Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
JCI Insight ; 8(5)2023 03 08.
Article in English | MEDLINE | ID: mdl-36692963

ABSTRACT

Most circulating endothelial cells are apoptotic, but rare circulating endothelial colony-forming cells (C-ECFCs), also known as blood outgrowth endothelial cells, with proliferative and vasculogenic activity can be cultured; however, the origin and naive function of these C-ECFCs remains obscure. Herein, detailed lineage tracing revealed murine C-ECFCs emerged in the early postnatal period, displayed high vasculogenic potential with enriched frequency of clonal proliferative cells compared with tissue-resident ECFCs, and were not committed to or derived from the BM hematopoietic system but from tissue-resident ECFCs. In humans, C-ECFCs were present in the CD34bright cord blood mononuclear subset, possessed proliferative potential and in vivo vasculogenic function in a naive or cultured state, and displayed a single cell transcriptome sharing some umbilical venous endothelial cell features, such as a higher protein C receptor and extracellular matrix gene expression. This study provides an advance for the field by identifying the origin, naive function, and antigens to prospectively isolate C-ECFCs for translational studies.


Subject(s)
Endothelial Cells , Extracellular Matrix , Humans , Animals , Mice , Prospective Studies , Clone Cells , Endothelial Protein C Receptor
2.
J Pediatr Surg ; 56(6): 1220-1225, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33745738

ABSTRACT

INTRODUCTION: Necrotizing enterocolitis (NEC) remains a devastating disease that affects the gastrointestinal tract of the preterm infant. Volatile organic compounds (VOCs) have emerged as a non-invasive biomarker in many diseases. We hypothesized that fecal VOC profiles would be significantly different between control and NEC pups in a NEC mouse model. METHODS: Experimental NEC was induced in five-day-old mice. Breastfed and formula-fed control groups were also studied. After four days, pups were euthanized and intestines were H&E stained and blindly scored. Stool microbiome analysis was performed via 16S rRNA sequencing. VOC analysis was assessed by the CyranoseⓇ 320 eNose device and p<0.05 was significant. RESULTS: NEC pups had severe intestinal injury when compared to controls. Microbiome analysis showed that both control groups had significantly higher microbial diversity and relative abundance of Lactobacillus than NEC, and lower relative abundance of Escherichia. Fecal VOC profile for NEC pups was significantly different from controls. CONCLUSIONS: Experimental NEC was associated with intestinal dysbiosis. Fecal VOC analysis by the CyranoseⓇ 320 eNose device can discriminate NEC pups from both breastfed and formula-fed controls. Further research is warranted to establish whether fecal VOCs can be used as a biomarker or predictive algorithm to diagnose NEC.


Subject(s)
Enterocolitis, Necrotizing , Microbiota , Volatile Organic Compounds , Animals , Enterocolitis, Necrotizing/diagnosis , Humans , Infant, Newborn , Infant, Premature , Mice , RNA, Ribosomal, 16S
3.
JCI Insight ; 4(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31672944

ABSTRACT

Retinopathy of prematurity (ROP) is a disorder of the developing retina of preterm infants. ROP can lead to blindness because of abnormal angiogenesis that is the result of suspended vascular development and vaso-obliteration leading to severe retinal stress and hypoxia. We tested the hypothesis that the use of the human progenitor cell combination, bone marrow-derived CD34+ cells and vascular wall-derived endothelial colony-forming cells (ECFCs), would synergistically protect the developing retinal vasculature in a mouse model of ROP, called oxygen-induced retinopathy (OIR). CD34+ cells alone, ECFCs alone, or the combination thereof were injected intravitreally at either P5 or P12 and pups were euthanized at P17. Retinas from OIR mice injected with ECFCs or the combined treatment revealed formation of the deep vascular plexus (DVP) while still in hyperoxia, with normal-appearing connections between the superficial vascular plexus (SVP) and the DVP. In addition, the combination of cells completely prevented aberrant retinal neovascularization and was more effective anatomically and functionally at rescuing the ischemia phenotype than either cell type alone. We show that the beneficial effects of the cell combination are the result of their ability to orchestrate an acceleration of vascular development and more rapid ensheathment of pericytes on the developing vessels. Lastly, our proteomic and transcriptomic data sets reveal pathways altered by the dual cell therapy, including many involved in neuroretinal maintenance, and principal component analysis (PCA) showed that cell therapy restored OIR retinas to a state that was closely associated with age-matched normal retinas. Together, these data herein support the use of dual cell therapy as a promising preventive treatment for the development of ROP in premature infants.


Subject(s)
Oxygen Inhalation Therapy/adverse effects , Retinal Neovascularization/etiology , Retinopathy of Prematurity/complications , Retinopathy of Prematurity/prevention & control , Stem Cells/cytology , Animals , Antigens, CD34/immunology , Disease Models, Animal , Mice , Retinopathy of Prematurity/pathology , Stem Cells/immunology
4.
Stem Cell Reports ; 9(5): 1573-1587, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29033304

ABSTRACT

Human endothelial colony-forming cells (ECFCs) represent a promising source of adult stem cells for vascular repair, yet their regenerative capacity is limited. Here, we set out to understand the molecular mechanism restricting the repair function of ECFCs. We found that key pro-angiogenic pathways are repressed in ECFCs due to the presence of bivalent (H3K27me3/H3K4me3) epigenetic marks, which decreases the cells' regenerative potential. Importantly, ex vivo treatment with a combination of epigenetic drugs that resolves bivalent marks toward the transcriptionally active H3K4me3 state leads to the simultaneous activation of multiple pro-angiogenic signaling pathways (VEGFR, CXCR4, WNT, NOTCH, SHH). This in turn results in improved capacity of ECFCs to form capillary-like networks in vitro and in vivo. Furthermore, restoration of perfusion is accelerated upon transplantation of drug-treated ECFCs in a model of hindlimb ischemia. Thus, ex vivo treatment with epigenetic drugs increases the vascular repair properties of ECFCs through transient activation of pro-angiogenic signaling pathways.


Subject(s)
Endothelial Progenitor Cells/metabolism , Epigenesis, Genetic , Neovascularization, Physiologic , Signal Transduction , Animals , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/transplantation , Female , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hindlimb/blood supply , Humans , Ischemia/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Stem Cell Transplantation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
5.
Blood ; 117(2): 440-50, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20966168

ABSTRACT

SIRT1 is a founding member of a sirtuin family of 7 proteins and histone deacetylases. It is involved in cellular resistance to stress, metabolism, differentiation, aging, and tumor suppression. SIRT1(-/-) mice demonstrate embryonic and postnatal development defects. We examined hematopoietic and endothelial cell differentiation of SIRT1(-/-) mouse embryonic stem cells (ESCs) in vitro, and hematopoietic progenitors in SIRT1(+/+)(+/-), and (-/-) mice. SIRT1(-/-) ESCs formed fewer mature blast cell colonies. Replated SIRT1(-/-) blast colony-forming cells demonstrated defective hematopoietic potential. Endothelial cell production was unaltered, but there were defects in formation of a primitive vascular network from SIRT1(-/-)-derived embryoid bodies. Development of primitive and definitive progenitors derived from SIRT1(-/-) ESCs were also delayed and/or defective. Differentiation delay/defects were associated with delayed capacity to switch off Oct4, Nanog and Fgf5 expression, decreased ß-H1 globin, ß-major globin, and Scl gene expression, and reduced activation of Erk1/2. Ectopic expression of SIRT1 rescued SIRT1(-/-) ESC differentiation deficiencies. SIRT1(-/-) yolk sacs manifested fewer primitive erythroid precursors. SIRT1(-/-) and SIRT1(+/-) adult marrow had decreased numbers and cycling of hematopoietic progenitors, effects more apparent at 5%, than at 20%, oxygen tension, and these progenitors survived less well in vitro under conditions of delayed growth factor addition. This suggests a role for SIRT1 in ESC differentiation and mouse hematopoiesis.


Subject(s)
Aging , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Sirtuin 1/metabolism , Animals , Blotting, Western , Cell Separation , Embryonic Stem Cells/cytology , Flow Cytometry , Gene Expression , Hematopoietic Stem Cells/cytology , Immunohistochemistry , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sirtuin 1/deficiency , Sirtuin 1/genetics
6.
Am J Pathol ; 176(5): 2394-404, 2010 May.
Article in English | MEDLINE | ID: mdl-20348241

ABSTRACT

Lysosomal acid lipase (LAL) is a key enzyme that cleaves cholesteryl esters and triglycerides to generate free fatty acids and cholesterol in lysosomes. Genetic ablation of the lal gene (lal(-/-)) in mice has resulted in a systemic increase of macrophages and neutrophils, causing severe inflammation and pathogenesis in multiple organs. We hypothesized that aberrant growth and differentiation of myeloid cells in lal(-/-) mice arises from dysregulated production of progenitor cells in the bone marrow. Indeed, lal(-/-) mice displayed increased numbers of primitive lin(-)Sca-1(+)c-Kit(+) (LSK) cells and granulocyte-macrophage precursors (GMP). Increased high proliferative potential colony-forming cells (HPP-CFC) were enumerated from cultured lal(-/-) bone marrow cells, as were significantly more CFU-GM, CFU-G, and CFU-M colonies. As a consequence, lal(-/-) mice developed significant myeloid infiltration, particularly with CD11b+/Gr-1+ myeloid-derived suppressive cells in multiple organs. Both decreased apoptosis and increased proliferation contribute to the systemic increase of myeloid cells in lal(-/-) myeloid cells. These lal(-/-) CD11b(+)/Gr-1(+) cells displayed suppressive activity on T cell proliferation and function in vitro. Bone marrow chimeras confirmed that the myeloproliferative disorder in lal(-/-) mice was primarily attributable to autonomous defects in myeloid progenitor cells, although the hematopoietic microenvironment in the lal(-/-) mice did not support hematopoiesis normally. These results provide evidence that LAL is an important regulator of myelopoiesis during hematopoietic development, differentiation, and homeostasis.


Subject(s)
Myelopoiesis , Sterol Esterase/physiology , Animals , Bone Marrow Cells/cytology , CD11b Antigen/biosynthesis , Cell Proliferation , Granulocytes/cytology , Homeostasis , Macrophages/cytology , Male , Mice , Mice, Transgenic , Stem Cells/cytology , Sterol Esterase/chemistry , T-Lymphocytes/cytology
7.
Blood ; 114(12): 2401-10, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19633199

ABSTRACT

Members of the tristetraprolin family of tandem CCCH finger proteins can bind to AU-rich elements in the 3'-untranslated region of mRNAs, leading to their deadenylation and subsequent degradation. Partial deficiency of 1 of the 4 mouse tristetraprolin family members, Zfp36l2, resulted in complete female infertility because of early embryo death. We have now generated mice completely deficient in the ZFP36L2 protein. Homozygous Zfp36l2 knockout (KO) mice died within approximately 2 weeks of birth, apparently from intestinal or other hemorrhage. Analysis of peripheral blood from KO mice showed a decrease in red and white cells, hemoglobin, hematocrit, and platelets. Yolk sacs from embryonic day 11.5 (E11.5) Zfp36l2 KO mice and fetal livers from E14.5 KO mice gave rise to markedly reduced numbers of definitive multilineage and lineage-committed hematopoietic progenitors. Competitive reconstitution experiments demonstrated that Zfp36l2 KO fetal liver hematopoietic stem cells were unable to adequately reconstitute the hematopoietic system of lethally irradiated recipients. These data establish Zfp36l2 as a critical modulator of definitive hematopoiesis and suggest a novel regulatory pathway involving control of mRNA stability in the life cycle of hematopoietic stem and progenitor cells.


Subject(s)
Embryo, Mammalian/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/pathology , Pancytopenia/genetics , RNA Stability/genetics , RNA-Binding Proteins/genetics , Tristetraprolin/physiology , Animals , Blotting, Northern , Blotting, Southern , Bone Marrow/metabolism , Embryo, Mammalian/cytology , Fetus/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunoenzyme Techniques , In Situ Hybridization , Liver/cytology , Liver/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Pancytopenia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/metabolism , Tissue Distribution
8.
Stem Cells Dev ; 14(1): 44-54, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15725743

ABSTRACT

Hematopoietic and endothelial cells may be derived from a common precursor cell (hemangioblast) during embryogenesis; however, some evidence suggests that hematopoietic cells may emerge from endothelial cells. The onset of definitive hematopoiesis at E8.25 in the murine embryo is marked by high-level CD41 expression. We questioned whether these hematopoietic cells were derived directly from mesoderm cells or emerged from endothelium. At 8.25 days post coitus (dpc), CD41 was coexpressed with CD31, CD34, and Flk1 in some intraluminal round cells that appeared to arise from flattened endothelial cells lining yolk sac capillary vessels. Cell-sorting studies revealed that all subpopulations of cells expressing CD41 possessed hematopoietic activity. Surprisingly, Tie2(+)Flk1(+) cells, a phenotype enriched in adult endothelial progenitors, also displayed some hematopoietic progenitor activity in vitro, but this activity was restricted to the CD41(+) fraction; only endothelial cells were derived from freshly isolated Tie2 (+)Flk1(bright) CD41() cells. Tie2(+)Flk1(dim)CD41() 8.25-dpc yolk sac cells devoid of hematopoietic progenitor activity gave rise to endothelial-like capillary networks in vitro and differentiated upon co-culture with OP9 stromal cells into definitive hematopoietic progenitors. These results demonstrate that CD41-expressing definitive hematopoietic cells appear to arise from endothelial cells lining nascent capillaries in vivo.


Subject(s)
Blood Cells/cytology , Cell Lineage , Endothelial Cells/cytology , Platelet Membrane Glycoprotein IIb , Stem Cells/cytology , Yolk Sac/cytology , Animals , Cells, Cultured , Hematopoiesis , Immunophenotyping , Mice , Mice, Inbred C57BL
9.
Exp Hematol ; 32(12): 1226-37, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15588947

ABSTRACT

OBJECTIVE: Murine hematopoietic stem cells (HSC) reside primarily in bone marrow but freely circulate throughout the systemic circulation with retention of transplantable hematopoietic repopulating ability. The mechanisms maintaining HSC potential during systemic circulation remain elusive. We hypothesized that vascular endothelial cells (EC) play an important role in maintaining circulating HSC repopulating ability. METHODS: Using Tie2-green fluorescence protein transgenic mice, we have isolated primary EC populations derived from several nonhematopoietic organs and cocultured bone marrow Sca1+c-Kit+lin- cells for 7 days in the presence or absence of growth factors. RESULTS: All cocultures promoted the growth of hematopoietic progenitor cells at day 7 of coculture in the presence of added growth factors. Compared to fresh sorted cells, brain and heart EC monolayers significantly increased, lung and liver EC monolayers maintained, and kidney EC monolayer markedly decreased the number of colony-forming unit-spleen day-8 colonies in the 7-day cocultures. HSC competitive repopulating unit activity was maintained during the heart and liver EC 7-day cocultures but was lost in the kidney EC coculture in vitro. CONCLUSION: These results demonstrate that some but not all primary EC isolated from nonhematopoietic organs support HSC function ex vivo.


Subject(s)
Bone Marrow/physiology , Endothelial Cells/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Animals , Antigens, Ly/metabolism , Brain/cytology , Cell Separation , Cells, Cultured , Coculture Techniques , Colony-Forming Units Assay , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Kidney/cytology , Liver/cytology , Lung/cytology , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Myocardium/cytology , Proto-Oncogene Proteins c-kit/metabolism
10.
Blood ; 102(13): 4345-53, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-12893772

ABSTRACT

The embryonic origin and development of hematopoietic and endothelial cells is highly interdependent. We hypothesized that primary endothelial cells from murine yolk sac and para-aortic splanchnopleura (P-Sp) may possess the capacity to expand hematopoietic stem cells (HSCs) and progenitor cells ex vivo. Using Tie2-GFP transgenic mice in combination with fluorochrome-conjugated monoclonal antibodies to vascular endothelial growth factor receptor-2 (Flk1) and CD41, we have successfully isolated pure populations of primary endothelial cells from 9.5-days after coitus (dpc) yolk sac and P-Sp. Adult murine bone marrow Sca-1+c-Kit+lin- cells were cocultured with yolk sac or P-Sp Tie2-GFP+Flk-1+CD41- endothelial cell monolayers for 7 days and the total number of nonadherent cells increased 47- and 295-fold, respectively, and hematopoietic progenitor counts increased 9.4- and 11.4-fold, respectively. Both the yolk sac and P-Sp endothelial cell cocultures facilitated long-term (> 6 months) HSC competitive repopulating ability (2.8- to 9.8-fold increases, respectively). These data suggest that 9.5-dpc yolk sac- and P-Sp-derived primary Tie2-GFP+Flk-1+CD41- endothelial cells possess the capacity to expand adult bone marrow hematopoietic progenitor cell and HSC repopulating ability ex vivo.


Subject(s)
Embryonic Structures/cytology , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Yolk Sac/cytology , Animals , Cell Differentiation , Cells, Cultured , Coculture Techniques , Green Fluorescent Proteins , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, TIE-2 , Recombinant Fusion Proteins/physiology
11.
Development ; 130(18): 4393-403, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12900455

ABSTRACT

The platelet glycoprotein IIb (alpha(IIb); CD41) constitutes the alpha subunit of a highly expressed platelet surface integrin protein. We demonstrate that CD41 serves as the earliest marker of primitive erythroid progenitor cells in the embryonic day 7 (E7.0) yolk sac and high-level expression identifies essentially all E8.25 yolk sac definitive hematopoietic progenitors. Some definitive hematopoietic progenitor cells in the fetal liver and bone marrow also express CD41. Hematopoietic stem cell competitive repopulating ability is present in CD41(dim) and CD41(lo/-) cells isolated from bone marrow and fetal liver cells, however, activity is enriched in the CD41(lo/-) cells. CD41(bright) yolk sac definitive progenitor cells co-express CD61 and bind fibrinogen, demonstrating receptor function. Thus, CD41 expression marks the onset of primitive and definitive hematopoiesis in the murine embryo and persists as a marker of some stem and progenitor cell populations in the fetal liver and adult marrow, suggesting novel roles for this integrin.


Subject(s)
Embryo, Mammalian/physiology , Erythroid Precursor Cells/metabolism , Hematopoiesis/physiology , Platelet Membrane Glycoprotein IIb/metabolism , Animals , B-Lymphocytes/metabolism , Biomarkers , Bone Marrow Cells/metabolism , Cell Lineage , Cell Transplantation , Embryo, Mammalian/anatomy & histology , Erythroid Precursor Cells/transplantation , Female , Fibrinogen/metabolism , Granulocytes/metabolism , Hemoglobins/genetics , Hemoglobins/metabolism , Integrin beta3/metabolism , Leukocyte Common Antigens/metabolism , Liver/cytology , Liver/embryology , Liver/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , T-Lymphocytes/metabolism , Transplantation Chimera , Vascular Endothelial Growth Factor Receptor-2/metabolism , Yolk Sac/cytology
12.
Blood ; 102(7): 2428-35, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-12791650

ABSTRACT

The first hematopoietic and endothelial progenitors are derived from a common embryonic precursor termed the hemangioblast. The genetic cascades that regulate the differentiation of the hemangioblast to hematopoietic and endothelial cells are largely unknown. In general, much of embryonic development is coordinately regulated by temporal and spatial expression of transcription factors, such as the Homeobox (Hox) gene family. We and others isolated a divergent homeobox gene termed Hex (or Prh) that is preferentially expressed in hematopoietic and endothelial cells. Using in vitro Hex-/- embryonic stem (ES) cell differentiation, in vivo yolk sac hematopoietic progenitor assays, and chimeric mouse analysis, we found that Hex is required for differentiation of the hemangioblast to definitive embryonic hematopoietic progenitors and to a lesser extent endothelial cells. Therefore, Hex is a novel regulator of hemangioblast differentiation to hematopoietic and endothelial cells.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Chimera , Endothelium/cytology , Fetus , Gene Expression Regulation, Developmental , Mice , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Transcription Factors , Up-Regulation/genetics , Yolk Sac/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...