Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Cell ; 187(12): 3090-3107.e21, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38749423

ABSTRACT

Platelet dysregulation is drastically increased with advanced age and contributes to making cardiovascular disorders the leading cause of death of elderly humans. Here, we reveal a direct differentiation pathway from hematopoietic stem cells into platelets that is progressively propagated upon aging. Remarkably, the aging-enriched platelet path is decoupled from all other hematopoietic lineages, including erythropoiesis, and operates as an additional layer in parallel with canonical platelet production. This results in two molecularly and functionally distinct populations of megakaryocyte progenitors. The age-induced megakaryocyte progenitors have a profoundly enhanced capacity to engraft, expand, restore, and reconstitute platelets in situ and upon transplantation and produce an additional platelet population in old mice. The two pools of co-existing platelets cause age-related thrombocytosis and dramatically increased thrombosis in vivo. Strikingly, aging-enriched platelets are functionally hyper-reactive compared with the canonical platelet populations. These findings reveal stem cell-based aging as a mechanism for platelet dysregulation and age-induced thrombosis.


Subject(s)
Aging , Blood Platelets , Cell Differentiation , Hematopoietic Stem Cells , Thrombosis , Animals , Hematopoietic Stem Cells/metabolism , Blood Platelets/metabolism , Thrombosis/pathology , Thrombosis/metabolism , Mice , Humans , Megakaryocytes/metabolism , Mice, Inbred C57BL , Megakaryocyte Progenitor Cells/metabolism , Male
2.
Development ; 149(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-35072209

ABSTRACT

Tissue-resident lymphoid cells (TLCs) span the spectrum of innate-to-adaptive immune function. Unlike traditional, circulating lymphocytes that are continuously generated from hematopoietic stem cells (HSCs), many TLCs are of fetal origin and poorly generated from adult HSCs. Here, we sought to further understand murine TLC development and the roles of Flk2 and IL7Rα, two cytokine receptors with known function in traditional lymphopoiesis. Using Flk2- and Il7r-Cre lineage tracing, we found that peritoneal B1a cells, splenic marginal zone B (MZB) cells, lung ILC2s and regulatory T cells (Tregs) were highly labeled. Despite high labeling, loss of Flk2 minimally affected the generation of these cells. In contrast, loss of IL7Rα, or combined deletion of Flk2 and IL7Rα, dramatically reduced the number of B1a cells, MZBs, ILC2s and Tregs, both in situ and upon transplantation, indicating an intrinsic and essential role for IL7Rα. Surprisingly, reciprocal transplants of wild-type HSCs showed that an IL7Rα-/- environment selectively impaired reconstitution of TLCs when compared with TLC numbers in situ. Taken together, our data defined Flk2- and IL7Rα-positive TLC differentiation paths, and revealed functional roles of Flk2 and IL7Rα in TLC establishment.


Subject(s)
Hematopoietic Stem Cells/immunology , Lymphopoiesis/genetics , Receptors, Interleukin-7/genetics , fms-Like Tyrosine Kinase 3/genetics , Adaptive Immunity/genetics , Animals , B-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation, Developmental/genetics , Hematopoietic Stem Cells/cytology , Immunity, Innate/genetics , Lymphocytes/cytology , Lymphocytes/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphopoiesis/immunology , Mice , Organ Specificity/genetics , T-Lymphocytes, Regulatory/immunology
3.
Stem Cells ; 41(5): 520-539, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36945732

ABSTRACT

Epigenetic mechanisms regulate the multilineage differentiation capacity of hematopoietic stem cells (HSCs) into a variety of blood and immune cells. Mapping the chromatin dynamics of functionally defined cell populations will shed mechanistic insight into 2 major, unanswered questions in stem cell biology: how does epigenetic identity contribute to a cell type's lineage potential, and how do cascades of chromatin remodeling dictate ensuing fate decisions? Our recent work revealed evidence of multilineage gene priming in HSCs, where open cis-regulatory elements (CREs) exclusively shared between HSCs and unipotent lineage cells were enriched for DNA binding motifs of known lineage-specific transcription factors. Oligopotent progenitor populations operating between the HSCs and unipotent cells play essential roles in effecting hematopoietic homeostasis. To test the hypothesis that selective HSC-primed lineage-specific CREs remain accessible throughout differentiation, we used ATAC-seq to map the temporal dynamics of chromatin remodeling during progenitor differentiation. We observed epigenetic-driven clustering of oligopotent and unipotent progenitors into distinct erythromyeloid and lymphoid branches, with multipotent HSCs and MPPs associating with the erythromyeloid lineage. We mapped the dynamics of lineage-primed CREs throughout hematopoiesis and identified both unique and shared CREs as potential lineage reinforcement mechanisms at fate branch points. Additionally, quantification of genome-wide peak count and size revealed overall greater chromatin accessibility in HSCs, allowing us to identify HSC-unique peaks as putative regulators of self-renewal and multilineage potential. Finally, CRISPRi-mediated targeting of ATACseq-identified putative CREs in HSCs allowed us to demonstrate the functional role of selective CREs in lineage-specific gene expression. These findings provide insight into the regulation of stem cell multipotency and lineage commitment throughout hematopoiesis and serve as a resource to test functional drivers of hematopoietic lineage fate.


Subject(s)
Chromatin , Hematopoiesis , Chromatin/genetics , Chromatin/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics
4.
Immunity ; 42(4): 665-78, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902481

ABSTRACT

Although classified as hematopoietic cells, tissue-resident macrophages (MFs) arise from embryonic precursors that seed the tissues prior to birth to generate a self-renewing population, which is maintained independently of adult hematopoiesis. Here we reveal the identity of these embryonic precursors using an in utero MF-depletion strategy and fate-mapping of yolk sac (YS) and fetal liver (FL) hematopoiesis. We show that YS MFs are the main precursors of microglia, while most other MFs derive from fetal monocytes (MOs). Both YS MFs and fetal MOs arise from erythro-myeloid progenitors (EMPs) generated in the YS. In the YS, EMPs gave rise to MFs without monocytic intermediates, while EMP seeding the FL upon the establishment of blood circulation acquired c-Myb expression and gave rise to fetal MOs that then seeded embryonic tissues and differentiated into MFs. Thus, adult tissue-resident MFs established from hematopoietic stem cell-independent embryonic precursors arise from two distinct developmental programs.


Subject(s)
Aging/immunology , Macrophages/immunology , Monocytes/immunology , Myeloid Progenitor Cells/immunology , Proto-Oncogene Proteins c-myb/immunology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage/immunology , Cell Tracking , Embryo, Mammalian , Female , Fetus , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Kidney/cytology , Kidney/immunology , Liver/cytology , Liver/immunology , Lung/cytology , Lung/immunology , Macrophages/cytology , Mice , Microglia/cytology , Microglia/immunology , Monocytes/cytology , Myeloid Progenitor Cells/cytology , Pregnancy , Primary Cell Culture , Proto-Oncogene Proteins c-myb/metabolism , Skin/cytology , Skin/immunology , Yolk Sac/cytology , Yolk Sac/immunology
5.
J Immunol ; 208(8): 1886-1900, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35365562

ABSTRACT

Our respiratory system is vital to protect us from the surrounding nonsterile environment; therefore, it is critical for a state of homeostasis to be maintained through a balance of inflammatory cues. Recent studies have shown that actively transcribed noncoding regions of the genome are emerging as key regulators of biological processes, including inflammation. lincRNA-Cox2 is one such example of an inflammatory inducible long intergenic noncoding RNA functioning to fine-tune immune gene expression. Using bulk and single-cell RNA sequencing, in addition to FACS, we find that lincRNA-Cox2 is most highly expressed in the lung and is most upregulated after LPS-induced lung injury (acute lung injury [ALI]) within alveolar macrophages, where it functions to regulate inflammation. We previously reported that lincRNA-Cox2 functions to regulate its neighboring protein Ptgs2 in cis, and in this study, we use genetic mouse models to confirm its role in regulating gene expression more broadly in trans during ALI. Il6, Ccl3, and Ccl5 are dysregulated in the lincRNA-Cox2-deficient mice and can be rescued to wild type levels by crossing the deficient mice with our newly generated lincRNA-Cox2 transgenic mice, confirming that this gene functions in trans. Many genes are specifically regulated by lincRNA-Cox2 within alveolar macrophages originating from the bone marrow because the phenotype can be reversed by transplantation of wild type bone marrow into the lincRNA-Cox2-deficient mice. In conclusion, we show that lincRNA-Cox2 is a trans-acting long noncoding RNA that functions to regulate immune responses and maintain homeostasis within the lung at baseline and on LPS-induced ALI.


Subject(s)
Acute Lung Injury , Cyclooxygenase 2 , Inflammation , Macrophages, Alveolar , RNA, Long Noncoding , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/metabolism , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
6.
Immunity ; 40(1): 91-104, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439267

ABSTRACT

Cardiac macrophages are crucial for tissue repair after cardiac injury but are not well characterized. Here we identify four populations of cardiac macrophages. At steady state, resident macrophages were primarily maintained through local proliferation. However, after macrophage depletion or during cardiac inflammation, Ly6c(hi) monocytes contributed to all four macrophage populations, whereas resident macrophages also expanded numerically through proliferation. Genetic fate mapping revealed that yolk-sac and fetal monocyte progenitors gave rise to the majority of cardiac macrophages, and the heart was among a minority of organs in which substantial numbers of yolk-sac macrophages persisted in adulthood. CCR2 expression and dependence distinguished cardiac macrophages of adult monocyte versus embryonic origin. Transcriptional and functional data revealed that monocyte-derived macrophages coordinate cardiac inflammation, while playing redundant but lesser roles in antigen sampling and efferocytosis. These data highlight the presence of multiple cardiac macrophage subsets, with different functions, origins, and strategies to regulate compartment size.


Subject(s)
Macrophages/immunology , Monocytes/physiology , Myocarditis/immunology , Myocardium/immunology , Animals , Antigen Presentation , Antigens, Ly/metabolism , Cell Death , Cell Differentiation , Cell Lineage , Cells, Cultured , Fetal Development , Heart/embryology , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/immunology , Phagocytosis , Receptors, CCR2/metabolism , Transcriptome , Yolk Sac/cytology
7.
Int J Mol Sci ; 24(23)2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38069208

ABSTRACT

Polycyclic aromatic hydrocarbon (PAH) pollutants and microbiome products converge on the aryl hydrocarbon receptor (AhR) to redirect selective rapid adherence of isolated bone marrow (BM) cells. In young adult mice, Cyp1b1-deficiency and AhR activation by PAH, particularly when prolonged by Cyp1a1 deletion, produce matching gene stimulations in these BM cells. Vascular expression of Cyp1b1 lowers reactive oxygen species (ROS), suppressing NF-κB/RelA signaling. PAH and allelic selectivity support a non-canonical AhR participation, possibly through RelA. Genes stimulated by Cyp1b1 deficiency were further resolved according to the effects of Cyp1b1 and Cyp1a1 dual deletions (DKO). The adherent BM cells show a cluster of novel stimulations, including select developmental markers; multiple re-purposed olfactory receptors (OLFR); and α-Defensin, a microbial disruptor. Each one connects to an enhanced specific expression of the catalytic RNA Pol2 A subunit, among 12 different subunits. Mesenchymal progenitor BMS2 cells retain these features. Cyp1b1-deficiency removes lymphocytes from adherent assemblies as BM-derived mesenchymal stromal cells (BM-MSC) expand. Cyp1b1 effects were cell-type specific. In vivo, BM-MSC Cyp1b1 expression mediated PAH suppression of lymphocyte progenitors. In vitro, OP9-MSC sustained these progenitors, while Csf1 induced monocyte progenitor expansion to macrophages. Targeted Cyp1b1 deletion (Cdh5-Cre; Cyp1b1fl/fl) established endothelium control of ROS that directs AhR-mediated suppression of B cell progenitors. Monocyte Cyp1b1 deletion (Lyz2-Cre; Cyp1b1fl/fl) selectively attenuated M1 polarization of expanded macrophages, but did not enhance effects on basal M2 polarization. Thus, specific sources of Cyp1b1 link to AhR and to an OLFR network to provide BM inflammatory modulation via diverse microbiome products.


Subject(s)
Mesenchymal Stem Cells , Polycyclic Aromatic Hydrocarbons , Receptors, Odorant , Animals , Mice , Bone Marrow/metabolism , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , Mesenchymal Stem Cells/metabolism , Oxygen , Polycyclic Aromatic Hydrocarbons/metabolism , Reactive Oxygen Species , Receptors, Aryl Hydrocarbon/metabolism
8.
Development ; 146(14)2019 07 22.
Article in English | MEDLINE | ID: mdl-31332039

ABSTRACT

The discovery of a fetal origin for tissue-resident macrophages (trMacs) has inspired an intense search for the mechanisms underlying their development. Here, we performed in vivo lineage tracing of cells with an expression history of IL7Rα, a marker exclusively associated with the lymphoid lineage in adult hematopoiesis. Surprisingly, we found that Il7r-Cre labeled fetal-derived, adult trMacs. Labeling was almost complete in some tissues and partial in others. The putative progenitors of trMacs, yolk sac (YS) erythromyeloid progenitors, did not express IL7R, and YS hematopoiesis was unperturbed in IL7R-deficient mice. In contrast, tracking of IL7Rα message levels, surface expression, and Il7r-Cre-mediated labeling across fetal development revealed dynamic regulation of Il7r mRNA expression and rapid upregulation of IL7Rα surface protein upon transition from monocyte to macrophage within fetal tissues. Fetal monocyte differentiation in vitro produced IL7R+ macrophages, supporting a direct progenitor-progeny relationship. Additionally, blockade of IL7R function during late gestation specifically impaired the establishment of fetal-derived trMacs in vivo These data provide evidence for a distinct function of IL7Rα in fetal myelopoiesis and identify IL7R as a novel regulator of trMac development.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Macrophages/physiology , Myelopoiesis/genetics , Receptors, Interleukin-7/physiology , Animals , Embryo, Mammalian , Female , Fetus/metabolism , Hematopoiesis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy
9.
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
10.
Am J Hematol ; 97(9): 1226-1235, 2022 09.
Article in English | MEDLINE | ID: mdl-35560111

ABSTRACT

Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas genome engineering has emerged as a powerful tool to modify precise genomic sequences with unparalleled accuracy and efficiency. Major advances in CRISPR technologies over the last 5 years have fueled the development of novel techniques in hematopoiesis research to interrogate the complexities of hematopoietic stem cell (HSC) biology. In particular, high throughput CRISPR based screens using various "flavors" of Cas coupled with sequencing and/or functional outputs are becoming increasingly efficient and accessible. In this review, we discuss recent achievements in CRISPR-mediated genomic engineering and how these new tools have advanced the understanding of HSC heterogeneity and function throughout life. Additionally, we highlight how these techniques can be used to answer previously inaccessible questions and the challenges to implement them. Finally, we focus on their translational potential to both model and treat hematological diseases in the clinic.


Subject(s)
CRISPR-Cas Systems , Hematologic Diseases , Bioengineering , Genomics/methods , Hematologic Diseases/genetics , Hematologic Diseases/therapy , Hematopoietic Stem Cells , Humans
11.
Stem Cells ; 38(9): 1159-1174, 2020 09.
Article in English | MEDLINE | ID: mdl-32442338

ABSTRACT

C-X-C motif chemokine ligand 12 (CXCL12; aka SDF1α) is a major regulator of a number of cellular systems, including hematopoiesis, where it influences hematopoietic cell trafficking, proliferation, and survival during homeostasis and upon stress and disease. A variety of constitutive, temporal, ubiquitous, and cell-specific loss-of-function models have documented the functional consequences on hematopoiesis upon deletion of Cxcl12. Here, in contrast to loss-of-function experiments, we implemented a gain-of-function approach by generating a doxycycline-inducible transgenic mouse model that enables spatial and temporal overexpression of Cxcl12. We demonstrated that ubiquitous CXCL12 overexpression led to an increase in multipotent progenitors in the bone marrow and spleen. The CXCL12+ mice displayed reduced reconstitution potential as either donors or recipients in transplantation experiments. Additionally, we discovered that Cxcl12 overexpression improved hematopoietic stem and progenitor cell mobilization into the blood, and conferred radioprotection by promoting quiescence. Thus, this new CXCL12+ mouse model provided new insights into major facets of hematopoiesis and serves as a versatile resource for studying CXCL12 function in a variety of contexts.


Subject(s)
Chemokine CXCL12/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Radiation Protection , Animals , Benzylamines/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Count , Cell Cycle/drug effects , Cyclams/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Neovascularization, Physiologic/drug effects
12.
Nature ; 512(7513): 198-202, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25079315

ABSTRACT

Haematopoietic stem cells (HSCs) self-renew for life, thereby making them one of the few blood cells that truly age. Paradoxically, although HSCs numerically expand with age, their functional activity declines over time, resulting in degraded blood production and impaired engraftment following transplantation. While many drivers of HSC ageing have been proposed, the reason why HSC function degrades with age remains unknown. Here we show that cycling old HSCs in mice have heightened levels of replication stress associated with cell cycle defects and chromosome gaps or breaks, which are due to decreased expression of mini-chromosome maintenance (MCM) helicase components and altered dynamics of DNA replication forks. Nonetheless, old HSCs survive replication unless confronted with a strong replication challenge, such as transplantation. Moreover, once old HSCs re-establish quiescence, residual replication stress on ribosomal DNA (rDNA) genes leads to the formation of nucleolar-associated γH2AX signals, which persist owing to ineffective H2AX dephosphorylation by mislocalized PP4c phosphatase rather than ongoing DNA damage. Persistent nucleolar γH2AX also acts as a histone modification marking the transcriptional silencing of rDNA genes and decreased ribosome biogenesis in quiescent old HSCs. Our results identify replication stress as a potent driver of functional decline in old HSCs, and highlight the MCM DNA helicase as a potential molecular target for rejuvenation therapies.


Subject(s)
Cellular Senescence/physiology , DNA Replication/physiology , Hematopoietic Stem Cells/pathology , Stress, Physiological , Animals , Cell Proliferation , Cellular Senescence/genetics , DNA Damage/genetics , DNA, Ribosomal/genetics , Female , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Histones/genetics , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Minichromosome Maintenance Proteins/genetics
13.
Nucleic Acids Res ; 46(10): e62, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29548006

ABSTRACT

RNA-sequencing (RNA-seq) is a powerful technique to investigate and quantify entire transcriptomes. Recent advances in the field have made it possible to explore the transcriptomes of single cells. However, most widely used RNA-seq protocols fail to provide crucial information regarding transcription start sites. Here we present a protocol, Tn5Prime, that takes advantage of the Tn5 transposase-based Smart-seq2 protocol to create RNA-seq libraries that capture the 5' end of transcripts. The Tn5Prime method dramatically streamlines the 5' capture process and is both cost effective and reliable. By applying Tn5Prime to bulk RNA and single cell samples, we were able to define transcription start sites as well as quantify transcriptomes at high accuracy and reproducibility. Additionally, similar to 3' end-based high-throughput methods like Drop-seq and 10× Genomics Chromium, the 5' capture Tn5Prime method allows the introduction of cellular identifiers during reverse transcription, simplifying the analysis of large numbers of single cells. In contrast to 3' end-based methods, Tn5Prime also enables the assembly of the variable 5' ends of the antibody sequences present in single B-cell data. Therefore, Tn5Prime presents a robust tool for both basic and applied research into the adaptive immune system and beyond.


Subject(s)
B-Lymphocytes/cytology , Sequence Analysis, RNA/methods , Transcription Initiation Site , Transposases/genetics , ADP-ribosyl Cyclase 1/genetics , Adult , Animals , B-Lymphocytes/physiology , Cell Line , Gene Expression Profiling , Gene Library , Humans , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Single-Cell Analysis/methods , Transposases/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
14.
Development ; 142(10): 1728-32, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25968308

ABSTRACT

In February 2015, over 200 scientists gathered for the Keystone Hematopoiesis meeting, which was held at the scenic Keystone Resort in Keystone, Colorado, USA. The meeting organizers, Patricia Ernst, Hanna Mikkola and Timm Schroeder, put together an exciting program, during which field leaders and new investigators presented discoveries that spanned developmental and adult hematopoiesis within both physiologic and pathologic contexts. Collectively, the program highlighted the increasing pace of new discoveries and the substantial progress made in the hematopoiesis field since the last Keystone meeting two years ago. In this Meeting Review, we highlight the main concepts discussed at the conference, with an emphasis on topics relevant to developmental biology.


Subject(s)
Altitude , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , Leukemia/metabolism
15.
Blood ; 128(24): 2765-2769, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27799163

ABSTRACT

Hematopoietic stem cells (HSCs) have long been considered the continuous source of all hematopoietic cells for the life of an individual. Recent findings have questioned multiple aspects of this view, including the ability of lifelong HSCs to contribute to tissue-resident immune cells. Here we discuss the most recent findings on the source of B1a cells, innatelike lymphocytes that primarily reside in serous cavities. Powerful experimental approaches including bar coding, single cell transplantation, in vivo lineage tracing, and HSC-specific pulse-chase labeling have provided novel insights on B1a-cell generation during ontogeny. We evaluate the evidence for fetal vs adult B1a-cell production capacity and the identity of putative cells of origin. Integrating these most recent findings with previous work, we propose a working model that encapsulates our current understanding of waves of immune development.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Lymphocytes/cytology , Animals , Cell Lineage , Fetus/cytology , Humans , Liver/cytology , Liver/embryology , Models, Biological
16.
EMBO J ; 32(19): 2535-47, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24022369

ABSTRACT

Haematopoietic stem cell (HSC) niches provide an environment essential for life-long HSC function. Intense investigation of HSC niches both feed off and drive technology development to increase our capability to assay functionally defined cells with high resolution. A major driving force behind the desire to understand the basic biology of HSC niches is the clear implications for clinical therapies. Here, with particular emphasis on cell type-specific deletion of SCL and CXCL12, we focus on unresolved issues on HSC niches, framed around some very recent advances and novel discoveries on the extrinsic regulation of HSC maintenance. We also provide ideas for possible paths forward, some of which are clearly within reach while others will require both novel tools and vision.


Subject(s)
Hematopoietic Stem Cells/physiology , Stem Cell Niche , Animals , Cell Movement , Chemokine CXCL12/physiology , Hematopoietic Stem Cells/cytology , Humans , Stem Cell Factor/physiology
17.
Cells ; 13(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667319

ABSTRACT

Platelets are the terminal progeny of megakaryocytes, primarily produced in the bone marrow, and play critical roles in blood homeostasis, clotting, and wound healing. Traditionally, megakaryocytes and platelets are thought to arise from multipotent hematopoietic stem cells (HSCs) via multiple discrete progenitor populations with successive, lineage-restricting differentiation steps. However, this view has recently been challenged by studies suggesting that (1) some HSC clones are biased and/or restricted to the platelet lineage, (2) not all platelet generation follows the "canonical" megakaryocytic differentiation path of hematopoiesis, and (3) platelet output is the default program of steady-state hematopoiesis. Here, we specifically investigate the evidence that in vivo lineage tracing studies provide for the route(s) of platelet generation and investigate the involvement of various intermediate progenitor cell populations. We further identify the challenges that need to be overcome that are required to determine the presence, role, and kinetics of these possible alternate pathways.


Subject(s)
Blood Platelets , Hematopoietic Stem Cells , Animals , Mice , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Differentiation , Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Humans
18.
Proc Natl Acad Sci U S A ; 107(23): 10520-5, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20498081

ABSTRACT

Formation of the vascular system within organs requires the balanced action of numerous positive and negative factors secreted by stromal and epithelial cells. Here, we used a genetic approach to determine the role of SLITs in regulating the growth and organization of blood vessels in the mammary gland. We demonstrate that vascularization of the gland is not affected by loss of Slit expression in the epithelial compartment. Instead, we identify a stromal source of SLIT, mural cells encircling blood vessels, and show that loss of Slit in the stroma leads to elevated blood vessel density and complexity. We examine candidate SLIT receptors, Robo1 and Robo4, and find that increased vessel angiogenesis is phenocopied by loss of endothelial-specific Robo4, as long as it is combined with the presence of an angiogenic stimulus such as preneoplasia or pregnancy. In contrast, loss of Robo1 does not affect blood vessel growth. The enhanced growth of blood vessels in Robo4(-/-) endothelium is due to activation of vascular endothelial growth factor (VEGF)-R2 signaling through the Src and FAK kinases. Thus, our studies present a genetic dissection of SLIT/ROBO signaling during organ development. We identify a stromal, rather than epithelial, source of SLITs that inhibits blood vessel growth by signaling through endothelial ROBO4 to down-regulate VEGF/VEGFR2 signaling.


Subject(s)
Mammary Glands, Animal/blood supply , Mammary Glands, Animal/metabolism , Neovascularization, Physiologic , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Receptors, Cell Surface , Receptors, Immunologic/deficiency , Vascular Endothelial Growth Factor Receptor-2/metabolism , Roundabout Proteins
19.
Sci Rep ; 12(1): 3156, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35210475

ABSTRACT

Hematopoietic stem cell (HSC) multipotency and self-renewal are typically defined through serial transplantation experiments. Host conditioning is necessary for robust HSC engraftment, likely by reducing immune-mediated rejection and by clearing limited HSC niche space. Because irradiation of the recipient mouse is non-specific and broadly damaging, there is a need to develop alternative models to study HSC performance at steady-state and in the absence of radiation-induced stress. We have generated and characterized two new mouse models where either all hematopoietic cells or only HSCs can be specifically induced to die in vivo or in vitro. Hematopoietic-specific Vav1-mediated expression of a loxP-flanked diphtheria-toxin receptor (DTR) renders all hematopoietic cells sensitive to diphtheria toxin (DT) in "Vav-DTR" mice. Crossing these mice to Flk2-Cre mice results in "HSC-DTR" mice which exhibit HSC-selective DT sensitivity. We demonstrate robust, rapid, and highly selective cell ablation in these models. These new mouse models provide a platform to test whether HSCs are required for long-term hematopoiesis in vivo, for understanding the mechanisms regulating HSC engraftment, and interrogating in vivo hematopoietic differentiation pathways and mechanisms regulating hematopoietic homeostasis.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Models, Animal , Animals , Cell Differentiation , Hematopoietic Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
Cell Rep ; 41(8): 111677, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36417858

ABSTRACT

Adult hematopoietic stem and progenitor cells (HSPCs) respond directly to inflammation and infection, causing both acute and persistent changes to quiescence, mobilization, and differentiation. Here we show that murine fetal HSPCs respond to prenatal inflammation in utero and that the fetal response shapes postnatal hematopoiesis and immune cell function. Heterogeneous fetal HSPCs show divergent responses to maternal immune activation (MIA), including changes in quiescence, expansion, and lineage-biased output. Single-cell transcriptomic analysis of fetal HSPCs in response to MIA reveals specific upregulation of inflammatory gene profiles in discrete, transient hematopoietic stem cell (HSC) populations that propagate expansion of lymphoid-biased progenitors. Beyond fetal development, MIA causes the inappropriate expansion and persistence of fetal lymphoid-biased progenitors postnatally, concomitant with increased cellularity and hyperresponsiveness of fetal-derived innate-like lymphocytes. Our investigation demonstrates how inflammation in utero can direct the output and function of fetal-derived immune cells by reshaping fetal HSC establishment.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells , Pregnancy , Female , Mice , Animals , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Fetus , Inflammation/metabolism , Fetal Development
SELECTION OF CITATIONS
SEARCH DETAIL