Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Blood ; 142(7): 658-674, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37267513

ABSTRACT

Myeloid cell heterogeneity is known, but whether it is cell-intrinsic or environmentally-directed remains unclear. Here, an inducible/reversible system pausing myeloid differentiation allowed the definition of clone-specific functions that clustered monocytes into subsets with distinctive molecular features. These subsets were orthogonal to the classical/nonclassical categorization and had inherent, restricted characteristics that did not shift under homeostasis, after irradiation, or with infectious stress. Rather, their functional fate was constrained by chromatin accessibility established at or before the granulocyte-monocyte or monocyte-dendritic progenitor level. Subsets of primary monocytes had differential ability to control distinct infectious agents in vivo. Therefore, monocytes are a heterogeneous population of functionally restricted subtypes defined by the epigenome of their progenitors that are differentially selected by physiologic challenges with limited plasticity to transition from one subset to another.


Subject(s)
Granulocytes , Monocytes , Myeloid Progenitor Cells , Epigenome , Epigenesis, Genetic , Cell Differentiation/genetics
2.
Nat Cardiovasc Res ; 1(1): 28-44, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35747128

ABSTRACT

Abnormal hematopoiesis advances cardiovascular disease by generating excess inflammatory leukocytes that attack the arteries and the heart. The bone marrow niche regulates hematopoietic stem cell proliferation and hence the systemic leukocyte pool, but whether cardiovascular disease affects the hematopoietic organ's microvasculature is unknown. Here we show that hypertension, atherosclerosis and myocardial infarction (MI) instigate endothelial dysfunction, leakage, vascular fibrosis and angiogenesis in the bone marrow, altogether leading to overproduction of inflammatory myeloid cells and systemic leukocytosis. Limiting angiogenesis with endothelial deletion of Vegfr2 (encoding vascular endothelial growth factor (VEGF) receptor 2) curbed emergency hematopoiesis after MI. We noted that bone marrow endothelial cells assumed inflammatory transcriptional phenotypes in all examined stages of cardiovascular disease. Endothelial deletion of Il6 or Vcan (encoding versican), genes shown to be highly expressed in mice with atherosclerosis or MI, reduced hematopoiesis and systemic myeloid cell numbers in these conditions. Our findings establish that cardiovascular disease remodels the vascular bone marrow niche, stimulating hematopoiesis and production of inflammatory leukocytes.

3.
Cell ; 184(5): 1348-1361.e22, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636128

ABSTRACT

Clonal hematopoiesis, a condition in which individual hematopoietic stem cell clones generate a disproportionate fraction of blood leukocytes, correlates with higher risk for cardiovascular disease. The mechanisms behind this association are incompletely understood. Here, we show that hematopoietic stem cell division rates are increased in mice and humans with atherosclerosis. Mathematical analysis demonstrates that increased stem cell proliferation expedites somatic evolution and expansion of clones with driver mutations. The experimentally determined division rate elevation in atherosclerosis patients is sufficient to produce a 3.5-fold increased risk of clonal hematopoiesis by age 70. We confirm the accuracy of our theoretical framework in mouse models of atherosclerosis and sleep fragmentation by showing that expansion of competitively transplanted Tet2-/- cells is accelerated under conditions of chronically elevated hematopoietic activity. Hence, increased hematopoietic stem cell proliferation is an important factor contributing to the association between cardiovascular disease and clonal hematopoiesis.


Subject(s)
Atherosclerosis/pathology , Clonal Hematopoiesis , Hematopoietic Stem Cells/pathology , Aging/pathology , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Bone Marrow/metabolism , Cell Proliferation , Clonal Evolution , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Sleep Deprivation/pathology
4.
Nat Med ; 25(11): 1761-1771, 2019 11.
Article in English | MEDLINE | ID: mdl-31700184

ABSTRACT

A sedentary lifestyle, chronic inflammation and leukocytosis increase atherosclerosis; however, it remains unclear whether regular physical activity influences leukocyte production. Here we show that voluntary running decreases hematopoietic activity in mice. Exercise protects mice and humans with atherosclerosis from chronic leukocytosis but does not compromise emergency hematopoiesis in mice. Mechanistically, exercise diminishes leptin production in adipose tissue, augmenting quiescence-promoting hematopoietic niche factors in leptin-receptor-positive stromal bone marrow cells. Induced deletion of the leptin receptor in Prrx1-creERT2; Leprfl/fl mice reveals that leptin's effect on bone marrow niche cells regulates hematopoietic stem and progenitor cell (HSPC) proliferation and leukocyte production, as well as cardiovascular inflammation and outcomes. Whereas running wheel withdrawal quickly reverses leptin levels, the impact of exercise on leukocyte production and on the HSPC epigenome and transcriptome persists for several weeks. Together, these data show that physical activity alters HSPCs via modulation of their niche, reducing hematopoietic output of inflammatory leukocytes.


Subject(s)
Atherosclerosis/therapy , Cardiovascular Diseases/therapy , Hematopoietic Stem Cells/metabolism , Inflammation/therapy , Physical Conditioning, Animal , Adipose Tissue/metabolism , Animals , Atherosclerosis/prevention & control , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/prevention & control , Epigenome/genetics , Exercise/physiology , Hematopoiesis/genetics , Hematopoiesis/physiology , Homeodomain Proteins/genetics , Humans , Inflammation/physiopathology , Leukocytes/metabolism , Leukocytosis/physiopathology , Leukocytosis/therapy , Mice , Receptors, Leptin/genetics , Sedentary Behavior , Transcriptome/genetics
6.
Nature ; 566(7744): 383-387, 2019 02.
Article in English | MEDLINE | ID: mdl-30760925

ABSTRACT

Sleep is integral to life1. Although insufficient or disrupted sleep increases the risk of multiple pathological conditions, including cardiovascular disease2, we know little about the cellular and molecular mechanisms by which sleep maintains cardiovascular health. Here we report that sleep regulates haematopoiesis and protects against atherosclerosis in mice. We show that mice subjected to sleep fragmentation produce more Ly-6Chigh monocytes, develop larger atherosclerotic lesions and produce less hypocretin-a stimulatory and wake-promoting neuropeptide-in the lateral hypothalamus. Hypocretin controls myelopoiesis by restricting the production of CSF1 by hypocretin-receptor-expressing pre-neutrophils in the bone marrow. Whereas hypocretin-null and haematopoietic hypocretin-receptor-null mice develop monocytosis and accelerated atherosclerosis, sleep-fragmented mice with either haematopoietic CSF1 deficiency or hypocretin supplementation have reduced numbers of circulating monocytes and smaller atherosclerotic lesions. Together, these results identify a neuro-immune axis that links sleep to haematopoiesis and atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Hematopoiesis/physiology , Sleep/physiology , Animals , Antigens, Ly/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Cells/metabolism , Female , Hematopoiesis/drug effects , Hypothalamic Area, Lateral/metabolism , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/deficiency , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Monocytes/drug effects , Monocytes/metabolism , Myelopoiesis/drug effects , Neutrophils/metabolism , Orexin Receptors/deficiency , Orexin Receptors/metabolism , Orexins/biosynthesis , Orexins/deficiency , Orexins/metabolism , Orexins/pharmacology , Sleep/drug effects , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Sleep Deprivation/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL