339
Views
0
CrossRef citations to date
0
Altmetric
Research Article

TLR4 signalling protects Sertoli cells from cell stress via reprogramming inflammasome and autophagy pathways in MAPK1/ERK2 dependent way

, , &
Article: 2268749 | Received 18 Jul 2023, Accepted 04 Oct 2023, Published online: 18 Oct 2023

References

  • Setchell BP. Hormones: what the testis really sees. Reprod Fertil Dev. 2004;16(5):1–14. doi: 10.10371/RD03048.
  • McCabe MJ, Allan CM, Foo CFH, et al. Androgen initiates sertoli cell tight junction formation in the hypogonadal (hpg) Mouse1. Biol Reprod. 2012;87(2):38. doi: 10.1095/biolreprod.111.094318.
  • Sarkar O, Mathur PP, Cheng CY, et al. Interleukin 1 alpha (IL1A) is a novel regulator of the blood-testis barrier in the rat. Biol Reprod. 2008;78(3):445–454. doi: 10.1095/biolreprod.107.064501.
  • Lie PPY, Cheng CY, Mruk DD. Interleukin-1alpha is a regulator of the blood-testis barrier. Faseb J. 2011;25(4):1244–1253. doi: 10.1096/fj.10-169995.
  • Al-Sadi R, Ye D, Said HM, et al. IL-1β-induced increase in intestinal epithelial tight junction permeability is mediated by MEKK-1 activation of canonical NF-κB pathway. Am J Pathol. 2010;177(5):2310–2322. doi: 10.2353/ajpath.2010.100371.
  • Al-Sadi R, Ye D, Dokladny K, et al. Mechanism of IL-1beta-induced increase in intestinal epithelial tight junction permeability. J Immunol. 2008;180(8):5653–5661. doi: 10.4049/jimmunol.180.8.5653.
  • Crépieux P, Marion S, Martinat N, et al. The ERK-dependent signalling is stage-specifically modulated by FSH, during primary sertoli cell maturation. Oncogene. 2001;20(34):4696–4709. doi: 10.1038/sj.onc.1204632.
  • Petersen C, Svechnikov K, Froysa B, et al. The p38 MAPK pathway mediates interleukin-1-induced sertoli cell proliferation. Cytokine. 2005;32(1):51–59. doi: 10.1016/j.cyto.2005.07.014.
  • Wong CH, Yan Cheng C. Mitogen-activated protein kinases, adherens junction dynamics, and spermatogenesis: a review of recent data. Dev Biol. 2005;286(1):1–15. doi: 10.1016/j.ydbio.2005.08.001.
  • Lu Y, Luo B, Li J, et al. Perfluorooctanoic acid disrupts the blood–testis barrier and activates the TNFα/p38 MAPK signaling pathway in vivo and in vitro. Arch Toxicol. 2016;90(4):971–983. doi: 10.1007/s00204-015-1492-y.
  • Zanatta AP, Gonçalves R, Zanatta L, et al. New ionic targets of 3,3′,5′-triiodothyronine at the plasma membrane of rat sertoli cells. Biochim Biophys Acta Biomembr. 2019;1861(4):748–759. doi: 10.1016/j.bbamem.2019.01.002.
  • Rato L, Socorro S, Cavaco JEB, et al. Tubular fluid secretion in the seminiferous epithelium: ion transporters and aquaporins in sertoli cells. J Membr Biol. 2010;236(2):215–224. doi: 10.1007/s00232-010-9294-x.
  • Pérez-García MT, Cidad P, López-López JR. The secret life of ion channels: kv1.3 potassium channels and proliferation. Am J Physiol Cell Physiol. 2018;314(1):C27–C42. doi: 10.1152/ajpcell.00136.2017.
  • Cidad P, Jiménez-Pérez L, García-Arribas D, et al. Kv1.3 channels can modulate cell proliferation during phenotypic switch by an ion-flux independent mechanism. Arterioscler Thromb Vasc Biol. 2012;32(5):1299–1307. doi: 10.1161/ATVBAHA.111.242727.
  • Hayrabedyan S, Todorova K, Jabeen A, et al. Sertoli cells have a functional NALP3 inflammasome that can modulate autophagy and cytokine production. Sci Rep. 2016;6(1):18896. doi: 10.1038/srep18896.
  • Walenta L, Schmid N, Schwarzer JU, et al. NLRP3 in somatic non-immune cells of rodent and primate testes. Reproduction. 2018;156(3):231–238. doi: 10.1530/REP-18-0111.
  • Hayrabedyan S, Todorova K. Nalp signalling is required in sertoli cells for tight-junction protein interaction. Acta Medica Bulgarica. 2015;42(1):12–17. doi: 10.1515/amb-2015-0002.
  • Hayrabedyan SB, Zasheva DY, Todorova KO. NLRs challenge impacts tight junction claudins in sertoli cells. Folia Med (Plovdiv). 2015;57(1):43–48. doi: 10.1515/folmed-2015-0018.
  • Di Persio S, Starace D, Capponi C, et al. TNF-α inhibits GDNF levels in sertoli cells, through a NF-κB-dependent, HES1-dependent mechanism. Andrology. 2021;9(3):956–964. doi: 10.1111/andr.12959.
  • Travassos LH, Carneiro LAM, Ramjeet M, et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat Immunol. 2010;11(1):55–62. doi: 10.1038/ni.1823.
  • Meroni SB, Galardo MN, Rindone G, et al. Molecular mechanisms and signaling pathways involved in sertoli cell proliferation. Front Endocrinol (Lausanne). 2019;10:224. doi: 10.3389/fendo.2019.00224.
  • Rassoulzadegan M, Paquis-Flucklinger V, Bertino B, et al. Transmeiotic differentiation of male germ cells in culture. Cell. 1993;75(5):997–1006. doi: 10.1016/0092-8674(93)90543-y.
  • Grada A, Otero-Vinas M, Prieto-Castrillo F, et al. Research techniques made simple: analysis of collective cell migration using the wound healing assay. J Invest Dermatol. 2017;137(2):e11–e16. doi: 10.1016/j.jid.2016.11.020.
  • Iwashita H, Torii S, Nagahora N, et al. Live cell imaging of mitochondrial autophagy with a novel fluorescent small molecule. ACS Chem Biol. 2017;12(10):2546–2551. doi: 10.1021/acschembio.7b00647.
  • Chih Chen Y, Rivera J, Fitzgerald M, et al. PreImplantation factor prevents atherosclerosis via its immunomodulatory effects without affecting serum lipids. Thromb Haemost. 2016;115(5):1010–1024. doi: 10.1160/TH15-08-0640.
  • Beacham DW, Blackmer T, Grady O’, et al. Cell-based potassium ion channel screening using the FluxORTM assay. J Biomol Screen. 2010;15(4):441–446. doi: 10.1177/1087057109359807.
  • Pike AF, Szabò I, Veerhuis R, et al. The potential convergence of NLRP3 inflammasome, potassium, and dopamine mechanisms in Parkinson’s disease. NPJ Parkinsons Dis. 2022;8(1):32. doi: 10.1038/s41531-022-00293-z.
  • Yuan X, Han S, Manyande A, et al. Spinal voltage-gated potassium channel Kv1.3 contributes to neuropathic pain via the promotion of microglial < scp > M1</scp > polarization and activation of the < scp > NLRP3</scp > inflammasome. Eur J Pain. 2023;27(2):289–302. doi: 10.1002/ejp.2059.
  • Tadokoro Y, Yomogida K, Ohta H, et al. Homeostatic regulation of germinal stem cell proliferation by the GDNF/FSH pathway. Mech Dev. 2002;113(1):29–39. doi: 10.1016/s0925-4773(02)00004-7.
  • Tzavlaki K, Moustakas A. TGF-β signaling. Biomolecules. 2020;10(3):487. doi: 10.3390/biom10030487.
  • Karin M. Nuclear factor-κB in cancer development and progression. Nature. 2006;441(7092):431–436. doi: 10.1038/nature04870.
  • Zhong Z, Umemura A, Sanchez-Lopez E, et al. NF-κB restricts inflammasome activation via elimination of damaged mitochondria. Cell. 2016;164(5):896–910. doi: 10.1016/j.cell.2015.12.057.
  • Laforge M, Rodrigues V, Silvestre R, et al. NF-κB pathway controls mitochondrial dynamics. Cell Death Differ. 2016;23(1):89–98. doi: 10.1038/cdd.2015.42.
  • Hobbs S, Reynoso M, Geddis AV, et al. LPS-stimulated NF-κB p65 dynamic response marks the initiation of TNF expression and transition to IL-10 expression in RAW 264.7 macrophages. Physiol Rep. 2018;6(21):e13914. doi: 10.14814/phy2.13914.
  • Lee JC, Laydon JT, McDonnell PC, et al. A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature. 1994;372(6508):739–746. doi: 10.1038/372739a0.
  • Nick JA, Avdi NJ, Young SK, et al. An intracellular signaling pathway linking lipopolysaccharide stimulation to cellular responses of the human neutrophil. Chest. 1999;116(1 Suppl):54S–55S. doi: 10.1378/chest.116.suppl_1.54S.
  • Fessler MB, Malcolm KC, Duncan MW, et al. A genomic and proteomic analysis of activation of the human neutrophil by lipopolysaccharide and its mediation by p38 mitogen-activated protein kinase. J Biol Chem. 2002;277(35):31291–31302. doi: 10.1074/jbc.M200755200.
  • Nyati KK, Masuda K, Zaman MMU, et al. TLR4-induced NF-κB and MAPK signaling regulate the IL-6 mRNA stabilizing protein Arid5a. Nucleic Acids Res. 2017;45(5):2687–2703. doi: 10.1093/nar/gkx064.
  • Saccani S, Pantano S, Natoli G. p38-dependent marking of inflammatory genes for increased NF-κB recruitment. Nat Immunol. 2002;3(1):69–75. doi: 10.1038/ni748.
  • Rajpoot S, Wary KK, Ibbott R, et al. TIRAP in the mechanism of inflammation. Front Immunol. 2021;12:697588. doi: 10.3389/fimmu.2021.697588.
  • Yang Y, Kim SC, Yu T, et al. Functional roles of p38 mitogen-activated protein kinase in macrophage-mediated inflammatory responses. Mediators Inflamm. 2014;2014:352371. doi: 10.1155/2014/352371.
  • Atsaves V, Leventaki V, Rassidakis GZ, et al. AP-1 transcription factors as regulators of immune responses in cancer. Cancers (Basel). 2019;11(7):1037. doi: 10.3390/cancers11071037.
  • Orth JM. The role of follicle-stimulating hormone in controlling sertoli cell proliferation in testes of fetal rats. Endocrinology. 1984;115(4):1248–1255. doi: 10.1210/endo-115-4-1248.
  • Michailidis G, Anastasiadou M, Guibert E, et al. Activation of innate immune system in response to lipopolysaccharide in chicken sertoli cells. Reproduction. 2014;148(3):259–270. doi: 10.1530/REP-14-0064.
  • Arato I, Milardi D, Giovagnoli S, et al. In “vitro” lps-stimulated sertoli cells pre-loaded with microparticles: intracellular activation pathways. Front Endocrinol (Lausanne). 2020;11:611932. doi: 10.3389/fendo.2020.611932.
  • Nakamura K, Kimple AJ, Siderovski DP, et al. PB1 domain interaction of p62/sequestosome 1 and MEKK3 regulates NF-κB activation. J Biol Chem. 2010;285(3):2077–2089. doi: 10.1074/jbc.M109.065102.
  • Craig EA, Stevens MV, Vaillancourt RR, et al. MAP3Ks as Central regulators of cell fate during development. Dev Dyn. 2008;237(11):3102–3114. doi: 10.1002/dvdy.21750.
  • Styles FL, Al-Owais MM, Scragg JL, et al. Kv1.3 voltage-gated potassium channels link cellular respiration to proliferation through a non-conducting mechanism. Cell Death Dis. 2021;12(4):372. doi: 10.1038/s41419-021-03627-6.
  • Teisseyre A, Palko-Labuz A, Sroda-Pomianek K, et al. Voltage-gated potassium channel Kv1.3 as a target in therapy of cancer. Front Oncol. 2019;9:933. doi: 10.3389/fonc.2019.00933.
  • Trocoli A, Djavaheri-Mergny M. The complex interplay between autophagy and NF-κB signaling pathways in cancer cells. Am J Cancer Res. 2011;1(5):629–649.
  • Xi H, Hu Z, Han S, et al. FSH-inhibited autophagy protects against oxidative stress in goat sertoli cells through p62-Nrf2 pathway. Theriogenology. 2023;195:103–114. doi: 10.1016/j.theriogenology.2022.10.022.