Thalidomide induces gamma-globin gene expression through increased reactive oxygen species-mediated p38 MAPK signaling and histone H4 acetylation in adult erythropoiesis

Wulin Aerbajinai, Jianqiong Zhu, Zhigang Gao, Kyung Chin, Griffin P Rodgers, Wulin Aerbajinai, Jianqiong Zhu, Zhigang Gao, Kyung Chin, Griffin P Rodgers

Abstract

Although thalidomide has been shown to improve anemia in some patients with myelodysplastic syndromes and stimulates erythropoietin in patients with multiple myeloma, thalidomide's specific effects on gamma-globin gene expression during erythroid differentiation have not been studied. Here, we investigated the effects of thalidomide on gamma-globin gene expression and the involved signaling pathway using an ex vivo culture system of primary human CD34+ cells. We found that thalidomide induced gamma-globin mRNA expression in a dose-dependent manner, but had no effect on beta-globin expression. We also demonstrated that intracellular reactive oxygen species (ROS) levels were increased by treatment with thalidomide for 48 hours (from day 3 to day 5). Western blot analysis demonstrated that thalidomide activated the p38 mitogen-activated protein kinase (MAPK) signaling pathway in a time- and dose-dependent manner and increased histone H4 acetylation. Pretreatment of cells with the antioxidant enzyme catalase and the intracellular hydroxyl scavenger dimethylthiourea (DMTU) abrogated the thalidomide-induced p38 MAPK activation and histone H4 acetylation. Moreover, pretreatment with catalase and DMTU diminished thalidomide-induced gamma-globin gene expression. These data indicate that thalidomide induces increased expression of the gamma-globin gene via ROS-dependent activation of the p38 MAPK signaling pathway and histone H4 acetylation.

Figures

Figure 1
Figure 1
Effects of thalidomide on cell growth and erythroid colony formation. (A) Human CD34+ cells were cultured in erythropoietin (EPO) medium with the indicated concentration of thalidomide for 2 weeks and counted on day 14. (B) Erythroid colonies (BFU-E + CFU-E) were calculated as numbers of colonies per well. CD34+ cells (1 × 103 cells/well) were cultured in methylcellulose with the indicated concentration of thalidomide. CFU-Es (▩) were evaluated at day 7 and BFU-Es (■) were evaluated at day 14. (C) Thalidomide (1-100 μM) was added to CD34+ cell cultures either for only the first week (days 0-7) or the second week (days 7-14), and the cells were counted on day 7 or day 14. Results are shown as means (± SD) from 3 different donors. *P < .05 versus untreated cells.
Figure 2
Figure 2
Effects of thalidomide on expression of γ- and β-globin mRNA in erythroid progenitor cells. CD34+ cells were cultured for 14 days in EPO and treated with different concentrations of thalidomide (0.01-100 μM) over the indicated time periods (days 0-14, days 0-6, or days 6-14). (A,C) γ-globin and (B,D) β-globin mRNA levels were quantitated by real-time PCR. Results are shown as means (± SD) from 3 different donors. *P < .05 versus untreated cells.
Figure 3
Figure 3
Thalidomide induces activation of p38 MAPK and acetylation of histone H4. CD34+ cells were cultured in EPO alone medium for 7 days prior to the addition of thalidomide. (A) Cells were treated with different concentrations of thalidomide for 30 minutes. (B) Cells were treated with 100 μM thalidomide for the indicated times. Cellular protein (30 μg) was analyzed by Western blot using phosphorylated p38 MAPK and acetyl-histone H4 antibodies. The membranes were stripped and reprobed with total p38 MAPK and histone H4 antibodies as indicated to confirm that similar amounts of protein extracts were analyzed in each lane.
Figure 4
Figure 4
Thalidomide induces intracellular ROS in erythroid progenitor cells. CD34+ cells were treated with the indicated concentration of thalidomide (A) or with 100 μM thalidomide in the presence or absence of catalase (4000 U/mL), DMTU (10 mM), or SB203580 (5 μM) (B) from day 3 to day 5 for 48 hours, followed by treatment with the DCFDA probe for 10 minutes. The intracellular reactive oxygen species (ROS) production was measured by flow cytometry. ROS levels induced by thalidomide were normalized to those of untreated cells as determined by monitoring the increased fluorescence in the cells. Results are shown as means (± SD) from 3 independent experiments. *P < .05 versus untreated cells.
Figure 5
Figure 5
Increased p38 MAPK phosporylation and acetylation of histone H4 by thalidomide inhibited by the antioxidants catalase and DMTU, and p38 inhibitor SB203580. CD34+ cells cultured in EPO medium for 7 days were pretreated with the antioxidants catalase (4000 U/mL) or DMTU (10 mM) (A) or with 5, 10, or 20 μM of the p38 MAPK inhibitor SB203580 (B) for 30 minutes, then coincubated with 100 μM thalidomide in the presence of indicated inhibitors for 30 minutes. Cell lysates (30 μg) were analyzed by Western blot using phosphorylated p38 MAPK and acetyl-histone H4 antibodies. Total p38 MAPK and histone H4 were analyzed as loading controls as described in Figure 3.
Figure 6
Figure 6
Thalidomide-induced γ-globin expression requires enhanced ROS and mediated p38 MAPK activation. CD34+ cells grown in EPO medium for 6 days were treated with 100 μM thalidomide in the presence or absence of the antioxidants catalase (4000 U/mL) or DMTU (10 mM) or the p38 MAPK inhibitor SB203580 (5 μM) from days 6 to 14. RNA extracted from cells harvested on day 14 was amplified by quantitative PCR to determine γ- and β-globin expression levels. (A) Average copy number of γ-globin molecules per 1 ng total RNA. (B) Average copy number of β-globin molecules per 1 ng total RNA. (C) Average ratio of γ/γ + β-globin percentages. Results are shown as means (± SD) from 3 independent donors that were analyzed in separate experiments. *P < .05 versus untreated cells.
Figure 7
Figure 7
Thalidomide induced fetal hemoglobin expression. Cells were cultured in EPO on days 0 to 6 and then were treated with the indicated concentration of thalidomide (A) or 100 μM thalidomide in the presence or absence of catalase (4000 U/mL), DMTU (10 mM), or SB203580 (5 μM) (B) from days 6 to 14. On day 14, cells were stained with fluorescently labeled anti-HbF antibodies and analyzed by flow cytometry. Panels represent fluorescence signal (y-axis) versus cell size (x-axis); the percentage of positive cells is shown in the top right corner of each panel. The panels shown are representative of the results for cultures from 3 separate donors.

References

    1. Lenz W, Knapp K. Thalidomide embryopathy. Arch Environ Health. 1962;5:100–105.
    1. Eriksson T, Bjorkman S, Hoglund P. Clinical pharmacology of thalidomide. Eur J Clin Pharmacol. 2001;57:365–376.
    1. Keifer JA, Guttridge DC, Ashburner BP, Baldwin AS., Jr Inhibition of NF-kappa B activity by thalidomide through suppression of IkappaB kinase activity. J Biol Chem. 2001;276:22382–22387.
    1. Stephens TD, Fillmore BJ. Hypothesis: thalidomide embryopathy-proposed mechanism of action. Teratology. 2000;61:189–195.
    1. Singhal S, Mehta J, Desikan R, et al. Anti-tumor activity of thalidomide in refractory multiple myeloma. N Engl J Med. 1999;341:1565–1571.
    1. Sampaio EP, Sarno EN, Galilly R, Cohn ZA, Kaplan G. Thalidomide selectively inhibits tumor necrosis factor alpha production by stimulated human monocytes. J Exp Med. 1991;173:699–703.
    1. Li X, Liu X, Wang J, et al. Thalidomide down-regulates the expression of VEGF and bFGF in cisplatin-resistant human lung carcinoma cells. Anticancer Res. 2003;23:2481–2487.
    1. Fujita J, Mestre JR, Zeldis JB, Subbaramaiah K, Dannenberg AJ. Thalidomide and its analogues inhibit lipopolysaccharide-mediated induction of cyclooxygenase-2. Clin Cancer Res. 2001;7:3349–3355.
    1. Sauer H, Günther J, Hescheler J, Wartenberg M. Thalidomide inhibits angiogenesis in embryoid bodies by the generation of hydroxyl radicals. Am J Pathol. 2000;156:151–158.
    1. Parman T, Wiley MJ, Wells PG. Free radical-mediated oxidative DNA damage in the mechanism of thalidomide teratogenicity. Nat Med. 1999;5:582–585.
    1. Hancock JT, Desikan R, Neill SJ. Role of reactive oxygen species in cell signaling pathways. Biochem Soc Trans. 2001;29:345–350.
    1. Musto P. Thalidomide therapy for myelodysplastic syndromes: current status and future perspectives. Leuk Res. 2004;28:325–332.
    1. Grzasko N, Dmoszynska A, Hus M, Soroka-Wojtaszko M. Stimulation of erythropoiesis by thalidomide in multiple myeloma patients: its influence on FasL, TRAIL and their receptors on erythroblasts. Haematologica. 2006;91:386–389.
    1. Raza A, Meyer P, Dutt D, et al. Thalidomide produces transfusion independence in long-standing refractory anemias of patients with myelodysplastic syndromes. Blood. 2001;98:958–965.
    1. Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. N Engl J Med. 1995;332:1317–1322.
    1. DeSimone J, Koshy M, Dorn L. Maintenance of elevated fetal hemoglobin levels by decitabine during dose interval treatment of sickle cell anemia. Blood. 2002;99:3905–3908.
    1. Cao H, Stamatoyannopoulos G, Jung M. Induction of human gamma-globin gene expression by histone deacetylase inhibitors. Blood. 2004;103:701–709.
    1. Rodgers GP, Saunthararajah Y. Advances in experimental treatment of beta-thalassaemia. Expert Opin Investig Drugs. 2001;10:925–934.
    1. Pace BS, Qian XH, Sangerman J, et al. p38 MAP kinase activation mediates gamma-globin gene induction in erythroid progenitors. Exp Hematol. 2003;31:1089–1096.
    1. Witt O, Monkemeyer S, Kanbach K, Pekrun A. Induction of fetal hemoglobin synthesis by valproate: modulation of MAP kinase pathways. Am J Hematol. 2002;71:45–46.
    1. Witt O, Monkemeyer S, Ronndahl B, Reinhardt D, Kanbach K, Pekrun A. Induction of fetal hemoglobin expression by the histone deacetylase inhibitor apicidin. Blood. 2003;101:2001–2007.
    1. Hsiao CH, Li W, Lou TF, Baliga BS, Pace BS. Fetal hemoglobin induction by histone deacetylase inhibitors involves generation of reactive oxygen species. Exp Hematol. 2006;34:264–273.
    1. Wojda U, Noel P, Miller JL. Fetal and adult hemoglobin production during adult erythropoiesis: coordinate expression correlates with cell proliferation. Blood. 2002;99:3005–3013.
    1. Koh KR, Janz M, Mapara MY, et al. Immunomodulatory derivative of thalidomide (IMiD CC-4047) induces a shift in lineage commitment by suppressing erythropoiesis and promoting myelopoiesis. Blood. 2005;105:3833–3840.
    1. Anderson G, Gries M, Kurihara N, et al. Thalidomide derivative CC-4047 inhibits osteoclast formation by down-regulation of PU 1. Blood. 2006;107:3098–3105.
    1. Dallalio G, Means RT., Jr Effects of oxidative stress on human erythroid colony formation: Modulation by gamma-interferon. J Lab Clin Med. 2003;141:395–400.
    1. Wojda U, Leigh KR, Njoroge JM, et al. Fetal hemoglobin modulation during human erythropoiesis: stem cell factor has “late” effects related to the expression pattern of CD117. Blood. 2003;101:492–497.
    1. Yamashita T, Wakao H, Miyajima A, Asano S. Differentiation inducers modulate cytokine signaling pathways in a murine erythroleukemia cell line. Cancer Res. 1998;58:556–61.
    1. Boosalis MS, Bandyopadhyay R, Bresnick EH, et al. Short-chain fatty acid derivatives stimulate cell proliferation and induce STAT-5 activation. Blood. 2001;97:3259–3267.
    1. Pace BS, Qian X, Sangerman J, et al. p38 MAP kinase is required for fetal hemoglobin induction by butyrate and trichostatin. Exp Hematol. 2003;11:1089–1096.
    1. Witt O, Sand K, Pekrum A. Butyrate-induced erythroid differentiation of human K562 leukemia cells involves inhibition of ERK and activation of p38 MAP kinase pathways. Blood. 2000;95:2391–2396.
    1. Johnson J, Hunter R, McElveen R, Qian XH, Baliga BS, Pace BS. Fetal hemoglobin induction by the histone deacetylase inhibitor, scriptaid. Cell Mol Biol. 2005;51:229–238.
    1. Varghese J, Chattopadhaya S, Sarin A. Inhibition of p38 kinase reveals a TNF-alpha-mediated, caspase-dependent, apoptotic death pathway in a human myelomonocyte cell line. J Immunol. 2001;166:6570–6577.
    1. Nguyen LT, Duncan GS, Mirtsos C, et al. TRAF2 deficiency results in hyperactivity of certain TNFR1 signals and impairment of CD40-mediated responses. Immunity. 1999;11:379–389.
    1. Nagata Y, Todokoro K. Requirement of activation of JNK and p38 for environmental stress-induced erythroid differentiation and apoptosis and of inhibition of ERK for apoptosis. Blood. 1999;94:853–863.
    1. Bulger M, Sawado T, Schubeler D, Groudine M. ChIPs of the beta-globin locus: unraveling gene regulation within an active domain. Curr Opin Genet Dev. 2002;12:170–177.
    1. Forsberg EC, Bresnick EH. Histone acetylation beyond promoters: long-range acetylation patterns in the chromatin world. Bioessays. 2001;23:820–830.
    1. Rahman I, Gilmour PS, Jimenez LA, MacNee W. Oxidative stress and TNF-alpha induce histone acetylation and NF-B/AP-1 activation in alveolar epithelial cells: potential mechanism in gene transcription in lung inflammation. Mol Cell Biochem. 2002;234/235:239–248.
    1. Tomita K, Barnes PJ, Adcock IM. The effect of oxidative stress on histone acetylation and IL-8 release. Biochem Biophys Res Commun. 2003;301:527–577.
    1. Anderson ME, Luo JL. Glutathione therapy: from prodrugs to genes. Semin Liver Dis. 1998;18:415–24.
    1. Iiyama M, Kakihana K, Kurosu T, Miura O. Reactive oxygen species generated by hematopoietic cytokines play roles in activation of receptor-mediated signaling and in cell cycle progression. Cell Signal. 2006;18:174–182.
    1. Sangerman J, Lee MS, Yao X, et al. Mechanism for fetal hemoglobin induction by histone deacetylase inhibitors involves γ-globin activation by CREB1 and ATF-2. Blood. 2006;108:3590–3599.
    1. Mankidy R, Faller DV, Mabaera R, et al. Short-chain fatty acids induce gamma-globin gene expression by displacement of a HDAC3-NCoR repressor complex. Blood. 2006;108:3179–3186.

Source: PubMed

3
Suscribir