Chronic variable stress activates hematopoietic stem cells

Timo Heidt, Hendrik B Sager, Gabriel Courties, Partha Dutta, Yoshiko Iwamoto, Alex Zaltsman, Constantin von Zur Muhlen, Christoph Bode, Gregory L Fricchione, John Denninger, Charles P Lin, Claudio Vinegoni, Peter Libby, Filip K Swirski, Ralph Weissleder, Matthias Nahrendorf, Timo Heidt, Hendrik B Sager, Gabriel Courties, Partha Dutta, Yoshiko Iwamoto, Alex Zaltsman, Constantin von Zur Muhlen, Christoph Bode, Gregory L Fricchione, John Denninger, Charles P Lin, Claudio Vinegoni, Peter Libby, Filip K Swirski, Ralph Weissleder, Matthias Nahrendorf

Abstract

Exposure to psychosocial stress is a risk factor for many diseases, including atherosclerosis. Although incompletely understood, interaction between the psyche and the immune system provides one potential mechanism linking stress and disease inception and progression. Known cross-talk between the brain and immune system includes the hypothalamic-pituitary-adrenal axis, which centrally drives glucocorticoid production in the adrenal cortex, and the sympathetic-adrenal-medullary axis, which controls stress-induced catecholamine release in support of the fight-or-flight reflex. It remains unknown, however, whether chronic stress changes hematopoietic stem cell activity. Here we show that stress increases proliferation of these most primitive hematopoietic progenitors, giving rise to higher levels of disease-promoting inflammatory leukocytes. We found that chronic stress induced monocytosis and neutrophilia in humans. While investigating the source of leukocytosis in mice, we discovered that stress activates upstream hematopoietic stem cells. Under conditions of chronic variable stress in mice, sympathetic nerve fibers released surplus noradrenaline, which signaled bone marrow niche cells to decrease CXCL12 levels through the β3-adrenergic receptor. Consequently, hematopoietic stem cell proliferation was elevated, leading to an increased output of neutrophils and inflammatory monocytes. When atherosclerosis-prone Apoe(-/-) mice were subjected to chronic stress, accelerated hematopoiesis promoted plaque features associated with vulnerable lesions that cause myocardial infarction and stroke in humans.

Figures

Figure 1. Chronic stress increases proliferation of…
Figure 1. Chronic stress increases proliferation of hematopoietic stem and progenitor cells in the bone marrow
a, Cohen’s perceived stress scale score in medical ICU residents (n = 10–15, Student’s t–test). b, Blood leukocytes in residents (n = 29, Wilcoxon test). c, Leukocytes in mouse blood and d, bone marrow after 3 weeks of stress (n = 9 per group, Student’s t–test). e, Gating for LSK and HSC. Proliferation was assessed after 3 weeks of stress (n = 5 per group, Mann–Whitney test). f, Bone marrow colony forming unit (CFU) assay after one week of stress (n = 5 per group, Mann–Whitney test). g, Bone marrow HSC and LSK after 3 weeks of stress (n = 10 per group, Student’s t–test). h, Long–term competitive repopulation assay using limiting dilutions of whole bone marrow from stressed or non–stressed mice (Poisson statistics for LT–HSC frequencies, P = 0.2 two–tailed t–test). i, Competitive reconstitution with 2×106 bone marrow cells from stressed or non–stressed controls co–injected with equal numbers of naive competitor cells, followed by assessment of blood chimerism (n = 10 mice per group, one–way ANOVA). Mean ± s.e.m., * P < 0.05.
Figure 2. Hematopoietic progenitors in the bone…
Figure 2. Hematopoietic progenitors in the bone marrow of stressed mice dilute DiD membrane dye faster
a, Intravital microscopy of the mouse calvarium after adoptive transfer of DiD–labelled LSK (white arrows) before and seven days after stress (n = 5 mice per group, Mann–Whitney test). Dotted lines outline bone. Scale bar indicates 50 μm. Single dots in graphs represent DiD+ cells’ target–to–background ratio before (upper panel) and after stress (lower panel). b, DiD fluorescence on day 0 and 7 days after adoptive transfer of DiD+ GFP+ LSK in non–stressed control (CT) or stressed mice (n = 5 per group). The bar graph illustrates the DiD fluorescence in GFP+ LSK (Mann–Whitney test). Mean ± s.e.m., * P < 0.05.
Figure 3. Stress–induced sympathetic nervous signaling regulates…
Figure 3. Stress–induced sympathetic nervous signaling regulates proliferation of bone marrow HSC via CXCL12
a, Noradrenaline ELISA after 3 weeks of stress (n = 8 per group, Student’s t–test). b, Immunoreactive staining for tyrosine hydroxylase (TH) in bone marrow. Scale bar indicates 10 μm. Bar graph shows TH–positive area (n = 5 mice per group, Mann–Whitney test). c, CXCL12 mRNA in bone marrow (n = 10 per group, one–way ANOVA). d, CXCL12 protein in bone marrow (n = 7 per group, one–way ANOVA). e, Dot plots and quantification of LSK and HSC (n = 5 per group, Mann–Whitney test). f, Effects of β3 adrenoreceptor blocker on blood leukocytes (n = 5 per group, Mann–Whitney test). Mean ± s.e.m., * P < 0.05.
Figure 4. Chronic stress increases inflammation in…
Figure 4. Chronic stress increases inflammation in mouse atherosclerotic plaques
a, Protease activity in aortic roots of ApoE−/− mice measured by FMT–CT after 6 weeks of stress. Circles indicate aortic root (n = 5 per group, Mann–Whitney test). b, Immunoreactive staining of aortic roots for CD11b and Ly6G. Bar graphs show percentage of positive area per root (n = 9–10 per group, Student’s t–test). Scale bar indicates 200 μm. c, Gating and quantification of aortic myeloid cells (n = 10 per group, Student’s t–test). d, qPCR for inflammatory genes in aorta (n = 9–10 per group, Student’s t–test). e, Masson trichrome staining (n = 9 per group, Student’s t–test). Scale bar indicates 50 μm, arrows point at fibrous cap covering necrotic core. Bar graphs show fibrous cap thickness and necrotic core area. Mean ± s.e.m., *P < 0.05.

References

    1. Black PH. The inflammatory response is an integral part of the stress response: Implications for atherosclerosis, insulin resistance, type II diabetes and metabolic syndrome X. Brain Behav Immun. 2003;17:350–364.
    1. Rosengren A, et al. Association of psychosocial risk factors with risk of acute myocardial infarction in 11119 cases and 13648 controls from 52 countries (the INTERHEART study): case-control study. Lancet. 2004;364:953–962.
    1. Glaser R, Kiecolt-Glaser JK. Stress-induced immune dysfunction: implications for health. Nat Rev Immunol. 2005;5:243–251.
    1. Powell ND, et al. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via beta-adrenergic induction of myelopoiesis. Proc Natl Acad Sci U S A. 2013;110:16574–16579.
    1. Cohen S, Kamarck T, Mermelstein R. A global measure of perceived stress. J Health Soc Behav. 1983;24:385–396.
    1. Lesage FX, Berjot S. Validity of occupational stress assessment using a visual analogue scale. Occup Med (Lond) 2011;61:434–436.
    1. Schweizer MC, Henniger MS, Sillaber I. Chronic mild stress (CMS) in mice: of anhedonia, ‘anomalous anxiolysis’ and activity. PLoS One. 2009;4:e4326.
    1. Nollet M, Guisquet AM, Belzung C. Models of depression: unpredictable chronic mild stress in mice. Curr Protoc Pharmacol. 2013 Chapter 5, Unit5.65.
    1. Yalcin I, Aksu F, Belzung C. Effects of desipramine and tramadol in a chronic mild stress model in mice are altered by yohimbine but not by pindolol. Eur J Pharmacol. 2005;514:165–174.
    1. Kiel MJ, et al. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121:1109–1121.
    1. Wilson A, et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell. 2008;135:1118–1129.
    1. Szilvassy SJ, Humphries RK, Lansdorp PM, Eaves AC, Eaves CJ. Quantitative assay for totipotent reconstituting hematopoietic stem cells by a competitive repopulation strategy. Proc Natl Acad Sci U S A. 1990;87:8736–8740.
    1. Essers MA, et al. IFNalpha activates dormant haematopoietic stem cells in vivo. Nature. 2009;458:904–908.
    1. Baldridge MT, King KY, Boles NC, Weksberg DC, Goodell MA. Quiescent haematopoietic stem cells are activated by IFN-gamma in response to chronic infection. Nature. 2010;465:793–797.
    1. Lo Celso C, Lin CP, Scadden DT. In vivo imaging of transplanted hematopoietic stem and progenitor cells in mouse calvarium bone marrow. Nat Protoc. 2011;6:1–14.
    1. Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452:442–447.
    1. Spiegel A, et al. Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34+ cells through Wnt signaling. Nat Immunol. 2007;8:1123–1131.
    1. Lucas D, et al. Chemotherapy-induced bone marrow nerve injury impairs hematopoietic regeneration. Nat Med. 2013;19:695–703.
    1. Zigmond RE, Ben-Ari Y. Electrical stimulation of preganglionic nerve increases tyrosine hydroxylase activity in sympathetic ganglia. Proc Natl Acad Sci U S A. 1977;74:3078–3080.
    1. Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014;505:327–334.
    1. Mendez-Ferrer S, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829–834.
    1. Ding L, Morrison SJ. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature. 2013;495:231–235.
    1. Eash KJ, Means JM, White DW, Link DC. CXCR4 is a key regulator of neutrophil release from the bone marrow under basal and stress granulopoiesis conditions. Blood. 2009;113:4711–4719.
    1. Tzeng YS, et al. Loss of Cxcl12/Sdf-1 in adult mice decreases the quiescent state of hematopoietic stem/progenitor cells and alters the pattern of hematopoietic regeneration after myelosuppression. Blood. 2011;117:429–439.
    1. Nie Y, Han YC, Zou YR. CXCR4 is required for the quiescence of primitive hematopoietic cells. J Exp Med. 2008;205:777–783.
    1. Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011;473:317–325.
    1. Randolph GJ. The fate of monocytes in atherosclerosis. J Thromb Haemost. 2009;7(Suppl 1):28–30.
    1. Rader DJ, Daugherty A. Translating molecular discoveries into new therapies for atherosclerosis. Nature. 2008;451:904–913.
    1. Swirski FK, Nahrendorf M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science. 2013;339:161–166.
    1. Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011;145:341–355.
    1. Libby P. Mechanisms of acute coronary syndromes and their implications for therapy. N Engl J Med. 2013;368:2004–2013.
    1. Adamsson Eryd S, Smith JG, Melander O, Hedblad B, Engstrom G. Incidence of coronary events and case fatality rate in relation to blood lymphocyte and neutrophil counts. Arterioscler Thromb Vasc Biol. 2012;32:533–539.
    1. Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874.
    1. Kaplan JR, et al. Social stress and atherosclerosis in normocholesterolemic monkeys. Science. 1983;220:733–735.
    1. Gu H, Tang C, Peng K, Sun H, Yang Y. Effects of chronic mild stress on the development of atherosclerosis and expression of toll-like receptor 4 signaling pathway in adolescent apolipoprotein E knockout mice. J Biomed Biotechnol. 2009;2009:613879.
    1. Bernberg E, Ulleryd MA, Johansson ME, Bergstrom GM. Social disruption stress increases IL-6 levels and accelerates atherosclerosis in ApoE-/- mice. Atherosclerosis. 2012;221:359–365.
    1. Wilbert-Lampen U, et al. Cardiovascular events during World Cup soccer. N Engl J Med. 2008;358:475–483.
    1. Wantha S, et al. Neutrophil-derived cathelicidin promotes adhesion of classical monocytes. Circ Res. 2013;112:792–801.
    1. Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17:1410–1422.
    1. Tall AR, Yvan-Charvet L, Westerterp M, Murphy AJ. Cholesterol efflux: a novel regulator of myelopoiesis and atherogenesis. Arterioscler Thromb Vasc Biol. 2012;32:2547–2552.
SUPPLEMENTARY REFERENCES
    1. Purton LE, Scadden DT. Limiting factors in murine hematopoietic stem cell assays. Cell Stem Cell. 2007;1:263–270.
    1. Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347:70–78.
    1. Dutta P, et al. Myocardial infarction accelerates atherosclerosis. Nature. 2012;487:325–329.
    1. Kobayashi M, Srour EF. Regulation of murine hematopoietic stem cell quiescence by Dmtf1. Blood. 2011;118:6562–6571.
    1. Shi C, et al. Bone marrow mesenchymal stem and progenitor cells induce monocyte emigration in response to circulating toll-like receptor ligands. Immunity. 2011;34:590–601.
    1. Westerterp M, et al. Regulation of hematopoietic stem and progenitor cell mobilization by cholesterol efflux pathways. Cell Stem Cell. 2012;11:195–206.
    1. Schepers K, et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell. 2013;13:285–299.
    1. Seimon TA, et al. Macrophage deficiency of p38alpha MAPK promotes apoptosis and plaque necrosis in advanced atherosclerotic lesions in mice. J Clin Invest. 2009;119:886–898.
    1. Zaheer A, et al. In vivo near-infrared fluorescence imaging of osteoblastic activity. Nat Biotechnol. 2001;19:1148–1154.
    1. Nahrendorf M, et al. Hybrid PET-optical imaging using targeted probes. Proc Natl Acad Sci U S A. 2010;107:7910–7915.

Source: PubMed

3
Subskrybuj