Redox Homeostasis in Poultry: Regulatory Roles of NF-κB
Abstract
:1. Introduction
2. Transcription Factor Nuclear Factor Kappa B
3. NF-κB and Oxidative Stress
4. Nrf2 and NF-ĸB Interplay in Oxidative Stress
5. NF-κB in Poultry Production
- Chicken p65 was shown to be approximately 55% identical to the mouse and human p65 proteins. Similar to its mammalian counterpart, chicken p65 contains the Rel homology domain (RHD) in its N-terminal consisting of 286 amino acids and the putative transactivation domain in its C-terminal region;
- It was proven that the RHD was highly conserved between the chicken and mammalian p65 proteins;
- The highest expression of a 2.6 kb transcript of p65 was detected in the spleen. It was also detected in other organs;
- A fusion protein containing the RHD of chicken p65 was reported to bind to a consensus kappa B-site;
- p65 was shown to form one or more complexes with various cellular proteins, including p50, p105, and c-Rel in chicken spleen cells [151].
6. Effect of Various Stress Factors on NF-κB Expression and Activity in Poultry
6.1. Thermal Stress
6.2. Mycotoxins
6.3. Mineral Dietary Excess and Heavy-Metal Contamination
6.3.1. Mn, Cu, and NF-κB
6.3.2. As and NF-κB
6.3.3. Cu, As, and NF-κB
6.3.4. Pb and NF-κB
6.3.5. Cd and NF-κB
6.4. Other Toxic Stress Factors
6.4.1. H2O2
6.4.2. NH3
6.4.3. H2S
6.5. LPS-Induced Stress
6.6. Diseases
6.6.1. Escherichia coli
6.6.2. Salmonella
6.6.3. Mycoplasma gallisepticum
6.6.4. Eimeria tenella
6.6.5. Clostridium perfringens
6.6.6. Chlamydia psittaci
6.6.7. Infectious Bursal Disease
6.6.8. Newcastle Disease
6.6.9. Other Viral Diseases
7. Nutritional Modulation of NF-κB in Poultry
7.1. Selenium
7.1.1. Se Deficiency
7.1.2. Se and Pb Toxicity
7.1.3. Se and Cd Toxicity
7.1.4. Se and LPS
7.2. Amino Acids
7.3. Phytogenic Supplements
7.4. Other Nutrients and Probiotics
8. NF-κB and Inflammation in Poultry Production
9. Conclusions
- Similar to mammalian species, in poultry, NF-κB plays a central role in the regulation of many physiological and pathological processes.
- In thermally stressed birds, NF-κB expression is condition-dependent, including temperature, exposure duration, and bird’s age.
- The effects of dietary AFB1 on NF-κB expression in chicken liver are also condition-dependent. In general, AFB1 was shown to compromise AO defenses and increase proinflammatory cytokine production via NF-κB induction.
- Mn or Cu excess in the chicken diet was shown to increase the expression of NF-κB in testes, heart, and immune organs.
- The proinflammatory effects of heavy metals (As, Cd and Pb) in chickens were shown to be mediated via NF-κB pathway activation in various tissues.
- Increased concentrations of NH3 and H2S (main environmental stressors in poultry production) in air during chicken housing were shown to impose oxidative stress and inflammatory responses via NF-κB activation.
- The stimulating effect of LPS on NF-κB expression was shown in vitro in model systems and in vivo with poultry.
- In many bacterial and viral diseases, NF-κB is activated to increase proinflammatory cytokine production and impose an inflammatory response to create a hostile environment for pathogens.
- The main bacterial pathogens causing various diseases in poultry production, including Escherichia coli, various Salmonella species, Mycoplasma gallisepticum, Eimeria tenella, Clostridium perfringens, and Chlamydia psittaci, were shown to induce proinflammatory responses in birds associated with increased NF-κB expression and activity.
- In model systems based on an investigation of gene expression changes due to various infections, it was proven that the development of viral diseases, including infectious bursal disease, Newcastle disease, Marek’s disease, and reovirus challenges, was associated with the induction of NF-κB and inflammatory responses.
- Nutritional modulation of NF-κB expression and activity was shown to be achieved by using various antioxidants, including selenium, various polyphenols, taurine, retinoic acid, vitamin E, and some probiotics. In fact, under various stress conditions, these nutrients can ameliorate (partly or completely) the increased NF-κB expression and activity imposed by stressors.
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
AFB1 | aflatoxin B1 |
AKT | protein kinase B |
ALT | alanine aminotransferase |
AO | antioxidant |
AP1 | transcription factor |
APEC | avian pathogenic Escherichia coli |
ARD | ankyrin repeat domain |
ARE | antioxidant response element |
ATF3 | activating transcription factor 3 |
CAT | catalase |
COX2 | cyclooxygenase-2 |
CXCLi2 | recombinant chicken chemotactic and angiogenic factor 2 |
Cyt C | cytochrome C |
DC | dendritic cells |
dpi | days post infection |
HDAC3 | histone deacetylase3 |
IBD | infectious bursal disease |
IBDV | infectious bursal disease virus |
IFIT5 | Interferon-induced protein with tetratricopeptide repeats 5 |
iNOS | inducible nitric oxide synthases |
IRF7 | interferon regulatory factor 7 |
LOX5 | lipoxygenase 5 |
NDV | Newcastle disease virus |
PGC-1α | peroxisome proliferator-activated receptor coactivator |
p53 | tumor protein p53 |
PPAR | peroxisome proliferator-activated receptor |
PTEN | phosphatase and tensin homolog |
PUFAs | polyunsaturated fatty acids |
ROS | reactive oxygen species |
RNS | reactive nitrogen species |
FHS | follicle-stimulating hormone |
FOXO3 | transcription factors |
FOXP3 | forkhead box protein P3 |
GCL | glutamyl-cysteine ligase |
GCLC | catalytic subunit of GCL |
GR | glutathione reductase |
Grx | glutaredoxin |
GSH | reduced glutathione |
GPx | glutathione peroxidase |
GST | glutathione S-transferase |
HAT | histone acetyl transferase |
HIF | hypoxia-inducible factor |
HO-1 | heme oxygenase 1 |
HS | heat stress |
HSP | heat-shock protein |
IκB | inhibitor of κB |
IKK | IκB kinase |
IL-1R | IL1 receptor |
IFN-γ | interferon gamma |
Keap1 | Kelch-like erythroid cell-derived protein with CNC homology (ECH)-associated protein 1 |
LPS | lipopolysaccharide |
MAPK | mitogen-activated protein kinase |
MDA | malondialdehyde |
MIP-1α | macrophage inflammatory protein |
MG | Mycoplasma gallisepticum |
MMP16 | matrix metalloproteinase 16 |
Msr | methionine sulfoxide reductase |
MT3 | metallothionein 3 |
NEMO | the regulatory subunit IKKγ |
NF-κB | nuclear factor-κB |
NLR | NOD-like receptor |
NRLC5 | NOD-like receptor family CARD domain containing 5 |
NQO1 | NAD(P)H:quinone dehydrogenase 1 |
Nrf2 | nuclear factor-erythroid-2 (NF-E2) and related factor 2 |
PCNA | proliferating cell nuclear antigen |
PGC-1α | peroxisome proliferator-activated receptor-gamma coactivator 1α |
PPARs | peroxisome proliferator-activated receptors |
Prx | peroxiredoxin |
PTGEs | prostaglandin E synthase |
PGE | prostaglandin E |
PRR | pattern recognition receptor |
RANKL | receptor activator of NF-κB ligand |
RHD | Rel homology domain |
RONS | reactive oxygen and nitrogen species |
ROS | reactive oxygen species |
SeMet | selenomethionine |
SOD | superoxide dismutase |
sPLA2 | secretory phospholipase 2 |
STAT3 | signal transducer and activator of transcription 3 |
TGF-β | transforming growth factor beta |
Th1 | T helper cell 1 |
TLR | Toll-like receptor |
TNF-α | tumor necrosis factor |
TNFR | TNF receptor |
TRAF6 | tumor necrosis factor receptor-associated factor 6 |
TRIF | TIR-domain-containing adapter-inducing interferon-β |
Trx | thioredoxin |
TrxR | thioredoxin reductase |
UCP2 | uncoupling protein 2 |
XOR | xanthine oxidoreductase |
Wnt4 | signaling molecule (Wingless-related MMTV integration site 4) |
References
- Surai, P.F. Vitagenes in Avian Biology and Poultry Health; Wageningen Academic Publishers: Wageningen, The Netherlands, 2020. [Google Scholar]
- Musaogullari, A.; Chai, Y.C. Redox Regulation by Protein S-Glutathionylation: From Molecular Mechanisms to Implications in Health and Disease. Int. J. Mol. Sci. 2020, 21, 8113. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Wang, X.; Saredy, J.; Yuan, Z.; Yang, X.; Wang, H. Innate-adaptive immunity interplay and redox regulation in immune response. Redox. Biol. 2020, 37, 101759. [Google Scholar] [CrossRef] [PubMed]
- Francioso, A.; Baseggio Conrado, A.; Mosca, L.; Fontana, M. Chemistry and Biochemistry of Sulfur Natural Compounds: Key Intermediates of Metabolism and Redox Biology. Oxid. Med. Cell Longev. 2020, 2020, 8294158. [Google Scholar] [CrossRef] [PubMed]
- Corkey, B.E.; Deeney, J.T. The Redox Communication Network as a Regulator of Metabolism. Front. Physiol. 2020, 11, 567796. [Google Scholar] [CrossRef]
- Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An Overview of Nrf2 Signaling Pathway and Its Role in Inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef]
- Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell. Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
- Carvalho, R.H.; Ida, E.I.; Madruga, M.S.; Martínez, S.L.; Shimokomaki, M.; Estévez, M. Underlying connections between the redox system imbalance, protein oxidation and impaired quality traits in pale, soft and exudative (PSE) poultry meat. Food Chem. 2017, 215, 129–137. [Google Scholar] [CrossRef]
- Pan, X.; Zhang, L.; Xing, T.; Li, J.; Gao, F. The impaired redox status and activated Nrf2/ARE pathway in wooden breast myopathy in broiler chickens. Asian-Australas. J. Anim. Sci. 2020, in press. [Google Scholar] [CrossRef] [Green Version]
- Estevez, M.; Petracci, M. Benefits of Magnesium Supplementation to Broiler Subjected to Dietary and Heat Stress: Improved Redox Status, Breast Quality and Decreased Myopathy Incidence. Antioxidants 2019, 8, 456. [Google Scholar] [CrossRef] [Green Version]
- Dalgaard, T.S.; Briens, M.; Engberg, R.M.; Lauridsen, C. The influence of selenium and selenoproteins on immune responses of poultry and pigs. Anim. Feed Sci. Technol. 2018, 238, 73–83. [Google Scholar] [CrossRef]
- Lauridsen, C. From oxidative stress to inflammation: Redox balance and immune system. Poult. Sci. 2019, 98, 4240–4246. [Google Scholar] [CrossRef] [PubMed]
- Surai, P.F. Selenium in Poultry Nutrition and Health; Wageningen Academic Publishers: Wageningen, The Netherlands, 2018. [Google Scholar]
- Surai, P.F.; Fisinin, V.I. Antioxidant-Prooxidant Balance in the Intestine: Applications in Chick Placement and Pig Weaning. J. Vet. Sci. Med. 2015, 3, 16. [Google Scholar]
- Mishra, B.; Jha, R. Oxidative Stress in the Poultry Gut: Potential Challenges and Interventions. Front. Vet. Sci. 2019, 6, 60. [Google Scholar] [CrossRef]
- Dong, Y.; Lei, J.; Zhang, B. Effects of dietary quercetin on the antioxidative status and cecal microbiota in broiler chickens fed with oxidized oil. Poult. Sci. 2020, 99, 4892–4903. [Google Scholar] [CrossRef] [PubMed]
- Kövesi, B.; Cserháti, M.; Erdélyi, M.; Zándoki, E.; Mézes, M.; Balogh, K. Long-Term Effects of Ochratoxin A on the Glutathione Redox System and Its Regulation in Chicken. Antioxidants 2019, 8, 178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Egresi, A.; Süle, K.; Szentmihályi, K.; Blázovics, A.; Fehér, E.; Hagymási, K.; Fébel, H. Impact of milk thistle (Silybum marianum) on the mycotoxin caused redox-homeostasis imbalance of ducks liver. Toxicon 2020, 187, 181–187. [Google Scholar] [CrossRef]
- Liu, Y.; Zhao, H.; Wang, Y.; Guo, M.; Mu, M.; Xing, M. Arsenic (III) and/or copper (II) induces oxidative stress in chicken brain and subsequent effects on mitochondrial homeostasis and autophagy. J. Inorg. Biochem. 2020, 211, 111201. [Google Scholar] [CrossRef]
- Surai, P.F.; Kochish, I.I. Nutritional modulation of the antioxidant capacities in poultry: The case of selenium. Poult. Sci. 2019, 98, 4231–4239. [Google Scholar] [CrossRef]
- Surai, P.F.; Kochish, I.I.; Romanov, M.N.; Griffin, D.K. Nutritional modulation of the antioxidant capacities in poultry: The case of vitamin E. Poult. Sci. 2019, 98, 4030–4041. [Google Scholar] [CrossRef]
- Surai, P.F.; Kochish, I.I. Carotenoids in Aviculture. In Pigments from Microalgae Handbook; Springer Nature Switzerland: Cham, Switzerland, 2020; pp. 515–540. [Google Scholar]
- Tolba, S.A.; Magnuson, A.D.; Sun, T.; Lei, X.G. Dietary supplemental microalgal astaxanthin modulates molecular profiles of stress, inflammation, and lipid metabolism in broiler chickens and laying hens under high ambient temperatures. Poult. Sci. 2020, 99, 4853–4860. [Google Scholar] [CrossRef]
- Surai, P.F.; Fisinin, V.I. Vitagenes in poultry production. Part 3. Vitagene concept development. Worlds Poult. Sci. J. 2016, 72, 793–804. [Google Scholar] [CrossRef]
- Surai, P.F.; Fisinin, V.I. Antioxidant system regulation: From vitamins to vitagenes. In Handbook of Cholesterol; Watson, R.R., de Meester, F., Eds.; Wageningen Academic Publishers: Wageningen, The Netherlands, 2016; pp. 451–481. [Google Scholar]
- Surai, P.F.; Kochish, I.I.; Fisinin, V.I. Antioxidant systems in poultry biology: Nutritional modulation of vitagenes. Eur. J. Poult. Sci. 2017, 81, 1612–9199. [Google Scholar]
- Surai, P.F.; Kochish, I.I.; Fisinin, V.I.; Kidd, M.T. Antioxidant Defence Systems and Oxidative Stress in Poultry Biology: An Update. Antioxidants 2019, 8, 235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Surai, P.F.; Fisinin, V.I. Vitagenes in poultry production. Part 1. Technological and environmental stresses. Worlds Poult. Sci. J. 2016, 72, 721–733. [Google Scholar] [CrossRef]
- Surai, P.F.; Fisinin, V.I. Vitagenes in poultry production. Part 2. Nutritional and internal stresses. Worlds Poult. Sci. J. 2016, 72, 761–772. [Google Scholar] [CrossRef]
- Surai, P.F.; Kochish, I.I.; Fisinin, V.I.; Grozina, A.A.; Shatskikh, E.V. Molecular Mechanisms of Chicken Gut Health Maintenance: Role of Microbiota; Agricultural Technologies: Moscow, Russia, 2018. [Google Scholar]
- Schijns, V.E.; van de Zande, S.; Lupiani, B.; Reddy, S.M. Practical aspects of poultry vaccination. In Avian immunology; Academic Press: Cambridge, MA, USA, 2014; pp. 345–362. [Google Scholar]
- Cervantes, H.M. Antibiotic-free poultry production: Is it sustainable? J. Appl. Poult. Res. 2015, 24, 91–97. [Google Scholar] [CrossRef]
- Kim, W.H.; Lillehoj, H.S. Immunity, immunomodulation, and antibiotic alternatives to maximize the genetic potential of poultry for growth and disease response. Anim. Feed Sci. Technol. 2019, 250, 41–50. [Google Scholar] [CrossRef]
- Desin, T.S.; Köster, W.; Potter, A.A. Salmonella vaccines in poultry: Past, present and future. Expert Rev. Vaccines 2013, 12, 87–96. [Google Scholar] [CrossRef]
- Guillén, S.; Marcén, M.; Álvarez, I.; Mañas, P.; Cebrián, G. Stress resistance of emerging poultry-associated Salmonella serovars. Int. J. Food Microb. 2020, 335, 108884. [Google Scholar] [CrossRef]
- Gast, R.K.; Porter Jr, R.E. Salmonella infections. In Diseases of Poultry; David, E.S., Boulianne, M., Logue, C.M., McDougald, L.R., Nair, V., Suarez, D.L., de Wit, S., Grimes, T., Johnson, D., Kromm, M., et al., Eds.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2020; pp. 717–753. [Google Scholar]
- Iannetti, L.; Neri, D.; Santarelli, G.A.; Cotturone, G.; Vulpiani, M.P.; Salini, R.; Antoci, S.; Di Serafino, G.; Di Giannatale, E.; Pomilio, F.; et al. Animal welfare and microbiological safety of poultry meat: Impact of different at-farm animal welfare levels on at-slaughterhouse Campylobacter and Salmonella contamination. Food Control 2020, 109, 106921. [Google Scholar] [CrossRef]
- Everest, H.; Hill, S.C.; Daines, R.; Sealy, J.E.; James, J.; Hansen, R.; Iqbal, M. The evolution, spread and global threat of H6Nx avian influenza viruses. Viruses 2020, 12, 673. [Google Scholar] [CrossRef] [PubMed]
- Wigley, P. Immunity to bacterial infection in the chicken. Dev. Comp. Immunol. 2013, 41, 413–417. [Google Scholar] [CrossRef] [PubMed]
- Korver, D.R. Implications of changing immune function through nutrition in poultry. Anim. Feed Sci. Technol. 2012, 173, 54–64. [Google Scholar] [CrossRef]
- Kaspers, B.; Göbel, T.W. The avian immune system. In Encyclopaedia of Immunobiology; Ratcliffe, M.J.H., Ed.; Elsevier: Amsterdam, The Netherlands, 2016; Volume 1, pp. 498–503. [Google Scholar]
- Swaggerty, C.L.; Callaway, T.R.; Kogut, M.H.; Piva, A.; Grilli, E. Modulation of the immune response to improve health and reduce foodborne pathogens in poultry. Microorganisms 2019, 7, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, C.; Li, J.; Zhang, W.; Shah, S.W.A.; Ishfaq, M. Mycoplasma gallisepticum triggers immune damage in the chicken thymus by activating the TLR-2/MyD88/NF-κB signaling pathway and NLRP3 inflammasome. Vet. Res. 2020, 51, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cardoso Dal Pont, G.; Farnell, M.; Farnell, Y.; Kogut, M.H. Dietary Factors as Triggers of Low-Grade Chronic Intestinal Inflammation in Poultry. Microorganisms 2020, 8, 139. [Google Scholar] [CrossRef] [Green Version]
- Sen, R.; Baltimore, D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell 1986, 46, 705–716. [Google Scholar] [CrossRef]
- Yu, H.; Lin, L.; Zhang, Z.; Zhang, H.; Hu, H. Targeting NF-κB pathway for the therapy of diseases: Mechanism and clinical study. Signal Transduct. Target Ther. 2020, 5, 209. [Google Scholar] [CrossRef]
- Lambrou, G.I.; Hatziagapiou, K.; Vlahopoulos, S. Inflammation and tissue homeostasis: The NF-κB system in physiology and malignant progression. Mol. Biol. Rep. 2020, 47, 4047–4063. [Google Scholar] [CrossRef]
- Chawla, M.; Roy, P.; Basak, S. Role of the NF-κB system in context-specific tuning of the inflammatory gene response. Cur. Opin. Immunol. 2020, 68, 21–27. [Google Scholar] [CrossRef]
- Kopitar-Jerala, N. Innate Immune Response in Brain, NF-Kappa B Signaling and Cystatins. Front. Mol. Neurosci. 2015, 8, 73. [Google Scholar] [CrossRef] [Green Version]
- Sehnert, B.; Burkhardt, H.; Dübel, S.; Voll, R.E. Cell-Type Targeted NF-kappaB Inhibition for the Treatment of Inflammatory Diseases. Cells 2020, 9, 1627. [Google Scholar] [CrossRef] [PubMed]
- Grilli, M.; Memo, M. Transcriptional pharmacology of neurodegenerative disorders: Novel venue towards neuroprotection against excitotoxicity? Mol. Psychiatry. 1997, 2, 192–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Zhao, Y.; Tian, B.; Jamaluddin, M.; Mitra, A.; Yang, J.; Rowicka, M.; Brasier, A.R.; Kudlicki, A. Modulation of gene expression regulated by the transcription factor NF-κB/RelA. J. Biol. Chem. 2014, 289, 11927–11944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Niederberger, E.; Geisslinger, G. Proteomics and NF-κB: An update. Expert Rev. Proteom. 2013, 10, 189–204. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Ding, J.; Yang, J.; Guo, X.; Zheng, Y. MicroRNA Roles in the Nuclear Factor Kappa B Signaling Pathway in Cancer. Front. Immunol. 2018, 9, 546. [Google Scholar] [CrossRef] [Green Version]
- Thoma, A.; Lightfoot, A.P. NF-kB and Inflammatory Cytokine Signalling: Role in Skeletal Muscle Atrophy. Adv. Exp. Med. Biol. 2018, 1088, 267–279. [Google Scholar]
- McGuire, C.; Prinz, M.; Beyaert, R.; van Loo, G. Nuclear factor kappa B (NF-κB) in multiple sclerosis pathology. Trends Mol. Med. 2013, 19, 604–613. [Google Scholar] [CrossRef]
- Awasthee, N.; Rai, V.; Chava, S.; Nallasamy, P.; Kunnumakkara, A.B.; Bishayee, A.; Chauhan, S.C.; Challagundla, K.B.; Gupta, S.C. Targeting IκappaB kinases for cancer therapy. Semin. Cancer Biol. 2019, 56, 12–24. [Google Scholar] [CrossRef]
- Baker, R.G.; Hayden, M.S.; Ghosh, S. NF-κB, inflammation, and metabolic disease. Cell. Metab. 2011, 13, 11–22. [Google Scholar] [CrossRef] [Green Version]
- Lingappan, K. NF-κB in oxidative stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Yousefzadeh, M.J.; Suh, Y.; Niedernhofer, L.J.; Robbins, P.D. Signal Transduction, Ageing and Disease. Subcell. Biochem. 2019, 91, 227–247. [Google Scholar] [PubMed]
- Patel, M.; Horgan, P.G.; McMillan, D.C.; Edwards, J. NF-κB pathways in the development and progression of colorectal cancer. Transl. Res. 2018, 197, 43–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sivandzade, F.; Prasad, S.; Bhalerao, A.; Cucullo, L. NRF2 and NF-κB interplay in cerebrovascular and neurodegenerative disorders: Molecular mechanisms and possible therapeutic approaches. Redox. Biol. 2019, 21, 101059. [Google Scholar] [CrossRef]
- Jones, S.V.; Kounatidis, I. Nuclear Factor-Kappa B and Alzheimer Disease, Unifying Genetic and Environmental Risk Factors from Cell to Humans. Front. Immunol. 2017, 8, 1805. [Google Scholar] [CrossRef] [Green Version]
- Hayden, M.S.; Ghosh, S. Regulation of NF-κB by TNF family cytokines. Semin. Immunol. 2014, 26, 253–266. [Google Scholar] [CrossRef] [Green Version]
- NF-kB Target Genes. Available online: www/bu.edu/nf-kb/gene-resources/target-genes/ (accessed on 1 December 2020).
- Scott, O.; Roifman, C.M. NF-κB pathway and the Goldilocks principle: Lessons from human disorders of immunity and inflammation. J. Allergy Clin. Immunol. 2019, 143, 1688–1701. [Google Scholar] [CrossRef]
- Zhang, L.; Xiao, X.; Arnold, P.R.; Li, X.C. Transcriptional and epigenetic regulation of immune tolerance: Roles of the NF-κB family members. Cell Mol. Immunol. 2019, 16, 315–323. [Google Scholar] [CrossRef] [Green Version]
- Park, Y.H. The nuclear factor-kappa B pathway and response to treatment in breast cancer. Pharmacogenomics 2017, 18, 1697–1709. [Google Scholar] [CrossRef]
- Sun, S.C. The non-canonical NF-κB pathway in immunity and inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef]
- Giridharan, S.; Srinivasan, M. Mechanisms of NF-κB p65 and strategies for therapeutic manipulation. J. Inflamm. Res. 2018, 11, 407–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rabie, N.S.; Amin Girh, Z. Bacterial vaccines in poultry. Bull. Natl. Res. Centre 2020, 44, 15. [Google Scholar] [CrossRef] [PubMed]
- Gimeno, I.M.; Schat, K.A. Virus-Induced Immunosuppression in Chickens. Avian Dis. 2018, 62, 272–285. [Google Scholar] [CrossRef] [PubMed]
- Kim, W.H.; Chaudhari, A.A.; Lillehoj, H.S. Involvement of T Cell Immunity in Avian Coccidiosis. Front. Immunol. 2019, 10, 2732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buelna-Chontal, M.; Zazueta, C. Redox activation of Nrf2 & NF-κB: A double end sword? Cell Signal. 2013, 25, 2548–2557. [Google Scholar]
- Pedruzzi, L.M.; Stockler-Pinto, M.B.; Leite, M., Jr.; Mafra, D. Nrf2-keap1 system versus NF-κB: The good and the evil in chronic kidney disease? Biochimie 2012, 94, 2461–2466. [Google Scholar] [CrossRef]
- Tkach, K.E.; Oyler, J.E.; Altan-Bonnet, G. Cracking the NF-κB code. Sci. Signal. 2014, 7, pe5. [Google Scholar] [CrossRef]
- Pal, S.; Bhattacharjee, A.; Ali, A.; Mandal, N.C.; Mandal, S.C. Chronic inflammation and cancer: Potential chemoprevention through nuclear factor kappa B and p53 mutual antagonism. J. Inflamm. 2014, 11, 23. [Google Scholar] [CrossRef] [Green Version]
- Salles, A.; Romano, A.; Freudenthal, R. Synaptic NF-kappa B pathway in neuronal plasticity and memory. J. Physiol. Paris 2014, 108, 256–262. [Google Scholar] [CrossRef]
- Tilborghs, S.; Corthouts, J.; Verhoeven, Y.; Arias, D.; Rolfo, C.; Trinh, X.B.; van Dam, P.A. The role of Nuclear Factor-kappa B signaling in human cervical cancer. Crit. Rev. Oncol. Hematol. 2017, 120, 141–150. [Google Scholar] [CrossRef]
- Hoffmann, A.; Baltimore, D. Circuitry of nuclear factor κB signaling. Immunol. Rev. 2006, 210, 171–186. [Google Scholar] [CrossRef] [PubMed]
- Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Jesús, T.J.; Ramakrishnan, P. NF-κB c-Rel Dictates the Inflammatory Threshold by Acting as a Transcriptional Repressor. iScience 2020, 23, 100876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nelson, R.H.; Nelson, D.E. Signal Distortion: How Intracellular Pathogens Alter Host Cell Fate by Modulating NF-κB Dynamics. Front. Immunol. 2018, 9, 2962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fusella, F.; Seclì, L.; Cannata, C.; Brancaccio, M. The one thousand and one chaperones of the NF-κB pathway. Cell. Mol. Life Sci. 2020, 77, 2275–2288. [Google Scholar] [CrossRef]
- Zhang, Q.; Lenardo, M.J.; Baltimore, D. 30 years of NF-κB: A blossoming of relevance to human pathobiology. Cell 2017, 168, 37–57. [Google Scholar] [CrossRef] [Green Version]
- Lepetsos, P.; Papavassiliou, K.A.; Papavassiliou, A.G. Redox and NF-κB signaling in osteoarthritis. Free Rad. Biol. Med. 2019, 132, 90–100. [Google Scholar] [CrossRef]
- Gloire, G.; Legrand-Poels, S.; Piette, J. NF-κB activation by reactive oxygen species: Fifteen years later. Biochem. Pharmacol. 2006, 72, 1493–1505. [Google Scholar] [CrossRef]
- Gloire, G.; Piette, J. Redox regulation of nuclear post-translational modifications during NF-kappaB activation. Antioxid. Redox Signal. 2009, 11, 2209–2222. [Google Scholar] [CrossRef]
- Herscovitch, M.; Comb, W.; Ennis, T.; Coleman, K.; Yong, S.; Armstead, B.; Kalaitzidis, D.; Chandani, S.; Gilmore, T.D. Intermolecular disulfide bond formation in the NEMO dimer requires Cys54 and Cys347. Biochem. Biophys. Res. Commun. 2008, 367, 103–108. [Google Scholar] [CrossRef] [Green Version]
- Wu, M.; Bian, Q.; Liu, Y.; Fernandes, A.F.; Taylor, A.; Pereira, P.; Shang, F. Sustained oxidative stress inhibits NF-κB activation partially via inactivating the proteasome. Free Rad. Biol. Med. 2009, 46, 62–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lushchak, V.I. Adaptive response to oxidative stress: Bacteria, fungi, plants and animals. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2011, 153, 175–190. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Hai, C. Novel insights into redox system and the mechanism of redox regulation. Mol. Biol. Rep. 2016, 43, 607–628. [Google Scholar] [CrossRef] [PubMed]
- Dayalan Naidu, S.; Kostov, R.V.; Dinkova-Kostova, A.T. Transcription factors Hsf1 and Nrf2 engage in crosstalk for cytoprotection. Trends Pharmacol. Sci. 2015, 36, 6–14. [Google Scholar] [CrossRef]
- Dengler, V.L.; Galbraith, M.; Espinosa, J.M. Transcriptional regulation by hypoxia inducible factors. Crit. Rev. Biochem. Mol. Biol. 2014, 49, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Cai, D.; Khor, S. “Hypothalamic Microinflammation” Paradigm in Aging and Metabolic Diseases. Cell Metab. 2019, 30, 19–35. [Google Scholar] [CrossRef]
- Yan, J.; Zhang, H.; Yin, Y.; Li, J.; Tang, Y.; Purkayastha, S.; Li, L.; Cai, D. Obesity- and aging-induced excess of central transforming growth factor-beta potentiates diabetic development via an RNA stress response. Nat. Med. 2014, 20, 1001–1008. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Zhang, G.; Zhang, H.; Karin, M.; Bai, H.; Cai, D. Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity. Cell 2008, 135, 61–73. [Google Scholar] [CrossRef] [Green Version]
- Meng, Q.; Cai, D. Defective hypothalamic autophagy directs the central pathogenesis of obesity via the IkappaB kinase beta (IKKbeta)/NF-kappaB pathway. J. Biol. Chem. 2011, 286, 32324–32332. [Google Scholar] [CrossRef] [Green Version]
- Kurata, S.; Matsumoto, M.; Tsuji, Y.; Nakajima, H. Lipopolysaccharide activates transcription of the heme oxygenase gene in mouse M1 cells through oxidative activation of nuclear factor kappa B. Eur. J. Biochem. 1996, 239, 566–571. [Google Scholar] [CrossRef]
- Tornatore, L.; Thotakura, A.K.; Bennett, J.; Moretti, M.; Franzoso, G. The nuclear factor kappa B signaling pathway: Integrating metabolism with inflammation. Trends Cell. Biol. 2012, 22, 557–566. [Google Scholar] [CrossRef] [PubMed]
- Mauro, C.; Leow, S.C.; Anso, E.; Rocha, S.; Thotakura, A.K.; Tornatore, L.; Moretti, M.; De Smaele, E.; Beg, A.A.; Tergaonkar, V.; et al. NF-κB controls energy homeostasis and metabolic adaptation by upregulating mitochondrial respiration. Nat. Cell Biol. 2011, 13, 1272–1279. [Google Scholar] [CrossRef] [PubMed]
- Wakabayashi, N.; Slocum, S.L.; Skoko, J.J.; Shin, S.; Kensler, T.W. When NRF2 talks, who’s listening? Antioxid. Redox Signal. 2020, 13, 1649–1663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soares, M.P.; Seldon, M.P.; Gregoire, I.P.; Vassilevskaia, T.; Berberat, P.O.; Yu, J.; Tsui, T.Y.; Bach, F.H. Hemeoxygenase-1 modulates the expression of adhesion molecules associated with endothelial cell activation. J. Immunol. 2004, 172, 3553–3563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yerra, V.G.; Negi, G.; Sharma, S.S.; Kumar, A. Potential therapeutic effects of the simultaneous targeting of the Nrf2 and NF-κB pathways in diabetic neuropathy. Redox Biol. 2013, 1, 394–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thimmulappa, R.K.; Lee, H.; Rangasamy, T.; Reddy, S.P.; Yamamoto, M.; Kensler, T.W.; Biswal, S. Nrf2 is a critical regulator of the innate immune response and survival during experimental sepsis. J. Clin. Investig. 2006, 116, 984–995. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.G.; Zhang, Y.Q.; Wu, Z.Z.; Hsieh, C.W.; Chu, C.S.; Wung, B.S. Peanut arachidin-1 enhances Nrf2-mediated protective mechanisms against TNF-α-induced ICAM-1 expression and NF-_B activation in endothelial cells. Int. J. Mol. Med. 2018, 41, 541–547. [Google Scholar]
- Bellezza, I.; Tucci, A.; Galli, F.; Grottelli, S.; Mierla, A.L.; Pilolli, F.; Minelli, A. Inhibition of NF-κB nuclear translocation via HO-1 activation underlies α-tocopheryl succinate toxicity. J. Nutr. Biochem. 2012, 23, 1583–1591. [Google Scholar] [CrossRef]
- Rushworth, S.A.; Shah, S.; MacEwan, D.J. TNF mediates the sustained activation of Nrf2 in human monocytes. J. Immunol. 2011, 187, 702–707. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, E.H.; Suzuki, T.; Funayama, R.; Nagashima, T.; Hayashi, M.; Sekine, H.; Tanaka, N.; Moriguchi, T.; Motohashi, H.; Nakayama, K.; et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat. Commun. 2016, 7, 11624. [Google Scholar] [CrossRef]
- Kim, S.W.; Lee, H.K.; Shin, J.H.; Lee, J.K. Up-down regulation of HO-1 and iNOS gene expressions by ethyl pyruvate via recruiting p300 to Nrf2 and depriving It from p65. Free Radic Biol Med. 2013, 65, 468–476. [Google Scholar] [CrossRef] [PubMed]
- Liu, G.H.; Qu, J.; Shen, X. NF-kappaB/p65 antagonizes Nrf2-ARE pathway by depriving CBP from Nrf2 and facilitating recruitment of HDAC3 to MafK. Biochim. Biophys. Acta 2008, 1783, 713–727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, D.F.; Kuo, H.P.; Liu, M.; Chou, C.K.; Xia, W.; Du, Y.; Shen, J.; Chen, C.T.; Huo, L.; Hsu, M.C.; et al. KEAP1 E3 ligase-mediated downregulation of NF-kappaB signaling by targeting IKKbeta. Mol. Cell 2009, 36, 131–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Banning, A.; Brigelius-Flohé, R. NF-kappaB, Nrf2, and HO-1 interplay in redox-regulated VCAM-1 expression. Antioxid. Redox Signal. 2005, 7, 889–899. [Google Scholar] [CrossRef]
- Brigelius-Flohé, R.; Flohé, L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid. Redox Signal. 2011, 15, 2335–2381. [Google Scholar] [CrossRef] [Green Version]
- Chuang, H.C.; Chang, C.W.; Chang, G.D.; Yao, T.P.; Chen, H. Histone deacetylase 3 binds to and regulates the GCMa transcription factor. Nucl. Acids Res. 2006, 34, 1459. [Google Scholar] [CrossRef] [Green Version]
- Hung, H.L.; Kim, A.Y.; Hong, W.; Rakowski, C.; Blobel, G.A. Stimulation of NF-E2 DNA binding by CREB-binding protein (CBP)-mediated acetylation. J. Biol. Chem. 2001, 276, 10715. [Google Scholar] [CrossRef] [Green Version]
- Ahmed, S.M.U.; Luo, L.; Namani, A.; Wang, X.J.; Tang, X. Nrf2 signaling pathway: Pivotal roles in inflammation. Biochim. Biophys. Acta 2017, 1863, 585–597. [Google Scholar] [CrossRef]
- Yang, H.; Magilnick, N.; Ou, X.; Lu, S.C. Tumour necrosis factor α induces co-ordinated activation of rat GSH synthetic enzymes via nuclear factor κB and activator protein-1. Biochem. J. 2005, 391, 399–408. [Google Scholar] [CrossRef] [Green Version]
- Rushworth, S.A.; Zaitseva, L.; Murray, M.Y.; Shah, N.M.; Bowles, K.M.; MacEwan, D.J. The high Nrf2 expression in human acute myeloid leukaemia is driven by NF-kappaB and underlies its chemo-resistance. Blood 2012, 120, 5188–5198. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.; Lin, Z.; Yan, Z.; Wang, Z.; Fu, X.; Yu, K. Sinomenine contributes to the inhibition of the inflammatory response and the improvement of osteoarthritis in mouse-cartilage cells by acting on the Nrf2/HO-1 and NF-κB signaling pathways. Int. Immunopharmacol. 2019, 75, 105715. [Google Scholar] [CrossRef] [PubMed]
- Lee, W.; Yang, S.; Lee, C.; Park, E.K.; Kim, K.M.; Ku, S.K.; Bae, J.S. Aloin reduces inflammatory gene iNOS via inhibition activity and p-STAT-1 and NF-κB. Food Chem. Toxicol. 2019, 126, 67–71. [Google Scholar] [CrossRef]
- Wang, J.; Chen, G.; Shi, T.; Wang, Y.; Guan, C. Possible treatment for cutaneous lichen planus: An in vitro anti-inflammatory role of Angelica polysaccharide in human keratinocytes HaCaT. Int. J. Immunopathol. Pharmacol. 2019, 33, 2058738418821837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, G.; Park, J.S.; Lee, E.J.; Ahn, J.H.; Kim, H.S. Anti-inflammatory and antioxidant mechanisms of urolithin B in activated microglia. Phytomedicine 2019, 55, 50–57. [Google Scholar] [CrossRef] [PubMed]
- Zhao, D.R.; Jiang, Y.S.; Sun, J.Y.; Li, H.H.; Luo, X.L.; Zhao, M.M. Anti-inflammatory Mechanism Involved in 4-Ethylguaiacol-Mediated Inhibition of LPS-Induced Inflammation in THP-1 Cells. J Agric. Food Chem. 2019, 67, 1230–1243. [Google Scholar] [CrossRef] [PubMed]
- Ren, J.; Li, L.; Wang, Y.; Zhai, J.; Chen, G.; Hu, K. Gambogic acid induces heme oxygenase-1 through Nrf2 signaling pathway and inhibits NF-κB and MAPK activation to reduce inflammation in LPS-activated RAW264.7 cells. Biomed. Pharmacother. 2019, 109, 555–562. [Google Scholar] [CrossRef] [PubMed]
- Gęgotek, A.; Ambrożewicz, E.; Jastrząb, A.; Jarocka-Karpowicz, I.; Skrzydlewska, E. Rutin and ascorbic acid cooperation in antioxidant and antiapoptotic effect on human skin keratinocytes and fibroblasts exposed to UVA and UVB radiation. Arch. Dermatol. Res. 2019, 311, 203–219. [Google Scholar] [CrossRef] [Green Version]
- Luo, Z.; Zheng, B.; Jiang, B.; Xue, X.; Xue, E.; Zhou, Y. Peiminine inhibits the IL-1β induced inflammatory response in mouse articular chondrocytes and ameliorates murine osteoarthritis. Food Funct. 2019, 10, 2198–2208. [Google Scholar] [CrossRef]
- Muhammad, T.; Ikram, M.; Ullah, R.; Rehman, S.U.; Kim, M.O. Hesperetin, a Citrus Flavonoid, Attenuates LPS-Induced Neuroinflammation, Apoptosis and Memory Impairments by Modulating TLR4/NF-κB Signaling. Nutrients 2019, 11, 648. [Google Scholar] [CrossRef] [Green Version]
- Jia, Y.N.; Peng, Y.L.; Zhao, Y.P.; Cheng, X.F.; Zhou, Y.; Chai, C.L.; Zeng, L.S.; Pan, M.H.; Xu, L. Comparison of the Hepatoprotective Effects of the Three Main Stilbenes from Mulberry Twigs. J Agric. Food Chem. 2019, 67, 5521–5529. [Google Scholar] [CrossRef]
- Zhang, H.F.; Wang, J.H.; Wang, Y.L.; Gao, C.; Gu, Y.T.; Huang, J.; Wang, J.H.; Zhang, Z. Salvianolic Acid A Protects the Kidney against Oxidative Stress by Activating the Akt/GSK-3β/Nrf2 Signaling Pathway and Inhibiting the NF-κB Signaling Pathway in 5/6 Nephrectomized Rats. Oxid. Med. Cell Longev. 2019, 2019, 2853534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abdel-Magied, N.; Shedid, S.M. The effect of naringenin on the role of nuclear factor (erythroid-derived 2)-like2 (Nrf2) and haem oxygenase 1 (HO-1) in reducing the risk of oxidative stress-related radiotoxicity in the spleen of rats. Environ. Toxicol. 2019, 34, 788–795. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.W.; Zhang, X.L.; Wu, Q.H.; Jin, Y.B.; Ning, C.T.; Wang, R.; Mao, J.X.; Chen, M. The hepatoprotective effects of Sedum sarmentosum extract and its isolated major constituent through Nrf2 activation and NF-κB inhibition. Phytomedicine 2019, 53, 263–273. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Xia, Z.; Yu, D.; Wang, J.; Jin, L.; Huang, D.; Ye, X.; Li, X.; Zhang, B. Hepatoprotective effects and structure-activity relationship of five flavonoids against lipopolysaccharide/d-galactosamine induced acute liver failure in mice. Int. Immunopharmacol. 2019, 68, 171–178. [Google Scholar] [CrossRef]
- Li, Z.; Feng, H.; Wang, Y.; Shen, B.; Tian, Y.; Wu, L.; Zhang, Q.; Jin, M.; Liu, G. Rosmarinic acid protects mice from lipopolysaccharide/d-galactosamine-induced acute liver injury by inhibiting MAPKs/NF-κB and activating Nrf2/HO-1 signaling pathways. Int. Immunopharmacol. 2019, 67, 465–472. [Google Scholar] [CrossRef]
- Liu, T.G.; Sha, K.H.; Zhang, L.G.; Liu, X.X.; Yang, F.; Cheng, J.Y. Protective effects of alpinetin on lipopolysaccharide/d-Galactosamine-induced liver injury through inhibiting inflammatory and oxidative responses. Microb. Pathog. 2019, 126, 239–244. [Google Scholar] [CrossRef]
- Tang, F.; Fan, K.; Wang, K.; Bian, C. Amygdalin attenuates acute liver injury induced by D-galactosamine and lipopolysaccharide by regulating the NLRP3, NF-κB and Nrf2/NQO1 signalling pathways. Biomed. Pharmacother. 2019, 111, 527–536. [Google Scholar] [CrossRef]
- Li, Q.; Tian, Z.; Wang, M.; Kou, J.; Wang, C.; Rong, X.; Li, J.; Xie, X.; Pang, X. Luteoloside attenuates neuroinflammation in focal cerebral ischemia in rats via regulation of the PPARγ/Nrf2/NF-κB signaling pathway. Int. Immunopharmacol. 2019, 66, 309–316. [Google Scholar] [CrossRef] [PubMed]
- Bian, X.; Liu, X.; Liu, J.; Zhao, Y.; Li, H.; Zhang, L.; Li, P.; Gao, Y. Hepatoprotective effect of chiisanoside from Acanthopanax sessiliflorus against LPS/D-GalN-induced acute liver injury by inhibiting NF-κB and activating Nrf2/HO-1 signaling pathways. J. Sci. Food Agric. 2019, 99, 3283–3290. [Google Scholar] [CrossRef]
- Ye, J.; Guan, M.; Lu, Y.; Zhang, D.; Li, C.; Zhou, C. Arbutin attenuates LPS-induced lung injury via Sirt1/ Nrf2/ NF-κBp65 pathway. Pulm. Pharmacol. Ther. 2019, 54, 53–59. [Google Scholar] [CrossRef]
- Ding, H.; Ci, X.; Cheng, H.; Yu, Q.; Li, D. Chicoric acid alleviates lipopolysaccharide-induced acute lung injury in mice through anti-inflammatory and anti-oxidant activities. Int. Immunopharmacol. 2019, 66, 169–176. [Google Scholar] [CrossRef] [PubMed]
- Alam, J.; Stewart, D.; Touchard, C.; Boinapally, S.; Choi, A.M.; Cook, J.L. Nrf2, a Cap’n’Collar transcription factor, regulates induction of the heme oxygenase-1 gene. J. Biol. Chem. 1999, 274, 26071–26078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lavrovsky, Y.; Schwartzman, M.L.; Levere, R.D.; Kappas, A.; Abraham, N.G. Identification of binding sites for transcription factors NF-kappa B and AP-2 in the promoter region of the human heme oxygenase 1 gene. Proc. Natl. Acad. Sci. USA 1994, 91, 5987–5991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mulcahy, R.T.; Wartman, M.A.; Bailey, H.H.; Gipp, J.J. Constitutive and beta-naphthoflavone-induced expression of the human gamma-glutamylcysteine synthetase heavy subunit gene is regulated by a distal antioxidant response element = TRE sequence. J. Biol. Chem. 1997, 272, 7445–7454. [Google Scholar] [CrossRef] [Green Version]
- Kimura, T.; Kawasaki, Y.; Okumura, F.; Sone, T.; Natsuki, R.; Isobe, M. Ethanol-induced expression of glutamate-cysteine ligase catalytic subunit gene is mediated by NF-kappaB. Toxicol. Lett. 2009, 185, 110–115. [Google Scholar] [CrossRef]
- Muri, J.; Kopf, M. Redox regulation of immunometabolism. Nat. Rev. Immunol. 2020, in press. [Google Scholar] [CrossRef] [PubMed]
- Kairisalo, M.; Korhonen, L.; Blomgren, K.; Lindholm, D. X-linked inhibitor of apoptosis protein increases mitochondrial antioxidants through NF-kappaB activation. Biochem. Biophys. Res. Commun. 2007, 364, 138–144. [Google Scholar] [CrossRef]
- Djavaheri-Mergny, M.; Javelaud, D.; Wietzerbin, J.; Besançon, F. NF-kappaB activation prevents apoptotic oxidative stress via an increase of both thioredoxin and MnSOD levels in TNFalpha-treated Ewing sarcoma cells. FEBS Lett. 2004, 578, 111–115. [Google Scholar] [CrossRef] [Green Version]
- Anrather, J.; Racchumi, G.; Iadecola, C. NF-kappaB regulates phagocytic NADPH oxidase by inducing the expression of gp91phox. J. Biol. Chem. 2006, 281, 5657–5667. [Google Scholar] [CrossRef] [Green Version]
- Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moldogazieva, N.T.; Mokhosoev, I.M.; Feldman, N.B.; Lutsenko, S.V. ROS and RNS signalling: Adaptive redox switches through oxidative/nitrosative protein modifications. Free Radic. Res. 2018, 52, 507–543. [Google Scholar] [CrossRef] [PubMed]
- Ikeda, T.; Honjo, K.; Hirota, Y.; Onodera, T. Isolation of the chicken NF-kappa B p65 subunit-encoding cDNA and characterization of its products. Gene 1993, 133, 237–242. [Google Scholar] [PubMed]
- Ikeda, T.; Hirota, Y.; Onodera, T. Isolation of a cDNA encoding the chicken p50B/p97 (Lyt-10) transcription factor. Gene 1994, 138, 193–196. [Google Scholar] [PubMed]
- Krishnan, V.A.; Schatzle, J.D.; Hinojos, C.M.; Bose, H.R., Jr. Structure and regulation of the gene encoding avian inhibitor of nuclear factor kappa B-alpha. Gene 1995, 166, 261–266. [Google Scholar] [CrossRef]
- Van Phi, L. Transcriptional activation of the chicken lysozyme gene by NF-kappa Bp65 (RelA) and c-Rel, but not by NF-kappa Bp50. Biochem. J. 1996, 313, 39–44. [Google Scholar] [CrossRef] [PubMed]
- Piffat, K.A.; Hrdlicková, R.; Nehyba, J.; Ikeda, T.; Liss, A.; Huang, S.; Sif, S.; Gilmore, T.D.; Bose, H.R., Jr. The chicken RelB transcription factor has transactivation sequences and a tissue-specific expression pattern that are distinct from mammalian RelB. Mol. Cell. Biol. Res. Commun. 2001, 4, 266–275. [Google Scholar] [CrossRef]
- Qiu, Y.; Shen, Y.; Li, X.; Ding, C.; Ma, Z. Molecular cloning and functional characterization of a novel isoform of chicken myeloid differentiation factor 88 (MyD88). Dev. Comp. Immunol. 2008, 32, 1522–1530. [Google Scholar] [CrossRef]
- Zhou, P.; Zeng, Y.; Rao, Z.; Li, Y.; Zheng, H.; Luo, R. Molecular characterization and functional analysis of duck IKKα. Dev. Comp. Immunol. 2021, 115, 103880. [Google Scholar] [CrossRef]
- Kim, Y.; Tian, M. NF-kappaB family of transcription factor facilitates gene conversion in chicken B cells. Mol. Immunol. 2009, 46, 3283–3291. [Google Scholar] [CrossRef]
- Chen, K.; Luo, Z.; Zheng, S.J. Gallus Heat shock cognate protein 70, a novel binding partner of Apoptin. Virol. J. 2011, 8, 324. [Google Scholar] [CrossRef] [Green Version]
- Kogut, M.H.; He, H.; Genovese, K.J. Bacterial toll-like receptor agonists induce sequential NF-κB-mediated leukotriene B4 and prostaglandin E2 production in chicken heterophils. Vet. Immunol. Immunopathol. 2012, 145, 159–170. [Google Scholar] [CrossRef]
- Shinohara, H.; Behar, M.; Inoue, K.; Hiroshima, M.; Yasuda, T.; Nagashima, T.; Kimura, S.; Sanjo, H.; Maeda, S.; Yumoto, N.; et al. Positive feedback within a kinase signaling complex functions as a switch mechanism for NF-κB activation. Science 2014, 344, 760–764. [Google Scholar] [CrossRef] [PubMed]
- Tang, X.; Zhang, L.; Wei, W. Roles of TRAFs in NF-κB signaling pathways mediated by BAFF. Immunol. Lett. 2018, 196, 113–118. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.L.; Feng, Z.Q.; Zeng, S.Q.; Li, S.M.; Zhu, Q.; Liu, Y.P. Molecular cloning and expression analysis of TRAF3 in chicken. Genet. Mol. Res. 2015, 14, 4408–4419. [Google Scholar] [CrossRef] [PubMed]
- Zhai, Y.; Luo, F.; Chen, Y.; Zhou, S.; Li, Z.; Liu, M.; Bi, D.; Jin, H. Molecular characterization and functional analysis of duck TRAF6. Dev. Comp. Immunol. 2015, 49, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Xu, Y.; Kang, X.; Meng, C.; Gu, D.; Zhou, Y.; Xiong, D.; Geng, S.; Jiao, X.; Pan, Z. Molecular cloning and functional analysis of TRAF6 from Yangzhou great white goose Anser anser. Dev. Comp. Immunol. 2019, 101, 103435. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Xu, Y.; Xiong, D.; Zhou, Y.; Kang, X.; Meng, C.; Gu, D.; Jiao, X.; Pan, Z. Molecular characterisation, expression and functional feature of TRAF6 in the King pigeon (Columba livia). Innate Immun. 2020, 26, 490–504. [Google Scholar] [CrossRef]
- Zhou, Y.; Kang, X.; Xiong, D.; Zhu, S.; Zheng, H.; Xu, Y.; Guo, Y.; Pan, Z.; Jiao, X. Molecular and functional characterization of pigeon (Columba livia) tumor necrosis factor receptor-associated factor 3. Dev. Comp. Immunol. 2017, 69, 51–59. [Google Scholar] [CrossRef]
- Kang, Y.; Nii, T.; Isobe, N.; Yoshimura, Y. Effects of TLR Ligands on the Expression of Cytokines and Possible Role of NFκB in its Process in the Theca of Chicken Follicles. J. Poult. Sci. 2018, 55, 288–300. [Google Scholar] [CrossRef]
- Chen, S.; Cheng, A.; Wang, M. Innate sensing of viruses by pattern recognition receptors in birds. Vet. Res. 2013, 44, 82. [Google Scholar] [CrossRef] [Green Version]
- Downing, T.; Lloyd, A.T.; O’Farrelly, C.; Bradley, D.G. The differential evolutionary dynamics of avian cytokine and TLR gene classes. J. Immunol. 2010, 184, 6993–7000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gillespie, M.; Shamovsky, V.; D’Eustachio, P. Human and chicken TLR pathways: Manual curation and computer-based orthology analysis. Mamm. Genome 2011, 22, 130–138. [Google Scholar] [CrossRef] [Green Version]
- Farnell, M.B.; Crippen, T.L.; He, H.; Swaggerty, C.L.; Kogut, M.H. Oxidative burst mediated by toll like receptors (TLR) and CD14 on avian heterophils stimulated with bacterial toll agonists. Dev. Comp. Immunol. 2003, 27, 423–429. [Google Scholar] [CrossRef]
- Kannaki, T.R.; Reddy, M.R.; Verma, P.C.; Shanmugam, M. Differential Toll-like receptor (TLR) mRNA expression patterns during chicken embryological development. Anim. Biotechnol. 2015, 26, 130–135. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Jiang, Y.; Yin, Q.; Liang, H.; She, R. Chicken intestine defensins activated murine peripheral blood mononuclear cells through the TLR4-NF-kappaB pathway. Vet. Immunol. Immunopathol. 2010, 133, 59–65. [Google Scholar] [CrossRef]
- Yu, H.; Lu, Y.; Qiao, X.; Wei, L.; Fu, T.; Cai, S.; Wang, C.; Liu, X.; Zhong, S.; Wang, Y. Novel Cathelicidins from Pigeon Highlights Evolutionary Convergence in Avain Cathelicidins and Functions in Modulation of Innate Immunity. Sci. Rep. 2015, 5, 11082. [Google Scholar] [CrossRef] [Green Version]
- Ishige, T.; Hara, H.; Hirano, T.; Kono, T.; Hanzawa, K. Basic characterization of avian NK-lysin (NKL) from the Japanese quail, Coturnix japonica. Anim. Sci. J. 2014, 85, 90–95. [Google Scholar] [CrossRef] [PubMed]
- Kamimura, T.; Isobe, N.; Yoshimura, Y. Effects of inhibitors of transcription factors, nuclear factor-κB and activator protein 1, on the expression of proinflammatory cytokines and chemokines induced by stimulation with Toll-like receptor ligands in hen vaginal cells. Poult. Sci. 2017, 96, 723–730. [Google Scholar] [CrossRef]
- Yong, Y.H.; Liu, S.F.; Hua, G.H.; Jia, R.M.; Gooneratne, R.; Zhao, Y.T.; Liao, M.; Ju, X.H. Goose toll-like receptor 3 (TLR3) mediated IFN-γ and IL-6 in anti-H5N1 avian influenza virus response. Vet. Immunol. Immunopathol. 2018, 197, 31–38. [Google Scholar] [CrossRef] [PubMed]
- Gao, M.; Guo, Y.; Du, J.; Song, Z.; Luo, X.; Wang, J.; Han, W. Evolutional conservation of molecular structure and antiviral function of a type I interferon, IFN-kappa, in poultry. Dev. Comp. Immunol. 2018, 89, 44–53. [Google Scholar] [CrossRef]
- Jia, H.; Li, G.; Li, J.; Tian, Y.; Wang, D.; Shen, J.; Tao, Z.; Xu, J.; Lu, L. Cloning, expression and bioinformatics analysis of the duck TLR 4 gene. Br. Poult. Sci. 2012, 53, 190–197. [Google Scholar] [CrossRef]
- Fang, Q.; Pan, Z.; Geng, S.; Kang, X.; Huang, J.; Sun, X.; Li, Q.; Cai, Y.; Jiao, X. Molecular cloning, characterization and expression of goose Toll-like receptor 5. Mol. Immunol. 2012, 52, 117–124. [Google Scholar] [CrossRef] [PubMed]
- Yong, Y.; Liu, S.; Hua, G.; Jia, R.; Zhao, Y.; Sun, X.; Liao, M.; Ju, X. Identification and functional characterization of Toll-like receptor 2-1 in geese. BMC Vet. Res. 2015, 11, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiong, D.; Song, L.; Pan, Z.; Jiao, X. Molecular cloning, characterization, and functional analysis of pigeon (Columba livia) Toll-like receptor 5. Poult. Sci. 2018, 97, 4031–4039. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Wang, Y.; Zhao, H.; Shao, Y.; Liu, J.; Xing, M. Characterization, functional and signaling elucidation of pigeon (Columba livia) interferon-α: Knockdown p53 negatively modulates antiviral response. Dev. Comp. Immunol. 2019, 90, 29–40. [Google Scholar] [CrossRef] [PubMed]
- Xiong, D.; Song, L.; Pan, Z.; Chen, X.; Geng, S.; Jiao, X. Identification and immune functional characterization of pigeon TLR7. Int. J. Mol. Sci. 2015, 16, 8364–8381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tao, Z.Y.; Zhu, C.H.; Shi, Z.H.; Song, C.; Xu, W.J.; Song, W.T.; Zou, J.M.; Qin, A.J. Molecular characterization, expression, and functional analysis of NOD1 in Qingyuan partridge chicken. Genet. Mol. Res. 2015, 14, 2691–2701. [Google Scholar] [CrossRef]
- Li, H.; Jin, H.; Li, Y.; Liu, D.; Foda, M.F.; Jiang, Y.; Luo, R. Molecular cloning and functional characterization of duck nucleotide-binding oligomerization domain 1 (NOD1). Dev. Comp. Immunol. 2017, 74, 82–89. [Google Scholar] [CrossRef]
- Truong, A.D.; Hong, Y.; Hoang, C.T.; Lee, J.; Hong, Y.H. Chicken IL-26 regulates immune responses through the JAK/STAT and NF-κB signaling pathways. Dev. Comp. Immunol. 2017, 73, 10–20. [Google Scholar] [CrossRef]
- Truong, A.D.; Hong, Y.; Rengaraj, D.; Lee, J.; Lee, K.; Hong, Y.H. Identification and functional characterization, including cytokine production modulation, of the novel chicken Interleukin-11. Dev. Comp. Immunol. 2018, 87, 51–63. [Google Scholar] [CrossRef]
- Hoang, C.T.; Hong, Y.; Truong, A.D.; Lee, J.; Lee, K.; Hong, Y.H. Molecular cloning of chicken interleukin-17B, which induces proinflammatory cytokines through activation of the NF-κB signaling pathway. Dev. Comp. Immunol. 2017, 74, 40–48. [Google Scholar] [CrossRef]
- Rohde, F.; Schusser, B.; Hron, T.; Farkašová, H.; Plachý, J.; Härtle, S.; Hejnar, J.; Elleder, D.; Kaspers, B. Characterization of Chicken Tumor Necrosis Factor-α, a Long Missed Cytokine in Birds. Front. Immunol. 2018, 9, 605. [Google Scholar] [CrossRef] [PubMed]
- Ji, G.G.; Shu, J.T.; Zhang, M.; Ju, X.J.; Shan, Y.J.; Liu, Y.F.; Tu, Y.J. Transcriptional regulatory region and DNA methylation analysis of TNNI1 gene promoters in Gaoyou duck skeletal muscle (Anas platyrhynchos domestica). Br. Poult. Sci. 2019, 60, 202–208. [Google Scholar] [CrossRef] [PubMed]
- Barjesteh, N.; Taha-Abdelaziz, K.; Kulkarni, R.R.; Sharif, S. Innate antiviral responses are induced by TLR3 and TLR4 ligands in chicken tracheal epithelial cells: Communication between epithelial cells and macrophages. Virology 2019, 534, 132–142. [Google Scholar] [CrossRef] [PubMed]
- Sutton, K.M.; Hu, T.; Wu, Z.; Siklodi, B.; Vervelde, L.; Kaiser, P. The functions of the avian receptor activator of NF-κB ligand (RANKL) and its receptors, RANK and osteoprotegerin, are evolutionarily conserved. Dev. Comp. Immunol. 2015, 51, 170–184. [Google Scholar] [CrossRef] [Green Version]
- Truong, A.D.; Hong, Y.; Nguyen, H.T.; Nguyen, C.T.; Chu, N.T.; Tran, H.T.T.; Dang, H.V.; Lillehoj, H.S.; Hong, Y.H. Molecular identification and characterisation of a novel chicken leukocyte immunoglobulin-like receptor A5. Br. Poult. Sci. 2020, in press. [Google Scholar]
- Paital, B.; Panda, S.K.; Hati, A.K.; Mohanty, B.; Mohapatra, M.K.; Kanungo, S.; Chainy, G.B. Longevity of animals under reactive oxygen species stress and disease susceptibility due to global warming. World J. Biol. Chem. 2016, 7, 110–127. [Google Scholar] [CrossRef]
- Vandana, G.D.; Sejian, V.; Lees, A.M.; Pragna, P.; Silpa, M.V.; Maloney, S.K. Heat stress and poultry production: Impact and amelioration. Int. J. Biometeorol. 2020, in press. [Google Scholar] [CrossRef]
- Wasti, S.; Sah, N.; Mishra, B. Impact of Heat Stress on Poultry Health and Performances, and Potential Mitigation Strategies. Animals 2020, 10, 1266. [Google Scholar] [CrossRef]
- Pu, S.; Usuda, K.; Nagaoka, K.; Gore, A.; Crews, D.; Watanabe, G. The relation between liver damage and reproduction in female Japanese quail (Coturnix japonica) exposed to high ambient temperature. Poult. Sci. 2020, 99, 4586–4597. [Google Scholar] [CrossRef]
- Nawab, A.; Li, G.; An, L.; Wu, J.; Chao, L.; Xiao, M.; Zhao, Y.; Birmani, M.W.; Ghani, M.W. Effect of curcumin supplementation on TLR4 mediated non-specific immune responses in liver of laying hens under high-temperature conditions. J. Therm. Biol. 2019, 84, 384–397. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Fu, C.; Yan, M.; Xie, H.; Li, S.; Yu, Q.; He, S.; He, J. Resveratrol modulates intestinal morphology and HSP70/90, NF-κB and EGF expression in the jejunal mucosa of black-boned chickens on exposure to circular heat stress. Food Funct. 2016, 7, 1329–1338. [Google Scholar] [CrossRef] [PubMed]
- Hangalapura, B.N.; Kaiser, M.G.; Poel, J.J.; Parmentier, H.K.; Lamont, S.J. Cold stress equally enhances in vivo pro-inflammatory cytokine gene expression in chicken lines divergently selected for antibody responses. Dev. Comp. Immunol. 2006, 30, 503–511. [Google Scholar] [CrossRef] [PubMed]
- Zhao, F.Q.; Zhang, Z.W.; Wang, C.; Zhang, B.; Yao, H.D.; Li, S.; Xu, S.W. The role of heat shock proteins in inflammatory injury induced by cold stress in chicken hearts. Cell Stress Chaperones 2013, 18, 773–783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, J.; Liu, C.P.; Zhang, Z.W.; Xing, M.W.; Xu, S.W. Influence of inflammatory pathway markers on oxidative stress induced by cold stress in intestine of quails. Res. Vet. Sci. 2013, 95, 495–501. [Google Scholar] [CrossRef] [PubMed]
- Ren, J.; Liu, C.; Zhao, D.; Fu, J. The role of heat shock protein 70 in oxidant stress and inflammatory injury in quail spleen induced by cold stress. Environ. Sci. Pollut. Res. Int. 2018, 25, 21011–21023. [Google Scholar] [CrossRef]
- Wei, H.; Zhang, R.; Su, Y.; Bi, Y.; Li, X.; Zhang, X.; Li, J.; Bao, J. Effects of Acute Cold Stress After Long-Term Cold Stimulation on Antioxidant Status, Heat Shock Proteins, Inflammation and Immune Cytokines in Broiler Heart. Front. Physiol. 2018, 9, 1589. [Google Scholar] [CrossRef] [Green Version]
- Surai, P.F.; Dvorska, J.E. Effects of Mycotoxins on Antioxidant Status and Immunity. In The Mycotoxin Blue Book; Diaz, D.E., Ed.; Nottingham University Press: Nottingham, UK, 2005; pp. 93–137. [Google Scholar]
- Gao, Y.; Meng, L.; Liu, H.; Wang, J.; Zheng, N. The Compromised Intestinal Barrier Induced by Mycotoxins. Toxins 2020, 12, 619. [Google Scholar] [CrossRef]
- Li, Y.; Ma, Q.G.; Zhao, L.H.; Wei, H.; Duan, G.X.; Zhang, J.Y.; Ji, C. Effects of lipoic acid on immune function, the antioxidant defense system, and inflammation-related genes expression of broiler chickens fed aflatoxin contaminated diets. Int. J. Mol. Sci. 2014, 15, 5649–5662. [Google Scholar] [CrossRef] [Green Version]
- Ma, Q.; Li, Y.; Fan, Y.; Zhao, L.; Wei, H.; Ji, C.; Zhang, J. Molecular Mechanisms of Lipoic Acid Protection against Aflatoxin B₁-Induced Liver Oxidative Damage and Inflammatory Responses in Broilers. Toxins 2015, 7, 5435–5447. [Google Scholar] [CrossRef] [Green Version]
- Rajput, S.A.; Sun, L.; Zhang, N.Y.; Khalil, M.M.; Ling, Z.; Chong, L.; Wang, S.; Rajput, I.R.; Bloch, D.M.; Khan, F.A.; et al. Grape Seed Proanthocyanidin Extract Alleviates AflatoxinB₁-Induced Immunotoxicity and Oxidative Stress via Modulation of NF-κB and Nrf2 Signaling Pathways in Broilers. Toxins 2019, 11, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Du, Y.; Zhu, Y.; Teng, X.; Zhang, K.; Teng, X.; Li, S. Toxicological Effect of Manganese on NF-κB/iNOS-COX-2 Signaling Pathway in Chicken Testes. Biol. Trace Elem. Res. 2015, 168, 227–234. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Zhao, H.; Wang, Y.; Shao, Y.; Li, J.; Liu, J.; Xing, M. The inflammatory responses in Cu-mediated elemental imbalance is associated with mitochondrial fission and intrinsic apoptosis in Gallus gallus heart. Chemosphere 2017, 189, 489–497. [Google Scholar] [CrossRef] [PubMed]
- Yang, F.; Liao, J.; Yu, W.; Pei, R.; Qiao, N.; Han, Q.; Hu, L.; Li, Y.; Guo, J.; Pan, J.; et al. Copper induces oxidative stress with triggered NF-κB pathway leading to inflammatory responses in immune organs of chicken. Ecotoxicol. Environ. Saf. 2020, 200, 110715. [Google Scholar] [CrossRef] [PubMed]
- Zhang, K.; Zhao, P.; Guo, G.; Guo, Y.; Tian, L.; Sun, X.; Li, S.; He, Y.; Sun, Y.; Chai, H.; et al. Arsenic Trioxide Attenuates NF-κB and Cytokine mRNA Levels in the Livers of Cocks. Biol. Trace Elem. Res. 2016, 170, 432–437. [Google Scholar] [CrossRef] [PubMed]
- Li, S.W.; Guo, Y.; He, Y.; Sun, X.; Zhao, H.J.; Wang, Y.; Wang, Y.J.; Xing, M.W. Assessment of arsenic trioxide toxicity on cock muscular tissue: Alterations of oxidative damage parameters, inflammatory cytokines and heat shock proteins. Ecotoxicology 2017, 26, 1078–1088. [Google Scholar] [CrossRef]
- Li, S.; Wang, Y.; Zhao, H.; He, Y.; Li, J.; Jiang, G.; Xing, M. NF-κB-mediated inflammation correlates with calcium overload under arsenic trioxide-induced myocardial damage in Gallus gallus. Chemosphere 2017, 185, 618–627. [Google Scholar] [CrossRef]
- Sun, X.; He, Y.; Guo, Y.; Li, S.; Zhao, H.; Wang, Y.; Zhang, J.; Xing, M. Arsenic affects inflammatory cytokine expression in Gallus gallus brain tissues. BMC Vet. Res. 2017, 13, 157. [Google Scholar] [CrossRef] [Green Version]
- Li, S.W.; Sun, X.; He, Y.; Guo, Y.; Zhao, H.J.; Hou, Z.J.; Xing, M.W. Assessment of arsenic trioxide in the heart of Gallus gallus: Alterations of oxidative damage parameters, inflammatory cytokines, and cardiac enzymes. Environ. Sci. Pollut. Res. Int. 2017, 24, 5781–5790. [Google Scholar] [CrossRef]
- Wang, Y.; Zhao, H.; Guo, M.; Shao, Y.; Liu, J.; Jiang, G.; Xing, M. Arsenite renal apoptotic effects in chickens co-aggravated by oxidative stress and inflammatory response. Metallomics 2018, 10, 1805–1813. [Google Scholar] [CrossRef]
- Zhao, H.; Wang, Y.; Shao, Y.; Liu, J.; Wang, S.; Xing, M. Oxidative stress-induced skeletal muscle injury involves in NF-κB/p53-activated immunosuppression and apoptosis response in copper (II) or/and arsenite-exposed chicken. Chemosphere 2018, 210, 76–84. [Google Scholar] [CrossRef]
- Sun, X.; Li, J.; Zhao, H.; Wang, Y.; Liu, J.; Shao, Y.; Xue, Y.; Xing, M. Synergistic effect of copper and arsenic upon oxidative stress, inflammation and autophagy alterations in brain tissues of Gallus gallus. J. Inorg. Biochem. 2018, 178, 54–62. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Zhao, H.; Wang, Y.; Shao, Y.; Zhang, L.; Xing, M. Impacts of simultaneous exposure to arsenic (III) and copper (II) on inflammatory response, immune homeostasis, and heat shock response in chicken thymus. Int. Immunopharmacol. 2018, 64, 60–68. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Zhao, H.; Wang, Y.; Shao, Y.; Li, J.; Xing, M. Alterations of antioxidant indexes and inflammatory cytokine expression aggravated hepatocellular apoptosis through mitochondrial and death receptor-dependent pathways in Gallus gallus exposed to arsenic and copper. Environ. Sci. Pollut. Res. Int. 2018, 25, 15462–15473. [Google Scholar] [CrossRef] [PubMed]
- Guo, M.; Zhao, H.; Wang, Y.; Liu, J.; Fei, D.; Yang, X.; Mu, M.; Xing, M. Elemental imbalance elicited by arsenic and copper exposures leads to oxidative stress and immunotoxicity in chicken gizzard, activating the protective effects of heat shock proteins. Environ. Sci. Pollut. Res. Int. 2019, 26, 36343–36353. [Google Scholar] [CrossRef]
- Liu, J.; Wang, Y.; Zhao, H.; Mu, M.; Guo, M.; Nie, X.; Sun, Y.; Xing, M. Arsenic (III) or/and copper (II) exposure induce immunotoxicity through trigger oxidative stress, inflammation and immune imbalance in the bursa of chicken. Ecotoxicol. Environ. Saf. 2020, 190, 110127. [Google Scholar] [CrossRef]
- Wang, D.; Zong, C.; Cheng, K. Chicken thalamic injury induced by copper (II) or / and arsenite exposure involves oxidative stress and inflammation-induced apoptosis. Ecotoxicol. Environ. Saf. 2020, 197, 110554. [Google Scholar] [CrossRef]
- Nie, X.; Wang, Y.; Zhao, H.; Guo, M.; Liu, Y.; Xing, M. As3+ or/and Cu2+ exposure triggers oxidative stress imbalance, induces inflammatory response and apoptosis in chicken brain. Ecotoxicol. Environ. Saf. 2020, 203, 110993. [Google Scholar] [CrossRef]
- Wang, H.; Li, S.; Teng, X. The antagonistic effect of selenium on lead-induced inflammatory factors and heat shock proteins mRNA expression in chicken livers. Biol. Trace Elem. Res. 2016, 171, 437–444. [Google Scholar] [CrossRef]
- Li, X.; Xing, M.; Chen, M.; Zhao, J.; Fan, R.; Zhao, X.; Cao, C.; Yang, J.; Zhang, Z.; Xu, S. Effects of selenium-lead interaction on the gene expression of inflammatory factors and selenoproteins in chicken neutrophils. Ecotoxicol. Environ. Saf. 2017, 139, 447–453. [Google Scholar] [CrossRef]
- Xie, W.; Ge, M.; Li, G.; Zhang, L.; Tang, Z.; Li, R.; Zhang, R. Astragalus Polysaccharide Protect against Cadmium-Induced Cytotoxicity through the MDA5/NF-κB Pathway in Chicken Peripheral Blood Lymphocytes. Molecules 2017, 22, 1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, M.; Li, X.; Fan, R.; Yang, J.; Jin, X.; Hamid, S.; Xu, S. Cadmium induces BNIP3-dependent autophagy in chicken spleen by modulating miR-33-AMPK axis. Chemosphere 2018, 194, 396–402. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Liu, J.; Chen, R.; Qi, M.; Tao, D.; Xu, S. The Antagonistic Effects of Selenium Yeast (SeY) on Cadmium-Induced Inflammatory Factors and the Heat Shock Protein Expression Levels in Chicken Livers. Biol. Trace Elem. Res. 2020, 198, 260–268. [Google Scholar] [CrossRef] [PubMed]
- Cao, H.; Gao, F.; Xia, B.; Zhang, M.; Liao, Y.; Yang, Z.; Hu, G.; Zhang, C. Alterations in trace element levels and mRNA expression of Hsps and inflammatory cytokines in livers of duck exposed to molybdenum or/and cadmium. Ecotoxicol. Environ. Saf. 2016, 125, 93–101. [Google Scholar] [CrossRef]
- Guo, H.; Deng, H.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wang, X.; Wu, B.; Chen, K. Nickel chloride (NiCl2)-caused inflammatory responses via activation of NF-κB pathway and reduction of anti-inflammatory mediator expression in the kidney. Oncotarget 2015, 6, 28607–28620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, W.; Lv, A.; Li, R.; Tang, Z.; Ma, D.; Huang, X.; Zhang, R.; Ge, M. Agaricus blazei Murill Polysaccharides Protect Against Cadmium-Induced Oxidative Stress and Inflammatory Damage in Chicken Spleens. Biol. Trace Elem. Res. 2018, 184, 247–258. [Google Scholar] [CrossRef]
- Yin, J.; Duan, J.; Cui, Z.; Ren, W.; Li, T.; Yin, Y. Hydrogen peroxide-induced oxidative stress activates NF-κB and Nrf2/Keap1 signals and triggers autophagy in piglets. RSC Advances 2015, 5, 15479–15486. [Google Scholar] [CrossRef]
- Chen, Z.; Xing, T.; Li, J.; Zhang, L.; Jiang, Y.; Gao, F. Hydrogen peroxide-induced oxidative stress impairs redox status and damages aerobic metabolism of breast muscle in broilers. Poult. Sci. 2020, in press. [Google Scholar] [CrossRef]
- Hofmann, T.; Schmucker, S.S.; Bessei, W.; Grashorn, M.; Stefanski, V. Impact of Housing Environment on the Immune System in Chickens: A Review. Animals 2020, 10, 1138. [Google Scholar] [CrossRef]
- Chen, X.; Zhang, L.; Li, J.; Gao, F.; Zhou, G. Hydrogen Peroxide-Induced Change in Meat Quality of the Breast Muscle of Broilers Is Mediated by ROS Generation, Apoptosis, and Autophagy in the NF-κB Signal Pathway. J. Agric. Food Chem. 2017, 65, 3986–3994. [Google Scholar] [CrossRef]
- Chen, X.; Gu, R.; Zhang, L.; Li, J.; Jiang, Y.; Zhou, G.; Gao, F. Induction of nuclear factor-κB signal-mediated apoptosis and autophagy by reactive oxygen species is associated with hydrogen peroxide-impaired growth performance of broilers. Animal 2018, 12, 2561–2570. [Google Scholar] [CrossRef] [PubMed]
- Sies, H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol. 2017, 11, 613–619. [Google Scholar] [CrossRef]
- Shi, Q.; Wang, W.; Chen, M.; Zhang, H.; Xu, S. Ammonia induces Treg/Th1 imbalance with triggered NF-κB pathway leading to chicken respiratory inflammation response. Sci. Total Environ. 2019, 659, 354–362. [Google Scholar] [CrossRef] [PubMed]
- An, Y.; Xing, H.; Zhang, Y.; Jia, P.; Gu, X.; Teng, X. The evaluation of potential immunotoxicity induced by environmental pollutant ammonia in broilers. Poult. Sci. 2019, 98, 3165–3175. [Google Scholar] [CrossRef] [PubMed]
- Han, Q.; Tong, J.; Sun, Q.; Teng, X.; Zhang, H.; Teng, X. The involvement of miR-6615-5p/Smad7 axis and immune imbalance in ammonia-caused inflammatory injury via NF-κB pathway in broiler kidneys. Poult. Sci. 2020, 99, 5378–5388. [Google Scholar] [CrossRef]
- Shah, S.; Ishfaq, M.; Nasrullah, M.; Qayum, A.; Akhtar, M.U.; Jo, H.; Hussain, M.; Teng, X. Ammonia inhalation-induced inflammation and structural impairment in the bursa of fabricius and thymus of broilers through NF-κB signaling pathway. Environ. Sci. Pollut. Res. Int. 2020, 27, 11596–11607. [Google Scholar] [CrossRef]
- Chen, D.; Ning, F.; Zhang, J.; Tang, Y.; Teng, X. NF-κB pathway took part in the development of apoptosis mediated by miR-15a and oxidative stress via mitochondrial pathway in ammonia-treated chicken splenic lymphocytes. Sci. Total Environ. 2020, 729, 139017. [Google Scholar] [CrossRef]
- Zhao, F.; Qu, J.; Wang, W.; Li, S.; Xu, S. The imbalance of Th1/Th2 triggers an inflammatory response in chicken spleens after ammonia exposure. Poult. Sci. 2020, 99, 3817–3822. [Google Scholar] [CrossRef]
- Wang, W.; Chen, M.; Jin, X.; Li, X.; Yang, Z.; Lin, H.; Xu, S. H(2)S induces Th1/Th2 imbalance with triggered NF-κB pathway to exacerbate LPS-induce chicken pneumonia response. Chemosphere 2018, 208, 241–246. [Google Scholar] [CrossRef]
- Chi, Q.; Chi, X.; Hu, X.; Wang, S.; Zhang, H.; Li, S. The effects of atmospheric hydrogen sulfide on peripheral blood lymphocytes of chickens: Perspectives on inflammation, oxidative stress and energy metabolism. Environ. Res. 2018, 167, 1–6. [Google Scholar] [CrossRef]
- Chi, Q.; Wang, D.; Hu, X.; Li, S.; Li, S. Hydrogen Sulfide Gas Exposure Induces Necroptosis and Promotes Inflammation through the MAPK/NF-κB Pathway in Broiler Spleen. Oxid. Med. Cell Longev. 2019, 2019, 8061823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, X.; Chi, Q.; Liu, Q.; Wang, D.; Zhang, Y.; Li, S. Atmospheric H2S triggers immune damage by activating the TLR-7/MyD88/NF-κB pathway and NLRP3 inflammasome in broiler thymus. Chemosphere 2019, 237, 124427. [Google Scholar] [CrossRef] [PubMed]
- Scott, T.; Owens, M.D. Thrombocytes respond to lipopolysaccharide through Toll-like receptor-4, and MAP kinase and NF-kappaB pathways leading to expression of interleukin-6 and cyclooxygenase-2 with production of prostaglandin E2. Mol. Immunol. 2008, 45, 1001–1008. [Google Scholar] [CrossRef] [PubMed]
- Winkler, C.; Ferdous, F.; Dimmick, M.; Scott, T. Lipopolysaccharide induced Interleukin-6 production is mediated through activation of ERK 1/2, p38 MAPK, MEK, and NFκB in chicken thrombocytes. Dev. Comp. Immunol. 2017, 73, 124–130. [Google Scholar] [CrossRef] [PubMed]
- Kogut, M.H.; Genovese, K.J.; He, H.; Kaiser, P. Flagellin and lipopolysaccharide up-regulation of IL-6 and CXCLi2 gene expression in chicken heterophils is mediated by ERK1/2-dependent activation of AP-1 and NF-kappaB signaling pathways. Innate Immun. 2008, 14, 213–222. [Google Scholar] [CrossRef] [Green Version]
- Ariyadi, B.; Isobe, N.; Yoshimura, Y. Toll-like receptor signaling for the induction of mucin expression by lipopolysaccharide in the hen vagina. Poult. Sci. 2014, 93, 673–679. [Google Scholar] [CrossRef]
- Liu, L.; Shen, J.; Zhao, C.; Wang, X.; Yao, J.; Gong, Y.; Yang, X. Dietary Astragalus polysaccharide alleviated immunological stress in broilers exposed to lipopolysaccharide. Int. J. Biol. Macromol. 2015, 72, 624–632. [Google Scholar] [CrossRef]
- Huang, X.Y.; Ansari, A.R.; Huang, H.B.; Zhao, X.; Li, N.Y.; Sun, Z.J.; Peng, K.M.; Zhong, J.; Liu, H.Z. Lipopolysaccharide mediates immuno-pathological alterations in young chicken liver through TLR4 signaling. BMC Immunol. 2017, 18, 12. [Google Scholar] [CrossRef] [Green Version]
- Li, N.; Ansari, A.R.; Sun, Z.; Huang, H.; Cui, L.; Hu, Y.; Zhao, X.; Zhong, J.; Abdel-Kafy, E.M.; Liu, H. Toll like receptor 4 signaling pathway participated in Salmonella lipopolysaccharide-induced spleen injury in young chicks. Microb. Pathog. 2017, 112, 288–294. [Google Scholar] [CrossRef]
- Chang, Y.; Yuan, L.; Liu, J.; Muhammad, I.; Cao, C.; Shi, C.; Zhang, Y.; Li, R.; Li, C.; Liu, F. Dihydromyricetin attenuates Escherichia coli lipopolysaccharide-induced ileum injury in chickens by inhibiting NLRP3 inflammasome and TLR4/NF-κB signalling pathway. Vet. Res. 2020, 51, 72. [Google Scholar] [CrossRef]
- Yang, L.; Liu, G.; Lian, K.; Qiao, Y.; Zhang, B.; Zhu, X.; Luo, Y.; Shang, Y.; Gu, X.L. Dietary leonurine hydrochloride supplementation attenuates lipopolysaccharide challenge-induced intestinal inflammation and barrier dysfunction by inhibiting the NF-κB/MAPK signaling pathway in broilers. J. Anim. Sci. 2019, 97, 1679–1692. [Google Scholar] [CrossRef] [PubMed]
- Velleman, S.G. Pectoralis Major (Breast) Muscle Extracellular Matrix Fibrillar Collagen Modifications Associated With the Wooden Breast Fibrotic Myopathy in Broilers. Front. Physiol. 2020, 11, 461. [Google Scholar] [CrossRef] [PubMed]
- Xing, T.; Luo, D.; Zhao, X.; Xu, X.; Li, J.; Zhang, L.; Gao, F. Enhanced cytokine expression and upregulation of inflammatory signaling pathways in broiler chickens affected by wooden breast myopathy. J. Sci. Food Agric. 2020, in press. [Google Scholar] [CrossRef] [PubMed]
- Fancher, C.A.; Zhang, L.; Kiess, A.S.; Adhikari, P.A.; Dinh, T.; Sukumaran, A.T. Avian Pathogenic Escherichia coli and Clostridium perfringens: Challenges in No Antibiotics Ever Broiler Production and Potential Solutions. Microorganisms 2020, 8, 1533. [Google Scholar] [CrossRef] [PubMed]
- Nhung, N.T.; Chansiripornchai, N.; Carrique-Mas, J.J. Antimicrobial Resistance in Bacterial Poultry Pathogens: A Review. Front. Vet. Sci. 2017, 4, 126. [Google Scholar] [CrossRef] [Green Version]
- Peng, L.Y.; Cui, Z.Q.; Wu, Z.M.; Fu, B.D.; Yi, P.F.; Shen, H.Q. RNA-seq profiles of chicken type II pneumocyte in response to Escherichia coli infection. PLoS ONE 2019, 14, e0217438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, L.Y.; Yuan, M.; Wu, Z.M.; Song, K.; Zhang, C.L.; An, Q.; Xia, F.; Yu, J.L.; Yi, P.F.; Fu, B.D.; et al. Anti-bacterial activity of baicalin against APEC through inhibition of quorum sensing and inflammatory responses. Sci. Rep. 2019, 9, 4063. [Google Scholar] [CrossRef]
- Peng, L.Y.; Yuan, M.; Song, K.; Yu, J.L.; Li, J.H.; Huang, J.N.; Yi, P.F.; Fu, B.D.; Shen, H.Q. Baicalin alleviated APEC-induced acute lung injury in chicken by inhibiting NF-κB pathway activation. Int. Immunopharmacol. 2019, 72, 467–472. [Google Scholar] [CrossRef]
- Jiao, J.; Yang, Y.; Liu, M.; Li, J.; Cui, Y.; Yin, S.; Tao, J. Artemisinin and Artemisia annua leaves alleviate Eimeria tenella infection by facilitating apoptosis of host cells and suppressing inflammatory response. Vet. Parasitol. 2018, 254, 172–177. [Google Scholar] [CrossRef]
- Yuan, M.; Peng, L.Y.; Wu, S.C.; Li, J.H.; Song, K.; Chen, S.; Huang, J.N.; Yu, J.L.; An, Q.; Yi, P.F.; et al. Schizandrin attenuates inflammation induced by avian pathogenic Escherichia coli in chicken type II pneumocytes. Int. Immunopharmacol. 2020, 81, 106313. [Google Scholar] [CrossRef]
- Wang, W.; Li, Z.; Han, Q.; Guo, Y.; Zhang, B.; D’inca, R. Dietary live yeast and mannan-oligosaccharide supplementation attenuate intestinal inflammation and barrier dysfunction induced by Escherichia coli in broilers. Br. J. Nutr. 2016, 116, 1878–1888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thames, H.T.; Sukumaran, A.T. A Review of Salmonella and Campylobacter in Broiler Meat: Emerging Challenges and Food Safety Measures. Foods 2020, 9, 776. [Google Scholar] [CrossRef]
- Keestra, A.M.; de Zoete, M.R.; van Aubel, R.A.; van Putten, J.P. Functional characterization of chicken TLR5 reveals species-specific recognition of flagellin. Mol. Immunol. 2008, 45, 1298–1307. [Google Scholar] [CrossRef] [PubMed]
- Chiang, H.I.; Berghman, L.R.; Zhou, H. Inhibition of NF-kB 1 (NF-kBp50) by RNA interference in chicken macrophage HD11 cell line challenged with Salmonellaenteritidis. Genet. Mol. Biol. 2009, 32, 507–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, H.; Chen, Z.; Xie, L.J.; Liu, G.F. Suppression of TLR4/NFκB signaling pathway improves cerebral ischemia-reperfusion injury in rats. Mol. Neurobiol. 2018, 55, 4311–4319. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Ma, T.; Wang, H.; Sheng, Z.; Dou, X.; Wang, K.; Li, Z.; Pan, Z.; Chang, G.; Chen, G. Up-regulation of NLRC5 and NF-κB signaling pathway in carrier chickens challenged with Salmonella enterica Serovar Pullorum at different persistence periods. Indian J. Biochem. Biophys. 2015, 52, 132–139. [Google Scholar] [PubMed]
- Geng, S.; Wang, Y.; Xue, Y.; Wang, H.; Cai, Y.; Zhang, J.; Barrow, P.; Pan, Z.; Jiao, X. The SseL protein inhibits the intracellular NF-κB pathway to enhance the virulence of Salmonella Pullorum in a chicken model. Microb. Pathog. 2019, 129, 1–6. [Google Scholar] [CrossRef]
- He, Y.; Yang, Y.; Dong, Y.; Yan, C.; Zhang, B. The Effects of Flavomycin and Colistin Sulfate Pre-Treatment on Ileal Bacterial Community Composition, the Response to Salmonella typhimurium and Host Gene Expression in Broiler Chickens. Microorganisms 2019, 7, 574. [Google Scholar] [CrossRef] [Green Version]
- He, Y.; Yang, Y.; Dong, Y.; Ito, K.; Zhang, B. Highly nutritious diet resists Salmonella Typhimurium infections by improving intestinal microbiota and morphology in broiler chickens. Poult. Sci. 2020, 99, 7055–7065. [Google Scholar] [CrossRef]
- Li, Q.; Xu, L.; Yin, C.; Liu, Z.; Li, Y.; Yuan, Y.; Hu, Y.; Jiao, X. The Invasion Plasmid Antigen J (IpaJ) from Salmonella Inhibits NF-κB Activation by Suppressing IκBα Ubiquitination. Infect. Immun. 2020, 88, e00875-19. [Google Scholar] [CrossRef]
- Ishfaq, M.; Hu, W.; Khan, M.Z.; Ahmad, I.; Guo, W.; Li, J. Current status of vaccine research, development, and challenges of vaccines for Mycoplasma gallisepticum. Poult. Sci. 2020, 99, 4195–4202. [Google Scholar] [CrossRef] [PubMed]
- Majumder, S.; Zappulla, F.; Silbart, L.K. Mycoplasma gallisepticum lipid associated membrane proteins up-regulate inflammatory genes in chicken tracheal epithelial cells via TLR-2 ligation through an NF-κB dependent pathway. PLoS ONE 2014, 9, e112796. [Google Scholar] [CrossRef] [PubMed]
- Tian, W.; Zhao, C.; Hu, Q.; Sun, J.; Peng, X. Roles of Toll-like receptors 2 and 6 in the inflammatory response to Mycoplasma gallisepticum infection in DF-1 cells and in chicken embryos. Dev. Comp. Immunol. 2016, 59, 39–47. [Google Scholar] [CrossRef] [PubMed]
- Sheedy, F.J. Turning 21: Induction of miR-21 as a Key Switch in the Inflammatory Response. Front. Immunol. 2015, 6, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krichevsky, A.M.; Gabriely, G. miR-21: A small multi-faceted RNA. J. Cell. Mol. Med. 2009, 13, 39–53. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Zou, M.; Sun, Y.; Zhang, K.; Peng, X. gga-miR-21 modulates Mycoplasma gallisepticum (HS strain)-Induced inflammation via targeting MAP3K1 and activating MAPKs and NF-κB pathways. Vet. Microbiol. 2019, 237, 108407. [Google Scholar] [CrossRef]
- Zhang, K.; Han, Y.; Wang, Z.; Zhao, Y.; Fu, Y.; Peng, X. gga-miR-146c Activates TLR6/MyD88/NF-κB Pathway through Targeting MMP16 to Prevent Mycoplasma Gallisepticum (HS Strain) Infection in Chickens. Cells 2019, 8, 501. [Google Scholar] [CrossRef] [Green Version]
- Zhang, K.; Han, Y.; Zhao, Y.; Sun, Y.; Zou, M.; Fu, Y.; Peng, X. Upregulated gga-miR-16-5p Inhibits the Proliferation Cycle and Promotes the Apoptosis of MG-Infected DF-1 Cells by Repressing PIK3R1-Mediated the PI3K/Akt/NF-κB Pathway to Exert Anti-Inflammatory Effect. Int. J. Mol. Sci. 2019, 20, 1036. [Google Scholar] [CrossRef] [Green Version]
- Yuan, B.; Zou, M.; Zhao, Y.; Zhang, K.; Sun, Y.; Peng, X. Up-Regulation of miR-130b-3p Activates the PTEN/PI3K/AKT/NF-κB Pathway to Defense against Mycoplasma gallisepticum (HS Strain) Infection of Chicken. Int. J. Mol. Sci. 2018, 19, 2172. [Google Scholar] [CrossRef] [Green Version]
- Yu, Y.; Chen, Y.; Wang, Y.; Li, Y.; Zhang, L.; Xin, J. TLR2/MyD88/NF-κB signaling pathway regulates IL-1β production in DF-1cells exposed to Mycoplasma gallisepticum LAMPs. Microb. Pathog. 2018, 117, 225–231. [Google Scholar] [CrossRef]
- Beaudet, J.; Tulman, E.R.; Pflaum, K.; Liao, X.; Kutish, G.F.; Szczepanek, S.M.; Silbart, L.K.; Geary, S.J. Transcriptional Profiling of the Chicken Tracheal Response to Virulent Mycoplasma gallisepticum Strain Rlow. Infect. Immun. 2017, 85, e00343-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zou, M.; Yang, W.; Niu, L.; Sun, Y.; Luo, R.; Wang, Y.; Peng, X. Polydatin attenuates Mycoplasma gallisepticum (HS strain)-induced inflammation injury via inhibiting the TLR6/ MyD88/NF-κB pathway. Microb. Pathog. 2020, 149, 104552. [Google Scholar] [CrossRef]
- Wu, Z.; Chen, C.; Miao, Y.; Liu, Y.; Zhang, Q.; Li, R.; Ding, L.; Ishfaq, M.; Li, J. Baicalin Attenuates Mycoplasma gallisepticum-Induced Inflammation via Inhibition of the TLR2-NF-κB Pathway in Chicken and DF-1 Cells. Infect. Drug Resist. 2019, 12, 3911–3923. [Google Scholar] [CrossRef] [Green Version]
- Ishfaq, M.; Chen, C.; Bao, J.; Zhang, W.; Wu, Z.; Wang, J.; Liu, Y.; Tian, E.; Hamid, S.; Li, R.; et al. Baicalin ameliorates oxidative stress and apoptosis by restoring mitochondrial dynamics in the spleen of chickens via the opposite modulation of NF-κB and Nrf2/HO-1 signaling pathway during Mycoplasma gallisepticum infection. Poult. Sci. 2019, 98, 6296–6310. [Google Scholar] [CrossRef] [PubMed]
- Zou, M.; Yang, L.; Niu, L.; Zhao, Y.; Sun, Y.; Fu, Y.; Peng, X. Baicalin ameliorates Mycoplasma gallisepticum-induced lung inflammation in chicken by inhibiting TLR6-mediated NF-κB signalling. Br. Poult. Sci. 2020, in press. [Google Scholar] [CrossRef] [PubMed]
- Niu, L.; Luo, R.; Zou, M.; Sun, Y.; Fu, Y.; Wang, Y.; Peng, X. Puerarin inhibits Mycoplasma gallisepticum (MG-HS)-induced inflammation and apoptosis via suppressing the TLR6/MyD88/NF-κB signal pathway in chicken. Int. Immunopharmacol. 2020, 88, 106993. [Google Scholar] [CrossRef] [PubMed]
- Fatoba, A.J.; Adeleke, M.A. Diagnosis and control of chicken coccidiosis: A recent update. J. Parasit. Dis. 2018, 42, 483–493. [Google Scholar] [CrossRef] [PubMed]
- Jin, H.; Haicheng, Y.; Caiyun, Z.; Yong, Z.; Jinrong, W. The Expression of NF-kB Signaling Pathway Was Inhibited by Silencing TGF-b4 in Chicken IECs Infected with E. tenella. Braz. J. Poult. Sci. 2020, in press. [Google Scholar] [CrossRef]
- Timbermont, L.; Haesebrouck, F.; Ducatelle, R.; Van Immerseel, F. (Necrotic enteritis in broilers: An updated review on the pathogenesis. Avian Pathol. 2011, 40, 341–347. [Google Scholar] [CrossRef]
- Mora, Z.V.; Macías-Rodríguez, M.E.; Arratia-Quijada, J.; Gonzalez-Torres, Y.S.; Nuño, K.; Villarruel-López, A. Clostridium perfringens as Foodborne Pathogen in Broiler Production: Pathophysiology and Potential Strategies for Controlling Necrotic Enteritis. Animals 2020, 10, 1718. [Google Scholar] [CrossRef]
- Van Immerseel, F.; De Buck, J.; Pasmans, F.; Huyghebaert, G.; Haesebrouck, F.; Ducatelle, R. Clostridium perfringens in poultry: An emerging threat for animal and public health. Avian Pathol. 2004, 33, 537–549. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.; Li, C.; Liu, D.; Guo, Y. Inflammatory responses to a Clostridium perfringens type A strain and α-toxin in primary intestinal epithelial cells of chicken embryos. Avian Pathol. 2015, 44, 81–91. [Google Scholar] [CrossRef] [PubMed]
- Athanasiadou, S.; Russell, K.M.; Kaiser, P.; Kanellos, T.; Burgess, S.T.; Mitchell, M.; Clutton, E.; Naylor, S.W.; Low, C.J.; Hutchings, M.R.; et al. Genome wide transcriptomic analysis identifies pathways affected by the infusion of Clostridium perfringens culture supernatant in the duodenum of broilers in situ. J. Anim. Sci. 2015, 93, 3152–3163. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.; Liu, D.; Zhang, B.; Li, Z.; Li, Y.; Ding, B.; Guo, Y. Two Lactobacillus Species Inhibit the Growth and α-Toxin Production of Clostridium perfringens and Induced Proinflammatory Factors in Chicken Intestinal Epithelial Cells in Vitro. Front. Microbiol. 2017, 8, 2081. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Wang, W.; Liu, D.; Guo, Y. Effects of Lactobacillus acidophilus on the growth performance and intestinal health of broilers challenged with Clostridium perfringens. J. Anim. Sci. Biotechnol. 2018, 9, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Knittler, M.R.; Sachse, K. Chlamydia psittaci: Update on an underestimated zoonotic agent. Pathog. Dis. 2015, 73, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Chu, J.; Li, X.; Qu, G.; Wang, Y.; Li, Q.; Guo, Y.; Hou, L.; Liu, J.; Eko, F.O.; He, C. Chlamydia psittaci PmpD-N Exacerbated Chicken Macrophage Function by Triggering Th2 Polarization and the TLR2/MyD88/NF-κB Signaling Pathway. Int. J. Mol. Sci. 2020, 21, 2003. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Li, Y.; Yan, X.; Sun, Z.; Wang, C.; Liu, S.; Xiao, J.; Lu, C.; Wu, Y. Chlamydia psittaci Plasmid-Encoded CPSIT_P7 Elicits Inflammatory Response in Human Monocytes via TLR4/Mal/MyD88/NF-κB Signaling Pathway. Front Microbiol. 2020, 11, 578009. [Google Scholar] [CrossRef]
- López-Osorio, S.; Chaparro-Gutiérrez, J.J.; Gómez-Osorio, L.M. Overview of Poultry Eimeria Life Cycle and Host-Parasite Interactions. Front. Vet. Sci. 2020, in press. [Google Scholar]
- Rehman, Z.U.; Meng, C.; Sun, Y.; Safdar, A.; Pasha, R.H.; Munir, M.; Ding, C. Oxidative Stress in Poultry: Lessons from the Viral Infections. Oxid. Med. Cell Longev. 2018, 2018, 5123147. [Google Scholar] [CrossRef]
- Khatri, M.; Sharma, J.M. Infectious bursal disease virus infection induces macrophage activation via p38 MAPK and NF-kappaB pathways. Virus Res. 2006, 118, 70–77. [Google Scholar] [CrossRef] [PubMed]
- Wong, R.T.; Hon, C.C.; Zeng, F.; Leung, F.C. Screening of differentially expressed transcripts in infectious bursal disease virus-induced apoptotic chicken embryonic fibroblasts by using cDNA microarrays. J. Gen. Virol. 2007, 88, 1785–1796. [Google Scholar] [CrossRef] [PubMed]
- Cubas-Gaona, L.L.; Diaz-Beneitez, E.; Ciscar, M.; Rodríguez, J.F.; Rodríguez, D. Exacerbated Apoptosis of Cells Infected with Infectious Bursal Disease Virus upon Exposure to Interferon Alpha. J. Virol. 2018, 92, e00364-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ou, C.; Wang, Q.; Wei, X.; Liu, M.; Liu, X.; He, C. Pro-apoptosis effects of protocatechuic acid in the early stage of infectious bursal disease virus infection. Microb. Pathog. 2018, 124, 216–222. [Google Scholar] [CrossRef] [PubMed]
- Bello, M.B.; Yusoff, K.; Ideris, A.; Hair-Bejo, M.; Peeters, B.P.H.; Omar, A.R. Diagnostic and Vaccination Approaches for Newcastle Disease Virus in Poultry: The Current and Emerging Perspectives. Biomed. Res. Int. 2018, 2018, 7278459. [Google Scholar] [CrossRef]
- Mayers, J.; Mansfield, K.L.; Brown, I.H. The role of vaccination in risk mitigation and control of Newcastle disease in poultry. Vaccine 2017, 35, 5974–5980. [Google Scholar] [CrossRef]
- Qu, Y.; Zhan, Y.; Yang, S.; Ren, S.; Qiu, X.; Rehamn, Z.U.; Tan, L.; Sun, Y.; Meng, C.; Song, C.; et al. Newcastle disease virus infection triggers HMGB1 release to promote the inflammatory response. Virology 2018, 525, 19–31. [Google Scholar] [CrossRef]
- Park, J.; Kim, M.; Na, G.; Jeon, I.; Kwon, Y.K.; Kim, J.H.; Youn, H.; Koo, Y. Glucocorticoids modulate NF-kappaB-dependent gene expression by up-regulating FKBP51 expression in Newcastle disease virus-infected chickens. Mol. Cell. Endocrinol. 2007, 278, 7–17. [Google Scholar] [CrossRef]
- Qian, J.; Xu, X.; Ding, J.; Yin, R.; Sun, Y.; Xue, C.; Wang, J.; Ding, C.; Yu, S.; Liu, X.; et al. Newcastle disease virus-like particles induce DC maturation through TLR4/NF-κB pathway and facilitate DC migration by CCR7-CCL19/CCL21 axis. Vet. Microbiol. 2017, 203, 158–166. [Google Scholar] [CrossRef]
- Chen, Y.; Liu, W.; Xu, H.; Liu, J.; Deng, Y.; Cheng, H.; Zhan, T.; Lu, X.; Liao, T.; Guo, L.; et al. Gga-miR-19b-3p Inhibits Newcastle Disease Virus Replication by Suppressing Inflammatory Response via Targeting RNF11 and ZMYND11. Front Microbiol. 2019, 10, 2006. [Google Scholar] [CrossRef] [Green Version]
- Li, J.J.; Yin, Y.; Yang, H.L.; Yang, C.W.; Yu, C.L.; Wang, Y.; Yin, H.D.; Lian, T.; Peng, H.; Zhu, Q.; et al. mRNA expression and functional analysis of chicken IFIT5 after infected with Newcastle disease virus. Infect. Genet. Evol. 2020, 86, 104585. [Google Scholar] [CrossRef] [PubMed]
- Bertzbach, L.D.; Conradie, A.M.; You, Y.; Kaufer, B.B. Latest Insights into Marek’s Disease Virus Pathogenesis and Tumorigenesis. Cancers 2020, 12, 647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, S.; Kunec, D.; Buza, J.J.; Chiang, H.I.; Zhou, H.; Subramaniam, S.; Pendarvis, K.; Cheng, H.H.; Burgess, S.C. Nuclear Factor kappa B is central to Marek’s disease herpesvirus induced neoplastic transformation of CD30 expressing lymphocytes in-vivo. BMC Syst. Biol. 2012, 6, 123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Gao, L.; Xu, Z.; Luo, D.; Zhang, Y.; Gao, Y.; Liu, C.; Zhang, Y.; Qi, X.; Cui, H.; et al. Marek’s Disease Virus RLORF4 Inhibits Type I Interferon Production by Antagonizing NF-κB Activation. J. Virol. 2019, 93, e01037-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benavente, J.; Martínez-Costas, J. Avian reovirus: Structure and biology. Virus Res. 2007, 123, 105–119. [Google Scholar] [CrossRef] [PubMed]
- De Carli, S.; Wolf, J.M.; Gräf, T.; Lehmann, F.K.; Fonseca, A.S.; Canal, C.W.; Lunge, V.R.; Ikuta, N. Genotypic characterization and molecular evolution of avian reovirus in poultry flocks from Brazil. Avian Pathol. 2020, 49, 611–620. [Google Scholar] [CrossRef] [PubMed]
- Lin, P.Y.; Liu, H.J.; Liao, M.H.; Chang, C.D.; Chang, C.I.; Cheng, H.L.; Lee, J.W.; Shih, W.L. Activation of PI 3-kinase/Akt/NF-kappaB and Stat3 signaling by avian reovirus S1133 in the early stages of infection results in an inflammatory response and delayed apoptosis. Virology 2010, 400, 104–114. [Google Scholar] [CrossRef] [Green Version]
- Xie, L.; Xie, Z.; Wang, S.; Huang, J.; Deng, X.; Xie, Z.; Luo, S.; Zeng, T.; Zhang, Y.; Zhang, M. Altered gene expression profiles of the MDA5 signaling pathway in peripheral blood lymphocytes of chickens infected with avian reovirus. Arch. Virol. 2019, 164, 2451–2458. [Google Scholar] [CrossRef]
- Yun, T.; Hua, J.; Ye, W.; Ni, Z.; Chen, L.; Zhang, C. The phosphoproteomic responses of duck (Cairna moschata) to classical/novel duck reovirus infections in the spleen tissue. Sci. Rep. 2020, 10, 15315. [Google Scholar] [CrossRef]
- Campbell, L.K.; Magor, K.E. Pattern Recognition Receptor Signaling and Innate Responses to Influenza A Viruses in the Mallard Duck, Compared to Humans and Chickens. Front. Cell. Infect. Microbiol. 2020, 10, 209. [Google Scholar] [CrossRef]
- Li, N.; Zhao, J.; Yang, Y.; Zeng, Y.; Liu, S. Innate immune responses to duck Tembusu virus infection. Vet. Res. 2020, 51, 87. [Google Scholar] [CrossRef] [PubMed]
- Nettleford, S.K.; Prabhu, K.S. Selenium and Selenoproteins in Gut Inflammation-A Review. Antioxidants 2018, 7, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.L.; Xu, B.; Huang, X.D.; Gao, Y.H.; Chen, Y.; Shan, A.S. Selenium Deficiency Affects the mRNA Expression of Inflammatory Factors and Selenoprotein Genes in the Kidneys of Broiler Chicks. Biol. Trace Elem. Res. 2016, 171, 201–207. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Qu, Y.; Wang, J.; Wu, R. Selenium Deficiency Attenuates Chicken Duodenal Mucosal Immunity via Activation of the NF-κb Signaling Pathway. Biol. Trace Elem. Res. 2016, 172, 465–473. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Liu, Z.; He, X.; Lian, S.; Liang, J.; Yu, D.; Sun, D.; Wu, R. Selenium deficiency induces duodenal villi cell apoptosis via an oxidative stress-induced mitochondrial apoptosis pathway and an inflammatory signaling-induced death receptor pathway. Metallomics 2018, 10, 1390–1400. [Google Scholar] [CrossRef] [PubMed]
- Zhang, R.; Guo, R.; Liu, Q.; Li, G.; Sun, B.; Huang, X. Selenium Deficiency via the TLR4/TRIF/NF-κB Signaling Pathway Leading to Inflammatory Injury in Chicken Spleen. Biol. Trace Elem. Res. 2020, in press. [Google Scholar] [CrossRef]
- Wang, Y.; Jiang, L.; He, J.; Hu, M.; Zeng, F.; Li, Y.; Tian, H.; Luo, X. The Adverse Effects of Se Toxicity on Inflammatory and Immune Responses in Chicken Spleens. Biol. Trace Elem. Res. 2018, 185, 170–176. [Google Scholar] [CrossRef] [PubMed]
- Huang, H.; Chen, J.; Sun, Q.; Liu, Y.; Tang, Y.; Teng, X. NLRP3 inflammasome is involved in the mechanism of mitigative effect of selenium on lead-induced inflammatory damage in chicken kidneys. Environ. Sci. Pollut. Res. Int. 2020, in press. [Google Scholar] [CrossRef]
- Jiayong, Z.; Shengchen, W.; Xiaofang, H.; Gang, S.; Shiwen, X. The antagonistic effect of selenium on lead-induced necroptosis via MAPK/NF-κB pathway and HSPs activation in the chicken spleen. Ecotoxicol. Environ. Saf. 2020, 204, 111049. [Google Scholar] [CrossRef]
- Zhang, J.; Hao, X.; Xu, S. Selenium Prevents Lead-Induced Necroptosis by Restoring Antioxidant Functions and Blocking MAPK/NF-κB Pathway in Chicken Lymphocytes. Biol. Trace Elem. Res. 2020, 198, 644–653. [Google Scholar] [CrossRef]
- Wang, X.; Bao, R.; Fu, J. The Antagonistic Effect of Selenium on Cadmium-Induced Damage and mRNA Levels of Selenoprotein Genes and Inflammatory Factors in Chicken Kidney Tissue. Biol. Trace Elem. Res. 2018, 181, 331–339. [Google Scholar] [CrossRef] [PubMed]
- Zhang, R.; Liu, Y.; Xing, L.; Zhao, N.; Zheng, Q.; Li, J.; Bao, J. The protective role of selenium against cadmium-induced hepatotoxicity in laying hens: Expression of Hsps and inflammation-related genes and modulation of elements homeostasis. Ecotoxicol. Environ. Saf. 2018, 159, 205–212. [Google Scholar] [CrossRef]
- Tang, K.K.; Li, H.Q.; Qu, K.C.; Fan, R.F. Selenium alleviates cadmium-induced inflammation and meat quality degradation via antioxidant and anti-inflammation in chicken breast muscles. Environ. Sci. Pollut. Res. Int. 2019, 26, 23453–23459. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Zhang, R.; Wang, T.; Jiang, H.; Guo, M.; Zhou, E.; Sun, Y.; Yang, Z.; Xu, S.; Cao, Y.; et al. Selenium inhibits LPS-induced pro-inflammatory gene expression by modulating MAPK and NF-κB signaling pathways in mouse mammary epithelial cells in primary culture. Inflammation 2014, 37, 478–485. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Wang, S.; Zhang, Q.; Li, X.; Xu, S. Selenomethionine alleviates LPS-induced chicken myocardial inflammation by regulating the miR-128-3p-p38 MAPK axis and oxidative stress. Metallomics 2020, 12, 54–64. [Google Scholar] [CrossRef]
- Qu, J.; Wang, W.; Zhang, Q.; Li, S. Inhibition of Lipopolysaccharide-Induced Inflammation of Chicken Liver Tissue by Selenomethionine via TLR4-NF-κB-NLRP3 Signaling Pathway. Biol. Trace Elem. Res. 2020, 195, 205–214. [Google Scholar] [CrossRef] [PubMed]
- Shi, X.; Wang, W.; Zheng, S.; Zhang, Q.; Xu, S. Selenomethionine relieves inflammation in the chicken trachea caused by LPS though inhibiting the NF-κB pathway. Biol. Trace Elem. Res. 2020, 194, 525–535. [Google Scholar] [CrossRef] [PubMed]
- Tan, J.; Liu, S.; Guo, Y.; Applegate, T.J.; Eicher, S.D. Dietary L-arginine supplementation attenuates lipopolysaccharide-induced inflammatory response in broiler chickens. Br. J. Nutr. 2014, 111, 1394–1404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bi, Y.; Nan, X.M.; Zheng, S.S.; Jiang, L.S.; Xiong, B.H. Effects of dietary threonine and immune stress on growth performance, carcass trait, serum immune parameters, and intestinal muc2 and NF-κb gene expression in Pekin ducks from hatch to 21 days. Poult. Sci. 2018, 97, 177–187. [Google Scholar] [CrossRef]
- Liu, S.Q.; Wang, L.Y.; Liu, G.H.; Tang, D.Z.; Fan, X.X.; Zhao, J.P.; Jiao, H.C.; Wang, X.J.; Sun, S.H.; Lin, H. Leucine alters immunoglobulin a secretion and inflammatory cytokine expression induced by lipopolysaccharide via the nuclear factor-κB pathway in intestine of chicken embryos. Animal 2018, 12, 1903–1911. [Google Scholar] [CrossRef]
- Surai, P.F. Polyphenol compounds in the chicken/animal diet: From the past to the future. J. Anim. Physiol. Anim. Nutr. 2014, 98, 19–31. [Google Scholar] [CrossRef] [PubMed]
- Surai, P.F. Silymarin as a Natural Antioxidant: An Overview of the Current Evidence and Perspectives. Antioxidants 2015, 4, 204–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, C.; Tian, Y.; Yan, F.; Kang, X.; Han, R.; Sun, G.; Zhang, H. Modulation of growth and immunity by dietary supplementation with resveratrol in young chickens receiving conventional vaccinations. Am. J. Vet. Res. 2014, 75, 752–759. [Google Scholar] [CrossRef] [PubMed]
- Sahin, K.; Orhan, C.; Akdemir, F.A.; Tuzcu, M.; Iben, C.; Sahin, N. Resveratrol protects quail hepatocytes against heat stress: Modulation of the Nrf2 transcription factor and heat shock proteins. J. Anim. Physiol. Anim. Nutr. 2012, 96, 66–74. [Google Scholar] [CrossRef] [PubMed]
- Fan, H.; Lv, Z.; Gan, L.; Guo, Y. Transcriptomics-Related Mechanisms of Supplementing Laying Broiler Breeder Hens with Dietary Daidzein to Improve the Immune Function and Growth Performance of Offspring. J. Agric. Food Chem. 2018, 66, 2049–2060. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.X.; Maria, T.C.; Zhou, B.; Xiao, F.L.; Wang, M.; Mao, Y.J.; Li, Y. Quercetin improves immune function in Arbor Acre broilers through activation of NF-κB signaling pathway. Poult. Sci. 2020, 99, 906–913. [Google Scholar] [CrossRef]
- Ge, C.; Zhang, C.; Ye, J.; Tang, X.; Wu, Y. Ginbaltosides promote proliferation of chicken primordial germ cells via PKC-involved activation of NF-kappaB. Cell. Biol. Int. 2007, 31, 1251–1256. [Google Scholar] [CrossRef]
- Lee, M.T.; Lin, W.C.; Wang, S.Y.; Lin, L.J.; Yu, B.; Lee, T.T. Evaluation of potential antioxidant and anti-inflammatory effects of Antrodia cinnamomea powder and the underlying molecular mechanisms via Nrf2- and NF-κB-dominated pathways in broiler chickens. Poult. Sci. 2018, 97, 2419–2434. [Google Scholar] [CrossRef]
- Chen, H.; Muhammad, I.; Zhang, Y.; Ren, Y.; Zhang, R.; Huang, X.; Diao, L.; Liu, H.; Li, X.; Sun, X.; et al. Antiviral Activity Against Infectious Bronchitis Virus and Bioactive Components of Hypericum perforatum L. Front. Pharmacol. 2019, 10, 1272. [Google Scholar] [CrossRef]
- Yu, M.; Ge, C.; Zeng, W.; Mi, Y.; Zhang, C. Retinoic acid promotes proliferation of chicken primordial germ cells via activation of PI3K/Akt-mediated NF-κB signalling cascade. Cell Biol. Int. 2012, 36, 705–712. [Google Scholar] [CrossRef]
- Zhang, X.; Zhong, X.; Zhou, Y.; Wang, G.; Du, H.; Wang, T. Dietary RRR-α-tocopherol succinate attenuates lipopolysaccharide-induced inflammatory cytokines secretion in broiler chicks. Br. J. Nutr. 2010, 104, 1796–1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhan, T.; Han, Y.; Tang, C.; Zhao, Q.; Sun, D.; Li, Y.; Jia, X.; Zhou, L.; Zhang, J. Metabolism and biological activity of α-tocopherol derived from vitamin E-enriched transgenic maize in broilers. J. Sci. Food. Agric. 2020, 100, 4319–4328. [Google Scholar] [CrossRef] [PubMed]
- Ma, Z.; Zhang, J.; Ma, H.; Dai, B.; Zheng, L.; Miao, J.; Zhang, Y. The influence of dietary taurine and reduced housing density on hepatic functions in laying hens. Poult. Sci. 2014, 93, 1724–1736. [Google Scholar] [CrossRef] [PubMed]
- Dai, B.; Zhang, Y.S.; Ma, Z.L.; Zheng, L.H.; Li, S.J.; Dou, X.H.; Gong, J.S.; Miao, J.F. Influence of dietary taurine and housing density on oviduct function in laying hens. J. Zhejiang Univ.-SCIENCE B 2015, 16, 456–464. [Google Scholar] [CrossRef] [Green Version]
- Ma, Z.L.; Gao, Y.; Ma, H.T.; Zheng, L.H.; Dai, B.; Miao, J.F.; Zhang, Y.S. Effects of taurine and housing density on renal function in laying hens. J. Zhejiang Univ.-SCIENCE B 2016, 17, 952–964. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.; Zhen, W.; Geng, Y.; Wang, Z.; Guo, Y. Pretreatment with probiotic Enterococcus faecium NCIMB 11181 ameliorates necrotic enteritis-induced intestinal barrier injury in broiler chickens. Sci. Rep. 2019, 9, 10256. [Google Scholar] [CrossRef]
- Sun, Y.; Qian, J.; Xu, X.; Tang, Y.; Xu, W.; Yang, W.; Jiang, Y.; Yang, G.; Ding, Z.; Cong, Y.; et al. Dendritic cell-targeted recombinant Lactobacilli induce DC activation and elicit specific immune responses against G57 genotype of avian H9N2 influenza virus infection. Vet. Microbiol. 2018, 223, 9–20. [Google Scholar] [CrossRef]
- Lan, D.; Xun, X.; Hu, Y.; Li, N.; Yang, C.; Jiang, X.; Liu, Y. Research on the Effect of Pediococcus pentosaceus on Salmonella enteritidis-Infected Chicken. BioMed Res. Int. 2020, 2020, 6416451. [Google Scholar] [CrossRef]
- Weavers, H.; Martin, P. The cell biology of inflammation: From common traits to remarkable immunological adaptations. J. Cell Biol. 2020, 219, e202004003. [Google Scholar] [CrossRef]
- Pflug, K.M.; Sitcheran, R. Targeting NF-κB-Inducing Kinase (NIK) in Immunity, Inflammation, and Cancer. Int. J. Mol. Sci. 2020, 21, 8470. [Google Scholar] [CrossRef]
- de Gregorio, E.; Colell, A.; Morales, A.; Marí, M. Relevance of SIRT1-NF-κB Axis as Therapeutic Target to Ameliorate Inflammation in Liver Disease. Int. J. Mol. Sci. 2020, 21, 3858. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Cui, C.; Ma, X.; Luo, W.; Zheng, S.G.; Qiu, W. Nuclear Factor κB (NF-κB)-Mediated Inflammation in Multiple Sclerosis. Front. Immunol. 2020, 11, 391. [Google Scholar] [CrossRef] [PubMed]
- Afonina, I.S.; Zhong, Z.; Karin, M.; Beyaert, R. Limiting inflammation-the negative regulation of NF-κB and the NLRP3 inflammasome. Nat. Immunol. 2017, 18, 861–869. [Google Scholar] [CrossRef]
- Kunnumakkara, A.B.; Shabnam, B.; Girisa, S.; Harsha, C.; Banik, K.; Devi, T.B.; Choudhury, R.; Sahu, H.; Parama, D.; Sailo, B.L.; et al. Inflammation, NF-κB, and Chronic Diseases: How are They Linked? Crit. Rev. Immunol. 2020, 40, 1–39. [Google Scholar] [CrossRef] [PubMed]
- Serhan, C.N. Treating inflammation and infection in the 21st century: New hints from decoding resolution mediators and mechanisms. FASEB J. 2017, 31, 1273–1288. [Google Scholar] [CrossRef] [Green Version]
- Rothschild, D.E.; McDaniel, D.K.; Ringel-Scaia, V.M.; Allen, I.C. Modulating inflammation through the negative regulation of NF-κB signaling. J. Leukoc. Biol. 2018, 103, 1131–1150. [Google Scholar] [CrossRef]
- Kogut, M.H.; Genovese, K.J.; Swaggerty, C.L.; He, H.; Broom, L. Inflammatory phenotypes in the intestine of poultry: Not all inflammation is created equal. Poult. Sci. 2018, 97, 2339–2346. [Google Scholar] [CrossRef]
- Julian, R.J. Production and growth-related disorders and other metabolic diseases of poultry—A review. Vet. J. 2005, 169, 350–369. [Google Scholar] [CrossRef]
- Kierończyk, B.; Rawski, M.; Józefiak, D.; Świątkiewicz, S. Infectious and non-infectious factors associated with leg disorders in poultry—A review. Ann. Anim. Sci. 2017, 17, 645–669. [Google Scholar] [CrossRef] [Green Version]
- Ducatelle, R.; Goossens, E.; De Meyer, F.; Eeckhaut, V.; Antonissen, G.; Haesebrouck, F.; Van Immerseel, F. Biomarkers for monitoring intestinal health in poultry: Present status and future perspectives. Vet. Res. 2018, 49, 43. [Google Scholar] [CrossRef] [Green Version]
- Golonka, R.M.; Xiao, X.; Abokor, A.A.; Joe, B.; Vijay-Kumar, M. Altered nutrient status reprograms host inflammation and metabolic health via gut microbiota. J. Nutr. Biochem. 2020, in press. [Google Scholar] [CrossRef] [PubMed]
- Yang, Q.; Wang, Y.; Jia, A.; Wang, Y.; Bi, Y.; Liu, G. The crosstalk between gut bacteria and host immunity in intestinal inflammation. J. Cell. Physiol. 2020, in press. [Google Scholar] [CrossRef] [PubMed]
- Tilg, H.; Zmora, N.; Adolph, T.E.; Elinav, E. The intestinal microbiota fuelling metabolic inflammation. Nat. Rev. Immunol. 2020, 20, 40–54. [Google Scholar] [CrossRef] [PubMed]
- Broom, L.J. Host–Microbe Interactions and Gut Health in Poultry—Focus on Innate Responses. Microorganisms 2019, 7, 139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oviedo-Rondón, E.O. Holistic view of intestinal health in poultry. Anim. Feed Sci. Technol. 2019, 250, 1–8. [Google Scholar] [CrossRef]
- Rius-Pérez, S.; Pérez, S.; Martí-Andrés, P.; Monsalve, M.; Sastre, J. NF-κB Signaling complexes in acute inflammation. Antioxid. Redox Signal. 2019, in press. [Google Scholar]
- Farooqui, A.A. Inflammation and Oxidative Stress in Neurological Disorders; Springer: Cham, Switzerland; Heidelberg, Germany; New York, NY, USA; Dordrecht, The Netherlands; London, UK, 2014. [Google Scholar]
- Kircheis, R.; Haasbach, E.; Lueftenegger, D.; Heyken, W.T.; Ocker, M.; Planz, O. NF-κB Pathway as a Potential Target for Treatment of Critical Stage COVID-19 Patients. Front. Immunol. 2020, 11, 598444. [Google Scholar] [CrossRef]
- Sha, W.C.; Liou, H.C.; Tuomanen, E.I.; Baltimore, D. Targeted disruption of the p50 subunit of NF-κB leads to multifocal defects in immune responses. Cell 1995, 80, 321–330. [Google Scholar] [CrossRef] [Green Version]
- Surai, P.F. Antioxidants in Poultry Nutrition and Reproduction: An Update. Antioxidants 2020, 9, 105. [Google Scholar] [CrossRef] [Green Version]
- Surai, P.F.; Kochish, I.I.; Kidd, M.T. Taurine in Poultry Nutrition. Anim. Feed Sci. Technol. 2020, 260, 114339. [Google Scholar] [CrossRef]
- Surai, P.F. Antioxidant action of carnitine: Molecular mechanisms and practical applications. EC Vet. Sci. 2015, 2, 66–84. [Google Scholar]
- Surai, P.F. Carnitine enigma: From antioxidant action to vitagene regulation. Part 1. Absorption, metabolism and antioxidant activities. J. Vet. Sci. Med. 2015, 3, 14. [Google Scholar] [CrossRef]
- Surai, P.F. Carnitine enigma: From antioxidant action to vitagene regulation. Part 2. Transcription factors and practical applications. J. Vet. Sci. Med. 2015, 3, 17. [Google Scholar] [CrossRef]
Mechanisms of Nrf2–NF-κB Interactions | References |
---|---|
Inhibiting effects of Nrf2 on NF-κB | |
Decreasing the intracellular ROS levels. This inhibits oxidative stress-mediated NF-κB activation | [103] |
Preventing the IκB-proteasomal degradation and inhibiting nuclear translocation of NF-κB. In Nrf2-deficient cells an inhibitor of NF-kB activity (IκB) is over-phosphorylated with rapid proteasomal degradation and increased NF-κB activity. Upregulation of Nrf2 induces increase heme oxygenase-1 (HO-1) levels and induce phase II enzymes expression blocking the degradation of IκB | [104,105,106,107] |
Reducing p50 and p65 DNA binding. Nrf2 silencing enhanced p50 and p65 DNA binding and tumour necrosis factor (TNF)-α-induced proinflammatory gene expression | [108] |
Preventing the recruitment of RNA polymerase II to start transcription of NF-κB-regulated genes. Nrf2 binds to regulatory regions of proinflammatory genes in an antioxidant-response element (ARE)-independent manner and prevents the recruitment of RNA polymerase II to start transcription of NF-κB-regulated genes | [109] |
Competition between Nrf2 and p65 for binding to the transcriptional co-activator CBP-p300 complex. Overexpression of p65 limits the availability of CBR for Nrf2 interaction. Knockdown of p65 promotes Nrf2 complex formation with CBR | [110,111] |
Degrading IKKβ through ubiquitination by Keap1 | [112] |
Inhibiting effects of NF-κB on Nrf2 | |
Inactivating Nrf2 by inducing cyclooxygenase 2 | [113,114] |
Recruiting MAF BZIP Transcription Factor K (MafK)-associated histone deacetylase 3 (HDAC3) activity to the HO-1 enhancer and deacetylating CBP leading to a suppression of its co-activator activity | [115,116] |
Interacting with CREB-binding protein, the competent Nrf2 coactivator, and inhibiting the transcription of genes regulated by Nrf2 | [111,117,118] |
Decreasing free CBP, a transcriptional co-activator of Nrf2, and promoting phosphorylation of p65. Overexpression of p65 limits the availability of CBR for Nrf2 interaction. Knockdown of p65 promotes Nrf2 complex formation with CBR | [111] |
κB sites in proximal promoter of Nrf2 are believed to be subject to binding and transcription initiation by p65 | [119] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Surai, P.F.; Kochish, I.I.; Kidd, M.T. Redox Homeostasis in Poultry: Regulatory Roles of NF-κB. Antioxidants 2021, 10, 186. https://doi.org/10.3390/antiox10020186
Surai PF, Kochish II, Kidd MT. Redox Homeostasis in Poultry: Regulatory Roles of NF-κB. Antioxidants. 2021; 10(2):186. https://doi.org/10.3390/antiox10020186
Chicago/Turabian StyleSurai, Peter F., Ivan I. Kochish, and Michael T. Kidd. 2021. "Redox Homeostasis in Poultry: Regulatory Roles of NF-κB" Antioxidants 10, no. 2: 186. https://doi.org/10.3390/antiox10020186
APA StyleSurai, P. F., Kochish, I. I., & Kidd, M. T. (2021). Redox Homeostasis in Poultry: Regulatory Roles of NF-κB. Antioxidants, 10(2), 186. https://doi.org/10.3390/antiox10020186