Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell ; 175(1): 43-56.e21, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30241615

ABSTRACT

Stem cell regulation and hierarchical organization of human skeletal progenitors remain largely unexplored. Here, we report the isolation of a self-renewing and multipotent human skeletal stem cell (hSSC) that generates progenitors of bone, cartilage, and stroma, but not fat. Self-renewing and multipotent hSSCs are present in fetal and adult bones and can also be derived from BMP2-treated human adipose stroma (B-HAS) and induced pluripotent stem cells (iPSCs). Gene expression analysis of individual hSSCs reveals overall similarity between hSSCs obtained from different sources and partially explains skewed differentiation toward cartilage in fetal and iPSC-derived hSSCs. hSSCs undergo local expansion in response to acute skeletal injury. In addition, hSSC-derived stroma can maintain human hematopoietic stem cells (hHSCs) in serum-free culture conditions. Finally, we combine gene expression and epigenetic data of mouse skeletal stem cells (mSSCs) and hSSCs to identify evolutionarily conserved and divergent pathways driving SSC-mediated skeletogenesis. VIDEO ABSTRACT.


Subject(s)
Bone Development/physiology , Bone and Bones/cytology , Hematopoietic Stem Cells/cytology , Animals , Bone and Bones/metabolism , Cartilage/cytology , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Signal Transduction , Single-Cell Analysis/methods , Stem Cells/cytology , Stromal Cells/cytology , Transcriptome/genetics
2.
Cell ; 160(1-2): 285-98, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25594184

ABSTRACT

How are skeletal tissues derived from skeletal stem cells? Here, we map bone, cartilage, and stromal development from a population of highly pure, postnatal skeletal stem cells (mouse skeletal stem cells, mSSCs) to their downstream progenitors of bone, cartilage, and stromal tissue. We then investigated the transcriptome of the stem/progenitor cells for unique gene-expression patterns that would indicate potential regulators of mSSC lineage commitment. We demonstrate that mSSC niche factors can be potent inducers of osteogenesis, and several specific combinations of recombinant mSSC niche factors can activate mSSC genetic programs in situ, even in nonskeletal tissues, resulting in de novo formation of cartilage or bone and bone marrow stroma. Inducing mSSC formation with soluble factors and subsequently regulating the mSSC niche to specify its differentiation toward bone, cartilage, or stromal cells could represent a paradigm shift in the therapeutic regeneration of skeletal tissues.


Subject(s)
Bone and Bones/cytology , Mesenchymal Stem Cells/cytology , Animals , Bone Morphogenetic Proteins/metabolism , Cartilage/cytology , Cell Lineage , Crosses, Genetic , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
3.
Nature ; 597(7875): 256-262, 2021 09.
Article in English | MEDLINE | ID: mdl-34381212

ABSTRACT

Loss of skeletal integrity during ageing and disease is associated with an imbalance in the opposing actions of osteoblasts and osteoclasts1. Here we show that intrinsic ageing of skeletal stem cells (SSCs)2 in mice alters signalling in the bone marrow niche and skews the differentiation of bone and blood lineages, leading to fragile bones that regenerate poorly. Functionally, aged SSCs have a decreased bone- and cartilage-forming potential but produce more stromal lineages that express high levels of pro-inflammatory and pro-resorptive cytokines. Single-cell RNA-sequencing studies link the functional loss to a diminished transcriptomic diversity of SSCs in aged mice, which thereby contributes to the transformation of the bone marrow niche. Exposure to a youthful circulation through heterochronic parabiosis or systemic reconstitution with young haematopoietic stem cells did not reverse the diminished osteochondrogenic activity of aged SSCs, or improve bone mass or skeletal healing parameters in aged mice. Conversely, the aged SSC lineage promoted osteoclastic activity and myeloid skewing by haematopoietic stem and progenitor cells, suggesting that the ageing of SSCs is a driver of haematopoietic ageing. Deficient bone regeneration in aged mice could only be returned to youthful levels by applying a combinatorial treatment of BMP2 and a CSF1 antagonist locally to fractures, which reactivated aged SSCs and simultaneously ablated the inflammatory, pro-osteoclastic milieu. Our findings provide mechanistic insights into the complex, multifactorial mechanisms that underlie skeletal ageing and offer prospects for rejuvenating the aged skeletal system.


Subject(s)
Aging/pathology , Bone and Bones/pathology , Cellular Senescence , Inflammation/pathology , Stem Cell Niche , Stem Cells/pathology , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Regeneration , Cell Lineage , Female , Fracture Healing , Hematopoiesis , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Myeloid Cells/cytology , Osteoclasts/cytology , Rejuvenation
4.
Nature ; 587(7835): 619-625, 2020 11.
Article in English | MEDLINE | ID: mdl-33208946

ABSTRACT

Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1-9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.


Subject(s)
Cells/classification , Cells/metabolism , Immunity , Lung/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome/genetics , Aged , Animals , Atlases as Topic , Biomarkers , Cell Communication , Cells/immunology , Chemokines/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Female , Humans , Lung/immunology , Male , Mice , Middle Aged , Receptors, Lymphocyte Homing/metabolism , Signal Transduction , Stromal Cells/metabolism
5.
Mol Cell Proteomics ; 22(12): 100677, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37949301

ABSTRACT

Proteins can be modified by lipids in various ways, for example, by myristoylation, palmitoylation, farnesylation, and geranylgeranylation-these processes are collectively referred to as lipidation. Current chemical proteomics using alkyne lipids has enabled the identification of lipidated protein candidates but does not identify endogenous lipidation sites and is not readily applicable to in vivo systems. Here, we introduce a proteomic methodology for global analysis of endogenous protein N-terminal myristoylation sites that combines liquid-liquid extraction of hydrophobic lipidated peptides with liquid chromatography-tandem mass spectrometry using a gradient program of acetonitrile in the high concentration range. We applied this method to explore myristoylation sites in HeLa cells and identified a total of 75 protein N-terminal myristoylation sites, which is more than the number of high-confidence myristoylated proteins identified by myristic acid analog-based chemical proteomics. Isolation of myristoylated peptides from HeLa digests prepared with different proteases enabled the identification of different myristoylated sites, extending the coverage of N-myristoylome. Finally, we analyzed in vivo myristoylation sites in mouse tissues and found that the lipidation profile is tissue-specific. This simple method (not requiring chemical labeling or affinity purification) should be a promising tool for global profiling of protein N-terminal myristoylation.


Subject(s)
Proteins , Proteomics , Humans , Animals , Mice , Myristic Acid/chemistry , Myristic Acid/metabolism , HeLa Cells , Proteins/metabolism , Peptides/metabolism , Liquid-Liquid Extraction , Protein Processing, Post-Translational
6.
Arterioscler Thromb Vasc Biol ; 43(7): 1262-1277, 2023 07.
Article in English | MEDLINE | ID: mdl-37051932

ABSTRACT

BACKGROUND: Peripheral vascular disease remains a leading cause of vascular morbidity and mortality worldwide despite advances in medical and surgical therapy. Besides traditional approaches, which can only restore blood flow to native arteries, an alternative approach is to enhance the growth of new vessels, thereby facilitating the physiological response to ischemia. METHODS: The ActinCreER/R26VT2/GK3 Rainbow reporter mouse was used for unbiased in vivo survey of injury-responsive vasculogenic clonal formation. Prospective isolation and transplantation were used to determine vessel-forming capacity of different populations. Single-cell RNA-sequencing was used to characterize distinct vessel-forming populations and their interactions. RESULTS: Two populations of distinct vascular stem/progenitor cells (VSPCs) were identified from adipose-derived mesenchymal stromal cells: VSPC1 is CD45-Ter119-Tie2+PDGFRa-CD31+CD105highSca1low, which gives rise to stunted vessels (incomplete tubular structures) in a transplant setting, and VSPC2 which is CD45-Ter119-Tie2+PDGFRa+CD31-CD105lowSca1high and forms stunted vessels and fat. Interestingly, cotransplantation of VSPC1 and VSPC2 is required to form functional vessels that improve perfusion in the mouse hindlimb ischemia model. Similarly, VSPC1 and VSPC2 populations isolated from human adipose tissue could rescue the ischemic condition in mice. CONCLUSIONS: These findings suggest that autologous cotransplantation of synergistic VSPCs from nonessential adipose tissue can promote neovascularization and represents a promising treatment for ischemic disease.


Subject(s)
Mesenchymal Stem Cells , Neovascularization, Physiologic , Mice , Humans , Animals , Neovascularization, Physiologic/physiology , Adipose Tissue , Neovascularization, Pathologic , Ischemia/therapy , Disease Models, Animal , Hindlimb/blood supply
7.
Nature ; 564(7736): 425-429, 2018 12.
Article in English | MEDLINE | ID: mdl-30518860

ABSTRACT

Haematopoiesis is an essential process that evolved in multicellular animals. At the heart of this process are haematopoietic stem cells (HSCs), which are multipotent and self-renewing, and generate the entire repertoire of blood and immune cells throughout an animal's life1. Although there have been comprehensive studies on self-renewal, differentiation, physiological regulation and niche occupation in vertebrate HSCs, relatively little is known about the evolutionary origin and niches of these cells. Here we describe the haematopoietic system of Botryllus schlosseri, a colonial tunicate that has a vasculature and circulating blood cells, and interesting stem-cell biology and immunity characteristics2-8. Self-recognition between genetically compatible B. schlosseri colonies leads to the formation of natural parabionts with shared circulation, whereas incompatible colonies reject each other3,4,7. Using flow cytometry, whole-transcriptome sequencing of defined cell populations and diverse functional assays, we identify HSCs, progenitors, immune effector cells and an HSC niche, and demonstrate that self-recognition inhibits allospecific cytotoxic reactions. Our results show that HSC and myeloid lineage immune cells emerged in a common ancestor of tunicates and vertebrates, and also suggest that haematopoietic bone marrow and the B. schlosseri endostyle niche evolved from a common origin.


Subject(s)
Hematopoiesis , Hematopoietic System/cytology , Mammals/blood , Phylogeny , Urochordata/cytology , Animals , Cell Differentiation , Cell Lineage , Cytotoxicity, Immunologic , Female , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Immunity, Cellular , Isoantigens/immunology , Male , Mammals/anatomy & histology , Myeloid Cells/cytology , Myeloid Cells/immunology , Phagocytosis/immunology , Stem Cell Niche , Transcriptome/genetics , Urochordata/anatomy & histology , Urochordata/genetics , Urochordata/immunology
8.
Allergol Int ; 73(2): 255-263, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38102028

ABSTRACT

BACKGROUND: In clinical research on multifactorial diseases such as atopic dermatitis, data-driven medical research has become more widely used as means to clarify diverse pathological conditions and to realize precision medicine. However, modern clinical data, characterized as large-scale, multimodal, and multi-center, causes difficulties in data integration and management, which limits productivity in clinical data science. METHODS: We designed a generic data management flow to collect, cleanse, and integrate data to handle different types of data generated at multiple institutions by 10 types of clinical studies. We developed MeDIA (Medical Data Integration Assistant), a software to browse the data in an integrated manner and extract subsets for analysis. RESULTS: MeDIA integrates and visualizes data and information on research participants obtained from multiple studies. It then provides a sophisticated interface that supports data management and helps data scientists retrieve the data sets they need. Furthermore, the system promotes the use of unified terms such as identifiers or sampling dates to reduce the cost of pre-processing by data analysts. We also propose best practices in clinical data management flow, which we learned from the development and implementation of MeDIA. CONCLUSIONS: The MeDIA system solves the problem of multimodal clinical data integration, from complex text data such as medical records to big data such as omics data from a large number of patients. The system and the proposed best practices can be applied not only to allergic diseases but also to other diseases to promote data-driven medical research.


Subject(s)
Biomedical Research , Dermatitis, Atopic , Humans , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/therapy , Data Management , Precision Medicine
9.
Nat Immunol ; 12(2): 144-50, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21170027

ABSTRACT

Transcription factor NF-κB and its activating kinase IKKß are associated with inflammation and are believed to be critical for innate immunity. Despite the likelihood of immune suppression, pharmacological blockade of IKKß-NF-κB has been considered as a therapeutic strategy. However, we found neutrophilia in mice with inducible deletion of IKKß (Ikkß(Δ) mice). These mice had hyperproliferative granulocyte-macrophage progenitors and pregranulocytes and a prolonged lifespan of mature neutrophils that correlated with the induction of genes encoding prosurvival molecules. Deletion of interleukin 1 receptor 1 (IL-1R1) in Ikkß(Δ) mice normalized blood cellularity and prevented neutrophil-driven inflammation. However, Ikkß(Δ)Il1r1(-/-) mice, unlike Ikkß(Δ) mice, were highly susceptible to bacterial infection, which indicated that signaling via IKKß-NF-κB or IL-1R1 can maintain antimicrobial defenses in each other's absence, whereas inactivation of both pathways severely compromises innate immunity.


Subject(s)
Bacterial Infections/immunology , Granulocyte-Macrophage Progenitor Cells/metabolism , I-kappa B Kinase/metabolism , Interleukin-1beta/metabolism , Neutrophils/metabolism , Animals , Cell Count , Cell Growth Processes/genetics , Cell Survival/genetics , Cells, Cultured , Disease Susceptibility , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/pathology , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Immunity, Innate/genetics , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Neutrophils/immunology , Neutrophils/pathology , Receptors, Interleukin-1 Type I/genetics , Up-Regulation/genetics
10.
Nature ; 530(7589): 223-7, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26863982

ABSTRACT

Haematopoietic stem cells (HSCs) are arguably the most extensively characterized tissue stem cells. Since the identification of HSCs by prospective isolation, complex multi-parameter flow cytometric isolation of phenotypic subsets has facilitated studies on many aspects of HSC biology, including self-renewal, differentiation, ageing, niche, and diversity. Here we demonstrate by unbiased multi-step screening, identification of a single gene, homeobox B5 (Hoxb5, also known as Hox-2.1), with expression in the bone marrow that is limited to long-term (LT)-HSCs in mice. Using a mouse single-colour tri-mCherry reporter driven by endogenous Hoxb5 regulation, we show that only the Hoxb5(+) HSCs exhibit long-term reconstitution capacity after transplantation in primary transplant recipients and, notably, in secondary recipients. Only 7-35% of various previously defined immunophenotypic HSCs are LT-HSCs. Finally, by in situ imaging of mouse bone marrow, we show that >94% of LT-HSCs (Hoxb5(+)) are directly attached to VE-cadherin(+) cells, implicating the perivascular space as a near-homogenous location of LT-HSCs.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Stem Cell Niche , Animals , Antigens, CD/metabolism , Biomarkers/analysis , Bone Marrow/metabolism , Cadherins/metabolism , Cell Self Renewal , Gene Expression Regulation , Genes, Reporter/genetics , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/genetics , Immunophenotyping , Male , Mice , Mice, Inbred C57BL
11.
Proc Natl Acad Sci U S A ; 116(4): 1447-1456, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30622181

ABSTRACT

While the aggregate differentiation of the hematopoietic stem cell (HSC) population has been extensively studied, little is known about the lineage commitment process of individual HSC clones. Here, we provide lineage commitment maps of HSC clones under homeostasis and after perturbations of the endogenous hematopoietic system. Under homeostasis, all donor-derived HSC clones regenerate blood homogeneously throughout all measured stages and lineages of hematopoiesis. In contrast, after the hematopoietic system has been perturbed by irradiation or by an antagonistic anti-ckit antibody, only a small fraction of donor-derived HSC clones differentiate. Some of these clones dominantly expand and exhibit lineage bias. We identified the cellular origins of clonal dominance and lineage bias and uncovered the lineage commitment pathways that lead HSC clones to different levels of self-renewal and blood production under various transplantation conditions. This study reveals surprising alterations in HSC fate decisions directed by conditioning and identifies the key hematopoiesis stages that may be manipulated to control blood production and balance.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/physiology , Cell Lineage , Clone Cells , Homeostasis/physiology , Mice , Mice, Inbred C57BL
12.
Proc Natl Acad Sci U S A ; 116(37): 18416-18422, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31451629

ABSTRACT

GABRR1 is a rho subunit receptor of GABA, the major inhibitory neurotransmitter in the mammalian brain. While most investigations of its function focused on the nervous system, its regulatory role in hematopoiesis has not been reported. In this study, we found GABRR1 is mainly expressed on subsets of human and mouse hematopoietic stem cells (HSCs) and megakaryocyte progenitors (MkPs). GABRR1-negative (GR-) HSCs led to higher donor-derived hematopoietic chimerism than GABRR1-positive (GR+) HSCs. GR+ but not GR- HSCs and MkPs respond to GABA in patch clamp studies. Inhibition of GABRR1 via genetic knockout or antagonists inhibited MkP differentiation and reduced platelet numbers in blood. Overexpression of GABRR1 or treatment with agonists significantly promoted MkP generation and megakaryocyte colonies. Thus, this study identifies a link between the neural and hematopoietic systems and opens up the possibility of manipulating GABA signaling for platelet-required clinical applications.


Subject(s)
Hematopoietic Stem Cells/metabolism , Megakaryocyte Progenitor Cells/metabolism , Receptors, GABA-A/metabolism , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Differentiation/physiology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans , Male , Megakaryocyte Progenitor Cells/cytology , Megakaryocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Receptors, GABA , Receptors, GABA-A/genetics , Transcriptome
13.
Proc Natl Acad Sci U S A ; 115(40): E9308-E9316, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30150396

ABSTRACT

Different combinations of transcription factors (TFs) function at each stage of hematopoiesis, leading to distinct expression patterns of lineage-specific genes. The identification of such regulators and their functions in hematopoiesis remain largely unresolved. In this study, we utilized screening approaches to study the transcriptional regulators of megakaryocyte progenitor (MkP) generation, a key step before platelet production. Promising candidate genes were generated from a microarray platform gene expression commons and individually manipulated in human hematopoietic stem and progenitor cells (HSPCs). Deletion of some of the candidate genes (the hit genes) by CRISPR/Cas9 led to decreased MkP generation during HSPC differentiation, while more MkPs were produced when some hit genes were overexpressed in HSPCs. We then demonstrated that overexpression of these genes can increase the frequency of mature megakaryocytic colonies by functional colony forming unit-megakaryocyte (CFU-Mk) assay and the release of platelets after in vitro maturation. Finally, we showed that the histone deacetylase inhibitors could also increase MkP differentiation, possibly by regulating some of the newly identified TFs. Therefore, identification of such regulators will advance the understanding of basic mechanisms of HSPC differentiation and conceivably enable the generation and maturation of megakaryocytes and platelets in vitro.


Subject(s)
Blood Platelets/metabolism , Cell Differentiation/physiology , Blood Platelets/cytology , CRISPR-Cas Systems , Cell Line , Humans , Megakaryocyte Progenitor Cells/cytology
14.
Proc Natl Acad Sci U S A ; 112(31): 9638-43, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195758

ABSTRACT

Determining the developmental pathway leading to erythrocytes and being able to isolate their progenitors are crucial to understanding and treating disorders of red cell imbalance such as anemia, myelodysplastic syndrome, and polycythemia vera. Here we show that the human erythrocyte progenitor (hEP) can be prospectively isolated from adult bone marrow. We found three subfractions that possessed different expression patterns of CD105 and CD71 within the previously defined human megakaryocyte/erythrocyte progenitor (hMEP; Lineage(-) CD34(+) CD38(+) IL-3Rα(-) CD45RA(-)) population. Both CD71(-) CD105(-) and CD71(+) CD105(-) MEPs, at least in vitro, still retained bipotency for the megakaryocyte (MegK) and erythrocyte (E) lineages, although the latter subpopulation is skewed in differentiation toward the erythroid lineage. Notably, the proliferative and differentiation output of the CD71(intermediate(int)/+) CD105(+) subset of cells within the MEP population was completely restricted to the erythroid lineage with the loss of MegK potential. CD71(+) CD105(-) MEPs are erythrocyte-biased MEPs (E-MEPs) and CD71(int/+) CD105(+) cells are EPs. These previously unclassified populations may facilitate further understanding of the molecular mechanisms governing human erythroid development and serve as potential therapeutic targets in disorders of the erythroid lineage.


Subject(s)
Cell Lineage , Cell Separation/methods , Erythroid Precursor Cells/cytology , Antigens, CD/metabolism , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Fractionation , Cell Lineage/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Erythrocytes/cytology , Erythrocytes/drug effects , Erythroid Precursor Cells/drug effects , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Immunophenotyping , Megakaryocytes/cytology , Megakaryocytes/drug effects , Models, Biological , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects
15.
Proc Natl Acad Sci U S A ; 111(7): 2698-703, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24497508

ABSTRACT

Emerging evidence indicates that myeloid cells are essential for promoting new blood vessel formation by secreting various angiogenic factors. Given that hypoxia-inducible factor (HIF) is a critical regulator for angiogenesis, we questioned whether HIF in myeloid cells also plays a role in promoting angiogenesis. To address this question, we generated a unique strain of myeloid-specific knockout mice targeting HIF pathways using human S100A8 as a myeloid-specific promoter. We observed that mutant mice where HIF-1 is transcriptionally activated in myeloid cells (by deletion of the von Hippel-Lindau gene) resulted in erythema, enhanced neovascularization in matrigel plugs, and increased production of vascular endothelial growth factor (VEGF) in the bone marrow, all of which were completely abrogated by either genetic or pharmacological inactivation of HIF-1. We further found that monocytes were the major effector producing VEGF and S100A8 proteins driving neovascularization in matrigel. Moreover, by using a mouse model of hindlimb ischemia we observed significantly improved blood flow in mice intramuscularly injected with HIF-1-activated monocytes. This study therefore demonstrates that HIF-1 activation in myeloid cells promotes angiogenesis through VEGF and S100A8 and that this may become an attractive therapeutic strategy to treat diseases with vascular defects.


Subject(s)
Calgranulin A/metabolism , Hypoxia-Inducible Factor 1/metabolism , Myeloid Cells/metabolism , Neovascularization, Physiologic/physiology , Transcriptional Activation/physiology , Vascular Endothelial Growth Factor A/metabolism , Analysis of Variance , Animals , Blotting, Western , Collagen , Crosses, Genetic , DNA Primers/genetics , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hindlimb/blood supply , Ischemia/physiopathology , Laminin , Mice , Mice, Transgenic , Polymerase Chain Reaction , Proteoglycans , Transcriptional Activation/genetics
16.
Genes Dev ; 23(20): 2376-81, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19833765

ABSTRACT

Common lymphoid progenitors (CLPs) clonally produce both B- and T-cell lineages, but have little myeloid potential in vivo. However, some studies claim that the upstream lymphoid-primed multipotent progenitor (LMPP) is the thymic seeding population, and suggest that CLPs are primarily B-cell-restricted. To identify surface proteins that distinguish functional CLPs from B-cell progenitors, we used a new computational method of Mining Developmentally Regulated Genes (MiDReG). We identified Ly6d, which divides CLPs into two distinct populations: one that retains full in vivo lymphoid potential and produces more thymocytes at early timepoints than LMPP, and another that behaves essentially as a B-cell progenitor.


Subject(s)
Antigens, Ly/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation , Lymphoid Progenitor Cells/cytology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Antigens, Ly/genetics , Cells, Cultured , GPI-Linked Proteins , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL
17.
Nature ; 467(7313): 338-42, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20720541

ABSTRACT

Epigenetic modifications must underlie lineage-specific differentiation as terminally differentiated cells express tissue-specific genes, but their DNA sequence is unchanged. Haematopoiesis provides a well-defined model to study epigenetic modifications during cell-fate decisions, as multipotent progenitors (MPPs) differentiate into progressively restricted myeloid or lymphoid progenitors. Although DNA methylation is critical for myeloid versus lymphoid differentiation, as demonstrated by the myeloerythroid bias in Dnmt1 hypomorphs, a comprehensive DNA methylation map of haematopoietic progenitors, or of any multipotent/oligopotent lineage, does not exist. Here we examined 4.6 million CpG sites throughout the genome for MPPs, common lymphoid progenitors (CLPs), common myeloid progenitors (CMPs), granulocyte/macrophage progenitors (GMPs), and thymocyte progenitors (DN1, DN2, DN3). Marked epigenetic plasticity accompanied both lymphoid and myeloid restriction. Myeloid commitment involved less global DNA methylation than lymphoid commitment, supported functionally by myeloid skewing of progenitors following treatment with a DNA methyltransferase inhibitor. Differential DNA methylation correlated with gene expression more strongly at CpG island shores than CpG islands. Many examples of genes and pathways not previously known to be involved in choice between lymphoid/myeloid differentiation have been identified, such as Arl4c and Jdp2. Several transcription factors, including Meis1, were methylated and silenced during differentiation, indicating a role in maintaining an undifferentiated state. Additionally, epigenetic modification of modifiers of the epigenome seems to be important in haematopoietic differentiation. Our results directly demonstrate that modulation of DNA methylation occurs during lineage-specific differentiation and defines a comprehensive map of the methylation and transcriptional changes that accompany myeloid versus lymphoid fate decisions.


Subject(s)
Cell Lineage , DNA Methylation , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Cell Line , Cell Lineage/genetics , CpG Islands/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Gene Expression Profiling , Genome/genetics , Hematopoiesis/genetics , Lymphocytes/cytology , Lymphocytes/metabolism , Metabolome , Metabolomics , Mice , Myeloid Cells/cytology , Myeloid Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
18.
Proc Natl Acad Sci U S A ; 110(27): 11103-8, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23690610

ABSTRACT

Mobilization of the T-cell response against cancer has the potential to achieve long-lasting cures. However, it is not known how to harness antigen-presenting cells optimally to achieve an effective antitumor T-cell response. In this study, we show that anti-CD47 antibody-mediated phagocytosis of cancer by macrophages can initiate an antitumor T-cell immune response. Using the ovalbumin model antigen system, anti-CD47 antibody-mediated phagocytosis of cancer cells by macrophages resulted in increased priming of OT-I T cells [cluster of differentiation 8-positive (CD8(+))] but decreased priming of OT-II T cells (CD4(+)). The CD4(+) T-cell response was characterized by a reduction in forkhead box P3-positive (Foxp3(+)) regulatory T cells. Macrophages following anti-CD47-mediated phagocytosis primed CD8(+) T cells to exhibit cytotoxic function in vivo. This response protected animals from tumor challenge. We conclude that anti-CD47 antibody treatment not only enables macrophage phagocytosis of cancer but also can initiate an antitumor cytotoxic T-cell immune response.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD47 Antigen/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Macrophages/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Blocking/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/pathology , Cytotoxicity, Immunologic/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagocytosis/immunology , Up-Regulation/genetics , Up-Regulation/immunology , Xenograft Model Antitumor Assays
19.
Proc Natl Acad Sci U S A ; 110(31): 12643-8, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23858471

ABSTRACT

Organs are composites of tissue types with diverse developmental origins, and they rely on distinct stem and progenitor cells to meet physiological demands for cellular production and homeostasis. How diverse stem cell activity is coordinated within organs is not well understood. Here we describe a lineage-restricted, self-renewing common skeletal progenitor (bone, cartilage, stromal progenitor; BCSP) isolated from limb bones and bone marrow tissue of fetal, neonatal, and adult mice. The BCSP clonally produces chondrocytes (cartilage-forming) and osteogenic (bone-forming) cells and at least three subsets of stromal cells that exhibit differential expression of cell surface markers, including CD105 (or endoglin), Thy1 [or CD90 (cluster of differentiation 90)], and 6C3 [ENPEP glutamyl aminopeptidase (aminopeptidase A)]. These three stromal subsets exhibit differential capacities to support hematopoietic (blood-forming) stem and progenitor cells. Although the 6C3-expressing subset demonstrates functional stem cell niche activity by maintaining primitive hematopoietic stem cell (HSC) renewal in vitro, the other stromal populations promote HSC differentiation to more committed lines of hematopoiesis, such as the B-cell lineage. Gene expression analysis and microscopic studies further reveal a microenvironment in which CD105-, Thy1-, and 6C3-expressing marrow stroma collaborate to provide cytokine signaling to HSCs and more committed hematopoietic progenitors. As a result, within the context of bone as a blood-forming organ, the BCSP plays a critical role in supporting hematopoiesis through its generation of diverse osteogenic and hematopoietic-promoting stroma, including HSC supportive 6C3(+) niche cells.


Subject(s)
Bone and Bones/metabolism , Cartilage/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Signal Transduction/physiology , Stem Cell Niche/physiology , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Bone and Bones/cytology , Cartilage/cytology , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/physiology , Hematopoietic Stem Cells/cytology , Mice , Mice, Transgenic , Stromal Cells/cytology , Stromal Cells/metabolism
20.
Biol Methods Protoc ; 9(1): bpae039, 2024.
Article in English | MEDLINE | ID: mdl-38884001

ABSTRACT

Mapping protein interaction complexes in their natural state in vivo is arguably the Holy Grail of protein network analysis. Detection of protein interaction stoichiometry has been an important technical challenge, as few studies have focused on this. This may, however, be solved by artificial intelligence (AI) and proteomics. Here, we describe the development of HaloTag-based affinity purification mass spectrometry (HaloMS), a high-throughput HaloMS assay for protein interaction discovery. The approach enables the rapid capture of newly expressed proteins, eliminating tedious conventional one-by-one assays. As a proof-of-principle, we used HaloMS to evaluate the protein complex interactions of 17 regulatory proteins in human adipocytes. The adipocyte interactome network was validated using an in vitro pull-down assay and AI-based prediction tools. Applying HaloMS to probe adipocyte differentiation facilitated the identification of previously unknown transcription factor (TF)-protein complexes, revealing proteome-wide human adipocyte TF networks and shedding light on how different pathways are integrated.

SELECTION OF CITATIONS
SEARCH DETAIL