-
LPS from Salmonella enterica serotype Typhimurium (S. Typhimurium, product number: L7261, Sigma-Aldrich Trading Co. Ltd., Shanghai, China) was dissolved in a phosphate-buffered solution (PBS) at 2.5 μg/mL (500 ng/egg). Quercetin (Product number: Q4591, Sigma-Aldrich Trading Co. Ltd., Shanghai, China) was dissolved in 100% ethanol at 50, 100, or 200 μmol/L (10, 20, or 40 nmol/egg).
Specific pathogen-free embryos (weight 56.76 ± 3.32 g) were provided by a chicken breeder (Jinan SAIS Poultry Co. Ltd., Jinan, Shandong, China). The fertilized eggs were individually weighed and divided into ten groups, each group consisting of 4 replicates with 3 eggs per replicate. There were untreated or injected with 0.2 mL/egg of PBS vehicle, LPS (500 ng/egg; 0.2 mL/egg), PBS + ethanol vehicle (0.2 mL each per egg), quercetin + LPS (10, 20, or 40 nmol + LPS 500 ng/egg), and quercetin (10, 20, or 40 nmol/egg) groups. The incubation, weighting, examination, injection of the treatment solution, and taken samples was the same as our previous study[28].
-
The protocols of histopathology were the same as our previous study[28].
-
The protocols of RT-qPCR were the same as our previous study[28]. A total of thirty-one genes were selected to show the cardiac inflammatory factors, autophagy, apoptosis, and pyroptosis (Table 1). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as the housekeeping gene. The relative levels of target mRNA expression were calculated using the 2−∆∆Ct method.
Table 1. Primer used in real-time quantitative polymerase chain reaction
Genes name Primer sequence (5’–3’) Gene bank ID APOA4 F:GAGGTACTTCACTGAGCTGGG R:TGAGCTGCTTGGTGATCTCAG NM_204938 AMPKα1 F:CGGCAGATAAACAGAAGCACGAG; R:CGATTCAGGATCTTCACTGCAAC NM_001039603 AMPKα2 F:CATGGACGTGTTGAAGAGGCAG; R:TTCTCTGGTTTCAGGTCCCTGTGG NM_001039605 ATG5 F:CCCATCCCTGGTCCGTAAC; R:CGGCGGCGTATACGAAGTA NM_001006409 ATG7 F:TGCAGTTTGCTCCCTTCAGT R:TGGGAAACCTGATGGATCGC NM_001396468 Bcl-2 F: TGGCTGCTTTACTCTTGGGG; R:TATCTCGCGGTTGTCGTAGC NM_205339 Beclin-1 F:CCGCTGAAGCTCGATACCTC R:TTCTGGCTGGTGGGATGAAC NM_001006332.1 CASP1 F:CACTTCCACTTCGGATGGCT; R:CCACGAGACAGTATCAGGCG XM_015295935 CASP3 F:ACCGAGATACCGGACTGTCA; R:GCCATGGCTTAGCAACACAC NM_204725 CASP12 F:AATAGTGGGCATCTGGGTCA; R:CGGTGTGATTTAGACCCGTAAGAC [97] Claudin 1 F:CTGGGTCTGGTTGGTGTGTT; R:CGAGCCACTCTGTTGCCATA NM_001013611 Fas F:GTCAGTGCTGCACGAAATGT; R:AACCTCCAAACCGAGTGCTT NM_001199487 Drp1 F: GGCAGTCACAGCAGCTAACA; R:GCATCCATGAGATCCAGCTT NM_001079722 GAPDH F: GAGAAACCAGCCAAGTATGATG; R: CACAGGAGACAACCTGGTCC NM_204305 IFNγ F:CTGACAAGTCAAAGCCGCAC; R:CTTCACGCCATCAGGAAGGT NM_205149 IL-1β F:GCTCAACATTGCGCTGTACC; R:AGGCGGTAGAAGATGAAGCG FJ537850 IL-6 F:ACGAGGAGAAATGCCTGACG; R:CTTCAGATTGGCGAGGAGGG NM_204628 IL-8 F:TGCCAGTGCATTAGCACTCA; R:TTGGCGTCAGCTTCACATCT HM179639 IL-10 F:TGCGAGAAGAGGAGCAAAGC R:AACTCCCCCATGGCTTTGTAG AJ621254 LC3B F:CTTCTTCCTCCTGGTGAACG; R:GCACTCCGAAAGTCTCCTGA NM_001031461 MMP9 F:ACACAGACTCTATGCTGCCTG; R:GAGAGTAGGGCGGGGAAAAT NM_204667 MMP3 F:ATCAGGCTCTACAGTGGTG; R:ATGGGATACATCAAGGCAC XM_025152201 MYD88 F:TTAGTCTTTCCCCAGGGGCT; R:GCCAGTCTTGTCCAGAACCA NM_001030962 NF-κB1 F:TCAACGCAGGACCTAAAGACAT; R:GCAGATAGCCAAGTTCAGGATG NM_001396396 Occludin F:TACATCATGGGCGTCAACCC R:CCAGATCTTACTGCGCGTCT NM_205128 PPARα F:AACGGAGTTCCAATCGCTGG R:GGATGCTGGTGAAAGGGTGT XM_025150258 RIPK1 F:GATCCATTTGCGAAGCTGCC R:CTTAGGCTAATGGCGCTGGT NM_204402 SGLT1 F:GGCCAGGGCTTACTATTGGT R:AGCTTCAGGTATCCGCACAT AJ236903 TLR4 F:GGCTCAACCTCACGTTGGTA; R:AGTCCGTTCTGAAATGCCGT KP410249 ZO-1 F:TATGAAGATCGTGCGCCTCC; R:GAGGTCTGCCATCGTAGCTC XM_015278977 TNFα F:CCCATCCCTGGTCCGTAAC; R:CGGCGGCGTATACGAAGTA MF000729 Note. APOA4: apolipoprotein 4; AMPKα1: Protein kinase AMP-activated catalytic subunit alpha 1; AMPKα2: Protein kinase AMP-activated catalytic subunit alpha 2; ATG5: Autophagy related gene 5; ATG7: Autophagy related gene 7; Bcl-2: B cell CLL/lymphoma 2; CASP1: Caspase 1; CASP3: Caspase 3; CASP12: Caspase 12; Drp1: dynamin 1 like; GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; Fas: Fas cell surface death receptor; IFNγ: Interferon gamma; IL-8: Interleukin-8; IL-1β: Interleukin-1β; IL-6: Interleukin-6; IL-10: Interleukin-10; LC3B: Microtubule associated protein 1 light chain 3 beta; MMP3: Matrix metallopeptidase 3; MMP9: Matrix metallopeptidase 9; MYD88: Myeloid differentiation primary response 88; NF-κB1: Nuclear factor kappa B subunit 1; RIPK1: receptor interacting serine/threonine kinase 1; PPARα: Peroxisome proliferator-activated receptor alpha; SGLT1: Sodium glucose co-transporter1; TLR4: Toll like receptor 4; ZO-1: Zonula occludens-1; TNFα: Tumor necrosis factor alpha. -
Immunohistochemical investigations were carried out using indirect method of peroxidase with a primary antibody specific for CASP1 (caspase 1, anti-CASP1, GB11383, Servicebio, Wuhan servicebio technology CO., LTD), CASP3 (caspase 3, anti-CASP3, GB11532, Servicebio), AMPKα2 (protein kinase AMP-activated catalytic subunit alpha 2, anti-AMPKα2, GB113685, Servicebio), Claudin 1 (anti-Claudin 1, GB113685, Servicebio), LC3-II/I (anti-LC3-II/I, GB11124, Servicebio), MMP9 (matrix metallopeptidase 9, anti-MMP9, GB11132, Servicebio), and TLR4 (anti-TLR4, GB11519, Servicebio). The protocols of immunohistochemiscal investigation were the same as our previous study[28].
-
The protocols of Western blotting analysis were the same as our previous study[28]. Specific antibodies were used to detect the following proteins: TLR4 (1:3000, GB11519, Servicebio), Interferon gamma (IFNγ, 1:3000, GB11107-1, Servicebio), MMP9 (1:3000, GB12132-1, Servicebio), MMP3 (matrix metallopeptidase 3, 1:3000, GB11131, Servicebio), LC3-II/I(1:3000, Servicebio) CASP1 (1:3000, Servicebio), CASP3 (1:3000, GB11383, Servicebio), AMPKα1(Protein kinase AMP- activated catalytic subunit alpha 1, 1:3000, 66536-1-IG, Proteintech, Wuhan, Hubei, China), GAPDH (1:2000, GB15002, Servicebio), and the secondary IgG HRP conjugated-antibody (1:5000; Servicebio). The expression levels of the target proteins were normalized to GAPDH.
-
All data were statistically analyzed using the paired t test and ANOVA method by the SPSS software (version 16.0, SPSS Inc., Chicago, Illinois, USA), and were presented as mean ± standard deviation. Differences at P < 0.01 were indicated as significant.
doi: 10.3967/bes2024.006
Quercetin Alleviates Lipopolysaccharide-Induced Cardiac Inflammation via Inhibiting Autophagy and Programmed Cell Death
-
Abstract:
Objective The aim of this study is to explore the potential modulatory role of quercetin against Endotoxin or lipopolysaccharide (LPS) induced septic cardiac dysfunction. Methods Specific pathogen-free chicken embryos (n = 120) were allocated untreated control, phosphate buffer solution (PBS) vehicle, PBS with ethanol vehicle, LPS (500 ng/egg), LPS with quercetin treatment (10, 20, or 40 nmol/egg, respectively), Quercetin groups (10, 20, or 40 nmol/egg). Fifteen-day-old embryonated eggs were inoculated with abovementioned solutions via the allantoic cavity. At embryonic day 19, the hearts of the embryos were collected for histopathological examination, RNA extraction, real-time polymerase chain reaction, immunohistochemical investigations, and Western blotting. Results They demonstrated that the heart presented inflammatory responses after LPS induction. The LPS-induced higher mRNA expressions of inflammation-related factors (TLR4, TNFα, MYD88, NF-κB1, IFNγ, IL-1β, IL-8, IL-6, IL-10, p38, MMP3, and MMP9) were blocked by quercetin with three dosages. Quercetin significantly decreased immunopositivity to TLR4 and MMP9 in the treatment group when compared with the LPS group. Quercetin significantly decreased protein expressions of TLR4, IFNγ, MMP3, and MMP9 when compared with the LPS group. Quercetin treatment prevented LPS-induced increase in the mRNA expression of Claudin 1 and ZO-1, and significantly decreased protein expression of claudin 1 when compared with the LPS group. Quercetin significantly downregulated autophagy-related gene expressions (PPARα, SGLT1, APOA4, AMPKα1, AMPKα2, ATG5, ATG7, Beclin-1, and LC3B) and programmed cell death (Fas, Bcl-2, CASP1, CASP12, CASP3, and RIPK1) after LPS induction. Quercetin significantly decreased immunopositivity to APOA4, AMPKα2, and LC3-II/LC3-I in the treatment group when compared with the LPS group. Quercetin significantly decreased protein expressions of AMPKα1, LC3-I, and LC3-II. Quercetin significantly decreased the protein expression to CASP1 and CASP3 by immunohistochemical investigation or Western blotting in treatment group when compared with LPS group. Conclusion Quercetin alleviates cardiac inflammation induced by LPS through modulating autophagy, programmed cell death, and myocardiocytes permeability. -
Key words:
- Quercetin /
- Lipopolysaccharide /
- Inflammation /
- Autophagy /
- Programmed cell death /
- Myocardiocytes permeability
The authors declare no conflict of interest.
注释:1) AUTHOR CONTRIBUTIONS: 2) CONFLICTS OF INTEREST: -
Figure 1. The histopathologic changes of the hearts induced by LPS and quercetin in chicken embryos A, control group; B, PBS group; C, PBS + ethanol group; D–F, 10 nmol, 20 nmol or 40 nmol quercetin; G, treatment group (500 ng/egg); H–J, 500 ng LPS +(10 nmol, 20 nmol, or 40 nmol). Scale bar: 40 μm. HB: Heterophilc banded cells, arrow.
S1. Quercetin alleviates the mRNA expressions of inflammatory associated factors induced by LPS in the hearts of chicken embryos. Data are presented as the mean ± SD. *Indicates the significant differences between the PBS group and the LPS group; #Indicates the significant differences between the LPS group and the (LPS + Q) group; ★Indicates the significant differences between the (PBS + ethanol) group and the (LPS + Q) group; ●Indicates the significant differences between the (PBS + ethanol) group and the Q (quercetin) group.
Figure 2. Quercetin alleviates the protein expression of TLR4, MMP9, Claudin 1, IFNγ, and MMP3 induced by LPS in the hearts of chicken embryos by immunohistochemistry and Western blotting. A, E, I: PBS group of TLR4, MMP9, Claudin 1, respectively; B, F, J: LPS group (500 ng LPS/egg) of TLR4, MMP9, Claudin 1, respectively; C, G, K, treatment group (500 ng LPS/egg + 40 nmol quercetin/egg) of TLR4, MMP9, Claudin 1, respectively; D, H, L, relative immunopositive area of TLR4, MMP9, Claudin 1, respectively. Immunopositivity to TLR4, MMP9, Claudin 1 (arrow, brown to yellow). Scale bar: 50 μm. M, the protein expression of TLR4, IFNγ, MMP3 and MMP9; N, relative protein expression of MMP9; O, relative protein expression of MMP3; P, relative protein expression of IFNγ; Q, relative protein expression of TLR4. Data were expressed as the mean ± SD. *Indicates the significant differences between the PBS group and the LPS group; **P < 0.01; ***P < 0.001. #indicates the significant differences between the LPS group and the (LPS + Q) group; Q, Quercetin; #P < 0.05; ##P < 0.01; ###P < 0.001.
S2. Qercetin ameliorates LPS-induced cardiac autophagy and programmed cell death in chicken embryos. The abbreviation and annotation is the same as Table 1 and Figure 2 Data are presented as the mean ± SD. *Indicates the significant differences between the PBS group and the LPS group; #Indicates the significant differences between the LPS group and the (LPS + Q) group; ★Indicates the significant differences between the (PBS + ethanol) group and the (LPS + Q) group; ●Indicates the significant differences between the (PBS + ethanol) group and the Q (quercetin) group.
Figure 3. Quercetin alleviates the protein expression of AMPKα1, AMPKα2, APOA4, LC3-II, and LC3-I induced by LPS in the hearts of chicken embryos by immunohistochemistry and Western blotting. A, E, I: PBS group of AMPKα2, APOA4, and LC3-II/I, respectively; B, F, J: LPS group (500 ng LPS/egg) of AMPKα2, APOA4, and LC3-II/I, respectively; C, G, K, treatment group (500 ng LPS/egg + 40 nmol quercetin/egg) of AMPKα2, APOA4, and LC3-II/I, respectively; D, H, L, relative immunopositive area of AMPKα2, APOA4, and LC3-II/I, respectively. Immunopositivity to AMPKα2, APOA4, and LC3-II/I (arrow, brown to yellow). Scale bar: 50 μm. M, the protein expression of AMPKα1, LC3-I, and LC3-II; N, relative protein expression of LC3-II; O, relative protein expression of LC3-I; P, ratio of relative protein expression of LC3-II/LC3-I; Q, relative protein expression of AMPKα1. Data were expressed as the mean ± SD. *Indicates the significant differences between the PBS group and the LPS group; **P < 0.01; ***P < 0.001. #Indicates the significant differences between the LPS group and the (LPS + Q) group; Q, Quercetin; #P < 0.05; ##P < 0.01; ###P < 0.001.
Figure 4. Quercetin alleviates the protein expression of CASP1 and CASP3 induced by LPS in the hearts of chicken embryos by immunohistochemistry and Western blotting. A, E: PBS group of CASP1 and CASP3, respectively; B, F: LPS group (500 ng LPS/egg) of CASP1 and CASP3, respectively; C, G, treatment group (500 ng LPS/egg + 40 nmol quercetin/egg) of CASP1 and CASP3, respectively; D, H, relative immunopositive area of CASP1 and CASP3, respectively. Immunopositivity to CASP1 and CASP3 (arrow, brown to yellow). Scale bar: 50 μm. I, the protein expression of CASP1 and CASP3; J, relative protein expression of CASP1; K, relative protein expression of CASP3. Data were expressed as the mean ± SD. *Indicates the significant differences between the PBS group and the LPS group; **P < 0.01; ***P < 0.001. #Indicates the significant differences between the LPS group and the (LPS + Q) group; Q, Quercetin; #P < 0.05; ##P < 0.01.
Table 1. Primer used in real-time quantitative polymerase chain reaction
Genes name Primer sequence (5’–3’) Gene bank ID APOA4 F:GAGGTACTTCACTGAGCTGGG R:TGAGCTGCTTGGTGATCTCAG NM_204938 AMPKα1 F:CGGCAGATAAACAGAAGCACGAG; R:CGATTCAGGATCTTCACTGCAAC NM_001039603 AMPKα2 F:CATGGACGTGTTGAAGAGGCAG; R:TTCTCTGGTTTCAGGTCCCTGTGG NM_001039605 ATG5 F:CCCATCCCTGGTCCGTAAC; R:CGGCGGCGTATACGAAGTA NM_001006409 ATG7 F:TGCAGTTTGCTCCCTTCAGT R:TGGGAAACCTGATGGATCGC NM_001396468 Bcl-2 F: TGGCTGCTTTACTCTTGGGG; R:TATCTCGCGGTTGTCGTAGC NM_205339 Beclin-1 F:CCGCTGAAGCTCGATACCTC R:TTCTGGCTGGTGGGATGAAC NM_001006332.1 CASP1 F:CACTTCCACTTCGGATGGCT; R:CCACGAGACAGTATCAGGCG XM_015295935 CASP3 F:ACCGAGATACCGGACTGTCA; R:GCCATGGCTTAGCAACACAC NM_204725 CASP12 F:AATAGTGGGCATCTGGGTCA; R:CGGTGTGATTTAGACCCGTAAGAC [97] Claudin 1 F:CTGGGTCTGGTTGGTGTGTT; R:CGAGCCACTCTGTTGCCATA NM_001013611 Fas F:GTCAGTGCTGCACGAAATGT; R:AACCTCCAAACCGAGTGCTT NM_001199487 Drp1 F: GGCAGTCACAGCAGCTAACA; R:GCATCCATGAGATCCAGCTT NM_001079722 GAPDH F: GAGAAACCAGCCAAGTATGATG; R: CACAGGAGACAACCTGGTCC NM_204305 IFNγ F:CTGACAAGTCAAAGCCGCAC; R:CTTCACGCCATCAGGAAGGT NM_205149 IL-1β F:GCTCAACATTGCGCTGTACC; R:AGGCGGTAGAAGATGAAGCG FJ537850 IL-6 F:ACGAGGAGAAATGCCTGACG; R:CTTCAGATTGGCGAGGAGGG NM_204628 IL-8 F:TGCCAGTGCATTAGCACTCA; R:TTGGCGTCAGCTTCACATCT HM179639 IL-10 F:TGCGAGAAGAGGAGCAAAGC R:AACTCCCCCATGGCTTTGTAG AJ621254 LC3B F:CTTCTTCCTCCTGGTGAACG; R:GCACTCCGAAAGTCTCCTGA NM_001031461 MMP9 F:ACACAGACTCTATGCTGCCTG; R:GAGAGTAGGGCGGGGAAAAT NM_204667 MMP3 F:ATCAGGCTCTACAGTGGTG; R:ATGGGATACATCAAGGCAC XM_025152201 MYD88 F:TTAGTCTTTCCCCAGGGGCT; R:GCCAGTCTTGTCCAGAACCA NM_001030962 NF-κB1 F:TCAACGCAGGACCTAAAGACAT; R:GCAGATAGCCAAGTTCAGGATG NM_001396396 Occludin F:TACATCATGGGCGTCAACCC R:CCAGATCTTACTGCGCGTCT NM_205128 PPARα F:AACGGAGTTCCAATCGCTGG R:GGATGCTGGTGAAAGGGTGT XM_025150258 RIPK1 F:GATCCATTTGCGAAGCTGCC R:CTTAGGCTAATGGCGCTGGT NM_204402 SGLT1 F:GGCCAGGGCTTACTATTGGT R:AGCTTCAGGTATCCGCACAT AJ236903 TLR4 F:GGCTCAACCTCACGTTGGTA; R:AGTCCGTTCTGAAATGCCGT KP410249 ZO-1 F:TATGAAGATCGTGCGCCTCC; R:GAGGTCTGCCATCGTAGCTC XM_015278977 TNFα F:CCCATCCCTGGTCCGTAAC; R:CGGCGGCGTATACGAAGTA MF000729 Note. APOA4: apolipoprotein 4; AMPKα1: Protein kinase AMP-activated catalytic subunit alpha 1; AMPKα2: Protein kinase AMP-activated catalytic subunit alpha 2; ATG5: Autophagy related gene 5; ATG7: Autophagy related gene 7; Bcl-2: B cell CLL/lymphoma 2; CASP1: Caspase 1; CASP3: Caspase 3; CASP12: Caspase 12; Drp1: dynamin 1 like; GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; Fas: Fas cell surface death receptor; IFNγ: Interferon gamma; IL-8: Interleukin-8; IL-1β: Interleukin-1β; IL-6: Interleukin-6; IL-10: Interleukin-10; LC3B: Microtubule associated protein 1 light chain 3 beta; MMP3: Matrix metallopeptidase 3; MMP9: Matrix metallopeptidase 9; MYD88: Myeloid differentiation primary response 88; NF-κB1: Nuclear factor kappa B subunit 1; RIPK1: receptor interacting serine/threonine kinase 1; PPARα: Peroxisome proliferator-activated receptor alpha; SGLT1: Sodium glucose co-transporter1; TLR4: Toll like receptor 4; ZO-1: Zonula occludens-1; TNFα: Tumor necrosis factor alpha. -
[1] Schlapbach LJ, Kissoon N, Alhawsawi A, et al. World Sepsis Day: a global agenda to target a leading cause of morbidity and mortality. Am J Physiol Lung Cell Mol Physiol, 2020; 319, L518−22. doi: 10.1152/ajplung.00369.2020 [2] Balayan S, Chauhan N, Chandra R, et al. Recent advances in developing biosensing based platforms for neonatal sepsis. Biosens Bioelectron, 2020; 169, 112552. doi: 10.1016/j.bios.2020.112552 [3] McNamara JF, Harris PNA, Chatfield MD, et al. Long term sepsis readmission, mortality and cause of death following Gram negative bloodstream infection: a propensity matched observational linkage study. Int J Infect Dis, 2022; 114, 34−44. doi: 10.1016/j.ijid.2021.10.047 [4] Chousalkar K, Gast R, Martelli F, et al. Review of egg-related salmonellosis and reduction strategies in United States, Australia, United Kingdom and New Zealand. Crit Rev Microbiol, 2018; 44, 290−303. doi: 10.1080/1040841X.2017.1368998 [5] Chroni A, Rallidis L, Vassou D, et al. Identification and characterization of a rare variant in apolipoprotein A-IV, p. (V336M), and evaluation of HDL functionality in a Greek cohort with extreme HDL cholesterol levels. Arch Biochem Biophys, 2020; 696, 108655. doi: 10.1016/j.abb.2020.108655 [6] Akkoyun DÇ, Akyuz A, Doğan M, et al. Quercetin inhibits heart injury in lipopolysaccharide-induced endotoxemic model by suppressing the effects of reactive oxygen species. Anal Quant Cytopathol Histopathol, 2016; 38, 183−8. [7] Wei XQ, Meng XL, Yuan YX, et al. Quercetin exerts cardiovascular protective effects in LPS-induced dysfunction in vivo by regulating inflammatory cytokine expression, NF-κB phosphorylation, and caspase activity. Mol Cell Biochem, 2018; 446, 43−52. doi: 10.1007/s11010-018-3271-6 [8] Liu G, Wu KJ, Zhang L, et al. Metformin attenuated endotoxin-induced acute myocarditis via activating AMPK. Int Immunopharmacol, 2017; 47, 166−72. doi: 10.1016/j.intimp.2017.04.002 [9] Panaro MA, Acquafredda A, Cavallo P, et al. Inflammatory responses in embryonal cardiomyocytes exposed to LPS challenge: an in vitro model of deciphering the effects of LPS on the heart. Curr Pharm Des, 2010; 16, 754−65. doi: 10.2174/138161210790883516 [10] Shen L, Weber CR, Turner JR. The tight junction protein complex undergoes rapid and continuous molecular remodeling at steady state. J Cell Biol, 2008; 181, 683−95. doi: 10.1083/jcb.200711165 [11] Mokhtari B, Badalzadeh R. The potentials of distinct functions of autophagy to be targeted for attenuation of myocardial ischemia/reperfusion injury in preclinical studies: an up-to-date review. J Physiol Biochem, 2021; 77, 377−404. doi: 10.1007/s13105-021-00824-x [12] Larsen KE, Sulzer D. Autophagy in neurons: a review. Histol Histopathol, 2002; 17, 897−908. [13] Zhang C, Syed TW, Liu RJ, et al. Role of endoplasmic reticulum stress, autophagy, and inflammation in cardiovascular disease. Front Cardiovasc Med, 2017; 4, 29. doi: 10.3389/fcvm.2017.00029 [14] Mechesso AF, Quah YX, Park SC. Ginsenoside Rg3 reduces the adhesion, invasion, and intracellular survival of Salmonella enterica serovar Typhimurium. J Ginseng Res, 2021; 45, 75−85. doi: 10.1016/j.jgr.2019.09.002 [15] Chiu B, Jantuan E, Shen F, et al. Autophagy-inflammasome interplay in heart failure: a systematic review on basics, pathways, and therapeutic perspectives. Ann Clin Lab Sci, 2017; 47, 243−52. [16] Ashrafizadeh M, Tavakol S, Ahmadi Z, et al. Therapeutic effects of kaempferol affecting autophagy and endoplasmic reticulum stress. Phytother Res, 2020; 34, 911−23. doi: 10.1002/ptr.6577 [17] Mohammadinejad R, Ahmadi Z, Tavakol S, et al. Berberine as a potential autophagy modulator. J Cell Physiol, 2019; 234, 14914−26. doi: 10.1002/jcp.28325 [18] Nagata S, Nakano H. Apoptotic and non-apoptotic cell death. Springer. 2017. [19] Mughal W, Dhingra R, Kirshenbaum LA. Striking a balance: autophagy, apoptosis, and necrosis in a normal and failing heart. Curr Hypertens Rep, 2012; 14, 540−7. doi: 10.1007/s11906-012-0304-5 [20] Cong L, Bai YP, Guo ZG. The crosstalk among autophagy, apoptosis, and pyroptosis in cardiovascular disease. Front Cardiovasc Med, 2022; 9, 997469. doi: 10.3389/fcvm.2022.997469 [21] Dong Y, Chen HW, Gao JL, et al. Molecular machinery and interplay of apoptosis and autophagy in coronary heart disease. J Mol Cell Cardiol, 2019; 136, 27−41. doi: 10.1016/j.yjmcc.2019.09.001 [22] Zheng XT, Chen WW, Gong FC, et al. The role and mechanism of pyroptosis and potential therapeutic targets in sepsis: a review. Front Immunol, 2021; 12, 711939. doi: 10.3389/fimmu.2021.711939 [23] Wei SQ, Feng M, Zhang SD. Molecular characteristics of cell pyroptosis and its inhibitors: a review of activation, regulation, and inhibitors. Int J Mol Sci, 2022; 23, 16115. doi: 10.3390/ijms232416115 [24] Jia Y, Li DZ, Yu J, et al. Potential diabetic cardiomyopathy therapies targeting pyroptosis: a mini review. Front Cardiovasc Med, 2022; 9, 985020. doi: 10.3389/fcvm.2022.985020 [25] Zhang P, Zang MR, Sang ZZ, et al. Vitamin C alleviates LPS-induced myocardial injury by inhibiting pyroptosis via the ROS-AKT/mTOR signalling pathway. BMC Cardiovasc Disord, 2022; 22, 561. doi: 10.1186/s12872-022-03014-9 [26] Li Q, Zhang MM, Zhao Y, et al. Irisin protects against LPS-stressed cardiac damage through inhibiting inflammation, apoptosis, and pyroptosis. SHOCK, 2021; 56, 1009−18. doi: 10.1097/SHK.0000000000001775 [27] Ouyang CH, You JY, Xie ZL. The interplay between autophagy and apoptosis in the diabetic heart. J Mol Cell Cardiol, 2014; 71, 71−80. doi: 10.1016/j.yjmcc.2013.10.014 [28] Yu JH, Hu GL, Cao HB, et al. Quercetin ameliorates lipopolysaccharide-induced duodenal inflammation through modulating autophagy, programmed cell death and intestinal mucosal barrier function in chicken embryos. Animals, 2022; 12, 3524. doi: 10.3390/ani12243524 [29] Afrin S, Gasparrini M, Forbes-Hernandez TY, et al. Protective effects of Manuka honey on LPS-treated RAW 264.7 macrophages. Part 1: Enhancement of cellular viability, regulation of cellular apoptosis and improvement of mitochondrial functionality. Food Chem Toxicol, 2018; 121, 203−13. doi: 10.1016/j.fct.2018.09.001 [30] Häkkinen SH, Kärenlampi SO, Heinonen IM, et al. Content of the flavonols quercetin, myricetin, and kaempferol in 25 edible berries. J Agric Food Chem, 1999; 47, 2274−9. doi: 10.1021/jf9811065 [31] Gasparrini M, Afrin S, Forbes-Hernández TY, et al. Protective effects of Manuka honey on LPS-treated RAW 264.7 macrophages. Part 2: Control of oxidative stress induced damage, increase of antioxidant enzyme activities and attenuation of inflammation. Food Chem Toxicol, 2018; 120, 578−87. doi: 10.1016/j.fct.2018.08.001 [32] Gasparrini M, Forbes-Hernandez TY, Giampieri F, et al. Anti-inflammatory effect of strawberry extract against LPS-induced stress in RAW 264.7 macrophages. Food Chem Toxicol, 2017; 102, 1−10. doi: 10.1016/j.fct.2017.01.018 [33] Ulusoy HG, Sanlier N. A minireview of quercetin: from its metabolism to possible mechanisms of its biological activities. Crit Rev Food Sci Nutr, 2020; 60, 3290−303. doi: 10.1080/10408398.2019.1683810 [34] Lotfi N, Yousefi Z, Golabi M, et al. The potential anti-cancer effects of quercetin on blood, prostate and lung cancers: an update. Front Immunol, 2023; 14, 1077531. doi: 10.3389/fimmu.2023.1077531 [35] Paskeh MDA, Entezari M, Clark C, et al. Targeted regulation of autophagy using nanoparticles: new insight into cancer therapy. Biochim Biophys Acta Mol Basis Dis, 2022; 1868, 166326. doi: 10.1016/j.bbadis.2021.166326 [36] Ashrafizadeh M, Paskeh MDA, Mirzaei S, et al. Targeting autophagy in prostate cancer: preclinical and clinical evidence for therapeutic response. J Exp Clin Cancer Res, 2022; 41, 105. doi: 10.1186/s13046-022-02293-6 [37] Liu J, Wang SC, Zhang QJ, et al. Selenomethionine alleviates LPS-induced chicken myocardial inflammation by regulating the miR-128-3p-p38 MAPK axis and oxidative stress. Metallomics, 2020; 12, 54−64. doi: 10.1039/c9mt00216b [38] Frantz S, Kobzik L, Kim YD, et al. Toll4 (TLR4) expression in cardiac myocytes in normal and failing myocardium. J Clin Invest, 1999; 104, 271−80. doi: 10.1172/JCI6709 [39] Zanoni I, Bodio C, Broggi A, et al. Similarities and differences of innate immune responses elicited by smooth and rough LPS. Immunol Lett, 2012; 142, 41−7. doi: 10.1016/j.imlet.2011.12.002 [40] Kogut M, He HQ, Kaiser P. Lipopolysaccharide binding protein/CD14/TLR4-dependent recognition of salmonella LPS induces the functional activation of chicken heterophils and up-regulation of pro-inflammatory cytokine and chemokine gene expression in these cells. Anim Biotechnol, 2005; 16, 165−81. doi: 10.1080/10495390500264896 [41] Panaro MA, Cianciulli A, Gagliardi N, et al. CD14 major role during lipopolysaccharide-induced inflammation in chick embryo cardiomyocytes. FEMS Immunol Med Microbiol, 2008; 53, 35−45. doi: 10.1111/j.1574-695X.2008.00397.x [42] Rossetti C, Peri F. The role of toll-like receptor 4 in infectious and non Infectious Inflammation. Springer. 2021. [43] De Vicente LG, Muñoz VR, Pinto AP, et al. TLR4 deletion increases basal energy expenditure and attenuates heart apoptosis and ER stress but mitigates the training-induced cardiac function and performance improvement. Life Sci, 2021; 285, 119988. doi: 10.1016/j.lfs.2021.119988 [44] Karnati HK, Pasupuleti SR, Kandi R, et al. TLR-4 signalling pathway: MyD88 independent pathway up-regulation in chicken breeds upon LPS treatment. Vet Res Commun, 2015; 39, 73−8. doi: 10.1007/s11259-014-9621-2 [45] Wang XP, Guo DQ, Li WL, et al. Danshen (Salvia miltiorrhiza) restricts MD2/TLR4-MyD88 complex formation and signalling in acute myocardial infarction-induced heart failure. J Cell Mol Med, 2020; 24, 10677−92. doi: 10.1111/jcmm.15688 [46] Zhang LL, Zhang C, Peng JP. Application of nanopore sequencing technology in the clinical diagnosis of infectious diseases. Biomed Environ Sci, 2022; 35, 381−92. [47] Kogut MH, Rothwell L, Kaiser P. Priming by recombinant chicken interleukin-2 induces selective expression of IL-8 and IL-18 mRNA in chicken heterophils during receptor-mediated phagocytosis of opsonized and nonopsonized Salmonella enterica serovar enteritidis. Mol Immunol, 2003; 40, 603−10. doi: 10.1016/j.molimm.2003.08.002 [48] Akseh S, Nemati M, Zamani-Gharehchamani E, et al. Amnion membrane proteins attenuate LPS-induced inflammation and apoptosis by inhibiting TLR4/NF-κB pathway and repressing MicroRNA-155 in rat H9c2 cells. Immunopharmacol Immunotoxicol, 2021; 43, 487−94. doi: 10.1080/08923973.2021.1945086 [49] Handley SA, Miller VL. General and specific host responses to bacterial infection in Peyer's patches: a role for stromelysin-1 (matrix metalloproteinase-3) during Salmonella enterica infection. Mol Microbiol, 2007; 64, 94−110. doi: 10.1111/j.1365-2958.2007.05635.x [50] Lalu MM, Csont T, Schulz R. Matrix metalloproteinase activities are altered in the heart and plasma during endotoxemia. Crit Care Med, 2004; 32, 1332−7. doi: 10.1097/01.CCM.0000127778.16609.EC [51] Iriti M, Kubina R, Cochis A, et al. Rutin, a quercetin glycoside, restores chemosensitivity in human breast cancer cells. Phytother Res, 2017; 31, 1529−38. doi: 10.1002/ptr.5878 [52] Liu XC, Zheng L, Liu M, et al. Protective effects of rutin on lipopolysaccharide-induced heart injury in mice. J Toxicol Sci, 2018; 43, 329−37. doi: 10.2131/jts.43.329 [53] Shu J, Gu YW, Jin L, et al. Matrix metalloproteinase 3 regulates angiotensin II-induced myocardial fibrosis cell viability, migration and apoptosis. Mol Med Rep, 2021; 23, 151. [54] Collins MM, Ryan AK. Manipulating claudin expression in avian embryos. Methods Mol Biol, 2011; 762, 195−212. [55] Simard A, Di Pietro E, Ryan AK. Gene expression pattern of Claudin-1 during chick embryogenesis. Gene Expr Patterns, 2005; 5, 553−60. doi: 10.1016/j.modgep.2004.10.009 [56] Simard A, Di Pietro E, Young CR, et al. Alterations in heart looping induced by overexpression of the tight junction protein Claudin-1 are dependent on its C-terminal cytoplasmic tail. Mech Dev, 2006; 123, 210−27. doi: 10.1016/j.mod.2005.12.004 [57] Kostin S. Zonula occludens-1 and connexin 43 expression in the failing human heart. J Cell Mol Med, 2007; 11, 892−5. doi: 10.1111/j.1582-4934.2007.00063.x [58] El Refaey M, Coles S, Musa H, et al. Altered expression of zonula occludens-1 affects cardiac Na+ channels and increases susceptibility to ventricular arrhythmias. Cells, 2022; 11, 665. doi: 10.3390/cells11040665 [59] Zhang JL, Vincent KP, Peter AK, et al. Cardiomyocyte expression of ZO-1 is essential for normal atrioventricular conduction but does not alter ventricular function. Circ Res, 2020; 127, 284−97. doi: 10.1161/CIRCRESAHA.119.315539 [60] Chen Y, Liu YQ, Dorn II GW. Mitochondrial fusion is essential for organelle function and cardiac homeostasis. Circ Res, 2011; 109, 1327−31. doi: 10.1161/CIRCRESAHA.111.258723 [61] Yu WC, Mei X, Zhang Q, et al. Yap overexpression attenuates septic cardiomyopathy by inhibiting DRP1-related mitochondrial fission and activating the ERK signaling pathway. J Recept Signal Transduct, 2019; 39, 175−86. doi: 10.1080/10799893.2019.1641822 [62] Bullon P, Cordero MD, Quiles JL, et al. Mitochondrial dysfunction promoted by Porphyromonas gingivalis lipopolysaccharide as a possible link between cardiovascular disease and periodontitis. Free Radic Biol Med, 2011; 50, 1336−43. doi: 10.1016/j.freeradbiomed.2011.02.018 [63] Xu T, Dong Q, Luo YX, et al. Porphyromonas gingivalis infection promotes mitochondrial dysfunction through Drp1-dependent mitochondrial fission in endothelial cells. Int J Oral Sci, 2021; 13, 28. doi: 10.1038/s41368-021-00134-4 [64] Tong MM, Zablocki D, Sadoshima J. The role of Drp1 in mitophagy and cell death in the heart. J Mol Cell Cardiol, 2020; 142, 138−45. doi: 10.1016/j.yjmcc.2020.04.015 [65] Zhao QC, Yan S, Lu J, et al. Drp1 regulates transcription of ribosomal protein genes in embryonic hearts. J Cell Sci, 2022; 135, jcs258956. doi: 10.1242/jcs.258956 [66] Sharp WW, Fang YH, Han M, et al. Dynamin-related protein 1 (Drp1)-mediated diastolic dysfunction in myocardial ischemia-reperfusion injury: therapeutic benefits of Drp1 inhibition to reduce mitochondrial fission. FASEB J, 2014; 28, 316−26. doi: 10.1096/fj.12-226225 [67] Ikeda Y, Shirakabe A, Maejima Y, et al. Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ Res, 2015; 116, 264−78. doi: 10.1161/CIRCRESAHA.116.303356 [68] Ren J, Sowers JR, Zhang YM. Autophagy and cardiometabolic diseases. Academic Press. 2018. [69] Chang X, He YH, Wang L, et al. Puerarin alleviates LPS-induced H9C2 cell injury by inducing mitochondrial autophagy. J Cardiovasc Pharmacol, 2022; 80, 600−8. [70] Ben-Shaul V, Lomnitski L, Nyska A, et al. The effect of natural antioxidants, NAO and apocynin, on oxidative stress in the rat heart following LPS challenge. Toxicol Lett, 2001; 123, 1−10. [71] Sul OJ, Ra SW. Quercetin prevents LPS-induced oxidative stress and inflammation by modulating NOX2/ROS/NF-kB in lung epithelial cells. Molecules, 2021; 26, 6949. doi: 10.3390/molecules26226949 [72] Lushnikova EL, Nepomnyashchikh LM, Pichigin VI, et al. Expression of mRNA of apolipoprotein E, apolipoprotein A-IV, and matricellular proteins in the myocardium and intensity of fibroplastic processes during experimental hypercholesterolemia. Bull Exp Biol Med, 2013; 156, 271−5. doi: 10.1007/s10517-013-2328-5 [73] Zhang WQ, Liu XH, Zhou JT, et al. Apolipoprotein A-IV restrains fat accumulation in skeletal and myocardial muscles by inhibiting lipogenesis and activating PI3K-AKT signalling. Arch Physiol Biochem, 2023, 1-11. [74] Wang Y, Zhang ZZ, Wu Y, et al. Quercetin postconditioning attenuates myocardial ischemia/reperfusion injury in rats through the PI3K/Akt pathway. Braz J Med Biol Res, 2013; 46, 861−7. doi: 10.1590/1414-431X20133036 [75] Ravindranath TM, Goto M, Bakr S, et al. LPS-induced changes in myocardial markers in neonatal rats. Biol Neonate, 2003; 84, 319−24. doi: 10.1159/000073641 [76] Lado-Abeal J, Martinez-Sánchez N, Cocho JA, et al. Lipopolysaccharide (LPS)-induced septic shock causes profound changes in myocardial energy metabolites in pigs. Metabolomics, 2018; 14, 131. doi: 10.1007/s11306-018-1433-x [77] Yamaguchi O. Autophagy in the Heart. Circ J, 2019; 83, 697−704. doi: 10.1253/circj.CJ-18-1065 [78] Drosatos K, Pollak NM, Pol CJ, et al. Cardiac myocyte KLF5 regulates Ppara expression and cardiac function. Circ Res, 2016; 118, 241−53. doi: 10.1161/CIRCRESAHA.115.306383 [79] Kumari R, Ray AG, Mukherjee D, et al. Downregulation of PTEN promotes autophagy via concurrent reduction in apoptosis in cardiac hypertrophy in PPAR α-/- mice. Front Cardiovasc Med, 2022; 9, 798639. doi: 10.3389/fcvm.2022.798639 [80] Saikia R, Joseph J. AMPK: a key regulator of energy stress and calcium-induced autophagy. J Mol Med, 2021; 99, 1539−51. doi: 10.1007/s00109-021-02125-8 [81] Eisenberg-Lerner A, Bialik S, Simon HU, et al. Life and death partners: apoptosis, autophagy and the cross-talk between them. Cell Death Differ, 2009; 16, 966−75. doi: 10.1038/cdd.2009.33 [82] Kajstura J, Mansukhani M, Cheng W, et al. Programmed cell death and expression of the protooncogene bcl-2 in myocytes during postnatal maturation of the heart. Exp Cell Res, 1995; 219, 110−21. doi: 10.1006/excr.1995.1211 [83] Grzegorzewska AK, Hrabia A, Paczoska-Eliasiewicz HE. Localization of apoptotic and proliferating cells and mRNA expression of caspases and Bcl-2 in gonads of chicken embryos. Acta Histochem, 2014; 116, 795−802. doi: 10.1016/j.acthis.2014.01.012 [84] Sun YX, Cai Y, Qian SH, et al. Beclin-1 improves mitochondria-associated membranes in the heart during endotoxemia. FASEB Bioadv, 2021; 3, 123−35. doi: 10.1096/fba.2020-00039 [85] Sun YX, Yao X, Zhang QJ, et al. Beclin-1-dependent autophagy protects the heart during sepsis. Circulation, 2018; 138, 2247−62. doi: 10.1161/CIRCULATIONAHA.117.032821 [86] Maejima Y, Isobe M, Sadoshima J. Regulation of autophagy by Beclin 1 in the heart. J Mol Cell Cardiol, 2016; 95, 19−25. doi: 10.1016/j.yjmcc.2015.10.032 [87] Haq S, Grondin J, Banskota S, et al. Autophagy: roles in intestinal mucosal homeostasis and inflammation. J Biomed Sci, 2019; 26, 19. doi: 10.1186/s12929-019-0512-2 [88] Liao ZH, Dai ZK, Cai CY, et al. Knockout of Atg5 inhibits proliferation and promotes apoptosis of DF-1 cells. In Vitro Cell Dev Biol Anim, 2019; 55, 341−8. doi: 10.1007/s11626-019-00342-7 [89] Zhu CM, Zhang SY, Liu D, et al. A novel gene prognostic signature based on differential DNA methylation in breast cancer. Front Genet, 2021; 12, 742578. doi: 10.3389/fgene.2021.742578 [90] Liu L, Chen ML, Lin K, et al. Inhibiting caspase-12 mediated inflammasome activation protects against oxygen-glucose deprivation injury in primary astrocytes. Int J Med Sci, 2020; 17, 1936−45. doi: 10.7150/ijms.44330 [91] Nakagawa T, Zhu H, Morishima N, et al. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-β. Nature, 2000; 403, 98−103. doi: 10.1038/47513 [92] Morishima N, Nakanishi K, Takenouchi H, et al. An endoplasmic reticulum stress-specific caspase cascade in apoptosis. Cytochrome c-independent activation of caspase-9 by caspase-12. J Biol Chem, 2002; 277, 34287−94. doi: 10.1074/jbc.M204973200 [93] Salvamoser R, Brinkmann K, O'Reilly LA, et al. Characterisation of mice lacking the inflammatory caspases-1/11/12 reveals no contribution of caspase-12 to cell death and sepsis. Cell Death Differ, 2019; 26, 1124−37. doi: 10.1038/s41418-018-0188-2 [94] Liu HH, Wang SJ, Gong LJ, et al. SIRT6 ameliorates LPS-induced apoptosis and tight junction injury in ARDS through the ERK1/2 pathway and autophagy. Int J Med Sci, 2023; 20, 581−94. doi: 10.7150/ijms.80920 [95] Wang JY, Luan YY, Fan EK, et al. TBK1/IKKε negatively regulate lps-induced neutrophil necroptosis and lung inflammation. SHOCK, 2021; 55, 338−48. doi: 10.1097/SHK.0000000000001632 [96] Liu YL, Hsu CC, Huang HJ, et al. Gallic acid attenuated LPS-induced neuroinflammation: protein aggregation and necroptosis. Mol Neurobiol, 2020; 57, 96−104. doi: 10.1007/s12035-019-01759-7 [97] Wang YC, Jiang L, Li YF, et al. Excessive selenium supplementation induced oxidative stress and endoplasmic reticulum stress in chicken spleen. Biol Trace Elem Res, 2016; 172, 481−7. doi: 10.1007/s12011-015-0596-9 -
23031+Supplementary Materials.pdf