Significance of NF-kappaB signaling and PARP1 activity in the TNF-induced inhibition of PHEX gene expression in human osteoblasts

  • Urszula Kędzierska Department of Animal Physiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
  • Łukasz Banasiak Department of Molecular Phylogenetics and Evolution, Institute of Botany, Faculty of Biology and Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland
  • Paweł Kiela Departments of Pediatrics and Immunobiology, Steele Children’s Research Center, University of Arizona, Tucson AZ 85724, USA
  • Paweł Marek Majewski Department of Animal Physiology, Institute of Zoology, Faculty of Biology, University of Warsaw Miecznikowa 1, 02-096 Warsaw

Abstract

Although loss of bone mineral density is a common symptom of chronic inflammatory diseases, its mechanisms are still poorly understood. The PHEX gene encodes a Zn-endopeptidase expressed in osteoblasts and contributes to bone mineralization. Data derived from rodents has indicated co-repression of the PHEX gene by the NF-κB pathway and poly(ADP-ribose) polymerase 1 (PARP1). The aim of this study was to determine the molecular mechanism involved in TNF-mediated downregulation of PHEX expression in human osteoblasts and human osteosarcoma cell line. We observed that activation of the NF-κB pathway by TNF was manifested as a nuclear increase in RELA and NFKB1 heterodimer. We found that TNF reduced PHEX expression and the proteasome inhibitor reversed this effect in osteosarcoma cell line. Contrary to the effects seen in rodents, inhibition of PARP1 enzymatic activity did not significantly reverse the effect of TNF on the human PHEX gene expression. EMSA studies showed that the number of adenines in the PHEX proximal promoter is crucial for the transcription factors’ interactions within that region. The obtained results support the hypothesis indicating the existence of a molecular mechanism of gene repression that involves a poly adenine-rich region of the proximal gene promoters and PARP1 transcriptional activity.

References

Al-Romaih K, Bayani J, Vorobyova J, Karaskova J, Park PC, Zielenska M, Squire JA (2003) Chromosomal instability in osteosarcoma and its association with centrosome abnormalities. Cancer Genet Cytogenet 144(2): 91-99. https://doi.org/10.1016/S0165-4608(02)00929-9

Benayahu D, Shur I, Marom R, Meller I, Issakov J (2002) Cellular and molecular properties associated with osteosarcoma cells. J Cell Biochem 84(1): 108-114. https://doi.org/10.1002/jcb.1270

Bowe AE, Finnegan R, Jan de Beur SM, Cho J, Levine MA, Kumar R, Schiavi SC (2001) FGF-23 inhibits renal tubular phosphate transport and is a PHEX substrate. Biochem Biophys Res Commun 284(4): 977-981. https://doi.org/10.1006/bbrc.2001.5084

Bu RF, Borysenko CW, Li YN, Cao LH, Sabokbar A, Blair HC (2003) Expression and function of TNF-family proteins and receptors in human osteoblasts. Bone 33(5): 760-770. https://doi.org/10.1016/j.bone.2003.07.006

Chang J, Wang Z, Tang E, Fan Z, McCauley L, Franceschi R, Wang CY (2009) Inhibition of osteoblastic bone formation by nuclear factor-kappaB. Nat Med 15(6): 682-689. https://doi.org/10.1038/nm.1954

Clowes JA, Riggs BL, Khosla S (2005) The role of the immune system in the pathophysiology of osteoporosis. Immunol Rev 208: 207-227. https://doi.org/10.1111/j.0105-2896.2005.00334.x

Collins JF, Ghishan FK (1994) Molecular cloning, functional expression, tissue distribution, and in situ hybridization of the renal sodium phosphate (Na+/P(i)) transporter in the control and hypophosphatemic mouse. FASEB J 8(11): 862-868. https://doi.org/10.1096/fasebj.8.11.8070635

Declercq, H., Van den Vreken, N., De Maeyer, E., Verbeeck, R., Schacht, E., De Ridder, L., & Cornelissen, M. (2004) Isolation, proliferation and differentiation of osteoblastic cells to study cell/biomaterial interactions: comparison of different isolation techniques and source. Biomaterials 25(5): 757-768. https://doi.org/10.1016/S0142-9612(03)00580-5

Dixon PH, Christie PT, Wooding C, Trump D, Grieff M, Holm I, Thakker RV (1998) Mutational analysis of PHEX gene in X-linked hypophosphatemia. J Clin Endocrinol Metab 83(10): 3615-3623.

https://doi.org/10.1210/jcem.83.10.5180

Drezner MK (2000) PHEX gene and hypophosphatemia. Kidney Int 57(1), 9-18. https://doi.org/10.1046/j.1523-1755.2000.00807.x

Du L, Desbarats M, Viel J, Glorieux FH, Cawthorn C, Ecarot B (1996) cDNA cloning of the murine Pex gene implicated in X-linked hypophosphatemia and evidence for expression in bone. Genomics 36(1): 22-28. https://doi.org/10.1006/geno.1996.0421

Francis F, Korn SHB, Reinhardt R, de Jong P, Poustka A, Lehrach H, Rowe PSN, Goulding JN, Summerfield T, Mountford R, Read AP, Popowska E, Pronicka E, Davies KE, O'Riordan JLH, Econs MJ, Nesbitt T, Drezner MK, Oudet C, Pannetier S, Hanauer A, Strom TM, Meindl A, Lorenz B, Cagnoli B, Mohnike KL, Murken J, Meitinger T (1995) A gene (PEX) with homologies to endopeptidases is mutated in patients with X-linked hypophosphatemic rickets. The HYP Consortium. Nat Genet 11(2): 130-136. https://doi.org/10.1038/ng1095-130

Ghishan FK, Kiela PR (2011) Advances in the understanding of mineral and bone metabolism in inflammatory bowel diseases. Am J Physiol Gastrointest Liver Physiol 300(2): G191-201. https://doi.org/10.1152/ajpgi.00496.2010

Hassa PO, Hottiger MO (2002) The functional role of poly(ADP-ribose)polymerase 1 as novel coactivator of NF-kappaB in inflammatory disorders. Cell Mol Life Sci 59(9): 1534-1553. https://doi.org/10.1007/s00018-002-8527-2

Hernando N, Biber J, Forster I, Murer H (2005) Recent advances in renal phosphate transport. Ther Apher Dial 9(4): 323-327. https://doi.org/10.1111/j.1744-9987.2005.00290.x

Ito N, Wijenayaka AR, Prideaux M, Kogawa M, Ormsby RT, Evdokiou A, Atkins GJ (2015) Regulation of FGF23 expression in IDG-SW3 osteocytes and human bone by pro-inflammatory stimuli. Mol Cell Endocrinol 399: 208-218. https://doi.org/10.1016/j.mce.2014.10.007

Johnell O, Kanis J (2005) Epidemiology of osteoporotic fractures. Osteoporos Int 16 Suppl 2: S3-7. https://doi.org/10.1007/s00198-004-1702-6

Johnell O, Kanis JA (2004) An estimate of the worldwide prevalence, mortality and disability associated with hip fracture. Osteoporos Int, 15(11): 897-902. https://doi.org/10.1007/s00198-004-1627-0

Johnell O, Kanis JA, Oden A, Sernbo I, Redlund-Johnell I, Petterson C, Jonsson B (2004) Mortality after osteoporotic fractures. Osteoporos Int 15(1): 38-42. https://doi.org/10.1007/s00198-003-1490-4

Kiela PR, Ghishan FK (2009) Recent advances in the renal-skeletal-gut axis that controls phosphate homeostasis. Lab Invest 89(1): 7-14. https://doi.org/10.1038/labinvest.2008.114

Krishnakumar R, Gamble MJ, Frizzell KM, Berrocal JG, Kininis M, Kraus WL (2008) Reciprocal binding of PARP-1 and histone H1 at promoters specifies transcriptional outcomes. Science 319(5864): 819-821. https://doi.org/10.1126/science.1149250

Larmonier CB, Shehab KW, Laubitz D, Jamwal DR, Ghishan FK, Kiela PR (2016) Transcriptional Reprogramming and Resistance to Colonic Mucosal Injury in Poly(ADP-ribose) Polymerase 1 (PARP1)-deficient Mice. J Biol Chem 291(17): 8918-8930. https://doi.org/10.1074/jbc.M116.714386

Majewski PM, Thurston RD, Ramalingam R, Kiela PR, Ghishan FK (2010) Cooperative role of NF-{kappa}B and poly(ADP-ribose) polymerase 1 (PARP-1) in the TNF-induced inhibition of PHEX expression in osteoblasts. J Biol Chem 285(45): 34828-34838.

https://doi.org/10.1074/jbc.M110.152868

Murer H, Hernando N, Forster I, Biber J (2003) Regulation of Na/Pi transporter in the proximal tubule. Annu Rev Physiol 65: 531-542. https://doi.org/ 10.1146/annurev.physiol.65.042902.092424

Nguewa PA, Fuertes MA, Valladares B, Alonso C, Perez JM (2005) Poly(ADP-ribose) polymerases: homology, structural domains and functions. Novel therapeutical applications. Prog Biophys Mol Biol 88(1): 143-172. https://doi.org/10.1016/j.pbiomolbio.2004.01.001

Novack DV (2011) Role of NF-kappaB in the skeleton. Cell Res 21(1): 169-182. https://doi.org/10.1038/cr.2010.159

Ogino H, Nozaki T, Gunji A, Maeda M, Suzuki H, Ohta T, Masutani M (2007) Loss of Parp-1 affects gene expression profile in a genome-wide manner in ES cells and liver cells. BMC Genomics 8: 41. https://doi.org/10.1186/1471-2164-8-41

Peralta-Leal A, Rodriguez-Vargas JM, Aguilar-Quesada R, Rodriguez MI, Linares JL, de Almodovar MR, Oliver FJ (2009) PARP inhibitors: new partners in the therapy of cancer and inflammatory diseases. Free Radic Biol Med 47(1): 13-26. https://doi.org/10.1016/j.freeradbiomed.2009.04.008

R Core Team (2016) R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. https://www.R-project.org/

Rodriguez-Bores L, Barahona-Garrido J, Yamamoto-Furusho JK (2007) Basic and clinical aspects of osteoporosis in inflammatory bowel disease. World J Gastroenterol 13(46): 6156-6165. https://doi.org/10.3748/wjg.v13.i46.6156

Slomiany BA, Kelly MM, Kurtz DT (2000) Extraction of nuclear proteins with increased DNA binding activity. Biotechniques 28(5): 938-942.

Sponholtz TR, Zhang X, Fontes JD, Meigs JB, Cupples LA, Kiel DP, McLean RR (2014) Association between inflammatory biomarkers and bone mineral density in a community-based cohort of men and women. Arthritis Care Res (Hoboken) 66(8): 1233-1240. https://doi.org/10.1002/acr.22270

Teti A, Zallone A (2009) Do osteocytes contribute to bone mineral homeostasis? Osteocytic osteolysis revisited. Bone 44(1): 11-16. https://doi.org/10.1016/j.bone.2008.09.017

Uno JK, Kolek OI, Hines ER, Xu H, Timmermann BN, Kiela PR, Ghishan FK (2006) The role of tumor necrosis factor alpha in down-regulation of osteoblast Phex gene expression in experimental murine colitis. Gastroenterology 131(2): 497-509.

https://doi.org/10.1053/j.gastro.2006.05.020

Viswanathan A, Sylvester FA (2008) Chronic pediatric inflammatory diseases: effects on bone. Rev Endocr Metab Disord 9(2): 107-122. https://doi.org/10.1007/s11154-007-9070-0

Published
2018-11-15
Section
Articles