Dynamics of hematopoiesis regulators content in bone marrow under the poultry embryonic tissue extract usage for the radiation-caused immune damage correction

  • М. С. Погоріла
  • М. М. Попов
  • О. А. Романова
  • О. М. Щербак
  • А. В. Мартинов
Keywords: poultry embryonic tissue extract, radiation-caused immune damage, bone marrow, mice, G-CSF, IL-6, Flt3-ligand, ELISA

Abstract

The aim of our study was to examine the effect of the poultry embryonic tissue extract (PETE) application at G-CSF, Flt3-ligand and IL-6 levels in the bone marrow cells supernatant of mice with radiation-caused immune disease. The PETE usage led to G-CSF levels increasing between 1st and 3rd day of observation, on the 7th day its level was at the normal rate. The comparative drug applying led to some stimulation of G-CSF levels, but compared to the PETE usage not so prolonged. During the early hours after depressive exposure under PETE treatment there was a significant IL-6 synthesis stimulation. At comparative drug applying its significant increase was registered only in the period from 16th to 24th hour. Influenced by PETE usage the Flt3-ligand level in mice with radiation-induced immune damage did not show such increase, which was observed in control animals – since the 3rd day their values were normal. At comparative drug applying the Flt3-ligand value did not reach normal rate on the 21st day of observation. Thus, due to the stimulation of the synthesis of blood formation positive regulators, which were determined in the bone marrow cells supernatant after radiation exposure, the PETE usage promoted successful and more rapid recovery of hematopoiesis, creating in turn the preconditions for effective immunogenesis.

Downloads

Download data is not yet available.

References

Bertho J.-M., Demarquay C., Douenat N. et al. Bone marrow stromal sells spontaneously produce Flt3-lagand: Influence of ionizing radiations and cytokine stimulation // International Journal of Radiation Biology. – 2008a. – Vol.84, issue 8. – Р. 659–667.

Bertho J.M., Prat M., Stefani J. et al. Correlation between plasma Flt3-ligand concentration and hematopoiesis during G-CSF-induced CD34+-cell mobilization // Stem Cells Dev. – 2008b. – No 17 (6). – Р. 1221–1225.

Bendall L.J., Bradstock K.F. G-CSF: From granulopoietic stimulant to bone marrow stem cell mobilizing agent // Cytokine and grows factor reviews – 2014. – Vol.25, issue 4. – Р. 355–367.

Chatterjee M., Osborne J., Bestetti G. et al. Viral IL-6-induced cell proliferation and immune evasion of interferon activity // Science. – 2002. – Vol.298, no 5597. – Р. 1432–1435.

Chen Yong Feng, Wu Zhong Min, Xie Cong et al. Expression level of IL-6 secreted by bone marrow stromal cells in mice with aplastic anemia // SRN Hematology. – 2013. – Vol.2013. – Article ID 986219.

De Haan I., Weersiong E., Dontje B. et al. In vitro generation of long-term repopulating hematopoietic stem cells by fibroblast growth factor-1 // Developmental Cell. – 2003. – Vol.4, issue 2. – P. 241–251.

Driessen R., Johnston H., Nisson S. Membrane bound stem cell factor is a key regulator in initial lodgment of stem cells within the endosteal marrow region // Exp. Hematol. – 2003. – Vol.31. – P. 1284–1291.

Eckert S., Bauer G. TGF-beta isoforms and fibroblasts growth factor exhibit analogous indirect antioncogenic activity through triggering of intracellular induction of apoptosis // Anticancer Res. – 1998. – No 18 (1A). – Р. 45–52.

European convention for the protection of vertebrate animals used for experimental and other scientific purposes // Strasburg. Council Treaty Series. – 1987. – No 123. – 52р.

Fu J.X., Zhang H., Yu F., Zhang X.G. Evaluation of serum and bone marrow positive- and negative- hematopoietic growth factors in patients with aplastic anemia and its implication on clinical outcome // Archives of Surgery. – 2000. – Vol.16. – Р. 554–557.

Gilbert L.A., Hemann M.T. Context-specific roles for paracrine IL-6 in lymphomagenesis // Genes & Dev. – 2012. – No 26. – Р. 1758–1768.

Gilliland D.G., Griffin J.D. The roles of FLT3 in hematopoiesis and leukemia // Blood. – 2002. – No 100 (5). – P. 1532–1542.

Herodin F., Mestries J.-C., Janodet D. et al. Recombinant glycosylated human interleukin-6 accelerates peripheral blood platelet count recovery in radiation-induced bone marrow depression in baboons // Blood. – 1992. – Vol.80, no 3. – P. 688–695.

Johns J.L., Christopher M.M. Extramedullary hematopoiesis: a new look at the underlying stem cell niche, theories of development, and occurrence in animals // Veterinary Pathology. – 2012. – Vol.49, no 3. – Р. 508–523.

Jung M., Kern F.G., Jorgensen T.J. Fibroblast growth factor-4 enhanced G2 arrest and cell survival following ionizing radiation // Cancer research. – 1994. – No 54. – Р. 5194–5197.

Karabagli H., Karabagli P., Ladher R.K., Shoenwolf G.C. Survey of fibroblast growth factor expression during chick organogenesis // The anatomical record. – 2002. – No 268. – Р. 1–6.

Kim D., Marchetti F., Chen Z. et al. Nanosensor dosimetry of mouse blood proteins after exposure to ionizing radiation // Sci. Rep. – 2013. – No 3. – Р. 2234.

Kulkarni S.S., Cary L.H., Gambles K. et al. Gamma-tocotrienol, a radiation prophylaxis agent, induces high levels of granulocyte colony-stimulating factor // Int. Immunopharmacol. – 2012. – No 14 (4). – Р. 495–503.
Radiation Proteomics: The effects of ionizing and non-ionizing radiation on cells and tissues / Ed. D.Leszczynski // Advances in Experimental Medicine and Biology. – Springer Science & Business Media, 2013. – Vol.990. – 130р.

Mickelsen A. Medical consequences of radiological and nuclear weapons. – Falls Church, Virginia: Office of the Surgeon General, United States Army, Washington, DC, U.S. G.P.O, 2012. – 260p.

Ossetrova N.I., Sandgren D.J., Blakeley W.F. Protein biomarkers for enhancement of radiation dose and injury assessment in nonhuman primate total-body irradiation model // Radiation Protection Dosimetry. – 2014. – Vol.159, issue 1–4. – Р. 61–76.

Prat M., Demarquay C., Frick J. et al. Radiation-induced increase in plasma Flt3-Ligand concentration in mice: Evidence for the implication of several cell types // Radiation Research. – 2005. – No 163. – Р. 408–417.

Prat M., Demarquay C., Frick J. et al. Use of Flt3 Ligand to evaluate residual hematopoiesis after heterogeneous irradiation in mice // Radiation Research. – 2006. – Vol.166, no 3. – Р. 504–511.

Singh V.K., Romaine P.L.P., Seed T.M. Medical countermeasures for radiation exposure and related injuries: characterization of medicines, FDA-approval status and inclusion into the strategic national stockpile // Health Phys. – 2015. – No 108(6). – Р. 607–630.

Zhang Z., Zhang M., Hang L. et al. Radiation effects of responsiveness of bone marrow to G-CSF // Adv. Exp. Med. Biol. – 2013. – Р. 273–280.

Zhao M., Ross G.T., Itkin T. et al. FGF signaling facilitates postinjury recovery of mouse hematopoietic system // Blood. – 2012. – Vol.120, no 9. – P. 1831–1842.
Cited
How to Cite
Погоріла, М. С., Попов, М. М., Романова, О. А., Щербак, О. М., & Мартинов, А. В. (1). Dynamics of hematopoiesis regulators content in bone marrow under the poultry embryonic tissue extract usage for the radiation-caused immune damage correction. The Journal of V.N.Karazin Kharkiv National University. Series «Biology», 27, 131-138. Retrieved from https://periodicals.karazin.ua/biology/article/view/8204
Section
PHYSIOLOGY OF HUMAN AND ANIMALS