Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 31: 285-316, 2013.
Article in English | MEDLINE | ID: mdl-23298209

ABSTRACT

Mesenchymal stem cells (MSCs) are self-renewing precursor cells that can differentiate into bone, fat, cartilage, and stromal cells of the bone marrow. Recent studies suggest that MSCs themselves are critical for forming a niche that maintains hematopoietic stem cells (HSCs). The ease by which human MSC-like and stromal progenitor cells can be isolated from the bone marrow and other tissues has led to the rapid development of clinical investigations exploring their anti-inflammatory properties, tissue preservation capabilities, and regenerative potential. However, the identity of genuine MSCs and their specific contributions to these various beneficial effects have remained enigmatic. In this article, we examine the definition of MSCs and discuss the importance of rigorously characterizing their stem cell activity. We review their role and that of other putative niche constituents in the regulation of bone marrow HSCs. Additionally, how MSCs and their stromal progeny alter immune function is discussed, as well as potential therapeutic implications.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Regenerative Medicine/methods , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Stem Cells/immunology , Stromal Cells/immunology , Stromal Cells/pathology , Stromal Cells/transplantation
2.
Cell ; 181(2): 219-222, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302564

ABSTRACT

Mounting evidence indicates that the nervous system plays a central role in cancer pathogenesis. In turn, cancers and cancer therapies can alter nervous system form and function. This Commentary seeks to describe the burgeoning field of "cancer neuroscience" and encourage multidisciplinary collaboration for the study of cancer-nervous system interactions.


Subject(s)
Neoplasms/metabolism , Nervous System/metabolism , Humans , Neurosciences
3.
Cell ; 178(5): 1102-1114.e17, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442403

ABSTRACT

Caloric restriction is known to improve inflammatory and autoimmune diseases. However, the mechanisms by which reduced caloric intake modulates inflammation are poorly understood. Here we show that short-term fasting reduced monocyte metabolic and inflammatory activity and drastically reduced the number of circulating monocytes. Regulation of peripheral monocyte numbers was dependent on dietary glucose and protein levels. Specifically, we found that activation of the low-energy sensor 5'-AMP-activated protein kinase (AMPK) in hepatocytes and suppression of systemic CCL2 production by peroxisome proliferator-activator receptor alpha (PPARα) reduced monocyte mobilization from the bone marrow. Importantly, we show that fasting improves chronic inflammatory diseases without compromising monocyte emergency mobilization during acute infectious inflammation and tissue repair. These results reveal that caloric intake and liver energy sensors dictate the blood and tissue immune tone and link dietary habits to inflammatory disease outcome.


Subject(s)
Caloric Restriction , Monocytes/metabolism , AMP-Activated Protein Kinases/metabolism , Adult , Animals , Antigens, Ly/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , PPAR alpha/deficiency , PPAR alpha/genetics , PPAR alpha/metabolism
4.
Nat Rev Mol Cell Biol ; 20(5): 303-320, 2019 05.
Article in English | MEDLINE | ID: mdl-30745579

ABSTRACT

The haematopoietic stem cell (HSC) microenvironment in the bone marrow, termed the niche, ensures haematopoietic homeostasis by controlling the proliferation, self-renewal, differentiation and migration of HSCs and progenitor cells at steady state and in response to emergencies and injury. Improved methods for HSC isolation, driven by advances in single-cell and molecular technologies, have led to a better understanding of their behaviour, heterogeneity and lineage fate and of the niche cells and signals that regulate their function. Niche regulatory signals can be in the form of cell-bound or secreted factors and other local physical cues. A combination of technological advances in bone marrow imaging and genetic manipulation of crucial regulatory factors has enabled the identification of several candidate cell types regulating the niche, including both non-haematopoietic (for example, perivascular mesenchymal stem and endothelial cells) and HSC-derived (for example, megakaryocytes, macrophages and regulatory T cells), with better topographical understanding of HSC localization in the bone marrow. Here, we review advances in our understanding of HSC regulation by niches during homeostasis, ageing and cancer, and we discuss their implications for the development of therapies to rejuvenate aged HSCs or niches or to disrupt self-reinforcing malignant niches.


Subject(s)
Aging/metabolism , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Homeostasis , Neoplasms/metabolism , Stem Cell Niche , Aging/pathology , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cellular Senescence , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hematopoietic Stem Cells/pathology , Humans , Neoplasms/pathology
5.
Cell ; 165(2): 262-3, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058656

ABSTRACT

Alterations of the circadian clock have been linked to cancer development. Puram et al. (in this issue) now uncover differential requirements between healthy hematopoietic and diseased leukemic stem cells for core circadian transcription factors, wherein leukemic cells depend on the clock machinery for survival and growth.


Subject(s)
Circadian Rhythm , Transcription Factors , Bombs , Circadian Clocks , Humans , Leukemia
6.
Immunity ; 53(2): 417-428.e4, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32735844

ABSTRACT

Psychological stress has adverse effects on various human diseases, including those of the cardiovascular system. However, the mechanisms by which stress influences disease activity remain unclear. Here, using vaso-occlusive episodes (VOEs) of sickle cell disease as a vascular disease model, we show that stress promotes VOEs by eliciting a glucocorticoid hormonal response that augments gut permeability, leading to microbiota-dependent interleukin-17A (IL-17A) secretion from T helper 17 (Th17) cells of the lamina propria, followed by the expansion of the circulating pool of aged neutrophils that trigger VOEs. We identify segmented filamentous bacteria as the commensal essential for the stress-induced expansion of aged neutrophils that enhance VOEs in mice. Importantly, the inhibition of glucocorticoids synthesis, blockade of IL-17A, or depletion of the Th17 cell-inducing gut microbiota markedly reduces stress-induced VOEs. These results offer potential therapeutic targets to limit the impact of psychological stress on acute vascular occlusion.


Subject(s)
Anemia, Sickle Cell/pathology , Gastrointestinal Microbiome/immunology , Interleukin-17/metabolism , Stress, Psychological/pathology , Th17 Cells/immunology , Anemia, Sickle Cell/psychology , Animals , Bacteria/immunology , Cell Line , Germ-Free Life , Glucocorticoids/biosynthesis , Granulocyte Colony-Stimulating Factor/metabolism , HEK293 Cells , Humans , Hypothalamo-Hypophyseal System/metabolism , Inflammation/immunology , Inflammation/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology
7.
Cell ; 153(5): 1025-35, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706740

ABSTRACT

Unique among leukocytes, neutrophils follow daily cycles of release from and migration back into the bone marrow, where they are eliminated. Because removal of dying cells generates homeostatic signals, we explored whether neutrophil elimination triggers circadian events in the steady state. Here, we report that the homeostatic clearance of neutrophils provides cues that modulate the physiology of the bone marrow. We identify a population of CD62L(LO) CXCR4(HI) neutrophils that have "aged" in the circulation and are eliminated at the end of the resting period in mice. Aged neutrophils infiltrate the bone marrow and promote reductions in the size and function of the hematopoietic niche. Modulation of the niche depends on macrophages and activation of cholesterol-sensing nuclear receptors and is essential for the rhythmic egress of hematopoietic progenitors into the circulation. Our results unveil a process that synchronizes immune and hematopoietic rhythms and expand the ascribed functions of neutrophils beyond inflammation. PAPERFLICK:


Subject(s)
Bone Marrow/physiology , Circadian Rhythm , Neutrophils/cytology , Neutrophils/physiology , Animals , Cell Movement , Cellular Senescence , Female , Hematopoietic Stem Cells/metabolism , Homeostasis , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Orphan Nuclear Receptors/metabolism
8.
Nature ; 607(7919): 578-584, 2022 07.
Article in English | MEDLINE | ID: mdl-35636458

ABSTRACT

The nervous and immune systems are intricately linked1. Although psychological stress is known to modulate immune function, mechanistic pathways linking stress networks in the brain to peripheral leukocytes remain poorly understood2. Here we show that distinct brain regions shape leukocyte distribution and function throughout the body during acute stress in mice. Using optogenetics and chemogenetics, we demonstrate that motor circuits induce rapid neutrophil mobilization from the bone marrow to peripheral tissues through skeletal-muscle-derived neutrophil-attracting chemokines. Conversely, the paraventricular hypothalamus controls monocyte and lymphocyte egress from secondary lymphoid organs and blood to the bone marrow through direct, cell-intrinsic glucocorticoid signalling. These stress-induced, counter-directional, population-wide leukocyte shifts are associated with altered disease susceptibility. On the one hand, acute stress changes innate immunity by reprogramming neutrophils and directing their recruitment to sites of injury. On the other hand, corticotropin-releasing hormone neuron-mediated leukocyte shifts protect against the acquisition of autoimmunity, but impair immunity to SARS-CoV-2 and influenza infection. Collectively, these data show that distinct brain regions differentially and rapidly tailor the leukocyte landscape during psychological stress, therefore calibrating the ability of the immune system to respond to physical threats.


Subject(s)
Brain , Fear , Leukocytes , Motor Neurons , Neural Pathways , Stress, Psychological , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Brain/cytology , Brain/physiology , COVID-19/immunology , Chemokines/immunology , Disease Susceptibility , Fear/physiology , Glucocorticoids/metabolism , Humans , Leukocytes/cytology , Leukocytes/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Monocytes/cytology , Monocytes/immunology , Motor Neurons/cytology , Motor Neurons/physiology , Neutrophils/cytology , Neutrophils/immunology , Optogenetics , Orthomyxoviridae Infections/immunology , Paraventricular Hypothalamic Nucleus/physiology , SARS-CoV-2/immunology , Stress, Psychological/immunology , Stress, Psychological/physiopathology
9.
Immunity ; 48(4): 632-648, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669248

ABSTRACT

Steady-state hematopoietic stem cells' (HSCs) self-renewal and differentiation toward their mature progeny in the adult bone marrow is tightly regulated by cues from the microenvironment. Recent insights into the cellular and molecular constituents have uncovered a high level of complexity. Here, we review emerging evidence showing how HSCs and their progeny are regulated by an interdependent network of mesenchymal stromal cells, nerve fibers, the vasculature, and also other hematopoietic cells. Understanding the interaction mechanisms in these intricate niches will provide great opportunities for HSC-related therapies and immune modulation.


Subject(s)
Bone Marrow Cells/physiology , Cell Self Renewal/physiology , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Stem Cell Niche/physiology , Animals , Cell Differentiation , Hematopoietic Stem Cells/physiology , Humans , Mesenchymal Stem Cells/physiology , Mice , Nerve Fibers/physiology
10.
Immunity ; 49(4): 627-639.e6, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30314756

ABSTRACT

The non-hematopoietic cell fraction of the bone marrow (BM) is classically identified as CD45- Ter119- CD31- (herein referred to as triple-negative cells or TNCs). Although TNCs are believed to contain heterogeneous stromal cell populations, they remain poorly defined. Here we showed that the vast majority of TNCs (∼85%) have a hematopoietic rather than mesenchymal origin. Single cell RNA-sequencing revealed erythroid and lymphoid progenitor signatures among CD51- TNCs. Ly6D+ CD44+ CD51- TNCs phenotypically and functionally resembled CD45+ pro-B lymphoid cells, whereas Ly6D- CD44+ CD51- TNCs were enriched in previously unappreciated stromal-dependent erythroid progenitors hierarchically situated between preCFU-E and proerythroblasts. Upon adoptive transfer, CD44+ CD51- TNCs contributed to repopulate the B-lymphoid and erythroid compartments. CD44+ CD51- TNCs also expanded during phenylhydrazine-induced acute hemolysis or in a model of sickle cell anemia. These findings thus uncover physiologically relevant new classes of stromal-associated functional CD45- hematopoietic progenitors.


Subject(s)
Bone Marrow Cells/immunology , Erythroid Cells/immunology , Lymphoid Progenitor Cells/immunology , Stromal Cells/immunology , Adoptive Transfer/methods , Animals , Blood Group Antigens/immunology , Blood Group Antigens/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/immunology , Cells, Cultured , Erythroid Cells/cytology , Erythroid Cells/metabolism , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
11.
Nature ; 589(7843): 591-596, 2021 01.
Article in English | MEDLINE | ID: mdl-33361809

ABSTRACT

Haematopoietic stem cells (HSCs) reside in specialized microenvironments in the bone marrow-often referred to as 'niches'-that represent complex regulatory milieux influenced by multiple cellular constituents, including nerves1,2. Although sympathetic nerves are known to regulate the HSC niche3-6, the contribution of nociceptive neurons in the bone marrow remains unclear. Here we show that nociceptive nerves are required for enforced HSC mobilization and that they collaborate with sympathetic nerves to maintain HSCs in the bone marrow. Nociceptor neurons drive granulocyte colony-stimulating factor (G-CSF)-induced HSC mobilization via the secretion of calcitonin gene-related peptide (CGRP). Unlike sympathetic nerves, which regulate HSCs indirectly via the niche3,4,6, CGRP acts directly on HSCs via receptor activity modifying protein 1 (RAMP1) and the calcitonin receptor-like receptor (CALCRL) to promote egress by activating the Gαs/adenylyl cyclase/cAMP pathway. The ingestion of food containing capsaicin-a natural component of chili peppers that can trigger the activation of nociceptive neurons-significantly enhanced HSC mobilization in mice. Targeting the nociceptive nervous system could therefore represent a strategy to improve the yield of HSCs for stem cell-based therapeutic agents.


Subject(s)
Autonomic Pathways , Cell Movement , Hematopoietic Stem Cells/cytology , Nociception/physiology , Nociceptors/physiology , Sympathetic Nervous System/cytology , Adenylyl Cyclases/metabolism , Animals , Autonomic Pathways/drug effects , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Receptor-Like Protein/metabolism , Capsaicin/pharmacology , Cell Movement/drug effects , Cyclic AMP/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Nociception/drug effects , Nociceptors/drug effects , Receptor Activity-Modifying Protein 1/metabolism , Signal Transduction/drug effects , Stem Cell Niche , Sympathetic Nervous System/drug effects
12.
Immunity ; 46(6): 979-981, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636968

ABSTRACT

Regulatory T (Treg) cells are well known to modulate inflammatory responses. In a recent issue of Cell, Ali et al. (2017) reveal a function for Treg cells in stem cell maintenance by showing that skin-resident Foxp3+ Treg cells preferentially localize to the hair follicle stem cell (HFSC) niche to control HFSC-mediated hair regeneration.


Subject(s)
Adult Stem Cells/immunology , Hair Follicle/immunology , Immune Tolerance , Stem Cell Niche/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Forkhead Transcription Factors/metabolism , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Mice , Mice, Knockout , Receptors, Notch/metabolism , Regeneration , Signal Transduction
13.
Blood ; 141(2): 194-199, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36315910

ABSTRACT

Sickle cell disease (SCD) is an inherited disorder resulting from a ß-globin gene mutation, and SCD patients experience erythrocyte sickling, vaso-occlusive episodes (VOE), and progressive organ damage. Chronic hemolysis, inflammation, and repeated red blood cell transfusions in SCD can disrupt iron homeostasis. Patients who receive multiple blood transfusions develop iron overload, and another subpopulation of SCD patients manifest iron deficiency. To elucidate connections between dietary iron, the microbiome, and SCD pathogenesis, we treated SCD mice with an iron-restricted diet (IRD). IRD treatment reduced iron availability and hemolysis, decreased acute VOE, and ameliorated chronic organ damage in SCD mice. Our results extend previous studies indicating that the gut microbiota regulate disease in SCD mice. IRD alters microbiota load and improves gut integrity, together preventing crosstalk between the gut microbiome and inflammatory factors such as aged neutrophils, dampening VOE, and organ damage. These findings provide strong evidence for the therapeutic potential of manipulating iron homeostasis and the gut microbiome to ameliorate SCD pathophysiology. Many treatments, which are under development, focus on lowering the systemic iron concentration to relieve disease complications, and our data suggest that iron-induced changes in microbiota load and gut integrity are related- and novel-therapeutic targets.


Subject(s)
Anemia, Sickle Cell , Vascular Diseases , Mice , Animals , Iron, Dietary , Iron , Hemolysis , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/therapy , Vascular Diseases/etiology , Vascular Diseases/prevention & control
14.
Immunity ; 45(3): 464-466, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27653597

ABSTRACT

In homeostasis, whether blood cells are derived from committed progenitor or mutipotent stem cell activity remains controversial. In this issue of Immunity, Sawai et al. (2016) describe murine HSCs as the major contributor to the maintenance of multilineage hematopoiesis, both in the steady state and during cytokine response.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/physiology , Cytokines/metabolism , Hematopoietic Stem Cells/metabolism , Homeostasis/physiology , Humans
16.
Nature ; 569(7755): 222-228, 2019 05.
Article in English | MEDLINE | ID: mdl-30971824

ABSTRACT

The bone marrow microenvironment has a key role in regulating haematopoiesis, but its molecular complexity and response to stress are incompletely understood. Here we map the transcriptional landscape of mouse bone marrow vascular, perivascular and osteoblast cell populations at single-cell resolution, both at homeostasis and under conditions of stress-induced haematopoiesis. This analysis revealed previously unappreciated levels of cellular heterogeneity within the bone marrow niche and resolved cellular sources of pro-haematopoietic growth factors, chemokines and membrane-bound ligands. Our studies demonstrate a considerable transcriptional remodelling of niche elements under stress conditions, including an adipocytic skewing of perivascular cells. Among the stress-induced changes, we observed that vascular Notch delta-like ligands (encoded by Dll1 and Dll4) were downregulated. In the absence of vascular Dll4, haematopoietic stem cells prematurely induced a myeloid transcriptional program. These findings refine our understanding of the cellular architecture of the bone marrow niche, reveal a dynamic and heterogeneous molecular landscape that is highly sensitive to stress and illustrate the utility of single-cell transcriptomic data in evaluating the regulation of haematopoiesis by discrete niche populations.


Subject(s)
Bone Marrow/blood supply , Cellular Microenvironment , Hematopoiesis , Hematopoietic Stem Cells , Single-Cell Analysis , Stem Cell Niche , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Differentiation , Cell Lineage , Endothelium, Vascular/cytology , Female , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Male , Mice , Myeloid Cells/cytology , Myeloid Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , RNA-Seq , Receptors, Notch/metabolism , Stem Cell Niche/genetics , Stress, Physiological/genetics , Transcriptome/genetics
17.
Nat Immunol ; 13(9): 888-99, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22797772

ABSTRACT

Although much progress has been made in the understanding of the ontogeny and function of dendritic cells (DCs), the transcriptional regulation of the lineage commitment and functional specialization of DCs in vivo remains poorly understood. We made a comprehensive comparative analysis of CD8(+), CD103(+), CD11b(+) and plasmacytoid DC subsets, as well as macrophage DC precursors and common DC precursors, across the entire immune system. Here we characterized candidate transcriptional activators involved in the commitment of myeloid progenitor cells to the DC lineage and predicted regulators of DC functional diversity in tissues. We identified a molecular signature that distinguished tissue DCs from macrophages. We also identified a transcriptional program expressed specifically during the steady-state migration of tissue DCs to the draining lymph nodes that may control tolerance to self tissue antigens.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Dendritic Cells/immunology , Transcription, Genetic , Cell Differentiation/genetics , Dendritic Cells/cytology , Gene Expression Profiling , Humans
18.
Angiogenesis ; 26(1): 129-166, 2023 02.
Article in English | MEDLINE | ID: mdl-36183032

ABSTRACT

Cancer cells are embedded within the tissue and interact dynamically with its components during cancer progression. Understanding the contribution of cellular components within the tumor microenvironment is crucial for the success of therapeutic applications. Here, we reveal the presence of perivascular GFAP+/Plp1+ cells within the tumor microenvironment. Using in vivo inducible Cre/loxP mediated systems, we demonstrated that these cells derive from tissue-resident Schwann cells. Genetic ablation of endogenous Schwann cells slowed down tumor growth and angiogenesis. Schwann cell-specific depletion also induced a boost in the immune surveillance by increasing tumor-infiltrating anti-tumor lymphocytes, while reducing immune-suppressor cells. In humans, a retrospective in silico analysis of tumor biopsies revealed that increased expression of Schwann cell-related genes within melanoma was associated with improved survival. Collectively, our study suggests that Schwann cells regulate tumor progression, indicating that manipulation of Schwann cells may provide a valuable tool to improve cancer patients' outcomes.


Subject(s)
Neoplasms , Neuroglia , Humans , Retrospective Studies , Neuroglia/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , Pericytes , Tumor Microenvironment/physiology , Neoplasms/pathology
19.
Immunity ; 38(4): 792-804, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601688

ABSTRACT

Despite accumulating evidence suggesting local self-maintenance of tissue macrophages in the steady state, the dogma remains that tissue macrophages derive from monocytes. Using parabiosis and fate-mapping approaches, we confirmed that monocytes do not show significant contribution to tissue macrophages in the steady state. Similarly, we found that after depletion of lung macrophages, the majority of repopulation occurred by stochastic cellular proliferation in situ in a macrophage colony-stimulating factor (M-Csf)- and granulocyte macrophage (GM)-CSF-dependent manner but independently of interleukin-4. We also found that after bone marrow transplantation, host macrophages retained the capacity to expand when the development of donor macrophages was compromised. Expansion of host macrophages was functional and prevented the development of alveolar proteinosis in mice transplanted with GM-Csf-receptor-deficient progenitors. Collectively, these results indicate that tissue-resident macrophages and circulating monocytes should be classified as mononuclear phagocyte lineages that are independently maintained in the steady state.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lung/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Adult , Animals , Bone Marrow Transplantation , Cell Proliferation , Cell Survival , Cells, Cultured , Homeostasis , Humans , Interleukin-4/metabolism , Macrophages/transplantation , Mice , Mice, Knockout , Mice, Mutant Strains , Parabiosis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
20.
Development ; 145(2)2018 01 22.
Article in English | MEDLINE | ID: mdl-29358215

ABSTRACT

Hematopoietic stem cells (HSCs) develop in discrete anatomical niches, migrating during embryogenesis from the aorta-gonad-mesonephros (AGM) region to the fetal liver, and finally to the bone marrow, where most HSCs reside throughout adult life. These niches provide supportive microenvironments that specify, expand and maintain HSCs. Understanding the constituents and molecular regulation of HSC niches is of considerable importance as it could shed new light on the mechanistic principles of HSC emergence and maintenance, and provide novel strategies for regenerative medicine. However, controversy exists concerning the cellular complexity of the bone marrow niche, and our understanding of the different HSC niches during development remains limited. In this Review, we summarize and discuss what is known about the heterogeneity of the HSC niches at distinct stages of their ontogeny, from the embryo to the adult bone marrow, drawing predominantly on data from mouse studies.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Stem Cell Niche/physiology , Aging/pathology , Aging/physiology , Animals , Aorta/embryology , Cell Lineage , Female , Gonads/embryology , Hematologic Neoplasms/pathology , Hematopoietic System/embryology , Humans , Male , Mesonephros/embryology , Mice , Placenta/cytology , Placenta/physiology , Pregnancy , Stromal Cells/cytology , Stromal Cells/physiology , Sympathetic Nervous System/embryology , Sympathetic Nervous System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL