#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Inflammasome expression is higher in ovarian tumors than in normal ovary


Autoři: Judith Luborsky aff001;  Animesh Barua aff002;  Seara Edassery aff004;  Janice M. Bahr aff005;  Seby L. Edassery aff004
Působiště autorů: Department of Pharmacology, Rush University Medical Center, Chicago, Illinois, United States of America aff001;  Department of Obstetrics & Gynecology, Rush University Medical Center, Chicago, Illinois, United States of America aff002;  Department of Pathology, Rush University Medical Center, Chicago, Illinois, United States of America aff003;  Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois, United States of America aff004;  Department of Animal Science, University of Illinois Urbana-Champaign, Champaign, Illinois, United States of America aff005
Vyšlo v časopise: PLoS ONE 15(1)
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pone.0227081

Souhrn

Chronic inflammation fundamentally influences cancer risk and development. A mechanism of chronic inflammation is the formation of inflammasome complexes which results in the sustained secretion of the pro-inflammatory cytokines IL1β and IL18. Inflammasome expression and actions vary among cancers. There is no information on inflammasome expression in ovarian cancer (OvCa). To determine if ovarian tumors express inflammasome components, mRNA and protein expression of NLRP3 (nucleotide-binding domain, leucine-rich repeat family, pyrin domain containing 3), caspase-1, IL1β, and IL18 expression in hen and human OvCa was assessed. Chicken (hen) OvCa a valid model of spontaneous human OvCa. Hens were selected into study groups with or without tumors using ultrasonography; tumors were confirmed by histology, increased cellular proliferation, and expression of immune cell marker mRNA. mRNA expression was higher for hallmarks of inflammasome activity (caspase-1, 5.9x increase, p = 0.04; IL1β, 4x increase, p = 0.04; and IL18, 7.8x increase, p = 0.0003) in hen OvCa compared to normal ovary. NLRP3, caspase-8 and caspase-11 mRNA did not differ significantly between tumor and non-tumor containing ovaries. Similar results occurred for human OvCa. Protein expression by immunohistochemistry paralleled mRNA expression and was qualitatively higher in tumors. Increased protein expression of caspase-1, IL1β, and IL18 occurred in surface epithelium, tumor cells, and immune cells. The aryl hydrocarbon receptor (AHR), a potential tumor suppressor and NLRP3 regulator, was higher in hen (2.4x increase, p = 0.002) and human tumors (1.8x increase, p = 0.038), suggesting a role in OvCa. Collectively, the results indicate that inflammasome expression is associated with hen and human OvCa, although the NLR sensor type remains to be determined.

Klíčová slova:

Immune cells – Ovaries – Epithelium – Cell staining – Immunohistochemistry techniques – Ovarian cancer – Cytoplasmic staining – Inflammasomes


Zdroje

1. Landskron G, De la Fuente M, Thuwajit P, Thuwajit C, Hermoso MA. Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res. 2014;2014:149185. doi: 10.1155/2014/149185 24901008

2. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860–7. doi: 10.1038/nature01322 [pii] 12490959

3. de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006;6(1):24–37. nrc1782 [pii] doi: 10.1038/nrc1782 16397525

4. Vendramini-Costa DB, Carvalho JE. Molecular link mechanisms between inflammation and cancer. Curr Pharm Des. 2012;18(26):3831–52. doi: 10.2174/138161212802083707 22632748

5. Lu A, Wu H. Structural mechanisms of inflammasome assembly. FEBS J. 2015;282(3):435–44. doi: 10.1111/febs.13133 25354325

6. Lin C, Zhang J. Inflammasomes in Inflammation-Induced Cancer. Front Immunol. 2017;8:271. doi: 10.3389/fimmu.2017.00271 28360909

7. Kantono M, Guo B. Inflammasomes and Cancer: The Dynamic Role of the Inflammasome in Tumor Development. Front Immunol. 2017;8:1132. doi: 10.3389/fimmu.2017.01132 28955343

8. Sharma D, Kanneganti TD. The cell biology of inflammasomes: Mechanisms of inflammasome activation and regulation. J Cell Biol. 2016;213(6):617–29. doi: 10.1083/jcb.201602089 27325789

9. Thi HTH, Hong S. Inflammasome as a Therapeutic Target for Cancer Prevention and Treatment. J Cancer Prev. 2017;22(2):62–73. doi: 10.15430/JCP.2017.22.2.62 28698859

10. He Y, Hara H, Nunez G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem Sci. 2016;41(12):1012–21. doi: 10.1016/j.tibs.2016.09.002 27669650

11. Cullen SP, Kearney CJ, Clancy DM, Martin SJ. Diverse Activators of the NLRP3 Inflammasome Promote IL-1beta Secretion by Triggering Necrosis. Cell Rep. 2015;11(10):1535–48. doi: 10.1016/j.celrep.2015.05.003 26027935

12. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7(2):99–109. doi: 10.1038/nrmicro2070 19148178

13. Karki R, Man SM, Kanneganti TD. Inflammasomes and Cancer. Cancer Immunol Res. 2017;5(2):94–9. doi: 10.1158/2326-6066.CIR-16-0269 28093447

14. Wei Q, Mu K, Li T, Zhang Y, Yang Z, Jia X, et al. Deregulation of the NLRP3 inflammasome in hepatic parenchymal cells during liver cancer progression. Lab Invest. 2014;94(1):52–62. doi: 10.1038/labinvest.2013.126 24166187

15. Allen IC, TeKippe EM, Woodford RM, Uronis JM, Holl EK, Rogers AB, et al. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med. 2010;207(5):1045–56. doi: 10.1084/jem.20100050 20385749

16. Jo EK, Kim JK, Shin DM, Sasakawa C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell Mol Immunol. 2016;13(2):148–59. doi: 10.1038/cmi.2015.95 26549800

17. Huai W, Zhao R, Song H, Zhao J, Zhang L, Zhang L, et al. Aryl hydrocarbon receptor negatively regulates NLRP3 inflammasome activity by inhibiting NLRP3 transcription. Nat Commun. 2014;5:4738. doi: 10.1038/ncomms5738 25141024

18. Vogel CFA, Haarmann-Stemmann T. The aryl hydrocarbon receptor repressor—More than a simple feedback inhibitor of AhR signaling: Clues for its role in inflammation and cancer. Curr Opin Toxicol. 2017;2:109–19. doi: 10.1016/j.cotox.2017.02.004 28971163

19. Wang K, Li Y, Jiang YZ, Dai CF, Patankar MS, Song JS, et al. An endogenous aryl hydrocarbon receptor ligand inhibits proliferation and migration of human ovarian cancer cells. Cancer Lett. 2013;340(1):63–71. doi: 10.1016/j.canlet.2013.06.026 23851185

20. Zudaire E, Cuesta N, Murty V, Woodson K, Adams L, Gonzalez N, et al. The aryl hydrocarbon receptor repressor is a putative tumor suppressor gene in multiple human cancers. J Clin Invest. 2008;118(2):640–50. doi: 10.1172/JCI30024 18172554

21. Hales DB, Zhuge Y, Lagman JA, Ansenberger K, Mahon C, Barua A, et al. Cyclooxygenases expression and distribution in the normal ovary and their role in ovarian cancer in the domestic hen (Gallus domesticus). Endocrine. 2008;33(3):235–44. doi: 10.1007/s12020-008-9080-z 18498063

22. Barua A, Abramowicz JS, Bahr JM, Bitterman P, Dirks A, Holub KA, et al. Detection of ovarian tumors in chicken by sonography: a step toward early diagnosis in humans? J Ultrasound Med. 2007;26(7):909–19. doi: 10.7863/jum.2007.26.7.909 17592054

23. Barua A, Bitterman P, Abramowicz JS, Dirks AL, Bahr JM, Hales DB, et al. Histopathology of ovarian tumors in laying hens: a preclinical model of human ovarian cancer. Int J Gynecol Cancer. 2009;19(4):531–9. doi: 10.1111/IGC.0b013e3181a41613 19509547

24. Bradaric MJ, Penumatsa K, Barua A, Edassery SL, Yu Y, Abramowicz JS, et al. Immune cells in the normal ovary and spontaneous ovarian tumors in the laying hen (Gallus domesticus) model of human ovarian cancer. PLoS One. 2013;8(9):e74147. doi: 10.1371/journal.pone.0074147 24040191

25. Barua A, Edassery SL, Bitterman P, Abramowicz JS, Dirks AL, Bahr JM, et al. Prevalence of antitumor antibodies in laying hen model of human ovarian cancer. Int J Gynecol Cancer. 2009;19(4):500–7. doi: 10.1111/IGC.0b013e3181a39db1 19509543

26. Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347(6220):1260419. doi: 10.1126/science.1260419 25613900

27. Uhlen M, Bjorling E, Agaton C, Szigyarto CA, Amini B, Andersen E, et al. A human protein atlas for normal and cancer tissues based on antibody proteomics. Mol Cell Proteomics. 2005;4(12):1920–32. doi: 10.1074/mcp.M500279-MCP200 16127175

28. Acadeny N. Guide for the Care and Use of Laboratory Animals. Washington (DC)2011.

29. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3(6):1101–8. doi: 10.1038/nprot.2008.73 18546601

30. Spitzer C, Barnow S, Volzke H, Wallaschofski H, John U, Freyberger HJ, et al. Association of posttraumatic stress disorder with low-grade elevation of C-reactive protein: evidence from the general population. J Psychiatr Res. 2009;44(1):15–21. S0022-3956(09)00126-5 [pii] doi: 10.1016/j.jpsychires.2009.06.002 19628221

31. Chang CC, Su KM, Lu KH, Lin CK, Wang PH, Li HY, et al. Key Immunological Functions Involved in the Progression of Epithelial Ovarian Serous Carcinoma Discovered by the Gene Ontology-Based Immunofunctionome Analysis. Int J Mol Sci. 2018;19(11). doi: 10.3390/ijms19113311 30356023

32. Guarda G, Zenger M, Yazdi AS, Schroder K, Ferrero I, Menu P, et al. Differential expression of NLRP3 among hematopoietic cells. J Immunol. 2011;186(4):2529–34. doi: 10.4049/jimmunol.1002720 21257968

33. Singh S, Mehta N, Lilan J, Budhthoki MB, Chao F, Yong L. Initiative action of tumor-associated macrophage during tumor metastasis. Biochim Open. 2017;4:8–18. doi: 10.1016/j.biopen.2016.11.002 29450136

34. Hagemann T, Wilson J, Burke F, Kulbe H, Li NF, Pluddemann A, et al. Ovarian cancer cells polarize macrophages toward a tumor-associated phenotype. J Immunol. 2006;176(8):5023–32. 176/8/5023 [pii] doi: 10.4049/jimmunol.176.8.5023 16585599

35. Shah CA, Allison KH, Garcia RL, Gray HJ, Goff BA, Swisher EM. Intratumoral T cells, tumor-associated macrophages, and regulatory T cells: association with p53 mutations, circulating tumor DNA and survival in women with ovarian cancer. Gynecol Oncol. 2008;109(2):215–9. S0090-8258(08)00015-2 [pii] doi: 10.1016/j.ygyno.2008.01.010 18314181

36. Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J Leukoc Biol. 2009;86(5):1065–73. jlb.0609385 [pii] doi: 10.1189/jlb.0609385 19741157

37. Murphy AJ, Kraakman MJ, Kammoun HL, Dragoljevic D, Lee MK, Lawlor KE, et al. IL-18 Production from the NLRP1 Inflammasome Prevents Obesity and Metabolic Syndrome. Cell Metab. 2016;23(1):155–64. doi: 10.1016/j.cmet.2015.09.024 26603191

38. Wei Y, Huang H, Qiu Z, Li H, Tan J, Ren G, et al. NLRP1 Overexpression Is Correlated with the Tumorigenesis and Proliferation of Human Breast Tumor. Biomed Res Int. 2017;2017:4938473. doi: 10.1155/2017/4938473 29214170

39. Tian X, Pascal G, Monget P. Evolution and functional divergence of NLRP genes in mammalian reproductive systems. BMC Evol Biol. 2009;9:202. doi: 10.1186/1471-2148-9-202 19682372

40. He Y, Zeng MY, Yang D, Motro B, Nunez G. NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature. 2016;530(7590):354–7. doi: 10.1038/nature16959 26814970

41. Pellegrini C, Antonioli L, Lopez-Castejon G, Blandizzi C, Fornai M. Canonical and Non-Canonical Activation of NLRP3 Inflammasome at the Crossroad between Immune Tolerance and Intestinal Inflammation. Front Immunol. 2017;8:36. doi: 10.3389/fimmu.2017.00036 28179906

42. Man SM, Hopkins LJ, Nugent E, Cox S, Gluck IM, Tourlomousis P, et al. Inflammasome activation causes dual recruitment of NLRC4 and NLRP3 to the same macromolecular complex. Proc Natl Acad Sci U S A. 2014;111(20):7403–8. doi: 10.1073/pnas.1402911111 24803432

43. Hahn ME. Aryl hydrocarbon receptors: diversity and evolution. Chem Biol Interact. 2002;141(1–2):131–60. doi: 10.1016/s0009-2797(02)00070-4 12213389

44. Hanieh H. Toward understanding the role of aryl hydrocarbon receptor in the immune system: current progress and future trends. Biomed Res Int. 2014;2014:520763. doi: 10.1155/2014/520763 24527450

45. Fan Y, Boivin GP, Knudsen ES, Nebert DW, Xia Y, Puga A. The aryl hydrocarbon receptor functions as a tumor suppressor of liver carcinogenesis. Cancer Res. 2010;70(1):212–20. doi: 10.1158/0008-5472.CAN-09-3090 19996281

46. Khorram O, Garthwaite M, Golos T. Uterine and ovarian aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) mRNA expression in benign and malignant gynaecological conditions. Mol Hum Reprod. 2002;8(1):75–80. doi: 10.1093/molehr/8.1.75 11756572

47. Yasui T, Kim EY, Iwata H, Franks DG, Karchner SI, Hahn ME, et al. Functional characterization and evolutionary history of two aryl hydrocarbon receptor isoforms (AhR1 and AhR2) from avian species. Toxicol Sci. 2007;99(1):101–17. doi: 10.1093/toxsci/kfm139 17556759

48. Lee JS, Kim EY, Nomaru K, Iwata H. Molecular and functional characterization of Aryl hydrocarbon receptor repressor from the chicken (Gallus gallus): interspecies similarities and differences. Toxicol Sci. 2011;119(2):319–34. doi: 10.1093/toxsci/kfq336 21047992

49. Xue P, Fu J, Zhou Y. The Aryl Hydrocarbon Receptor and Tumor Immunity. Front Immunol. 2018;9:286. doi: 10.3389/fimmu.2018.00286 29487603


Článok vyšiel v časopise

PLOS One


2020 Číslo 1
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#