLRs which are residing on surface mammary epithelial cells [56]. Upon activation, the TLRs further engage myeloid differentiation factor 88 (MyD88) [57] and c-Jun N-terminal kinase (JNK) [58], which triggers NF-κB [32] and mitogen-activated protein kinase (MAPK) signaling. The translocation of NF-κB and MAPK signaling further regulates the production of target inflammatory genes [59,60,61,62]. The mechanism of NF-κB signaling activated by S. aureus and E. coli during mastitis is shown in Figure 2.

5.2. Mechanism of NF-κB Signaling Activation by Inflammatory Cytokines

Besides S. aureus and E. coli, various inflammatory cytokines activate NF-κB signaling regulation in mammary epithelial cells. The NF-κB and MAPK pathways activate pro-inflammatory cytokines interleukin 6 (IL-6), IL-1β and TNF-α [63]. Nuclear factor-κB is a nuclear transcription factor that exists in an inactive form in the cytoplasm and is bound to its inhibitor IκB [64,65]. Once activated, the NF-κB unit p65 separates from IκB and translocates from the cytoplasm to the nucleus, where it regulates inflammatory gene expression [66]. The pathogenic message usually causes the liberation of NF-κB from IκB [65]. The regulation of the inflammation through NF-κB by pro-inflammatory cytokines is shown in Figure 3. The promoter of the inflammatory genes contains binding sites for NF-κB, and thus mostly depends on NF-κB for its regulation [67]. It has been reported that active NF-κB complexes cannot be detected in healthy cow milk cells, while the NF-κB elevated level was noticed in the milk cells of cows with acute mastitis. In addition, the activity of NF-κB in milk cells varies from low to high in chronic mastitis [67]. Stimulation of LPS causes mammary epithelial cells to produce cytokines TNF-α, IL-6 and IL-1β [68]. The increased levels of TNF-α, IL-6 and IL-1β have been observed in LPS-infused mammary glands [69]. Furthermore, Blum et al. reported the high level of cytokines (TNF-α, IL-6 and IL-17), somatic cell count (SCC), and up-regulation of TLR4 expression in leukocytes of the milk of an E. coli-induced mastitic cow [70]. In the mammary glands, inflammation is associated with an increased level of neutrophil chemo-attractants and the cytokines IL-1ß, IL-6, IL8 and TNF-α [71,72]. The expression level reported for IL8 and TNF-α in E. coli induced-mastitis in bovine mammary epithelial cells (BMECs) was much higher than for S. aureus, which is due to the weak Lipoteichoic acid (LTA) induction of TNF-α, or inactivation of NF-κB signaling [73]. Boulanger et al. observed that NF-κB was highly associated with the level of the expression of interleukin-8 and granulocyte/macrophage colony-stimulating factors, two NF-κB-dependent cytokines critically linked to the regulation and continuation of neutrophilic inflammation. Altogether, these findings suggested the crucial role of NF-κB in the pathogenesis of mastitis.

5.3. Bovine Myeloid Differentiation Primary Response 88 (MYD88), NFKBIA and TRAPPC9 Role as a Regulator of Lipopolysaccharide (LPS)-Induced NF-κB Signaling Pathways

MYD88 is the main adopter molecule for TLR2, 4, 5, 7, 8 and 9 signaling [74]. The TLRs, when activated by mastitis-induced bacteria, pass the signal to MYD88, which is considered the critical immune regulator adapter molecule against various pathogens [75,76]. MYD88 acts as the key regulator of NF-κB by causing the degradation of IKB. Wang and his co-authors compared the expression level of MYD88 in healthy and mastitic cows. It was observed that MYD88 expression, which works as a bridge between TLRs and NF-κB, was elevated in mastitic cows compared to healthy ones [69,77,78]. It was noticed in a study that inhibition of MYD88, along with its inhibitor, Pepinh-MYD, significantly reduced the level of NF-κB [63].
Another essential protein is the nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (NFKBIA), which encodes IκB and is responsible for the negative activation of NF-κB transcription factors. It has been shown in a report that LPS cause the degradation of IκBα; they facilitate the translocation of NF-κB in the nucleus, which in response accelerates the re-synthesis of IκBα [79]. Fang et al. noticed the up-regulation of NFKBIA in S. aureus-induced mastitis [80]. The trafficking protein particle complex 9 (TRAPPC9), also called NIK-and-IKK2-binding protein (NIBP), is a key regulator of NF-κB signaling [72,81,82]. An in-vitro study revealed NIBP low expression results in the down-regulation of TNF-α-induced NF-κB [83]. Wang et al. noticed through a genome-wide association study (GWAS) that the mutation in TRAPPC9 is associated with milk SCS [84]. The high expression level of the TRAPPC9 gene was reported in mammary epithelial cells infected with S. aureus. Furthermore, it was revealed that the TRAPPC9 gene might be considered a potential marker against mastitis [85]. The above-published studies showed that MYD88, NFKBIA and TRAPPC9 might work as a bridge between cell surface receptors and NF-κB. Thus, any change in these genes may disturb NF-κB signaling, which facilitates the pathogenesis of mastitis.

5.4. NF-κB Regulates the Immunity and Inflammatory Linked Genes during Mastitis

When NF-κB signaling is activated by external stimuli, such as bacteria or cytokines, it starts to regulate the production of inflammatory chemokines (IL-8, CXCL1, CXCL10, etc.), cytokines (IL-6, TNF-α, IFN-gamma and IL-1β), adhesion molecules (ICAM-1 and MMPs), growth factors (CSF) and apoptotic associated genes [76,86,87]. For the site of infection, many proteins are required; these proteins are: adhesion factors, such as ICAM-1 and VCAM-1, which facilitate neutrophil margination, diapedesis and transepithelial migration; chemokines, such as interleukin (IL)-8, which are responsible for chemotactic of neutrophils; IL-1β and TNF-α, which regulate neutrophils [67]. When bacteria enter the teat, the mammary epithelial cells secrete chemokines (CXCL8 and CXCL20) and cytokines (TNF-α and IL-1β). Production of cytokines and chemokines in the milk of the mastitic mammary gland is considered the key player of inflammation [72,88]. The TNF-α and fatty acid synthetase (FAS) mRNA expression was significantly up-regulated in LPS-challenged quarters [89]. A study reported the up-regulation of CXCL8 and TNF-α in E. coli induced mastitis in mammary epithelial cells [88]. In addition, the high expression of CXCL10, CCL2, CCL5 and CCL20 was noticed in bovine mammary epithelial cells in E. coli induced mastitis, which is essential for the recruitment of leucocytes [90]. The expression levels of IL-6, complement factor 3 (C3), NFKBIA and MMP9 were also elevated during mammary gland infection [90]. It has been reported that monocytes, natural killer cells and activated lymphocytes are majorly regulated by the chemokines CXCL10 and CCL5 [91]. Apart from the above functions, CXCL10 directs the recruitment and activation of neutrophils towards LPS-infection spots in mice and humans [83,92,93]. In addition, CXCL10 was reported as a highly expressed gene in response to E. coli infection in mammary glands [94]. The levels of CXCL10 and ICAM1 were noticed to be significantly elevated in the S. aureus-mastitic mammary glands of cows [95]. Similarly, the high expression of CCl5 has also reported in E. coli-induced mastitis in BMECs [96]. The expression levels of CXCL8, IL6 and CSF3 were higher in S. aureus challenged BMECs [80]. Additionally, many other immunity and inflammatory associated genes, such as SAA3, CCL5, C3 and CSF3, were also documented in mastitis-infected mammary glands [69]. Furthermore, the high expressions of CXCL10, IL6, CXCL8, IFN-gamma and IL-1β induced by LPS in BMECs are able to regulate inflammation [97]. It has been demonstrated in previous reports that inflammatory cytokines and chemokines create protection against foreign invading pathogens in bovine mammary glands, by increasing the movement of leucocytes from the blood into the mammary tissue [98]. Similarly, a study reported the protecting role of IL-1β by recruiting neutrophils into the mammary gland [99].

5.5. Research Progress on Target of NF-κB Signaling as a Therapeutic in Mastitis Control

It is well known that TLRs, upon recognition of external stimuli, activate NF-κB regulation to produce inflammatory linked genes to eliminate the cause of infection in mammary epithelial cells. TLR4, a pro-inflammatory cytokine, and LPS, a component of the cell wall of bacteria, are common inducers of NF-κB signaling. The LPS-induced inflammation in mammary epithelial cells causes the up-regulation of TLR4 [100,101]. Recently, NF-κB signaling is being widely targeted as a therapeutic choice against mastitis resistance. A study proved, experimentally, that selenium restricts S. aureus-induced mastitis through inhibition of the MAPK and NF-κB pathways and TLR2 [102]. Cytokines, an important group of inflammatory mediators, play a major role in the process of inflammation [103]. Stimulation by LPS causes mammary epithelial cells to produce the cytokines TNF-α, IL-6 and IL-1β [60]. Increased levels of TNF-α, IL-6 and IL-1β have been observed in LPS-infused mammary glands. Similarly, Akhter et al. [104] noticed the up-regulation of pro-inflammatory cytokines in S. aureus-induced mammary epithelial cells. Further, they proved that the expression levels of genes associated with TLR2/TLR4-mediated NF-κB/MAPKs pathways were higher in S. aureus-infected mammary epithelial cells. The excessive expression of pro-inflammatory IL1β may lead to pathological conditions [105]. Dai et al. noticed that methionine and arginine attenuated the proinflammatory action by preventing the regulation of NF-κB. Furthermore, methionine and arginine down-regulated the levels of TLR4 and IL1β in LPS-induced mastitis, which caused the excessive regulation of inflammatory changes, and thus damaged the cells [106]. Taken together, it has been concluded here that methionine and arginine, being blockers of NF-κB, can be considered as prophylactic agents of mastitis.
Exogenous hydrogen sulfide has the ability to suppress inflammatory cytokine production, ROS [107,108,109], and promotes anti-inflammatory proteins [110]. The high level of ROS is associated with the imbalance between cellular redox states and oxidative stress, which has a significant role in the promotion of inflammation [111]. It was noticed that LPS alone diminished cell viability and caused inflammatory changes in mammary epithelial cells. However, it was found that the hydrogen sulfide (H2S) combined with LPS restored the viability of the cells [112]. Sun et al. revealed that H2S, after entry into the cells, first blocked the TLR4 and ROS, and thereby no signal was given for NF-κB to produce a high level of inflammatory proteins in mammary epithelial cells [113]. In addition, the mRNA expression of TNF-α, IL-1β, IL-8 and IL-6 was also very low in H2S-treated mammary epithelial cells.
Morin has anti-inflammatory properties [114] and inhibits the release of the inflammatory cytokines IL-6 and IL-8 and tumor necrosis factor (TNF) from mast cells [115]. It was experimentally proved that morin is associated with inhibition of TNF-α, IL-6 and IL-1β in LPS-induced bovine mammary epithelial cells (bMECs). To suppress the level of cytokines, morin down-regulates the levels of MAPK and NF-κB pathways in LPS-induced mammary epithelial cells [78]. NLRP3 inflammasome is the key regulator of IL-1β, while a recent study noticed that morin significantly down-regulated the level of IL-1β [116] in LPS-induced bovine mammary epithelial cells by suppression of NF-κB and nucleotide-binding domain, leucine-rich repeat-containing family, pyrin domain containing 3 (NLRP3) inflammasome [117]. Furthermore, it has been noticed that morin also maintained the integrity of the tight junction from the action of the inflammatory cytokines regulated by NF-κB [63]. Likewise, polydatin has anti-inflammatory efficiency and can be used to control S. aureus-induced mastitis. The most in-depth mechanism showed that polydatin decreased the expression of TLR2 and MyD88, which further suppressed the level of NF-κB in mammary epithelial cells of S. aureus- induced mastitis [60].
Tea tree oil (TTO) is an essential oil which has antibacterial and anti-inflammatory properties and promotes the movement of polymorphonuclear leukocytes towards the infection. TTO also inhibits NFKBIA and TNF-α [118]. In addition, TTO act as an inhibitor of the NF-κB pathway, which is essential for the regulation of immunity and inflammatory responses in mammary glands. Nucleotide-binding oligomerization domain (NOD) is a type of PRR that plays an important role in the regulation of innate immunity [119]. Recently, it was documented that by blocking NOD1/NF-κB signaling, LPS stimulation reduced neutrophil migration and phagocytic killing ability. Further, it was proved that the activation of NOD1/NF-κB in vitro restricted the action of LPS by promoting the functional capacity of neutrophil [60]. Chlorogenic acid has anti-inflammatory and antibacterial effects [120,121]. A study reported that chlorogenic acid inhibits cytokine production in LPS-stimulated RAW264.7 cells by suppressing the phosphorylation of NF-κB [122]. Similarly, chlorogenic acid was noticed to reduce the level of cytokines followed by inhibition of TLR4 and phosphorylation of NF-κB in LPS-induced mastitic mammary epithelial cells [123]. Thymol was found to be very effective in mastitis treatment. The mechanism for the association of thymol was tested in BMECs. The western blot result showed that thymol treatment significantly inhibited the production of IL-6 and TNF-α, followed by suppression of the NF-κB pathway [124]. In Table 1, we summarized those studies which targeted the NF-κB signaling to control mastitis.
From the above discussion, it has been cleared that NF-κB signaling plays a role of backbone in the pathogenesis of mastitis by promoting cytokine production. Thus, by targeting NF-κB, mastitis can be effectively controlled [124].

6. Conclusions

Overall, the current review, based on published studies, revealed that activation of NF-κB resulted in decreased of milk and apoptotic signaling, which could be minimized through selective modulation of NF-κB signals. Furthermore, the review suggested that NF-κB is a vital regulator of milk loss during mammary gland involution and infection, and recognized the NF-κB signaling pathway as a possible target for preventing mastitis-induced milk loss in dairy cattle. In addition, based on published literature, we concluded that TLR4, IL-1β, IL-6, TNF-α and MYD88 are key players in NF-κB signaling and also have an essential role in mastitis development. From the literature studies, it was revealed that S. aureus and E. coli, after attachment with TLRs, used NF-κB pathway for pathogenesis. Thus, the utilization of NF-κB as a therapeutic target in mastitis control showed successful outcomes. In addition, TLR4, IL-1β, IL-6, TNF-α, MYD88 and NF-κB might be a useful addition as markers in mastitis control strategies.

Author Contributions

Conceptualization, M.Z.K. and Z.C.; writing—original draft preparation, M.Z.K. and Z.C.; editing and technical review, A.K., J.M., Y.M., T.C., D.S., J.X. and Z.C.; visualization, Z.C.; supervision, Z.C. All authors have read and agreed to the published version of the manuscript.

Funding

The review was supported by the National Key Research and Development Program of China (2018YFD0501600). The funder had no role in the study design, data collection, analysis, decision to publish, and preparation of the manuscript.

Acknowledgments

We acknowledge the financial support of the National Key Research and Development Program of China (2018YFD0501600). We also acknowledge the China Agricultural University, Beijing, China, for providing us with an environment of learning. Without this platform, the completion of this work would not have been an easy task.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gomes, F.; Henriques, M. Control of Bovine Mastitis: Old and Recent Therapeutic Approaches. Curr. Microbiol. 2016, 72, 377–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Zhao, X.; Lacasse, P. Mammary tissue damage during bovine mastitis: Causes and control. J. Anim. Sci. 2008, 86, 57–65. [Google Scholar] [CrossRef] [PubMed]
  3. Abebe, R.; Hatiya, H.; Abera, M.; Megersa, B.; Asmare, K. Bovine mastitis: Prevalence, risk factors and isolation of Staphylococcus aureus in dairy herds at Hawassa milk shed, South Ethiopia. BMC Vet. Res. 2016, 12, 270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Heikkilä, A.M.; Liski, E.; Pyörälä, S.; Taponen, S. Pathogen-specific production losses in bovine mastitis. J. Dairy Sci. 2018, 101, 9493–9504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Skarbye, A.P.; Krogh, M.A.; Sørensen, J.T. The effect of individual quarter dry-off in management of subclinical mastitis on udder condition and milk production in organic dairy herds: A randomized field trial. J. Dairy Sci. 2018, 101, 11186–11198. [Google Scholar] [CrossRef] [PubMed]
  6. Havixbeck, J.J.; Rieger, A.M.; Wong, M.E.; Hodgkinson, J.W.; Barreda, D.R. Neutrophil contributions to the induction and regulation of the acute inflammatory response in teleost fish. J. Leukoc. Biol. 2016, 99, 241–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Pisanu, S.; Cubeddu, T.; Pagnozzi, D.; Rocca, S.; Cacciotto, C.; Alberti, A.; Marogna, G.; Uzzau, S.; Addis, M.F. Neutrophil extracellular traps in sheep mastitis. Vet. Res. 2015, 46, 59. [Google Scholar] [CrossRef] [Green Version]
  8. Lavon, Y.; Leitner, G.; Kressel, Y.; Ezra, E.; Wolfenson, D. Comparing effects of bovine Streptococcus and Escherichia coli mastitis on impaired reproductive performance. J. Dairy Sci. 2019, 102, 10587–10598. [Google Scholar] [CrossRef]
  9. Poutrel, B.; Bareille, S.; Lequeux, G.; Leboeuf, F. Prevalence of Mastitis Pathogens in France: Antimicrobial Susceptibility of Staphylococcus aureus, Streptococcus uberis and Escherichia coli. J. Vet. Sci. Technol. 2018, 9, 2. [Google Scholar] [CrossRef]
  10. Biswas, S.; Chakravarti, S.; Barui, A. Emergence of coagulase positive methicillin resistant Staphylococcus Aureus isolated from buffalo mastitis milk samples. Explor. Anim. Med. Res. 2018, 8, 190–196. [Google Scholar]
  11. Li, T.; Lu, H.; Wang, X.; Gao, Q.; Dai, Y.; Shang, J.; Li, M. Molecular characteristics of Staphylococcus aureus causing bovine mastitis between 2014 and 2015. Front. Cell. Infect. Microbiol. 2017, 7, 127. [Google Scholar] [CrossRef] [PubMed]
  12. Jensen, K.; Günther, J.; Talbot, R.; Petzl, W.; Zerbe, H.; Schuberth, H.J.; Seyfert, H.M.; Glass, E.J. Escherichia coli- and Staphylococcus aureus-induced mastitis differentially modulate transcriptional responses in neighbouring uninfected bovine mammary gland quarters. BMC Genom. 2013, 14, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Jing, X.Q.; Zhao, Y.Q.; Shang, C.C.; Yao, Y.L.; Tian, T.T.; Li, J.; Chen, D.K. Dynamics of cytokines associated with IL-17 producing cells in serum and milk in mastitis of experimental challenging with Staphylococcus aureus and Escherichia coli in dairy goats. J. Anim. Vet. Adv. 2012, 11, 475–479. [Google Scholar]
  14. Pumipuntu, N.; Kulpeanprasit, S.; Santajit, S.; Tunyong, W.; Kong-ngoen, T.; Hinthong, W.; Indrawattana, N. Screening method for Staphylococcus aureus identification in subclinical bovine mastitis from dairy farms. Vet. World 2017, 10, 721–726. [Google Scholar] [CrossRef] [Green Version]
  15. Wellnitz, O.; Bruckmaier, R.M. The innate immune response of the bovine mammary gland to bacterial infection. Vet. J. 2012, 192, 148–152. [Google Scholar] [CrossRef]
  16. Brenaut, P.; Lefèvre, L.; Rau, A.; Laloë, D.; Pisoni, G.; Moroni, P.; Bevilacqua, C.; Martin, P. Contribution of mammary epithelial cells to the immune response during early stages of a bacterial infection to Staphylococcus aureus. Vet. Res. 2014, 45, 16. [Google Scholar] [CrossRef] [Green Version]
  17. Kumar, H.; Kawai, T.; Akira, S. Pathogen recognition by the innate immune system. Int. Rev. Immunol. 2011, 30, 16–34. [Google Scholar] [CrossRef]
  18. Albiger, B.; Dahlberg, S.; Henriques-Normark, B.; Normark, S. Role of the innate immune system in host defence against bacterial infections: Focus on the Toll-like receptors. J. Intern. Med. 2007, 261, 511–528. [Google Scholar] [CrossRef]
  19. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [Green Version]
  20. Kawai, T.; Akira, S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int. Immunol. 2009, 21, 317–337. [Google Scholar] [CrossRef] [Green Version]
  21. Bhattarai, D.; Worku, T.; Dad, R.; Rehman, Z.U.; Gong, X.; Zhang, S. Mechanism of pattern recognition receptors (PRRs) and host pathogen interplay in bovine mastitis. Microb. Pathog. 2018, 120, 64–70. [Google Scholar] [CrossRef] [PubMed]
  22. Takeuchi, O.; Akira, S. Pattern Recognition Receptors and Inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ren, C.; Zhang, Q.; Haan, B.J.; De Zhang, H.; Faas, M.M. Identification of TLR2 / TLR6 signalling lactic acid bacteria for supporting immune regulation. Sci. Rep. 2016, 6, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Guo, W.; Liu, B.; Hu, G.; Kan, X.; Li, Y.; Gong, Q.; Xu, D.; Ma, H.; Cao, Y.; Huang, B.; et al. Vanillin protects the blood–milk barrier and inhibits the inflammatory response in LPS-induced mastitis in mice. Toxicol. Appl. Pharmacol. 2019, 365, 9–18. [Google Scholar] [CrossRef]
  25. Doyle, S.L.; O’Neill, L.A.J. Toll-like receptors: From the discovery of NF-κB to new insights into transcriptional regulations in innate immunity. Biochem. Pharmacol. 2006, 72, 1102–1113. [Google Scholar] [CrossRef]
  26. Liu, C.Y.; Gao, X.X.; Chen, L.; You, Q.X. Rapamycin suppresses Abeta25–35- or LPSinduced neuronal inflammation via modulation of NF-kappaB signaling. Neuroscience 2017, 355, 188–199. [Google Scholar] [CrossRef]
  27. Ghosh, S.; May, M.J.; Kopp, E.B. NF-kB and Rel proteins: Evolutionary conserved mediators of the immune response. Annu. Rev. Immunol. 1998, 16, 225–260. [Google Scholar] [CrossRef]
  28. Tripathi, P.; Aggarwal, A. NF-kB transcription factor: A key player in the generation of immune response. Curr. Sci. 2006, 90, 25. [Google Scholar]
  29. Fitzgerald, D.C.; Meade, K.G.; McEvoy, A.N.; Lillis, L.; Murphy, E.P.; MacHugh, D.E.; Baird, A.W. Tumour necrosis factor-α (TNF-α) increases nuclear factor κB (NFκB) activity in and interleukin-8 (IL-8) release from bovine mammary epithelial cells. Vet. Immunol. Immunopathol. 2007, 116, 59–68. [Google Scholar] [CrossRef]
  30. Renard, P.; Zachary, M.D.; Bougelet, C.; Mirault, M.E.; Haegeman, G.; Remacle, J.; Raes, M. Effects of antioxidant enzyme modulations on interleukin-1-induced nuclear factor kappa B activation. Biochem. Pharmacol. 1997, 53, 149–160. [Google Scholar] [CrossRef]
  31. Chandel, N.S.; Trzyna, W.C.; McClintock, D.S.; Schumacker, P.T. Role of Oxidants in NF-κB Activation and TNF-α Gene Transcription Induced by Hypoxia and Endotoxin. J. Immunol. 2000, 165, 1013–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Oliveira-Nascimento, L.; Massari, P.; Wetzler, L.M. The Role of TLR2 in Infection and Immunity. Front. Immunol. 2012, 18, 3–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ghosh, S.; Hayden, M.S. New regulators of NF-κB in inflammation. Nat. Rev. Immunol. 2008, 8, 837–848. [Google Scholar] [CrossRef] [PubMed]
  34. Oeckinghaus, A.; Ghosh, S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb. Perspect. Biol. 2009, 1, a000034. [Google Scholar] [CrossRef]
  35. Fu, Z.H.; Liu, S.Q.; Qin, M.B.; Huang, J.A.; Xu, C.Y.; Wu, W.H.; Zhu, L.Y.; Qin, N.; Lai, M.Y. NIK- and IKKβ-binding protein contributes to gastric cancer chemoresistance by promoting epithelial-mesenchymal transition through the NF-κB signaling pathway. Oncol. Rep. 2018, 39, 2721–2730. [Google Scholar] [CrossRef] [Green Version]
  36. Chen, X.; Zheng, X.; Zhang, M.; Yin, H.; Jiang, K.; Wu, H.; Dai, A.; Yang, S. Nuciferine alleviates LPS-induced mastitis in mice via suppressing the TLR4-NF-κB signaling pathway. Inflamm. Res. 2018, 67, 903–911. [Google Scholar] [CrossRef]
  37. Stelwagen, K.; Singh, K. The role of tight junctions in mammary gland function. J. Mammary Gland Biol. Neoplasia 2014, 19, 131–138. [Google Scholar] [CrossRef]
  38. Quesnell, R.R.; Erickson, J.; Schultz, D.B. Apical electrolyte concentration modulates barrier function and tight junction protein localization in bovine mammary epithelium. Am. J. Physiol. Cell. Physiol. 2007, 292, C305–C318. [Google Scholar] [CrossRef] [Green Version]
  39. Song, X.; Zhang, W.; Wang, T.; Jiang, H.; Zhang, Z.; Fu, Y.; Yang, Z.Y.; Cao, Y.; Zhang, N. Geniposide plays an anti-inflammatory role via regulating TLR4 and downstream signaling pathways in lipopolysaccharide-induced mastitis in mice. Inflammation 2014, 37, 1588–1598. [Google Scholar] [CrossRef]
  40. Guo, W.; Liu, B.; Yin, Y.; Kan, X.; Gong, Q.; Li, Y.; Cao, Y.; Wang, J.; Xu, D.; Ma, H.; et al. Licochalcone A protects the blood-milk barrier integrity and relieves the inflammatory response in LPS-induced mastitis. Front. Immunol. 2019, 10, 287. [Google Scholar] [CrossRef] [Green Version]
  41. Glynn, D.J.; Hutchinson, M.R.; Ingman, W.V. Toll-Like Receptor 4 Regulates Lipopolysaccharide-Induced Inflammation and Lactation Insufficiency in a Mouse Model of Mastitis1. Biol. Reprod. 2014, 90, 1–11. [Google Scholar] [CrossRef] [PubMed]
  42. Hu, G.; Hong, D.; Zhang, T.; Duan, H.; Wei, P.; Guo, X.; Mu, X. Cynatratoside-C from Cynanchum atratum displays anti-inflammatory effect via suppressing TLR4 mediated NF-κB and MAPK signaling pathways in LPS-induced mastitis in mice. Chem. Biol. Interact. 2018, 279, 187–195. [Google Scholar] [CrossRef]
  43. Notebaert, S.; Carlsen, H.; Janssen, D.; Vandenabeele, P.; Blomhoff, R.; Meyer, E. In vivo imaging of NF-κB activity during Escherichia coli-induced mammary gland infection. Cell. Microbiol. 2008, 10, 1249–1258. [Google Scholar] [CrossRef] [PubMed]
  44. Wu, J.; Li, L.; Sun, Y.; Huang, S.; Tang, J.; Yu, P.; Wang, G. Altered Molecular Expression of the TLR4 / NF- κ B Signaling Pathway in Mammary Tissue of Chinese Holstein Cattle with Mastitis. PLoS ONE 2015, 10, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Xiao, H.B.; Wang, C.R.; Liu, Z.K.; Wang, J.Y. LPS induces pro-inflammatory response in mastitis mice and mammary epithelial cells: Possible involvement of NF-κB signaling and OPN. Pathol. Biol. 2015, 63, 11–16. [Google Scholar] [CrossRef]
  46. Connelly, L.; Barham, W.; Pigg, R.; Saint-Jean, L.; Sherrill, T.; Cheng, D.S.; Chodosh, L.A.; Blackwell, T.S.; Yull, F.E. Activation of nuclear factor kappa B in mammary epithelium promotes milk loss during mammary development and infection. J. Cell. Physiol. 2010, 222, 73–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Brantley, D.M.; Chen, C.L.; Muraoka, R.S.; Bushdid, P.B.; Bradberry, J.L.; Kittrell, F.; Medina, D.; Matrisian, L.M.; Kerr, L.D.; Yull, F.E. Nuclear factor-kappaB (NF-kappaB) regulates proliferation and branching in mouse mammary epithelium. Mol. Biol. Cell. 2001, 12, 1445–1455. [Google Scholar] [CrossRef]
  48. Brantley, D.M.; Yull, F.E.; Muraoka, R.S.; Hicks, D.J.; Cook, C.M.; Kerr, L.D. Dynamic expression and activity of NF-kappaB during post-natal mammary gland morphogenesis. Mech. Dev. 2000, 97, 149–155. [Google Scholar] [CrossRef]
  49. Baxter, F.O.; Neoh, K.; Tevendale, M.C. The beginning of the end: Death signaling in early involution. J. Mammary. Gland Biol. Neoplasia. 2007, 12, 3–13. [Google Scholar] [CrossRef]
  50. Karin, M.; Ben-Neriah, Y. Phosphorylation meets ubiquitination: The control of NF-[kappa]B activity. Annu. Rev. Immunol. 2000, 18, 621–663. [Google Scholar] [CrossRef]
  51. Clarkson, R.W.; Heeley, J.L.; Chapman, R.; Aillet, F.; Hay, R.T.; Wyllie, A.; Watson, C.J. NF-kappaB inhibits apoptosis in murine mammary epithelia. J. Biol. Chem. 2000, 275, 12737–12742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Friedrichsen, S.; Harper, C.V.; Semprini, S.; Wilding, M.; Adamson, A.; Spiller, D.G.; Nelson, G.; Mullins, J.J.; White, M.R.H.; Davis, J.R.E. Tumor Necrosis Factor-α Activates the Human Prolactin Gene Promoter via Nuclear Factor-κB Signaling. Endocrinology 2006, 147, 773–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Terzidou, V.; Lee, Y.; Lindstrom, T.; Johnson, M.; Thornton, S.; Phillip, R.B. Regulation of the human oxytocin receptor by nuclear factor-kB and CCAAT/enhancer-binding protein-b. J. Clin. Endocrinol. Metab. 2006, 91, 2317–2326. [Google Scholar] [CrossRef] [PubMed]
  54. Boutet, P.; Sulon, J.; Closset, R.; Detilleux, J.; Beckers, J.F.; Bureau, F.; Lekeux, P. Prolactin-induced activation of nuclear factor kappaB in bovine mammary epithelial cells: Role in chronic mastitis. J. Dairy Sci. 2007, 90, 155–164. [Google Scholar] [CrossRef] [Green Version]
  55. Notebaert, S.; Demon, D.; Vanden Berghe, T.; Vandenabeele, P.; Meyer, E. Inflammatory mediators in Escherichia coli-induced mastitis in mice. Comp. Immunol. Microbiol. Infect. Dis. 2008, 31, 551–565. [Google Scholar] [CrossRef]
  56. Atabai, K.; Matthay, M.A. The pulmonary physician in critical care 5: Acute lung injury and the acute respiratory distress syndrome: Definitions and epidemiology. Thorax 2002, 57, 452–458. [Google Scholar] [CrossRef] [Green Version]
  57. Barbalat, R.; Lau, L.; Locksley, R.M.; Barton, G.M. Toll-like receptor 2 on inflammatory monocytes induces type i interferon in response to viral but not bacterial ligands. Nat. Immunol. 2009, 10, 1200–1209. [Google Scholar] [CrossRef]
  58. Fu, Y.; Gao, R.; Cao, Y.; Guo, M.; Wei, Z.; Zhou, E.; Li, Y.; Yao, M.; Yang, Z.; Zhang, N. Curcumin attenuates inflammatory responses by suppressing TLR4-mediated NF-κB signaling pathway in lipopolysaccharide-induced mastitis in mice. Int. Immunopharmacol. 2014, 20, 54–58. [Google Scholar] [CrossRef]
  59. Guo, Y.F.; Xu, N.N.; Sun, W.; Zhao, Y.; Li, C.Y.; Guo, M.Y. Luteolin reduces inflammation in Staphylococcus aureus-induced mastitis by inhibiting NF-kB activation and MMPs expression. Oncotarget 2017, 8, 28481–28493. [Google Scholar] [CrossRef] [Green Version]
  60. Jiang, K.F.; Zhao, G.; Deng, G.Z.; Wu, H.C.; Yin, N.N.; Chen, X.Y.; Qiu, C.W.; Peng, X.L. Polydatin ameliorates Staphylococcus aureus-induced mastitis in mice via inhibiting TLR2-mediated activation of the p38 MAPK/NF-κB pathway. Acta Pharmacol. Sin. 2017, 38, 211–222. [Google Scholar] [CrossRef]
  61. Li, D.; Fu, Y.; Zhang, W.; Su, G.; Liu, B.; Guo, M.; Li, F.; Liang, D.; Liu, Z.; Zhang, X.; et al. Salidroside attenuates inflammatory responses by suppressing nuclear factor-kappaB and mitogen activated protein kinases activation in lipopolysaccharide-induced mastitis in mice. Inflamm. Res. 2013, 62, 9–15. [Google Scholar] [CrossRef] [PubMed]
  62. Watters, T.M.; Kenny, E.F.; O’Neill, L.A. Structure, function and regulation of the Toll/IL-1 receptor adaptor proteins. Immunol. Cell Biol. 2007, 85, 411–41910. [Google Scholar] [CrossRef] [PubMed]
  63. Jiang, A.; Zhang, Y.; Zhang, X.; Wu, D.; Liu, Z.; Li, S.; Yang, Z. Morin alleviates LPS-induced mastitis by inhibiting the PI3K/AKT, MAPK, NF-κB and NLRP3 signaling pathway and protecting the integrity of blood-milk barrier. Int. Immunopharmacol. 2020, 78, 105972. [Google Scholar] [CrossRef] [PubMed]
  64. Baeuerle, P.A.; Baichwal, V.R. NF-κB as a frequent target for immunosuppressive and anti-inflammatory molecules. Adv. Immunol. 1997, 65, 111–137. [Google Scholar]
  65. Scheidereit, C. IkappaB kinase complexes: Gateways to NF-kappaB activation and transcription. Oncogene 2006, 25, 6685–6705. [Google Scholar] [CrossRef] [Green Version]
  66. Hoshino, K.; Takeuchi, O.; Kawai, T.; Sanjo, H.; Ogawa, T.; Takeda, Y.; Takeda, K.; Akira, S. Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: Evidence for TLR4 as the Lps gene product. J. Immunol. 1999, 162, 3749–3752. [Google Scholar]
  67. Boulanger, D.; Bureau, F.; Mélotte, D.; Mainil, J.; Lekeux, P. Increased nuclear factor κB activity in milk cells of mastitis-affected cows. J. Dairy Sci. 2003, 86, 1259–1267. [Google Scholar] [CrossRef]
  68. Wellnitz, O.; Kerr, D.E. Cryopreserved bovine mammary cells to model epithelial response to infection. Vet. Immunol. Immunopathol. 2004, 101, 191–202. [Google Scholar] [CrossRef]
  69. Wang, X.G.; Ju, Z.H.; Hou, M.H.; Jiang, Q.; Yang, C.H.; Zhang, Y.; Sun, Y.; Li, R.L.; Wang, C.F.; Zhong, J.F.; et al. Correction: Deciphering Transcriptome and Complex Alternative Splicing Transcripts in Mammary Gland Tissues from Cows Naturally Infected with Staphylococcus aureus Mastitis. PLoS ONE 2016, 11, e0167666. [Google Scholar] [CrossRef]
  70. Blum, S.E.; Heller, E.D.; Jacoby, S.; Krifucks, O.; Leitner, G. Comparison of the immune responses associated with experimental bovine mastitis caused by different strains of Escherichia coli. J. Dairy Res. 2017, 84, 190–197. [Google Scholar] [CrossRef]
  71. Porcherie, A.; Cunha, P.; Trotereau, A.; Roussel, P.; Gilbert, F.B.; Rainard, P.; Germon, P. Repertoire of Escherichia coli agonists sensed by innate immunity receptors of the bovine udder and mammary epithelial cells. Vet. Res. 2012, 43, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Riollet, C.; Rainard, P.; Poutrel, B. Differential induction of complement fragment C5a and inflammatory cytokines during intramammary infections with Escherichia coli and Staphylococcus aureus. Clin. Diagn. Lab. Immunol. 2000, 7, 161–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yang, W.; Zerbe, H.; Petzl, W.; Brunner, R.M.; Günther, J.; Draing, C.; von Aulock, S.; Schuberth, H.J.; Seyfert, H.M. Bovine TLR2 and TLR4 properly transduce signals from Staphylococcus aureus and E. coli, but S. aureus fails to both activate NF-κB in mammary epithelial cells and to quickly induce TNFα and interleukin-8 (CXCL8) expression in the udder. Mol. Immunol. 2008, 45, 1385–1397. [Google Scholar] [CrossRef]
  74. Takeda, K.; Akira, S. TLR signaling pathways. Semin. Immunol. 2004, 16, 3–9. [Google Scholar] [CrossRef]
  75. O’Neill, L.A.; Bowie, A.G. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat. Rev. Immunol. 2007, 7, 353–364. [Google Scholar] [CrossRef] [PubMed]
  76. Takeuchi, O.; Akira, S. Toll-like receptors; their physiological role and signal transduction system. Int. Immunopharmacol. 2001, 1, 625–635. [Google Scholar] [CrossRef]
  77. Cates, E.A.; Connor, E.E.; Mosser, D.M.; Bannerman, D.D. Functional characterization of bovine TIRAP and MyD88 in mediating bacterial lipopolysaccharide-induced endothelial NF-κB activation and apoptosis. Comp. Immunol. Microbiol. Infect. Dis. 2009, 32, 477–490. [Google Scholar] [CrossRef] [Green Version]
  78. Wang, J.; Guo, C.; Wei, Z.; He, X.; Kou, J.; Zhou, E.; Yang, Z.; Fu, Y. Morin suppresses inflammatory cytokine expression by downregulation of nuclear factorkappaB and mitogen-activated protein kinase (MAPK) signaling pathways in lipopolysaccharide-stimulated primary bovine mammary epithelial cells. J. Dairy. Sci. 2016, 99, 3016–3022. [Google Scholar] [CrossRef] [Green Version]
  79. Vallabhapurapu, S.; Karin, M. Regulation and Function of NF-κB Transcription Factors in the Immune System. Annu. Rev. Immunol. 2009, 27, 693–733. [Google Scholar] [CrossRef]
  80. Fang, L.; Hou, Y.; An, J.; Li, B.; Song, M.; Wang, X.; Sørensen, P.; Dong, Y.; Liu, C.; Wang, Y.; et al. Genome-wide transcriptional and post-transcriptional regulation of innate immune and defense responses of bovine mammary gland to Staphylococcus aureus. Front. Cell. Infect. Microbiol. 2016, 6, 193. [Google Scholar] [CrossRef] [Green Version]
  81. Bodnar, B.; DeGruttola, A.; Zhu, Y.; Lin, Y.; Zhang, Y.; Mo, X.; Hu, W. Emerging role of NIK/IKK2-binding protein (NIBP)/trafficking protein particle complex 9 (TRAPPC9) in nervous system diseases. Transl. Res. 2020, 224, 55–70. [Google Scholar] [CrossRef]
  82. Mir, A.; Kaufman, L.; Noor, A.; Motazacker, M.M.; Jamil, T.; Azam, M.; Kahrizi, K.; Rafiq, M.A.; Weksberg, R.; Nasr, T.; et al. Identification of Mutations in TRAPPC9, which Encodes the NIK- and IKK-β-Binding Protein, in Nonsyndromic Autosomal-Recessive Mental Retardation. Am. J. Hum. Genet. 2009, 85, 909–915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Qin, M.; Zhang, J.; Xu, C.; Peng, P.; Tan, L.; Liu, S.; Huang, J. Knockdown of NIK and IKKβ-binding protein (NIBP) reduces colorectal cancer metastasis through down-regulation of the canonical NF-κB signaling pathway and suppression of MAPK signaling mediated through ERK and JNK. PLoS ONE 2017, 12, e0170595. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, X.; Ma, P.; Liu, J.; Zhang, Q.; Zhang, Y.; Ding, X.; Jiang, L.; Wang, Y.; Zhang, Y.; Sun, D. Genome-wide association study in Chinese Holstein cows reveal two candidate genes for somatic cell score as an indicator for mastitis susceptibility. BMC Genetics 2015, 16, 111. [Google Scholar] [CrossRef] [PubMed]
  85. Song, M.; Wei, Y.; Khan, Z.M.; Wang, X.; Yu, Y. Molecular marker study of inflammatory reaction in Bovine mammary epithelium cell line induced by methicillin-resistant staphylococcus areus (MRSA). Acta Veterin Zootecn Sina 2016, 47, 1995–2010. [Google Scholar]
  86. Wu, D.; Zhang, X.; Liu, L.; Guo, Y. Key CMM Combinations in Prescriptions for Treating Mastitis and Working Mechanism Analysis Based on Network Pharmacology. Evid. Based Complement. Altern. Med. 2019, 2019. [Google Scholar] [CrossRef]
  87. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal. Transduct. Target. Ther. 2017, 2, 1–9. [Google Scholar] [CrossRef] [Green Version]
  88. Bannerman, D.D.; Paape, M.J.; Lee, J.W.; Zhao, X.; Hope, J.C.; Rainard, P. Escherichia coli and Staphylococcus aureus elicit differential innate immune responses following intramammary infection. Clin. Diagn. Lab. Immunol. 2004, 11, 463–472. [Google Scholar] [CrossRef] [Green Version]
  89. Bruckmaier, R.M. Gene expression of factors related to the immune reaction in response to intramammary Escherichia coli lipopolysaccharide challenge. J. Dairy Res. 2005, 72, 120–124. [Google Scholar] [CrossRef] [Green Version]
  90. Gilbert, F.B.; Cunha, P.; Jensen, K.; Glass, E.J.; Foucras, G.; Robert-Granié, C.; Rupp, R.; Rainard, P. Differential response of bovine mammary epithelial cells to Staphylococcus aureus or Escherichia coli agonists of the innate immune system. Vet. Res. 2013, 44, 40. [Google Scholar] [CrossRef] [Green Version]
  91. Jia, T.; Leiner, I.; Dorothee, G.; Brandl, K.; Pamer, E.G. MyD88 and Type I Interferon Receptor-Mediated Chemokine Induction and Monocyte Recruitment during Listeria monocytogenes Infection. J. Immunol. 2009, 183, 1271–1278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Kelly-Scumpia, K.M.; Scumpia, P.O.; Delano, M.J.; Weinstein, J.S.; Cuenca, A.G.; Wynn, J.L.; Moldawer, L.L. Type I interferon signaling in hematopoietic cells is required for survival in mouse polymicrobial sepsis by regulating CXCL10. J. Exp. Med. 2010, 207, 319–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zeng, X.; Moore, T.A.; Newstead, M.W.; Deng, J.C.; Lukacs, N.W.; Standiford, T.J. IP-10 mediates selective mononuclear cell accumulation and activation in response to intrapulmonary transgenic expression and during adenovirus-induced pulmonary inflammation. J. Interf. Cytokine Res. 2005, 25, 103–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zheng, J.; Watson, A.D.; Kerr, D.E. Genome-wide expression analysis of lipopolysaccharide-induced mastitis in a mouse model. Infect. Immun. 2006, 74, 1907–1915. [Google Scholar] [CrossRef] [Green Version]
  95. Kosciuczuk, E.M.; Lisowski, P.; Jarczak, J.; Majewska, A.; Rzewuska, M.; Zwierzchowski, L.; Bagnicka, E. Transcriptome profiling of Staphylococci-infected cow mammary gland parenchyma. BMC Vet. Res. 2017, 13, 1–12. [Google Scholar] [CrossRef]
  96. Griesbeck-Zilch, B.; Osman, M.; Kühn, C.; Schwerin, M.; Bruckmaier, R.H.; Pfaffl, M.W.; Hammerle-Fickinger, A.; Meyer, H.H.D.; Wellnitz, O. Analysis of key molecules of the innate immune system in mammary epithelial cells isolated from marker-assisted and conventionally selected cattle. J. Dairy Sci. 2009, 92, 4621–4633. [Google Scholar] [CrossRef] [Green Version]
  97. Islam, M.A.; Takagi, M.; Fukuyama, K.; Komatsu, R.; Albarracin, L.; Nochi, T.; Suda, Y.; Ikeda-Ohtsubo, W.; Rutten, V.; van Eden, W.; et al. Transcriptome analysis of the inflammatory responses of bovine mammary epithelial cells: Exploring immunomodulatory target genes for bovine mastitis. Pathogens 2020, 9, 200. [Google Scholar] [CrossRef]
  98. Rainard, P.; Riollet, C. Innate immunity of the bovine mammary gland. Vet. Res. 2006, 37, 369–400. [Google Scholar] [CrossRef] [Green Version]
  99. Waller, K.P. Modulation of endotoxin-induced inflammation in the bovine teat using antagonists inhibitors to leukotrienes, platelet activating factor and interleukin 1beta. Vet. Immunol. Immunopathol. 1997, 57, 239–251. [Google Scholar] [CrossRef]
  100. Bulgari, O.; Dong, X.; Roca, A.L.; Caroli, A.M.; Loor, J.J. Innate immune responses induced by lipopolysaccharide and lipoteichoic acid in primary goat mammary epithelial cells. J. Anim. Sci. Biotechnol. 2017, 8, 29. [Google Scholar] [CrossRef]
  101. Wang, W.; Hu, X.; Shen, P.; Zhang, N.; Fu, Y. Sodium houttuyfonate inhibits LPS-induced inflammatory response via suppressing TLR4/NF-ĸB signaling pathway in bovine mammary epithelial cells. Microb. Pathog. 2017, 107, 12–16. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, H.; Bi, C.; Wang, Y.; Sun, J.; Meng, X.; Li, J. Selenium ameliorates Staphylococcus aureus-induced inflammation in bovine mammary epithelial cells by inhibiting activation of TLR2, NF-ΚB and MAPK signaling pathways. BMC Vet. Res. 2018, 14, 197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Sordillo, L.M.; Streicher, K.L. Mammary gland immunity and mastitis susceptibility. J. Mammary Gland Biol. Neoplasia 2002, 7, 135–146. [Google Scholar] [CrossRef]
  104. Akhtar, M.; Guo, S.; Guo, Y.; Zahoor, A.; Shaukat, A.; Chen, Y.; Guo, M. Upregulated-gene expression of Pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) via TLRs following NF-κB and MAPKs in bovine mastitis. Acta Tropica 2020, 207, 105458. [Google Scholar] [CrossRef] [PubMed]
  105. Ren, K.; Torres, R. Role of interleukin-1β during pain and inflammation. Brain Res. Rev. 2009, 60, 57. [Google Scholar] [CrossRef] [Green Version]
  106. Dai, H.; Coleman, D.N.; Hu, L.; Martinez-Cortés, I.; Wang, M.; Parys, C.; Shen, X.; Loor, J.J. Methionine and arginine supplementation alter inflammatory and oxidative stress responses during lipopolysaccharide challenge in bovine mammary epithelial cells in vitro. J. Dairy Sci. 2020, 103, 676–689. [Google Scholar] [CrossRef]
  107. Zimmermann, K.K.; Spassov, S.G.; Strosing, K.M.; Ihle, P.M.; Engelstaedter, H.; Hoetzel, A.; Faller, S. Hydrogen Sulfide Exerts Anti-oxidative and Anti-inflammatory Effects in Acute Lung Injury. Inflammation 2018, 41, 249–259. [Google Scholar] [CrossRef]
  108. Benedetti, F.; Curreli, S.; Krishnan, S.; Davinelli, S.; Cocchi, F.; Scapagnini, G.; Gallo, R.C.; Zella, D. Anti-inflammatory effects of H2S during acute bacterial infection: A review. J. Transl. Med. 2017, 15, 100. [Google Scholar] [CrossRef]
  109. Zhi-Zhong, X.; Yang, L.; Jin-Song, B. Hydrogen sulfide and cellular redox homeostasis. Oxidative Med. Cell. Longev. 2016, 1–12. [Google Scholar] [CrossRef] [Green Version]
  110. Liu, W.; Xu, C.; You, X.; Olson, D.M.; Chemtob, S.; Gao, L.; Ni, X. Hydrogen sulfide delays LPS-Induced preterm birth in mice via anti-inflammatory pathways. PLoS ONE 2016, 11, e0152838. [Google Scholar] [CrossRef]
  111. Ahire, J.J.; Mokashe, N.U.; Patil, H.J.; Chaudhari, B.L. Antioxidative potential of folate producing probiotic Lactobacillus helveticus CD6. J. Food Sci. Technol. 2013, 50, 26–34. [Google Scholar] [CrossRef] [PubMed]
  112. Liu, P.; Yang, C.; Lin, S.; Zhao, G.; Zhang, T.; Guo, S.; Jiang, K.; Wu, H.; Qiu, C.; Guo, M.; et al. Sodium houttuyfonate inhibits LPS-induced mastitis in mice via the NF-κB signalling pathway. Mol. Med. Rep. 2019, 19, 2279–2286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Sun, L.; Chen, L.; Wang, F.; Zheng, X.; Yuan, C.; Niu, Q.; Li, Z.; Deng, L.; Zheng, B.; Li, C.; et al. Exogenous hydrogen sulfide prevents lipopolysaccharide-induced inflammation by blocking the TLR4/NF-κB pathway in MAC-T cells. Gene 2019, 710, 114–121. [Google Scholar] [CrossRef] [PubMed]
  114. Gálvez, J.; Coelho, G.; Crespo, M.E.; Cruz, T.; Rodríguez-Cabezas, M.E.; Concha, A.; Gonzalez, M.; Zarzuelo, A. Intestinal anti-inflammatory activity of morin on chronic experimental colitis in the rat. Aliment. Pharmacol. Ther. 2001, 15, 2027–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kempuraj, D.; Madhappan, B.; Christodoulou, S.; Boucher, W.; Cao, J.; Papadopoulou, N.; Cetrulo, C.L.; Theoharides, T.C. Flavonols inhibit pro-inflammatory mediator release, intracellular calcium ion levels and protein kinase C theta phosphorylation in human mast cells. Br. J. Pharmacol. 2005, 145, 934–944. [Google Scholar] [CrossRef] [Green Version]
  116. van de Veerdonk, F.L.; Netea, M.G.; Dinarello, C.A.; Joosten, L.A.B. Inflammasome activation and IL-1β and IL-18 processing during infection. Trends Immunol. 2011, 32, 110–116. [Google Scholar] [CrossRef]
  117. Yu, S.; Liu, X.; Yu, D.; Changyong, E.; Yang, J. Morin Protects LPS-Induced Mastitis via Inhibiting NLRP3 Inflammasome and NF-κB Signaling Pathways. Inflammation 2020, 43, 1293–1303. [Google Scholar] [CrossRef]
  118. Zhan, K.; Yang, T.; Feng, B.; Zhu, X.; Chen, Y.; Huo, Y.; Zhao, G. The protective roles of tea tree oil extracts in bovine mammary epithelial cells and polymorphonuclear leukocytes. J. Anim. Sci. Biotechnol. 2020, 11, 62. [Google Scholar] [CrossRef]
  119. Mahla, R.S.; Reddy, M.C.; Vijaya Raghava Prasad, D.; Kumar, H. Sweeten PAMPs: Role of sugar complexed PAMPs in innate immunity and vaccine biology. Front. Immunol. 2013, 4, 248. [Google Scholar] [CrossRef] [Green Version]
  120. Zhao, M.; Wang, H.; Yang, B.; Tao, H. Identification of cyclodextrin inclusion complex of chlorogenic acid and its antimicrobial activity. Food Chem. 2010, 120, 1138–1142. [Google Scholar] [CrossRef]
  121. Sato, Y.; Itagaki, S.; Kurokawa, T.; Ogura, J.; Kobayashi, M.; Hirano, T.; Sugawara, M.; Iseki, K. In vitro and in vivo antioxidant properties of chlorogenic acid and caffeic acid. Int. J. Pharm. 2011, 403, 136–138. [Google Scholar] [CrossRef] [PubMed]
  122. Shan, J.; Fu, J.; Zhao, Z.; Kong, X.; Huang, H.; Luo, L.; Yin, Z. Chlorogenic acid inhibits lipopolysaccharide-induced cyclooxygenase-2 expression in RAW264.7 cells through suppressing NF-κB and JNK/AP-1 activation. Int. Immunopharmacol. 2009, 9, 1042–1048. [Google Scholar] [CrossRef] [PubMed]
  123. Ruifeng, G.; Yunhe, F.; Zhengkai, W.; Ershun, Z.; Yimeng, L.; Minjun, Y.; Xiaojing, S.; Zhengtao, Y.; Naisheng, Z. Chlorogenic acid attenuates lipopolysaccharide-induced mice mastitis by suppressing TLR4-mediated NF-κB signaling pathway. Eur. J. Pharmacol. 2014, 729, 54–58. [Google Scholar] [CrossRef] [PubMed]
  124. Liang, D.; Li, F.; Fu, Y.; Cao, Y.; Song, X.; Wang, T.; Wang, W.; Guo, M.; Zhou, E.; Li, D.; et al. Thymol inhibits LPS-stimulated inflammatory response via down-regulation of NF-κB and MAPK signaling pathways in mouse mammary epithelial cells. Inflammation 2014, 37, 214–222. [Google Scholar] [CrossRef] [PubMed]
  125. Garcia, M.T.H.; Elsasser, H.T.; Biswas, D.; Moyes, M.K. The effect of citrus-derived oil on bovine blood neutrophil function and gene expression in vitro. J. Dairy Sci. 2014, 98, 1–99127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Li, J.; Yin, P.; Gong, P.; Lv, A.; Zhang, Z.; Liu, F. 8-Methoxypsoralen protects bovine mammary epithelial cells against lipopolysaccharide-induced inflammatory injury via suppressing JAK/STAT and NF-κB pathway. Microbiol.Immunol. 2019, 63, 427–437. [Google Scholar] [CrossRef]
  127. Yang, Z.; Yin, R.; Cong, Y.; Yang, Z.; Zhou, E.; Wei, Z.; Zhang, N. Oxymatrine Lightened the Inflammatory Response of LPS-Induced Mastitis in Mice through Affecting NF-κB and MAPKs Signaling Pathways. Inflammation 2014, 37, 2047–2055. [Google Scholar] [CrossRef]
  128. Ershun, Z.; Yunhe, F.; Zhengkai, W.; Yongguo, C.; Naisheng, Z.; Zhengtao, Y. Cepharanthine Attenuates Lipopolysaccharide-Induced Mice Mastitis by Suppressing the NF-κB Signaling Pathway. Inflammation 2014, 32, 331–337. [Google Scholar] [CrossRef]
  129. Su, S.; Xiaoyu, L.; Siting, L.; Pengfei, M.; Yingying, H.; Yanli, D.; Hongyan, D.; Shibin, F.; Jinchun, L.; Xichun, W.; et al. Rutin protects against lipopolysaccharide-induced mastitis by inhibiting the activation of the NF-κB signaling pathway and attenuating endoplasmic reticulum stress. Inflammopharmacology 2018, 27, 77–88. [Google Scholar] [CrossRef]
  130. Li, D.; Zhang, N.; Cao, Y.; Zhang, W.; Su, G.; Sun, Y.; Yang, Z. Emodin ameliorates lipopolysaccharide-induced mastitis in mice by inhibiting activation of NF-κB and MAPKs signal pathways. Eur. J. Pharmacol. 2013, 705, 79–85. [Google Scholar] [CrossRef]
  131. He, X.; Liu, W.; Shi, M.; Yang, Z.; Zhang, X.; Gong, P. Docosahexaenoic acid attenuates LPS-stimulated inflammatory response by regulating the PPARγ/NF-κB pathways in primary bovine mammary epithelial cells. Res. Vet. Sci. 2017, 112, 7–12. [Google Scholar] [CrossRef] [PubMed]
  132. He, X.; Wei, Z.; Zhou, E.; Chen, L.; Kou, J.; Wang, J.; Yang, Z. Baicalein attenuates inflammatory responses by suppressing TLR4 mediated NF-κB and MAPK signaling pathways in LPS-induced mastitis in mice. Internat. Immunopharmacol. 2015, 28, 470–476. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The key inducers of the NF-κB pathway and regulation of immunity, inflammation, cell survival and apoptosis by NF-κB signaling.
Figure 1. The key inducers of the NF-κB pathway and regulation of immunity, inflammation, cell survival and apoptosis by NF-κB signaling.
Animals 10 01625 g001
Figure 2. The interactive mechanism of E. coli and S. aureus with TLR2, TLR4 and TLR6, and the regulation of NF-κB signaling to activate the inflammatory genes.
Figure 2. The interactive mechanism of E. coli and S. aureus with TLR2, TLR4 and TLR6, and the regulation of NF-κB signaling to activate the inflammatory genes.
Animals 10 01625 g002
Figure 3. The regulation of the inflammation process by pro-inflammatory cytokines through NF-κB signaling; the cytokines, after attachment with receptors, cause the degradation of IKB from NF-κB. Upon activation, NF-κB directly binds to the promoters of target genes on DNA in the nucleus and regulates the specific inflammatory proteins.
Figure 3. The regulation of the inflammation process by pro-inflammatory cytokines through NF-κB signaling; the cytokines, after attachment with receptors, cause the degradation of IKB from NF-κB. Upon activation, NF-κB directly binds to the promoters of target genes on DNA in the nucleus and regulates the specific inflammatory proteins.
Animals 10 01625 g003
Table 1. Chemicals and their anti-inflammatory effect in mastitis by suppression NF-κB signaling.
Table 1. Chemicals and their anti-inflammatory effect in mastitis by suppression NF-κB signaling.
AuthorsAgent FunctionTargets
Sun et al. [113]H2S Block TLR4, ROS, NF-κB
Garcia et al. [125]Citrus oilsAntibacterialDown-regulate TLR2, NFKBIA, IL8, TNF-α
Wang et al. [78]MorinAnti-inflammatoryInhibit IL-6, TNF-α, IL-1β, suppress NF-κB phosphorylation
Li et al. [126]8-MethoxypsoralenAnti-inflammatoryInhibit IL-6, TNF-α, IL-8, IL-1β, suppress NF-κB phosphorylation
Chen et al. [36]NuciferineAnti-inflammatoryInhibit TLR4, TNF-α, IL-1β, suppress NF-κB phosphorylation
Yang et al. [127]OxymatrineAnti-inflammatorySuppress NF-κB phosphorylation
Ershun et al. [128]CepharanthineAnti-inflammatoryInhibit IL6, TNF-α, IL-1β, suppress NF-κB phosphorylation
Su et al. [129]Rutin Decrease level of IL-1β, IL-6, and TNF-α, suppress NF-κB phosphorylation
Liu et al. [112]Sodium houttuyfonateAntinflammatoryInhibit NF-κB phosphorylation
Li et al. [130]Emodin amelioratesAnti-inflammatory, antibacterial Decrease level of IL-1β, IL-6, and TNF-α, suppress NF-κB phosphorylation
Hu et al. [42]Cynatratoside-C from Cynanchum atratumAnti-inflammatorySuppress TLR4, inhibit NF-κB phosphorylation
He et al. [131]Docosahexaenoic acidAnti-inflammatoryDecrease level of IL-1β, IL-6, and TNF-α, suppress NF-κB phosphorylation
He et al. [132]BaicaleinAnti-inflammatorySuppress TLR4, inhibit NF-κB phosphorylation

Share and Cite

MDPI and ACS Style

Khan, M.Z.; Khan, A.; Xiao, J.; Ma, J.; Ma, Y.; Chen, T.; Shao, D.; Cao, Z. Overview of Research Development on the Role of NF-κB Signaling in Mastitis. Animals 2020, 10, 1625. https://doi.org/10.3390/ani10091625

AMA Style

Khan MZ, Khan A, Xiao J, Ma J, Ma Y, Chen T, Shao D, Cao Z. Overview of Research Development on the Role of NF-κB Signaling in Mastitis. Animals. 2020; 10(9):1625. https://doi.org/10.3390/ani10091625

Chicago/Turabian Style

Khan, Muhammad Zahoor, Adnan Khan, Jianxin Xiao, Jiaying Ma, Yulin Ma, Tianyu Chen, Dafu Shao, and Zhijun Cao. 2020. "Overview of Research Development on the Role of NF-κB Signaling in Mastitis" Animals 10, no. 9: 1625. https://doi.org/10.3390/ani10091625

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