Differential stem- and progenitor-cell trafficking by prostaglandin E2

Jonathan Hoggatt, Khalid S Mohammad, Pratibha Singh, Amber F Hoggatt, Brahmananda R Chitteti, Jennifer M Speth, Peirong Hu, Bradley A Poteat, Kayla N Stilger, Francesca Ferraro, Lev Silberstein, Frankie K Wong, Sherif S Farag, Magdalena Czader, Ginger L Milne, Richard M Breyer, Carlos H Serezani, David T Scadden, Theresa A Guise, Edward F Srour, Louis M Pelus, Jonathan Hoggatt, Khalid S Mohammad, Pratibha Singh, Amber F Hoggatt, Brahmananda R Chitteti, Jennifer M Speth, Peirong Hu, Bradley A Poteat, Kayla N Stilger, Francesca Ferraro, Lev Silberstein, Frankie K Wong, Sherif S Farag, Magdalena Czader, Ginger L Milne, Richard M Breyer, Carlos H Serezani, David T Scadden, Theresa A Guise, Edward F Srour, Louis M Pelus

Abstract

To maintain lifelong production of blood cells, haematopoietic stem cells (HSCs) are tightly regulated by inherent programs and extrinsic regulatory signals received from their microenvironmental niche. Long-term repopulating HSCs reside in several, perhaps overlapping, niches that produce regulatory molecules and signals necessary for homeostasis and for increased output after stress or injury. Despite considerable advances in the specific cellular or molecular mechanisms governing HSC-niche interactions, little is known about the regulatory function in the intact mammalian haematopoietic niche. Recently, we and others described a positive regulatory role for prostaglandin E2 (PGE2) on HSC function ex vivo. Here we show that inhibition of endogenous PGE2 by non-steroidal anti-inflammatory drug (NSAID) treatment in mice results in modest HSC egress from the bone marrow. Surprisingly, this was independent of the SDF-1-CXCR4 axis implicated in stem-cell migration. Stem and progenitor cells were found to have differing mechanisms of egress, with HSC transit to the periphery dependent on niche attenuation and reduction in the retentive molecule osteopontin. Haematopoietic grafts mobilized with NSAIDs had superior repopulating ability and long-term engraftment. Treatment of non-human primates and healthy human volunteers confirmed NSAID-mediated egress in other species. PGE2 receptor knockout mice demonstrated that progenitor expansion and stem/progenitor egress resulted from reduced E-prostanoid 4 (EP4) receptor signalling. These results not only uncover unique regulatory roles for EP4 signalling in HSC retention in the niche, but also define a rapidly translatable strategy to enhance transplantation therapeutically.

Figures

Figure 1. NSAIDs mobilize hematopoietic stem and…
Figure 1. NSAIDs mobilize hematopoietic stem and progenitor cells
Meloxicam enhances mobilization of HPC, a, and HSC b, into blood (n=4–5 mice/group/experiment; 3 experiments). c, Chimerism; d, competitive repopulating units (CRU); and e, LT-HSC frequency (Poisson distribution) 36 weeks after limiting dilution competitive transplants of peripheral blood mononuclear cells (PBMC) from mice treated with G-CSF and combination regimens (n=8 mice/group, assayed individually). Mice were treated with G-CSF or a staggered regimen of G-CSF + Meloxicam and PBMC transplanted into lethally irradiated mice. f, Neutrophil and g, platelet recovery were monitored for 90 days. *P<0.05, ** P<0.01, ***P<0.001; unpaired two-tailed t-test. All error bars represent mean ± s.e.m.
Figure 2. Non-human primates and healthy human…
Figure 2. Non-human primates and healthy human volunteers mobilize HSC/HPC in response to NSAID treatment
a, Four baboons were treated with G-CSF +/- Meloxicam in a cross-over design and b, CD34+ cells and c, CFU-GM in peripheral blood (PB) determined. d, CD34+ cells and e, CFU-GM in PB determined pre- and post-5 days of meloxicam alone treatment. Seven healthy human volunteers were treated with 15 mg/day p.o. for 5 days, and were assessed for f, CD34+ cells; g, CFU-GM; h, BFU-E, and i, CFU-GEMM pre- and post-treatment. Statistics represent paired, two-tailed t-test.
Figure 3. Prostaglandin E 2 EP4 receptor…
Figure 3. Prostaglandin E2 EP4 receptor antagonism/knockout expands bone marrow HPC and enhances mobilization
a, HPC mobilization with G-CSF, G-CSF + meloxicam, G-CSF + EP receptor antagonists, or G-CSF + meloxicam + EP receptor agonists (n=5 mice/group, assayed individually). b, The EP4 antagonist L-161,982 enhanced HSC mobilization (n=4 mice/group, assayed individually), and c, long-term reconstitution 16 weeks post-transplant with d, multi-lineage reconstitution (n=5 mice/group, assayed individually). e, Meloxicam enhances mobilization of HPC, and f, SLAM SKL cells in WT littermates, but not in EP4 conditional knockouts (n=3,4 mice/group, assayed individually). *P<0.05, ** P<0.01, ***P<0.001; unpaired two-tailed t-test. †P<0.05 compared to G-CSF + meloxicam. All error bars represent mean ± s.e.m.
Figure 4. NSAIDs attenuate hematopoietic supportive molecules…
Figure 4. NSAIDs attenuate hematopoietic supportive molecules and differentially mobilize HSC and HPC in OPN knockout and EP4 conditional knockout mice
a, b, Assessment of Col2.3-GFP cells after vehicle or meloxicam demonstrates reduced c, percentages and d, number of osteolineage cells (n=4 mice/group, assayed individually). e, Immunohistochemical staining of hematopoietic supportive molecules after treatment with meloxicam (400X). f, Meloxicam enhances mobilization of HPC in OPN −/− mice, with g,h, no enhancement in long-term reconstitution 16 weeks post-transplant. i, Representation of chimera generation allowing conditional knockout of donor hematopoietic cells, or recipient stromal cells. EP4 was deleted with tamoxifen 8 weeks post-transplant and mice treated with G-CSF or G-CSF + meloxicam. j, Enhanced mobilization of HPC by meloxicam when EP4 is expressed on hematopoietic cells and k, enhanced mobilization of HSC when EP4 is expressed by stromal cells (n=4 mice/group, assayed individually). *P<0.05, **P<0.01, ***P<0.001; unpaired two-tailed t-test. Error bars represent mean ± s.e.m.

References

    1. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841.
    1. Ding L, Saunders TL, Enikolopov G, Morrison SJ. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature. 2012;481:457.
    1. Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829.
    1. Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464:852.
    1. Zhang J, Niu C, Ye L, Huang H, He X, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425:836.
    1. Hoggatt J, Singh P, Sampath J, Pelus LM. Prostaglandin E2 enhances hematopoietic stem cell homing, survival, and proliferation. Blood. 2009;113:5444.
    1. North TE, Goessling W, Walkley CR, Lengerke C, Kopani KR, et al. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature. 2007;447:1007.
    1. Ahmed M, Khanna D, Furst DE. Meloxicam in rheumatoid arthritis. Expert. Opin. Drug Metab Toxicol. 2005;1:739.
    1. Rinder HM, Tracey JB, Souhrada M, Wang C, Gagnier RP, et al. Effects of meloxicam on platelet function in healthy adults: a randomized, double-blind, placebo-controlled trial. J. Clin. Pharmacol. 2002;42:881.
    1. Breyer RM, Bagdassarian CK, Myers SA, Breyer MD. Prostanoid receptors: subtypes and signaling. Annu. Rev. Pharmacol. Toxicol. 2001;41:661.
    1. Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124:407.
    1. Bethel M, Srour EF, Kacena MA. Hematopoietic cell regulation of osteoblast proliferation and differentiation. Curr. Osteoporos. Rep. 2011;9:96.
    1. Hoggatt J, Pelus LM. Many mechanisms mediating mobilization: an alliterative review. Curr. Opin. Hematol. 2011;18:231.
    1. Mundy GR, Yoneda T, Hiraga T. Preclinical studies with zoledronic acid and other bisphosphonates: impact on the bone microenvironment. Semin. Oncol. 2001;28:35.
    1. Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116:4815.
    1. Chow A, Lucas D, Hidalgo A, Mendez-Ferrer S, Hashimoto D, et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J. Exp Med. 2011;208:261.
    1. Christopher MJ, Rao M, Liu F, Woloszynek JR, Link DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J. Exp Med. 2011;208:251.
    1. Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 2002;109:625.
    1. Chitteti BR, Cheng YH, Poteat B, Rodriguez-Rodriguez S, Goebel WS, et al. Impact of interactions of cellular components of the bone marrow microenvironment on hematopoietic stem and progenitor cell function. Blood. 2010;115:3239.
    1. Nakamura Y, Arai F, Iwasaki H, Hosokawa K, Kobayashi I, et al. Isolation and characterization of endosteal niche cell populations that regulate hematopoietic stem cells. Blood. 2010;116:1422.
    1. Stier S, Ko Y, Forkert R, Lutz C, Neuhaus T, et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J. Exp. Med. 2005;201:1781.
    1. Grassinger J, Haylock DN, Storan MJ, Haines GO, Williams B, et al. Thrombin-cleaved osteopontin regulates hemopoietic stem and progenitor cell functions through interactions with alpha9beta1 and alpha4beta1 integrins. Blood. 2009;114:49.
    1. Nie Y, Waite J, Brewer F, Sunshine MJ, Littman DR, et al. The role of CXCR4 in maintaining peripheral B cell compartments and humoral immunity. J. Exp. Med. 2004;200:1145.
    1. Kennedy CR, Zhang Y, Brandon S, Guan Y, Coffee K, et al. Salt-sensitive hypertension and reduced fertility in mice lacking the prostaglandin EP2 receptor. Nat. Med. 1999;5:217.
    1. Guan Y, Zhang Y, Wu J, Qi Z, Yang G, et al. Antihypertensive effects of selective prostaglandin E2 receptor subtype 1 targeting. J. Clin. Invest. 2007;117:2496.
    1. Schneider A, Guan Y, Zhang Y, Magnuson MA, Pettepher C, et al. Generation of a conditional allele of the mouse prostaglandin EP4 receptor. Genesis. 2004;40:7.
    1. Mignone JL, Kukekov V, Chiang AS, Steindler D, Enikolopov G. Neural stem and progenitor cells in nestin-GFP transgenic mice. J. Comp Neurol. 2004;469:311.
    1. Kalajzic Z, Liu P, Kalajzic I, Du Z, Braut A, et al. Directing the expression of a green fluorescent protein transgene in differentiated osteoblasts: comparison between rat type I collagen and rat osteocalcin promoters. Bone. 2002;31:654.
    1. Chen XS, Sheller JR, Johnson EN, Funk CD. Role of leukotrienes revealed by targeted disruption of the 5-lipoxygenase gene. Nature. 1994;372:179.
    1. King AG, Horowitz D, Dillon SB, Levin R, Farese AM, et al. Rapid mobilization of murine hematopoietic stem cells with enhanced engraftment properties and evaluation of hematopoietic progenitor cell mobilization in rhesus monkeys by a single injection of SB-251353, a specific truncated form of the human CXC chemokine GRObeta. Blood. 2001;97:1534.
    1. Pelus LM, Broxmeyer HE, Kurland JI, Moore MA. Regulation of macrophage and granulocyte proliferation. Specificities of prostaglandin E and lactoferrin. J. Exp. Med. 1979;150:277.
    1. Broxmeyer HE, Mejia JA, Hangoc G, Barese C, Dinauer M, et al. SDF-1/CXCL12 enhances in vitro replating capacity of murine and human multipotential and macrophage progenitor cells. Stem Cells Dev. 2007;16:589.
    1. Fukuda S, Bian H, King AG, Pelus LM. The chemokine GRObeta mobilizes early hematopoietic stem cells characterized by enhanced homing and engraftment. Blood. 2007;110:860.
    1. Hoggatt AF, Hoggatt J, Honerlaw M, Pelus LM. A spoonful of sugar helps the medicine go down: a novel technique to improve oral gavage in mice. J. Am. Assoc. Lab Anim Sci. 2010;49:329.
    1. Sutherland DR, Anderson L, Keeney M, Nayar R, Chin-Yee I. The ISHAGE guidelines for CD34+ cell determination by flow cytometry. International Society of Hematotherapy and Graft Engineering. J. Hematother. 1996;5:213.
    1. Mohammad KS, Chen CG, Balooch G, Stebbins E, McKenna CR, et al. Pharmacologic inhibition of the TGF-beta type I receptor kinase has anabolic and anti-catabolic effects on bone. PLoS. One. 2009;4:e5275.
    1. Rowe PS, Matsumoto N, Jo OD, Shih RN, Oconnor J, et al. Correction of the mineralization defect in hyp mice treated with protease inhibitors CA074 and pepstatin. Bone. 2006;39:773.
    1. Parfitt AM, Drezner MK, Glorieux FH, Kanis JA, Malluche H, et al. Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee. J. Bone Miner. Res. 1987;2:595.
    1. Murali G, Milne GL, Webb CD, Stewart AB, McMillan RP, et al. Fish oil and indomethacin in combination potently reduce dyslipidemia and hepatic steatosis in LDLR−/−) mice. J. Lipid Res. 2012;53:2186.
    1. Liu T, Laidlaw TM, Feng C, Xing W, Shen S, et al. Prostaglandin E2 deficiency uncovers a dominant role for thromboxane A2 in house dust mite-induced allergic pulmonary inflammation. Proc. Natl. Acad. Sci. U. S. A. 2012;109:12692.

Source: PubMed

3
S'abonner