DOI QR코드

DOI QR Code

Effects of intrauterine growth restriction during late pregnancy on the cell growth, proliferation, and differentiation in ovine fetal thymuses

  • Zi, Yang (College of Animal Science, Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University) ;
  • Ma, Chi (College of Animal Science, Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University) ;
  • He, Shan (College of Animal Science, Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University) ;
  • Yang, Huan (College of Animal Science, Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University) ;
  • Zhang, Min (College of Animal Science, Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University) ;
  • Gao, Feng (College of Animal Science, Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University) ;
  • Liu, Yingchun (College of Life Science, Inner Mongolia Key Laboratory of Biomanufacturing, Inner Mongolia Agricultural University)
  • Received : 2021.09.11
  • Accepted : 2022.01.12
  • Published : 2022.07.01

Abstract

Objective: This study investigated the effects of intrauterine growth restriction (IUGR) during late pregnancy on the cell growth, proliferation, and differentiation in ovine fetal thymuses. Methods: Eighteen time-mated Mongolian ewes with singleton fetuses were allocated to three groups at d 90 of pregnancy: restricted group 1 (RG1, 0.18 MJ ME/body weight [BW]0.75/d, n = 6), restricted group 2 (RG2, 0.33 MJ ME/BW0.75/d, n = 6) and control group (CG, ad libitum, 0.67 MJ ME/BW0.75/d, n = 6). Fetuses were recovered at slaughter on d 140. Results: The G0/G1 phase cell number in fetal thymus of the RG1 group was increased but the proliferation index and the expression of proliferating cell nuclear antigen (PCNA) were reduced compared with the CG group (p<0.05). Fetuses in the RG1 group exhibited decreased growth hormone receptor (GHR), insulin-like growth factor 2 receptor (IGF-2R), and their mRNA expressions (p<0.05). For the RG2 fetuses, there were no differences in the proliferation index and PCNA expression (p>0.05), but growth hormone (GH) and the mRNA expression of GHR were lower than those of the CG group (p<0.05). The thymic mRNA expressions of cyclin-dependent protein kinases (CDKs including CDK1, CDK2, and CDK4), CCNE, E2-factors (E2F1, E2F2, and E2F5) were reduced in the RG1 and RG2 groups (p<0.05), and decreased mRNA expressions of E2F4, CCNA, CCNB, and CCND were occurred in the RG1 fetuses (p<0.05). The decreased E-cadherin (E-cad) as a marker for epithelial-mesenchymal transition (EMT) was found in the RG1 and RG2 groups (p<0.05), but the OB-cadherin which is a marker for activated fibroblasts was increased in fetal thymus of the RG1 group (p<0.05). Conclusion: These results indicate that weakened GH/IGF signaling system repressed the cell cycle progression in G0/G1 phase in IUGR fetal thymus, but the switch from reduced E-cad to increased OB-cadherin suggests that transdifferentiation process of EMT associated with fibrogenesis was strengthened. The impaired cell growth, retarded proliferation and modified differentiation were responsible for impaired maturation of IUGR fetal thymus.

Keywords

Acknowledgement

This work was supported by National Natural Science Foundation of China (31760677 and 31460612, Beijing, China) and Inner Mongolia Natural Science Foundation (2021MS03015 and 2020MS03042 Hohhot, China).

References

  1. Savino W, Dardenne M, Velloso LA, Silva-Barbosa SD. The thymus is a common target in malnutrition and infection. Br J Nutr 2007;98:S11-6. https://doi.org/10.1017/s0007114507832880
  2. Ortiz R, Cortes L, Cortes E, Medina H. Malnutrition alters the rates of apoptosis in splenocytes and thymocyte subpo pulations of rats. Clin Exp Immunol 2009;155:96-106. https://doi.org/10.1111/j.1365-2249.2008.03796.x
  3. Manley NR. Thymus organogenesis and molecular mechanisms of thymic epithelial cell differentiation. Semin Immunol 2000;12:421-8. https://doi.org/10.1006/smim.2000.0263
  4. Gao F, Hou XZ, Liu YC. Study of the effect of IUGR during late pregnancy on the immune capability of neonatal lambs. Prog Nat Sci China (chinese) 2006;16:1336-40.
  5. Palmer AC. Nutritionally mediated programming of the developing immune system. Adv Nutr 2011;2:377-95. https://doi.org/10.3945/an.111.000570
  6. Robinson JJ, Sinclair KD, McEvoy TG. Nutritional effects on foetal growth. Anim Sci 1999;68:315-31. https://doi.org/10.1017/S1357729800050323
  7. McMillen IC, Adams MB, Ross JT, et al. Fetal growth restriction: adaptations and consequences. Reproduction 2001;122:195-204. https://doi.org/10.1530/rep.0.1220195
  8. Osgerby JC, Wathes DC, Howard D, Gadd TS. The effect of maternal undernutrition on ovine fetal growth. J Endocrinol 2002;173:131-41. https://doi.org/10.1677/joe.0.1730131
  9. Greenwood PL, Bell AW. Consequences of intra-uterine growth retardation for postnatal growth, metabolism and pathophysiology. Reprod Suppl 2003;61:195-206.
  10. Cromi A, Ghezzi F, Raffaelli R, Bergamini V, Siesto G, Bolis P. Ultrasonographic measurement of thymus size in IUGR fetuses: a marker of the fetal immunoendocrine response to malnutrition. Ultrasound Obstet Gynecol 2009;33:421-6. https://doi.org/10.1002/uog.6320
  11. Ekin A, Gezer C, Taner CE, Solmaz U, Gezer NS, Ozeren M. Prognostic value of fetal thymus size in intrauterine growth restriction. J Ultrasound Med 2016;35:511-7. https://doi.org/10.7863/ultra.15.05039
  12. Olearo E, Oberto M, Ogge G, et al. Thymic volume in healthy, small for gestational age and growth restricted fetuses. Prenat Diagn 2012;32:662-7. https://doi.org/10.1002/pd.3883
  13. Contreras YM, Yu X, Hale MA, et al. Intrauterine growth restriction alters T-lymphocyte cell number and dual specificity phosphatase 1 levels in the thymus of newborn and juvenile rats. Pediatr Res 2011;70:123-9. http://doi.org/10.1203/PDR.0b013e31821f6e75
  14. Liu Y, He S, Zhang Y, et al. Effects of intrauterine growth restriction during late pregnancy on the development of the ovine fetal thymus and the T-lymphocyte subpopulation. Am J Reprod Immunol 2015;74:26-37. https://doi.org/10.1111/aji.12371
  15. Morrison JL. Sheep models of intrauterine growth restriction: fetal adaptations and consequences. Clin Exp Pharmacol Physiol 2008;35:730-43. https://doi.org/10.1111/j.1440-1681.2008.04975.x
  16. The State Science and Technology Commission of China: Regulations for administration of affairs concerning experimental animal. Beijing, China: The China Legal System Publishing House Press; 1988.
  17. Symonds ME, Budge H, Stephenson T, McMillen IC. Fetal endocrinology and development-manipulation and adaptation to long-term nutritional and environmental challenges. Reproduction 2001;121:853-62. https://doi.org/10.1530/rep.0.1210853
  18. Gao F, Liu YC, Zhang C, Zhang ZH, Song SS. Effect of intrauterine growth restriction during late pregnancy on the growth performance, blood components, immunity and anti-oxidation capability of ovine fetus. Livest Sci 2013;155:435-41. https://doi.org/10.1016/j.livsci.2013.04.016
  19. Liu YC, Ma C, Li H, Li L, Gao F, Ao C. Effects of intrauterine growth restriction during late pregnancy on the cell apoptosis and related gene expression in ovine fetal liver. Theriogenology 2017;90:204-9. https://doi.org/10.1016/j.theriogenology.2016.11.030
  20. Sambrook J, Russell DW. Molecular cloning: a laboratory manual (3nd ed.).; Cold Spring Harbor, NY, USA: Cold Spring Harbor Laboratory Press; 2001.
  21. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 2001;25:402-8. https://doi.org/10.1006/meth.2001.1262
  22. SAS Institute, Inc. SAS/STAT user's guide. Cary, NC, USA: SAS Institute, Inc.; 2001.
  23. Bacon WA, Hamilton RS, Yu Z, et al. Single-cell analysis identifies thymic maturation delay in growth-restricted neonatal mice. Front Immunol 2018;9:2523. https://doi.org/10.3389/fimmu.2018.02523
  24. Mitsumori K, Takegawa K, Shimo T, Onodera H, Yasuhara K, Takahashi M. Morphometric and immunohistochemical studies on atrophic changes in lympho-hematopoietic organs of rats treated with piperonyl butoxide or subjected to dietary restriction. Arch Toxicol 1996;70:809-14. https://doi.org/10.1007/s002040050343
  25. Urrego D, Tomczak AP, Zahed F, Stuhmer W, Pardo LA. Potassium channels in cell cycle and cell proliferation. Philos Trans R Soc Lond B Biol Sci 2014;369:20130094. http://doi.org/10.1098/rstb.2013.0094
  26. Hindley C, Philpott A. The cell cycle and pluripotency. Biochem J 2013;451:135-43. https://doi.org/10.1042/BJ20121627
  27. Kalucka J, Missiaen R, Georgiadou M, et al. Metabolic control of the cell cycle. Cell Cycle 2015;14:3379-88 https://doi.org/10.1080/15384101.2015.1090068
  28. Massague J. G1 cell-cycle control and cancer. Nature 2004;432:298-306. https://doi.org/10.1038/nature03094
  29. Zonis S, Ljubimov VA. Mahgerefteh M, Pechnick RN, Wawrowsky K, Chesnokova V. p21Cip restrains hippocampal neurogenesis and protects neuronal progenitors from apoptosis during acute systemic inflammation. Hippocampus 2013;23:1383-94. https://doi.org/10.1002/hipo.22192
  30. Bretones G, Delgado D, Leon J. Myc and cell cycle control. Biochim Biophys Acta 2015;1849:506-16. https://doi.org/10.1016/j.bbagrm.2014.03.013
  31. Satyanarayana A, Kaldis P. Mammalian cell-cycle regulation: several Cdks, numerous cyclins and diverse compensatory mechanisms. Oncogene 2009;28:2925-39. https://doi.org/10.1038/onc.2009.170
  32. Kitagawa M, Kitagawa K, Kotake Y, Niida H, Ohhata T. Cell cycle regulation by long non-coding RNAs. Cell Mol Life Sci 2013;70:4785-94. https://doi.org/10.1007/s00018-013-1423-0
  33. Lim SH, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 2013;140;3079-93. https://doi.org/10.1242/dev.091744
  34. Wong JV, Dong P, Nevins JR, Mathey-Prevot B, You L. Network calisthenics: control of E2F dynamics in cell cycle entry. Cell Cycle 2011;10:3086-94. https://doi.org/10.4161/cc.10.18.17350
  35. Jing J, Xiong S, Li Z, et al. A feedback regulatory loop involving p53/miR-200 and growth hormone endocrine axis controls embryo size of zebrafish. Sci Rep 2015;5:15906. https://doi.org/10.1038/srep15906
  36. Savino W. Neuroendocrine control of T cell development in mammals: role of growth hormone in modulating thymocyte migration. Exp Physiol 2007;92:813-7. https://doi.org/10.1113/expphysiol.2007.038422
  37. Lins MP, de Araujo Vieira LF, Rosa AA, Smaniotto S. Growth hormone in the presence of laminin modulates interaction of human thymic epithelial cells and thymocytes in vitro. Biol Res 2016;49:37. https://doi.org/10.1186/s40659-016-0097-0
  38. Chentoufi AA, Geenen V. Thymic self-antigen expression for the design of a negative/tolerogenic self-vaccine against type 1 diabetes. Clin Dev Immunol 2011;2011:349368. https://doi.org/10.1155/2011/349368
  39. Geenen V. The thymic insulin-like growth factor axis: involvement in physiology and disease. Horm Metab Res 2003;35:656-63. https://doi.org/10.1055/s-2004-814161
  40. Boguszewski CL, Boguszewski MC, Kopchick JJ. Growth hormone, insulin-like growth factor system and carcinogenesis. Endokrynol Pol 2016;67:414-26. https://doi.org/10.5603/EP.a2016.0053
  41. Hay ED, Zuk A. Transformations between epithelium and mesenchyme: normal, pathological, and experimentally induced. Am J Kidney Dis 1995;26:678-90. https://doi.org/10.1016/0272-6386(95)90610-x
  42. Manley NR, Richie ER, Blackburn CC, Condie BG, Sage J. Structure and function of the thymic microenvironment. Front Biosci (Landmark Ed) 2011;16:2461-77. https://doi.org/10.2741/3866
  43. Sun L, Li H, Luo H, Zhao Y. Thymic epithelial cell development and its dysfunction in human diseases. Biomed Res Int 2014;2014:206929. https://doi.org/10.1155/2014/206929
  44. Larue L, Bellacosa A. Epithelial-mesenchymal transition in development and cancer: role of phosphatidylinositol 3' kinase/AKT pathways. Oncogene 2005;24:7443-54. https://doi.org/10.1038/sj.onc.1209091
  45. Thompson EW, Newgreen DF. Carcinoma invasion and metastasis: a role for epithelial-mesenchymal transition? Cancer Res 2005;65:5991-5. https://doi.org/10.1158/0008-5472.CAN-05-0616
  46. Strutz F, Zeisberg M, Ziyadeh FN, et al. Role of basic fibroblast growth factor-2 in epithelial-mesenchymal transformation. Kidney Int 2002;61:1714-28. https://doi.org/10.1046/j.1523-1755.2002.00333.x
  47. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 2014;15:178-96. https://doi.org/10.1038/nrm3758