Ineffective erythropoiesis with reduced red blood cell survival in serotonin-deficient mice

Pascal Amireault, Sarah Hatia, Elisa Bayard, Florence Bernex, Corinne Collet, Jacques Callebert, Jean-Marie Launay, Olivier Hermine, Elke Schneider, Jacques Mallet, Michel Dy, Francine Côté, Pascal Amireault, Sarah Hatia, Elisa Bayard, Florence Bernex, Corinne Collet, Jacques Callebert, Jean-Marie Launay, Olivier Hermine, Elke Schneider, Jacques Mallet, Michel Dy, Francine Côté

Abstract

Serotonin (5-HT) has long been recognized as a neurotransmitter in the central nervous system, where it modulates a variety of behavioral functions. Availability of 5-HT depends on the expression of the enzyme tryptophan hydroxylase (TPH), and the recent discovery of a dual system for 5-HT synthesis in the brain (TPH2) and periphery (TPH1) has renewed interest in studying the potential functions played by 5-HT in nonnervous tissues. Moreover, characterization of the TPH1 knockout mouse model (TPH1(-/-)) led to the identification of unsuspected roles for peripheral 5-HT, revealing the importance of this monoamine in regulating key physiological functions outside the brain. Here, we present in vivo data showing that mice deficient in peripheral 5-HT display morphological and cellular features of ineffective erythropoiesis. The central event occurs in the bone marrow where the absence of 5-HT hampers progression of erythroid precursors expressing 5-HT(2A) and 5-HT(2B) receptors toward terminal differentiation. In addition, red blood cells from 5-HT-deficient mice are more sensitive to macrophage phagocytosis and have a shortened in vivo half-life. The combination of these two defects causes TPH1(-/-) animals to develop a phenotype of macrocytic anemia. Direct evidence for a 5-HT effect on erythroid precursors is provided by supplementation of the culture medium with 5-HT that increases the proliferative capacity of both 5-HT-deficient and normal cells. Our thorough analysis of TPH1(-/-) mice provides a unique model of morphological and functional aberrations of erythropoiesis and identifies 5-HT as a key factor for red blood cell production and survival.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Fig. 1.
Fig. 1.
TPH1−/− mice display a phenotype of aregenerative macrocytic anemia. (AC) A significant decrease in the number of RBCs (7.5 ± 0.2 vs. 8.8 ± 0.1 × 106 cells/μL), hematocrit (41.5 ± 0.8 vs. 45.0 ± 0.7%), and hemoglobin levels (15.1 ± 0.3 vs. 16.6 ± 0.2 g/dL) is seen in TPH1−/− vs. WT mice and indicates an anemic phenotype. (D and E) A significant increase in mean corpuscular volume (MCV) (55.0 ± 0.3 vs. 51.2 ± 0.2 fl), associated with a general increase in RBC cell size, is observed in TPH1−/− vs. WT mice and is consistent with macrocytosis. (F) Mean corpuscular hemoglobin (MCH) is increased significantly (20.2 ± 0.2 vs. 18.8 ± 0.1 pg), but (G) no difference is seen in mean corpuscular hemoglobin concentration (MCHC) (36.7 ± 0.2 vs. 36.8 ± 0.3 g/dL) in TPH1−/− vs. WT mice. (H) A significant increase in EPO level is observed in plasma of TPH1−/− mice (541 ± 81 pg/mL, n = 30) compared with WT mice (229 ± 49 pg/mL, n = 17) and indicates a response to the anemia. (I) No reticulocytosis is observed in TPH1−/− mice (184,000 ± 20,000 reticulocytes/mL, n = 23) compared with WT mice (164,000 ± 18,000 reticulocytes/mL, n = 17) and points to aregenerative anemia. P was calculated using an unpaired t test in G and I, an unpaired t test with Welch's correction in B and H, and a MannWhitney test in A, C, D, and F. P < 0.05 was considered significant. In AD, F, and G each dot represents a single blood count (WT, n = 42; TPH1−/−, n = 56; **P < 0.01; ***P < 0.001).
Fig. 2.
Fig. 2.
Decreased RBC survival and increased splenic iron deposits in TPH1−/− mice. (A) Flow cytometry analysis shows that a higher proportion of TPH1−/− RBCs binds annexin V and thus exposes PS at the surface. (B) TPH1−/− RBCs also have an increased sensitivity to osmotic shock because incubation in a hypertonic solution exacerbates the difference in annexin V binding between WT and TPH1−/− RBCs. (C) TPH1−/− RBCs show a marked susceptibility to in vitro phagocytosis compatible with previous studies reporting the enhanced clearance of PS-exposing cells by macrophages. (D) RBCs obtained from TPH1−/− mice show a significantly reduced half-life (10.3 ± 0.6 d, n = 12) compared with WT controls (14.4 ± 0.4 d, n = 12). (E) Decreased half-life of TPH1−/− RBCs could result from their ingestion by splenic macrophages because iron deposits are visualized by Perls’ staining. (F) Howell–Jolly bodies (arrowheads) also are seen on RBC smears of TPH1−/− mice, suggesting improper enucleation. P was calculated using an unpaired t test in B and C and an unpaired t test with Welch's correction in A. *P < 0.05; **P < 0.01.
Fig. 3.
Fig. 3.
Decreased number of erythroid precursors in the BM of TPH1−/− mice. (A) (Left and Center) Comparative analysis of femoral histology sections shows that TPH1−/− mice have a decreased hematopoietic cellularity in the BM. (Right) Total cell counts of BM cells confirm this observation, given that femur and tibia of TPH1−/− mice contain significantly fewer nucleated cells (13.2 ± 0.9 × 106 cells, n = 21) than those of WT mice (18.6 ± 0.9 × 106 cells, n = 16). (B) The frequency of the erythroid progenitor BFU-E is increased significantly in 5-HT–deficient mice (Left), but the total number of CFU-E is maintained (Right), suggesting a compensatory response to the anemia. (WT, n = 6; TPH1−/− mice, n = 6.) (C) No difference in the profile of Ter119+CD71+ cells (erythroblast population) is observed between TPH1−/− and WT cells isolated from the BM (Left and Center), but the total number of Ter119+CD71+ cells is significantly decreased in TPH1−/− compared with WT mice (Right) (3.5 ± 0.2 × 106 cells, n = 24, vs. 5.3 ± 0.3 × 106 cells, n = 20). P was calculated using an unpaired t test in A and B and a Mann–Whitney test in C. P < 0.05 was considered significant. **P < 0.01; ***P < 0.001.
Fig. 4.
Fig. 4.
Abnormal proliferation of erythroid precursors in TPH1−/− mice. WT and TPH1−/− mice were injected with BrdU, and Ter119+CD71+ cells were isolated from BM and analyzed by flow cytometry. (A) A typical flow cytometry profile reveals no difference in the DNA content, but (B) the percentage of cells incorporating BrdU is reduced significantly in 5-HT–deficient vs. WT mice (19.3 ± 1.4%, n = 11, vs. 24.7 ± 1.4%, n = 11). (C) BM aspirate smear from a TPH1−/− mouse showing binucleated erythroblasts (arrow) indicative of a cell-division defect. In B, P was calculated using a Mann–Whitney test. *P < 0.05.
Fig. 5.
Fig. 5.
Proliferation defect in TPH1−/− erythroid precursors rescued by 5-HT supplementation. Proerythroblasts obtained from WT (open square) or TPH1−/− (open circle) BM were induced to differentiate in vitro. Proliferative capacity was measured at 24, 48, and 72 h following the induction of proerythroblasts into a differentiation medium containing EPO and transferrin only (dotted line) or supplemented with 5-HT (solid line). (A) The significant decrease in proliferative capacity observed in 5-HT–deficient erythroid precursors was rescued by 5-HT supplementation (filled circles). Supplementation with 5-HT also increased the proliferative capacity of WT precursors (filled squares). (B) The significant increase in proliferative capacity when 5-HT is added to the differentiation medium (filled squares) also is observed when PNU 22394, a 5-HT2 agonist, is added to the medium (filled diamonds). Results are representative of two independent experiments (n = 6).

References

    1. Socolovsky M, et al. Ineffective erythropoiesis in Stat5a(-/-)5b(-/-) mice due to decreased survival of early erythroblasts. Blood. 2001;98:3261–3273.
    1. Lee JM, Chan K, Kan YW, Johnson JA. Targeted disruption of Nrf2 causes regenerative immune-mediated hemolytic anemia. Proc Natl Acad Sci USA. 2004;101:9751–9756.
    1. Leguit RJ, van den Tweel JG. The pathology of bone marrow failure. Histopathology. 2010;57:655–670.
    1. Cantor AB, Orkin SH. Transcriptional regulation of erythropoiesis: An affair involving multiple partners. Oncogene. 2002;21:3368–3376.
    1. Koury MJ, Sawyer ST, Brandt SJ. New insights into erythropoiesis. Curr Opin Hematol. 2002;9:93–100.
    1. Hoyer D, Hannon JP, Martin GR. Molecular, pharmacological and functional diversity of 5-HT receptors. Pharmacol Biochem Behav. 2002;71:533–554.
    1. Raymond JR, et al. Multiplicity of mechanisms of serotonin receptor signal transduction. Pharmacol Ther. 2001;92:179–212.
    1. Liu Y, Suzuki YJ, Day RM, Fanburg BL. Rho kinase-induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res. 2004;95:579–586.
    1. Sonier B, Arseneault M, Lavigne C, Ouellette RJ, Vaillancourt C. The 5-HT2A serotonergic receptor is expressed in the MCF-7 human breast cancer line and reveals a mitogenic effect of serotonin. Biochem Biophys Res Commun. 2006;43:1053–1059.
    1. Collet C, et al. The serotonin 5-HT2B receptor controls bone mass via osteoblast recruitment and proliferation. FASEB J. 2008;22:418–427.
    1. Nebigil CG, Etienne N, Messaddeq N, Maroteaux L. Serotonin is a novel survival factor of cardiomyocytes: Mitochondria as a target of 5-HT2B receptor signaling. FASEB J. 2003;17:1373–1375.
    1. Fitzpatrick PF. Tetrahydropterin-dependent amino acid hydroxylases. Annu Rev Biochem. 1999;68:355–381.
    1. Fitzpatrick PF. Tetrahydropterin-dependent amino acid hydroxylases. Annu Rev Biochem. 1999;68:355–381.
    1. Côté F, et al. Disruption of the nonneuronal tph1 gene demonstrates the importance of peripheral serotonin in cardiac function. Proc Natl Acad Sci USA. 2003;100:13525–13530.
    1. Walther DJ, et al. Synthesis of serotonin by a second tryptophan hydroxylase isoform. Science. 2003;299:76.
    1. Gutknecht L, Kriegebaum C, Waider J, Schmitt A, Lesch KP. Spatio-temporal expression of tryptophan hydroxylase isoforms in murine and human brain: Convergent data from Tph2 knockout mice. Eur Neuropsychopharmacol. 2009;19:266–282.
    1. Gershon MD, Tack J. The serotonin signaling system: From basic understanding to drug development for functional GI disorders. Gastroenterology. 2007;132:397–414.
    1. Matsuda M, et al. Serotonin regulates mammary gland development via an autocrine-paracrine loop. Dev Cell. 2004;6:193–203.
    1. Côté F, et al. Maternal serotonin is crucial for murine embryonic development. Proc Natl Acad Sci USA. 2007;104:329–334.
    1. Lesurtel M, et al. Platelet-derived serotonin mediates liver regeneration. Science. 2006;312:104–107.
    1. Yadav VK, et al. Lrp5 controls bone formation by inhibiting serotonin synthesis in the duodenum. Cell. 2008;135:825–837.
    1. Paulmann N, et al. Intracellular serotonin modulates insulin secretion from pancreatic beta-cells by protein serotonylation. PLoS Biol. 2009;7:e1000229.
    1. Lowy PH, Keighley G, Cohen NS. Stimulation by serotonin of erythropoietin-dependent erythropoiesis in mice. Br J Haematol. 1970;19:711–718.
    1. Noveck RJ, Fisher JW. Erythropoietic effects of 5-hydroxytryptamine. Proc Soc Exp Biol Med. 1971;138:103–107.
    1. Yang M, et al. Promoting effects of serotonin on hematopoiesis: Ex vivo expansion of cord blood CD34+ stem/progenitor cells, proliferation of bone marrow stromal cells, and antiapoptosis. Stem Cells. 2007;25:1800–1806.
    1. Goodnough LT, Skikne B, Brugnara C. Erythropoietin, iron, and erythropoiesis. Blood. 2000;96:823–833.
    1. Wagner KU, et al. Conditional deletion of the Bcl-x gene from erythroid cells results in hemolytic anemia and profound splenomegaly. Development. 2000;127:4949–4958.
    1. Pretsch W, Merkle S, Favor J, Werner T. A mutation affecting the lactate dehydrogenase locus Ldh-1 in the mouse. II. Mechanism of the LDH-A deficiency associated with hemolytic anemia. Genetics. 1993;135:161–170.
    1. McEvoy L, Williamson P, Schlegel RA. Membrane phospholipid asymmetry as a determinant of erythrocyte recognition by macrophages. Proc Natl Acad Sci USA. 1986;83:3311–3315.
    1. Van Putten LM. The life span of red cells in the rat and the mouse as determined by labeling with DFP32 in vivo. Blood. 1958;13:789–794.
    1. Connor J, Pak CC, Schroit AJ. Exposure of phosphatidylserine in the outer leaflet of human red blood cells. Relationship to cell density, cell age, and clearance by mononuclear cells. J Biol Chem. 1994;269:2399–2404.
    1. de Jong K, et al. Short survival of phosphatidylserine-exposing red blood cells in murine sickle cell anemia. Blood. 2001;98:1577–1584.
    1. Lin CS, Lim SK, D'Agati V, Costantini F. Differential effects of an erythropoietin receptor gene disruption on primitive and definitive erythropoiesis. Genes Dev. 1996;10:154–164.
    1. Koury ST, Koury MJ, Bondurant MC. Morphological changes in erythroblasts during erythropoietin-induced terminal differentiation in vitro. Exp Hematol. 1988;16:758–763.
    1. Wickramasinghe SN. Diagnosis of megaloblastic anaemias. Blood Rev. 2006;20:299–318.
    1. Kinross KM, Clark AJ, Iazzolino RM, Humbert PO. E2f4 regulates fetal erythropoiesis through the promotion of cellular proliferation. Blood. 2006;108:886–895.
    1. Tallack MR, Keys JR, Humbert PO, Perkins AC. EKLF/KLF1 controls cell cycle entry via direct regulation of E2f2. J Biol Chem. 2009;284:20966–20974.
    1. Li FX, Zhu JW, Hogan CJ, DeGregori J. Defective gene expression, S phase progression, and maturation during hematopoiesis in E2F1/E2F2 mutant mice. Mol Cell Biol. 2003;23:3607–3622.
    1. Koury MJ, Ponka P. New insights into erythropoiesis: The roles of folate, vitamin B12, and iron. Annu Rev Nutr. 2004;24:105–131.
    1. Koury MJ, Price JO, Hicks GG. Apoptosis in megaloblastic anemia occurs during DNA synthesis by a p53-independent, nucleoside-reversible mechanism. Blood. 2000;96:3249–3255.
    1. Dolznig H, et al. Erythroid progenitor renewal versus differentiation: Genetic evidence for cell autonomous, essential functions of EpoR, Stat5 and the GR. Oncogene. 2006;25:2890–2900.
    1. Dolznig H, et al. Establishment of normal, terminally differentiating mouse erythroid progenitors: Molecular characterization by cDNA arrays. FASEB J. 2001;15:1442–1444.
    1. Shuey DL, Sadler TW, Lauder JM. Serotonin as a regulator of craniofacial morphogenesis: Site specific malformations following exposure to serotonin uptake inhibitors. Teratology. 1992;46:367–378.
    1. Buznikov GA, Lambert HW, Lauder JM. Serotonin and serotonin-like substances as regulators of early embryogenesis and morphogenesis. Cell Tissue Res. 2001;305:177–186.

Source: PubMed

3
Subscribe