Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion

B Kumar, M Garcia, L Weng, X Jung, J L Murakami, X Hu, T McDonald, A Lin, A R Kumar, D L DiGiusto, A S Stein, V A Pullarkat, S K Hui, N Carlesso, Y-H Kuo, R Bhatia, G Marcucci, C-C Chen, B Kumar, M Garcia, L Weng, X Jung, J L Murakami, X Hu, T McDonald, A Lin, A R Kumar, D L DiGiusto, A S Stein, V A Pullarkat, S K Hui, N Carlesso, Y-H Kuo, R Bhatia, G Marcucci, C-C Chen

Abstract

Little is known about how leukemia cells alter the bone marrow (BM) niche to facilitate their own growth and evade chemotherapy. Here, we provide evidence that acute myeloid leukemia (AML) blasts remodel the BM niche into a leukemia growth-permissive and normal hematopoiesis-suppressive microenvironment through exosome secretion. Either engrafted AML cells or AML-derived exosomes increased mesenchymal stromal progenitors and blocked osteolineage development and bone formation in vivo. Preconditioning with AML-derived exosomes 'primed' the animals for accelerated AML growth. Conversely, disruption of exosome secretion in AML cells through targeting Rab27a, an important regulator involved in exosome release, significantly delayed leukemia development. In BM stromal cells, AML-derived exosomes induced the expression of DKK1, a suppressor of normal hematopoiesis and osteogenesis, thereby contributing to osteoblast loss. Conversely, treatment with a DKK1 inhibitor delayed AML progression and prolonged survival in AML-engrafted mice. In addition, AML-derived exosomes induced a broad downregulation of hematopoietic stem cell-supporting factors (for example, CXCL12, KITL and IGF1) in BM stromal cells and reduced their ability to support normal hematopoiesis. Altogether, this study uncovers novel features of AML pathogenesis and unveils how AML cells create a self-strengthening leukemic niche that promotes leukemic cell proliferation and survival, while suppressing normal hematopoiesis through exosome secretion.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
AML engraftment results in expansion of LT-HSCs, altered stromal compartments, reduced osteoblast number and bone architecture changes. (a) Neonatal DKO mice were injected with vehicle control or 2 million AML cells (MV411, KG1A, NB4 cell lines or COH101, COH103 patient samples) intrahepatically. The litter was killed when transplanted mice were moribund or 4 weeks after transplantation. Spleen weight of control and AML-transplanted (MV411, KG1A, COH101) DKO mice (n=5–7 mice per group in at least three independent experiments). (b) Frequency of the LT-HSC population in marrow of control and AML-transplanted (MV411, KG1A, NB4, COH101) DKO mice (n=5–8 mice per group in at least three independent experiments). (c) Frequency of Sca1+, CD146+ and CD166+ cells in the stromal compartment of control and AML-transplanted DKO mice (n=7–13 mice per group in at least three independent experiments). (d) Trichrome stain of bone sections from control and AML-transplanted (MV411, KG1A) DKO mice. Osteoblasts are marked with yellow arrows. (e) Immunofluorescence staining of bone sections from control or MLL-AF9-transplanted B6 mice. Staining of Sca1 and OCN (left) are shown. Osteoblasts are marked with yellow arrows. Number of Sca1+ cells in the view field determined by immunofluorescent staining (right). (f) Representative μ-CT 2D cross-section of trabecular bone region in femurs of control or AML-transplanted (KG1A, MV411) DKO mice. (g) Femurs of control or AML-transplanted (KG1A, MV411) DKO mice (n=6–8 in two independent experiments). Quantitative μ-CT analysis of cortical wall thickness in compact bone region, relative bone volume (BV/TV), thickness of trabecular bone, number of trabeculae and space between trabeculae in trabecular bone region. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Figure 2
Figure 2
AML-derived exosomes are elevated and associated with reduced OCN levels in AML patients, are internalized by BM cells, induce splenomegaly, increase LT-HSC population and alter stromal compartment in the BM microenvironment. (a) Plasma exosome numbers measured by NanoSight (left) and plasma OCN levels were determined by ELISA (right) in healthy control (n=12) and AML patients (n=43). (b) Plasma exosome count in AML patients with low (<10 ng/ml, n=29) or high (>10ng/ml, n=14) plasma OCN levels. (c) Overall, 100 μg CFSE-labeled exosomes were co-cultured with BM cells for 4 h. Uptake of CFSE-labeled exosomes by marrow cells was analyzed by fluorescence microscopy. (d) Uptake of CFSE-labeled exosomes by marrow cells was analyzed by FACS analysis for internalization of CFSE-labeled exosomes in indicated BM subpopulations (left). Percent CSFE+ cells within indicated BM subpopulations were summarized (right, n=3 mice in three independent experiments). (e) Six- to eight-week-old B6 mice were injected with either normal human PBMC-derived or AML-derived exosomes. Mice were euthanized 30 days after initial injection. (f) Mouse spleen is weight is summarized (n=5–6 mice per group in at least two independent experiments). (g) Frequency of LT-HSC in BM (n=6–8 mice per group in at least three independent experiments). (h) Frequency of Sca1+, CD146+ and CD166+ cells in the stromal compartment (n=6–8 mice per group in at least three independent experiments). ns, not significant, *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Figure 3
Figure 3
AML-derived exosomes modulate gene expression in BM stroma and suppress osteogenic differentiation of mesenchymal stromal progenitors; the block is reversed by DKK1 inhibitor. (a) qRT-PCR showing expression levels of indicated genes (n=4–12 in at least three independent experiments). BM Sca1+, CD146+ or CD166+ stromal cells from B6 mice treated with normal PBMC-derived or corresponding AML-derived exosomes. (b, c) Sorted Sca1+ stromal cells from BM of normal B6 mice were treated with normal PBMC-derived or AML (MV411, KG1A)-derived exosomes. Treated cells were divided and cultured in osteogenic, adipogenic or chondrogenic conditions for 21 days. Alizarin Red staining (b) and OCN expression (c) in stroma after osteogenic induction. (d) qRT-PCR of OCN expression of sorted Sca1+ cells from BM of normal B6 mice were cultured with PBMC-derived or AML (MV411)-derived exosomes, with or without DKK1 inhibitor (Way-262611). (e) NSG mice were injected with four doses of DKK1 inhibitor (Way-262611) or DMSO prior to AML transplantation and one dose after. Kaplan–Meier survival curve analysis of treated mice (lower, n=12–13 mice per group in two independent experiments). ns, not significant, *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Figure 4
Figure 4
AML-derived exosomes reduce stem cell activities, limit ability of stroma to support normal HSCs and accelerate AML progression; disruption of exosome production delays AML progression. (a) Repopulation ability of marrow from normal PBMC-derived or AML (KG1A)-derived exosome-treated donors was assayed 22 weeks after transplantation into congenic recipients (n=5–6 mice per group in two independent experiments). (b) CD45.1 HSCs were co-cultured with the AML cell/exosome-modified stroma for 48 h and transplanted (100 per mouse) into lethally irradiated WT CD45.2 recipients along with 200 000 unfractionated CD45.2 helper BM cells. Ability of control and AML cell/exosome co-cultured HSCs to repopulate the blood of recipient mice was assayed 19 weeks after transplantation (n=6–8 mice per group in three independent experiments). (c) Three weekly doses of AML-derived or normal PBMC-derived exosomes, or PBS vehicle control, were injected intravenously into 6- to 8-week-old DKO mice. Mice were irradiated and injected with 20 million KG1A cells. AML cell engraftment in peripheral blood was assayed 20 days later (middle). Kaplan–Meier survival analysis of control or treated mice (right; n=8–13 mice in at least two independent experiments). (d) MV411 AML cells were transduced with lentiviral shRNA against Rab27a, a protein involved in exosome release. qRT-PCR of Rab27a levels (left) and total exosome protein quantification (middle). Kaplan–Meier survival curve for mice transplanted with lentiviral shRab27a-transduced MV411 cells (right; n=8–10 mice in two independent experiments). ns, not significant, *P<0.05, **P<0.01, ***P<0.001.

References

    1. Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature 2014; 505: 327–334.
    1. Lane SW, Scadden DT, Gilliland DG. The leukemic stem cell niche: current concepts and therapeutic opportunities. Blood 2009; 114: 1150–1157.
    1. Walkley CR, Olsen GH, Dworkin S, Fabb SA, Swann J, McArthur GA et al. A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency. Cell 2007; 129: 1097–1110.
    1. Walkley CR, Shea JM, Sims NA, Purton LE, Orkin SH. Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment. Cell 2007; 129: 1081–1095.
    1. Wang L, Zhang H, Rodriguez S, Cao L, Parish J, Mumaw C et al. Notch-dependent repression of miR-155 in the bone marrow niche regulates hematopoiesis in an NF-kappaB-dependent manner. Cell Stem Cell 2014; 15: 51–65.
    1. Raaijmakers MHGP, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 2010; 464: 852–857.
    1. Kode A, Manavalan JS, Mosialou I, Bhagat G, Rathinam CV, Luo N et al. Leukaemogenesis induced by an activating beta-catenin mutation in osteoblasts. Nature 2014; 506: 240–244.
    1. Zhang B, Ho YW, Huang Q, Maeda T, Lin A, Lee SU et al. Altered microenvironmental regulation of leukemic and normal stem cells in chronic myelogenous leukemia. Cancer Cell 2012; 21: 577–592.
    1. Schepers K, Pietras EM, Reynaud D, Flach J, Binnewies M, Garg T et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell 2013; 13: 285–299.
    1. Medyouf H, Mossner M, Jann JC, Nolte F, Raffel S, Herrmann C et al. Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit. Cell Stem Cell 2014; 14: 824–837.
    1. Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 2008; 10: 1470–1476.
    1. Kowal J, Tkach M, Thery C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol 2014; 29: 116–125.
    1. Taylor DD, Gercel-Taylor C. Exosomes/microvesicles: mediators of cancer-associated immunosuppressive microenvironments. Semin Immunopathol 2011; 33: 441–454.
    1. Lee TH, D'Asti E, Magnus N, Al-Nedawi K, Meehan B, Rak J. Microvesicles as mediators of intercellular communication in cancer—the emerging science of cellular 'debris'. Semin Immunopathol 2011; 33: 455–467.
    1. Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, Dvorak P et al. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery. Leukemia 2006; 20: 847–856.
    1. Melo SA, Sugimoto H, O'Connell JT, Kato N, Villanueva A, Vidal A et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell 2014; 26: 707–721.
    1. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 2012; 18: 883–891.
    1. Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014; 25: 501–515.
    1. Corrado C, Raimondo S, Saieva L, Flugy AM, De Leo G, Alessandro R. Exosome-mediated crosstalk between chronic myelogenous leukemia cells and human bone marrow stromal cells triggers an interleukin 8-dependent survival of leukemia cells. Cancer Lett 2014; 348: 71–76.
    1. Huan J, Hornick NI, Shurtleff MJ, Skinner AM, Goloviznina NA, Roberts CT Jr et al. RNA trafficking by acute myelogenous leukemia exosomes. Cancer Res 2013; 73: 918–929.
    1. Huan J, Hornick NI, Goloviznina NA, Kamimae-Lanning AN, David LL, Wilmarth PA et al. Coordinate regulation of residual bone marrow function by paracrine trafficking of AML exosomes. Leukemia 2015; 29: 2285–2295.
    1. Park CY, Majeti R, Weissman IL. In vivo evaluation of human hematopoiesis through xenotransplantation of purified hematopoietic stem cells from umbilical cord blood. Nat Protoc 2008; 3: 1932–1940.
    1. Krause DS, Fulzele K, Catic A, Sun CC, Dombkowski D, Hurley MP et al. Differential regulation of myeloid leukemias by the bone marrow microenvironment. Nat Med 2013; 19: 1513–1517.
    1. Hanoun M, Zhang D, Mizoguchi T, Pinho S, Pierce H, Kunisaki Y et al. Acute myelogenous leukemia-induced sympathetic neuropathy promotes malignancy in an altered hematopoietic stem cell niche. Cell Stem Cell 2014; 15: 365–375.
    1. Hu X, Garcia M, Weng L, Jung X, Murakami JL, Kumar B et al. Identification of a common mesenchymal stromal progenitor for the adult haematopoietic niche. Nat Commun 2016; 7: 13095.
    1. Zhang X, Yuan X, Shi H, Wu L, Qian H, Xu W. Exosomes in cancer: small particle, big player. J Hematol Oncol 2015; 8: 83.
    1. Sheldon H, Heikamp E, Turley H, Dragovic R, Thomas P, Oon CE et al. New mechanism for Notch signaling to endothelium at a distance by Delta-like 4 incorporation into exosomes. Blood 2010; 116: 2385–2394.
    1. Thery C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol 2006; Chapter 3: Unit 3.22.
    1. Pinzone JJ, Hall BM, Thudi NK, Vonau M, Qiang YW, Rosol TJ et al. The role of Dickkopf-1 in bone development, homeostasis, and disease. Blood 2009; 113: 517–525.
    1. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol 2010; 12: 19–30.
    1. Camussi G, Deregibus MC, Bruno S, Grange C, Fonsato V, Tetta C. Exosome/microvesicle-mediated epigenetic reprogramming of cells. Am J Cancer Res 2011; 1: 98–110.
    1. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015; 527: 329–335.
    1. Wang W, Zimmerman G, Huang X, Yu S, Myers J, Wang Y et al. Aberrant Notch signaling in the bone marrow microenvironment of acute lymphoid leukemia suppresses osteoblast-mediated support of hematopoietic niche function. Cancer Res 2016; 76: 1641–1652.
    1. Frisch BJ, Ashton JM, Xing L, Becker MW, Jordan CT, Calvi LM. Functional inhibition of osteoblastic cells in an in vivo mouse model of myeloid leukemia. Blood 2012; 119: 540–550.
    1. Krevvata M, Silva BC, Manavalan JS, Galan-Diez M, Kode A, Matthews BG et al. Inhibition of leukemia cell engraftment and disease progression in mice by osteoblasts. Blood 2014; 124: 2834–2846.
    1. Bowers M, Zhang B, Ho Y, Agarwal P, Chen CC, Bhatia R. Osteoblast ablation reduces normal long-term hematopoietic stem cell self-renewal but accelerates leukemia development. Blood 2015; 125: 2678–2688.
    1. Massenkeil G, Fiene C, Rosen O, Michael R, Reisinger W, Arnold R. Loss of bone mass and vitamin D deficiency after hematopoietic stem cell transplantation: standard prophylactic measures fail to prevent osteoporosis. Leukemia 2001; 15: 1701–1705.
    1. Ganguly S, Divine CL, Aljitawi OS, Abhyankar S, McGuirk JP, Graves L. Prophylactic use of zoledronic acid to prevent early bone loss is safe and feasible in patients with acute myeloid leukemia undergoing allogeneic stem cell transplantation. Clin Transplant 2012; 26: 447–453.
    1. Drake MT. Osteoporosis and cancer. Curr Osteoporos Rep 2013; 11: 163–170.
    1. Mao B, Wu W, Li Y, Hoppe D, Stannek P, Glinka A et al. LDL-receptor-related protein 6 is a receptor for Dickkopf proteins. Nature 2001; 411: 321–325.
    1. Fleming HE, Janzen V, Lo Celso C, Guo J, Leahy KM, Kronenberg HM et al. Wnt signaling in the niche enforces hematopoietic stem cell quiescence and is necessary to preserve self-renewal in vivo. Cell Stem Cell 2008; 2: 274–283.
    1. Gustafson B, Eliasson B, Smith U. Thiazolidinediones increase the wingless-type MMTV integration site family (WNT) inhibitor Dickkopf-1 in adipocytes: a link with osteogenesis. Diabetologia 2010; 53: 536–540.
    1. Christodoulides C, Laudes M, Cawthorn WP, Schinner S, Soos M, O'Rahilly S et al. The Wnt antagonist Dickkopf-1 and its receptors are coordinately regulated during early human adipogenesis. J Cell Sci 2006; 119 (Pt 12): 2613–2620.
    1. Cheng SL, Shao JS, Cai J, Sierra OL, Towler DA. Msx2 exerts bone anabolism via canonical Wnt signaling. J Biol Chem 2008; 283: 20505–20522.
    1. Granchi D, Baglio SR, Amato I, Giunti A, Baldini N. Paracrine inhibition of osteoblast differentiation induced by neuroblastoma cells. International journal of cancer. J Int Cancer 2008; 123: 1526–1535.
    1. Gregory CA, Singh H, Perry AS, Prockop DJ. The Wnt signaling inhibitor dickkopf-1 is required for reentry into the cell cycle of human adult stem cells from bone marrow. J Biol Chem 2003; 278: 28067–28078.
    1. Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B et al. The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med 2003; 349: 2483–2494.
    1. Bu G, Lu W, Liu CC, Selander K, Yoneda T, Hall C et al. Breast cancer-derived Dickkopf1 inhibits osteoblast differentiation and osteoprotegerin expression: implication for breast cancer osteolytic bone metastases. International journal of cancer. J Int Cancer 2008; 123: 1034–1042.
    1. Voorzanger-Rousselot N, Goehrig D, Journe F, Doriath V, Body JJ, Clezardin P et al. Increased Dickkopf-1 expression in breast cancer bone metastases. Br J Cancer 2007; 97: 964–970.
    1. Wang Y, Krivtsov AV, Sinha AU, North TE, Goessling W, Feng Z et al. The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML. Science 2010; 327: 1650–1653.
    1. Lane SW, Wang YJ, Lo Celso C, Ragu C, Bullinger L, Sykes SM et al. Differential niche and Wnt requirements during acute myeloid leukemia progression. Blood 2011; 118: 2849–2856.
    1. Yeung J, Esposito MT, Gandillet A, Zeisig BB, Griessinger E, Bonnet D et al. beta-Catenin mediates the establishment and drug resistance of MLL leukemic stem cells. Cancer Cell 2010; 18: 606–618.
    1. Conklin KA. Dietary polyunsaturated fatty acids: impact on cancer chemotherapy and radiation. Altern Med Rev 2002; 7: 4–21.
    1. Ye H, Adane B, Khan N, Sullivan T, Minhajuddin M, Gasparetto M et al. Leukemic stem cells evade chemotherapy by metabolic adaptation to an adipose tissue niche. Cell Stem Cell 2016; 19: 23–37.
    1. Frost ML, Fogelman I, Blake GM, Marsden PK, Cook G Jr. Dissociation between global markers of bone formation and direct measurement of spinal bone formation in osteoporosis. J Bone Miner Res 2004; 19: 1797–1804.
    1. Yagi M, Arentsen L, Shanley RM, Rosen CJ, Kidder LS, Sharkey LC et al. A dual-radioisotope hybrid whole-body micro-positron emission tomography/computed tomography system reveals functional heterogeneity and early local and systemic changes following targeted radiation to the murine caudal skeleton. Calcif Tissue Int 2014; 94: 544–552.

Source: PubMed

3
Abonnieren