Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Hepatology ; 56(3): 1086-96, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22488442

ABSTRACT

UNLABELLED: Although the vasculogenic potential of circulating and cord blood (CB)-derived endothelial colony-forming cells (ECFC) has been demonstrated in vitro and in vivo, little is known about the inherent biologic ability of these cells to home to different organs and contribute to tissue-specific cell populations. Here we used a fetal sheep model of in utero transplantation to investigate and compare the intrinsic ability of human CB-derived ECFC to migrate to the liver and to the intestine, and to define ECFC's intrinsic ability to integrate and contribute to the cytoarchitecture of these same organs. ECFCs were transplanted by an intraperitoneal or intrahepatic route (IH) into fetal sheep at concentrations ranging from 1.1-2.6 × 10(6) cells/fetus. Recipients were evaluated at 85 days posttransplant for donor (human) cells using flow cytometry and confocal microscopy. We found that, regardless of the route of injection, and despite the IH delivery of ECFC, the overall liver engraftment was low, but a significant percentage of cells were located in the perivascular regions and retained the expression of hallmark endothelial makers. By contrast, ECFC migrated preferentially to the intestinal crypt region and contributed significantly to the myofibroblast population. Furthermore, ECFC expressing CD133 and CD117 lodged in areas where endogenous cells expressed those same phenotypes. CONCLUSION: ECFC inherently constitute a potential source of cells for the treatment of intestinal diseases, but strategies to increase the numbers of ECFC persisting within the hepatic parenchyma are needed in order to enhance ECFC therapeutic potential for this organ.


Subject(s)
Cell Movement , Endothelial Cells/physiology , Fetal Blood , Intestines/cytology , Liver/cytology , Animals , Endothelial Cells/transplantation , Humans , Sheep
2.
Radiat Res ; 176(2): 208-16, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21663393

ABSTRACT

Endothelial colony-forming cells (ECFCs) are endothelial progenitor cells that circulate at low concentration in human umbilical cord and adult peripheral blood and are largely resident in blood vessels. ECFCs not only appear to be critical for normal vascular homeostasis and repair but may also contribute to tumor angiogenesis and response to therapy. To begin to characterize the potential role of ECFCs during the treatment of tumors in children and adults with radiation, we characterized the X-ray sensitivity of cord and adult blood-derived ECFCs. We found both cord blood and adult ECFCs to be highly radiation sensitive (3 Gy resulted in >90% killing without induction of apoptosis). The X-ray survival curves suggested reduced potential for repair capacity, but X-ray fractionation studies demonstrated that all the ECFCs exhibited repair when the radiation was fractionated. Finally, the mechanisms of X-ray-induced cell death for cord blood and adult ECFCs were different at low and high dose. At low dose, all ECFCs appear to die by mitotic death/catastrophe. However, at high radiation doses (≥ 10 Gy) cord blood ECFCs underwent p53 stabilization and Bax-dependent apoptosis as well as p21-dependent G1 and G2/M cell cycle checkpoints. By contrast, after 10 Gy adult ECFCs undergo only large-scale radiation-induced senescence, which is a cellular phenotype linked to premature development of atherosclerosis and vasculopathies. These data demonstrate that the ECFC response to radiation is dose-dependent and developmentally regulated and may provide potential mechanistic insight into their role in tumor and normal tissue response after ionizing radiation treatment.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/radiation effects , Apoptosis/radiation effects , Endothelial Cells/cytology , Fetal Blood/cytology , Adult , Animals , Cell Cycle/radiation effects , Cell Separation , Dose-Response Relationship, Drug , Humans , Mice , Radiation Tolerance/radiation effects , Time Factors , X-Rays/adverse effects
3.
Cytometry A ; 77(9): 831-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20803735

ABSTRACT

Defining whether human circulating proangiogenic cells represent a subset of the hematopoietic system and express CD45 or are hematopoietic derivatives that do not express CD45 (and are called endothelial progenitor cells) remains controversial. We have previously developed a polychromatic flow cytometry (PFC) protocol to isolate subsets of hematopoietic cells and we now identify the circulating pool of CD34(+)CD45(dim) cells representing functional circulating hematopoietic stem and progenitor cells (CHSPCs) that can be separated on the basis of AC133 expression and report that the AC133(+) subset of the CHSPCs enhances the growth of tumor blood vessels in vivo in immunodeficient mice. In addition, the ratio of AC133(+) proangiogenic CHSPCs to AC133(-) nonangiogenic CHSPCs unambiguously correlates with the severity of the clinical state of patients with peripheral arterial disease. In sum, a PFC protocol validated via in vitro and in vivo analyses, can be used to interrogate the roles of human hematopoietic elements in the growth and maintenance of the vasculature.


Subject(s)
Flow Cytometry/methods , Hematopoietic Stem Cells/cytology , Peripheral Arterial Disease/pathology , Staining and Labeling/methods , AC133 Antigen , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/analysis , Antigens, CD34/analysis , Female , Glycoproteins/analysis , Humans , Infant, Newborn , Leukocyte Common Antigens/analysis , Male , Mice , Mice, SCID , Middle Aged , Monocytes/cytology , Neovascularization, Pathologic/pathology , Peptides/analysis , Severity of Illness Index , Young Adult
4.
J Pediatr ; 157(4): 540-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20542287

ABSTRACT

OBJECTIVE: To evaluate whether counts of circulating colony forming unit-endothelial cells (CFU-ECs), cells co-expressing CD34, CD133, and CD31 (CD34+CD133+CD31+), and CD34+CD45- cells are altered in adolescents with type 1 diabetes and if the changes in counts correlate with endothelial dysfunction. STUDY DESIGN: Adolescents with diabetes (ages 18 to 22 years) and race- and sex-matched control subjects were studied. We assessed circulating CFU-ECs, using colony assays, and CD34+CD133+CD31+ and CD34+CD45- cells, using poly-chromatic flow cytometry. CFU-ECs and CD34+CD133+CD31+ are hematopoietic-derived progenitors that inversely correlate with cardiovascular risk in adults. CD34+CD45- cells are enriched for endothelial cells with robust vasculogenic potential. Vascular reactivity was tested by laser Doppler iontophoresis. RESULTS: Subjects with diabetes had lower CD34+CD133+CD31+ cells, a trend toward reduced CFU-ECs, and increased CD34+CD45- cells compared with control subjects. Endothelium-dependent vasodilation was impaired in subjects with diabetes, which correlated with reductions in circulating CD34+CD133+CD31+ cells. CONCLUSIONS: Long-term sequelae of type 1 diabetes include vasculopathies. Endothelial progenitor cells promote vascular health by facilitating endothelial integrity and function. Lower CD34+CD133+CD31+ cells may be a harbinger of future macrovascular disease risk. Higher circulating CD34+CD45- cells may reflect ongoing endothelial damage. These cells are potential biomarkers to guide therapeutic interventions to enhance endothelial function and to prevent progression to overt vascular disease.


Subject(s)
Antigens, CD/immunology , Biomarkers/blood , Diabetes Mellitus, Type 1/physiopathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Adolescent , Blood Flow Velocity , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/immunology , Endothelium, Vascular/immunology , Female , Humans , Male , Vasodilation/physiology , Young Adult
5.
Am J Physiol Heart Circ Physiol ; 296(5): H1675-82, 2009 May.
Article in English | MEDLINE | ID: mdl-19252096

ABSTRACT

Cell therapy with endothelial progenitor cells (EPCs) is an emerging therapeutic option to promote angiogenesis or endothelial repair. Although the release of angiogenic paracrine factors is known to contribute to their therapeutic effect, little is known about their release of proinflammatory factors and expression of proinflammatory adhesion molecules. "Early" EPCs and "late" EPCs were isolated from human peripheral blood and their release of chemokines and thromboinflammatory mediators as well as their expression of the proinflammatory adhesion molecules was assessed at baseline and with stimulation. The effect of simvastatin on monocyte chemoattractant protein-1 (MCP-1) secretion by late EPCs from patients with vascular disease was also evaluated. All groups of EPCs released chemokines and thromboinflammatory mediators. Early EPCs primarily released thromboinflammatory mediators such as tissue factor (0.5 +/- 0.1 ng/million cells, P < 0.05), whereas adult late EPCs primarily released chemokines such as MCP-1 (287 +/- 98 ng/million cells, P < 0.05). Stimulation with tumor necrosis factor (TNF)-alpha augmented the expression of proinflammatory adhesion molecules and paracrine factors by all EPC subtypes. The release of MCP-1 by late EPCs was markedly reduced by simvastatin treatment of the cells. All EPC subtypes expressed proinflammatory paracrine factors and adhesion molecules involved in atherosclerosis. Future clinical studies should therefore not only assess the efficacy of EPCs but also monitor inflammatory activation following EPC transplantation in patients. Pharmacological modulation of EPCs before and after transplantation may represent a novel approach to improve their safety.


Subject(s)
Adult Stem Cells/immunology , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Endothelial Cells/immunology , Fetal Stem Cells/immunology , Inflammation Mediators/metabolism , Paracrine Communication , Adult , Adult Stem Cells/drug effects , Cell Proliferation , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Endothelial Cells/drug effects , Fetal Stem Cells/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/metabolism , Leukocyte Common Antigens/metabolism , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Simvastatin/pharmacology , Thromboplastin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
6.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2C.1, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18770637

ABSTRACT

Circulating endothelial progenitor cells (EPCs) in adult human peripheral blood were originally identified in 1997 by Asahara et al., which challenged the paradigm that vasculogenesis is a process restricted to embryonic development. Since their original identification, EPCs have been extensively studied as biomarkers to assess the risk of cardiovascular disease in human subjects and as a potential cell therapeutic for vascular regeneration. Endothelial colony-forming cells (ECFCs), which are a subtype of EPCs, were recently identified from circulating adult and human umbilical cord blood. In contrast to other types of EPCs, which display various monocyte/macrophage phenotypes and functions, ECFCs are characterized by robust proliferative potential, secondary and tertiary colony formation upon replating, and de novo blood vessel formation in vivo when transplanted into immunodeficient mice. In this unit, we describe detailed methodologies for isolation and characterization of ECFCs from both human peripheral and umbilical cord blood.


Subject(s)
Blood Cells/cytology , Cell Separation/methods , Endothelial Cells/cytology , Stem Cells/cytology , Adipose Tissue/cytology , Animals , Antigens, Surface/metabolism , Cell Proliferation , Cell Transplantation , Clone Cells , Collagen/metabolism , Colony-Forming Units Assay , Cryopreservation , Drug Combinations , Humans , Immunohistochemistry , Laminin/metabolism , Lipoproteins, LDL/metabolism , Mice , Organ Specificity , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proteoglycans/metabolism , Stromal Cells/cytology , Umbilical Cord/cytology
7.
Hum Mol Genet ; 17(15): 2336-44, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18442999

ABSTRACT

Neurofibromatosis type I (NF1) is a genetic disorder caused by mutations in the NF1 tumor suppressor gene. Neurofibromin is encoded by NF1 and functions as a negative regulator of Ras activity. Somatic mutations in the residual normal NF1 allele within cancers of NF1 patients is consistent with NF1 functioning as a tumor-suppressor. However, the prevalent non-malignant manifestations of NF1, including learning and bone disorders emphasize the importance of dissecting the cellular and biochemical effects of NF1 haploinsufficiency in multiple cell lineages. One of the least studied complications of NF1 involves cardiovascular disorders, including arterial occlusions that result in cerebral and visceral infarcts. NF1 vasculopathy is characterized by vascular smooth muscle cell (VSMC) accumulation in the intima area of vessels resulting in lumen occlusion. We recently showed that Nf1 haploinsufficiency increases VSMC proliferation and migration via hyperactivation of the Ras-Erk pathway, which is a signaling axis directly linked to neointima formation in diverse animal models of vasculopathy. Given this observation, we tested whether heterozygosity of Nf1 would lead to vaso-occlusive disease in genetically engineered mice in vivo. Strikingly, Nf1+/- mice have increased neointima formation, excessive vessel wall cell proliferation and Erk activation after vascular injury in vivo. Further, this effect is directly dependent on a Gleevec sensitive molecular pathway. Therefore, these studies establish an Nf1 model of vasculopathy, which mirrors features of human NF1 vaso-occlusive disease, identifies a potential therapeutic target and provides a platform to further dissect the effect of Nf1 haploinsufficiency in cardiovascular disease.


Subject(s)
Arterial Occlusive Diseases/genetics , Cardiovascular Diseases/genetics , Cerebrovascular Disorders/genetics , Drug Resistance, Neoplasm/genetics , Genes, Neurofibromatosis 1 , Neurofibromatosis 1/complications , Animals , Antineoplastic Agents/pharmacology , Arterial Occlusive Diseases/etiology , Arterial Occlusive Diseases/pathology , Benzamides , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cell Proliferation , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/pathology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Imatinib Mesylate , Mice , Mice, Mutant Strains , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Tunica Intima/drug effects , Tunica Intima/pathology
8.
Pediatr Res ; 64(1): 68-73, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18360305

ABSTRACT

Endothelial progenitor cells (EPCs) are used for angiogenic therapies and as biomarkers of cardiovascular disease. Human umbilical cord blood (UCB) is a rich source of endothelial colony forming cells (ECFCs), which are EPCs with robust proliferative potential that may be useful for clinical vascular regeneration. Previous studies show that hematopoietic progenitor cells are increased in premature UCB compared with term controls. Based on this paradigm, we hypothesized that premature UCB would be an enriched source of ECFCs. Thirty-nine UCB samples were obtained from premature infants (24-37 wk gestational age (GA)) and term controls. ECFC colonies were enumerated, clonally isolated, and identified by expression of endothelial cell surface antigens and functional analysis. GA of 33-36 wk UCB yielded predominantly ECFC colonies at equivalent numbers to term infants. UCB from 24 to 28 wk GA infants had significantly fewer ECFCs. Surprisingly, 24-28 wk GA UCB yielded predominantly mesenchymal stem cell (MSC) colonies, capable of differentiating into adipocytes, chondrocytes, and osteocytes. MSCs were rarely identified in 37-40 wk GA UCB. These studies demonstrate that circulating MSCs and ECFCs appear at different GA in the human UCB, and that 24-28 wk GA UCB may be a novel source of MSCs for therapeutic use in human diseases.


Subject(s)
Endothelial Cells/physiology , Fetal Blood/cytology , Fetal Stem Cells/physiology , Mesenchymal Stem Cells/physiology , Adipocytes/physiology , Antigens, CD/analysis , Cell Differentiation , Cell Proliferation , Cell Shape , Cells, Cultured , Chondrocytes/physiology , Endothelial Cells/immunology , Fetal Stem Cells/immunology , Gestational Age , Humans , Infant, Newborn , Infant, Premature , Mesenchymal Stem Cells/immunology , Neovascularization, Physiologic , Osteocytes/physiology , Phenotype
9.
Diabetes ; 57(3): 724-31, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18086900

ABSTRACT

OBJECTIVE: Emerging data demonstrate that maternal diabetes has long-term health consequences for offspring, including the development of hypertension. In adults, circulating endothelial progenitor cells (EPCs) participate in vascular repair, and EPC numbers and function inversely correlate with the risk of developing vascular disease. Therefore, our objectives were to determine whether hyperglycemia or exposure to a diabetic intrauterine environment alters EPC function. RESEARCH DESIGN AND METHODS: We used well-established clonogenic endothelial colony-forming cell (ECFC) assays and murine transplantation experiments to examine human vasculogenesis. RESULTS: Both in vitro hyperglycemia and a diabetic intrauterine environment reduced ECFC colony formation, self-renewal capacity, and capillary-like tube formation in matrigel. This cellular phenotype was linked to premature senescence and reduced proliferation. Further, cord blood ECFCs from diabetic pregnancies formed fewer chimeric vessels de novo after transplantation into immunodeficient mice compared with neonatal ECFCs harvested from uncomplicated pregnancies. CONCLUSIONS; Collectively, these data demonstrate that hyperglycemia or exposure to a diabetic intrauterine environment diminishes neonatal ECFC function both in vitro and in vivo, providing potential mechanistic insights into the long-term cardiovascular complications observed in newborns of diabetic pregnancies.


Subject(s)
Diabetes, Gestational , Endothelial Cells/drug effects , Hyperglycemia , Stem Cells/drug effects , Adult , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Female , Glucose/pharmacology , Humans , Infant, Newborn , Pregnancy
10.
FEBS Lett ; 581(25): 4927-31, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17904123

ABSTRACT

Recent studies have highlighted the importance of paracrine growth factors as mediators of pro-angiogenic effects by endothelial progenitor cells (EPCs), but little is known about the release of lipid-based factors like endocannabinoids by EPCs. In the current study, the release of the endocannabinoids anandamide and 2-arachidonoylglycerol by distinct human EPC sub-types was measured using HPLC/tandem mass-spectrometry. Anandamide release was highest by adult blood colony-forming EPCs at baseline and they also demonstrated increased 2-arachidonoylglycerol release with TNF-alpha stimulation. Treatment of mature endothelial cells with endocannabinoids significantly reduced the induction of the pro-inflammatory adhesion molecule CD106 (VCAM-1) by TNF-alpha.


Subject(s)
Arachidonic Acids/biosynthesis , Cannabinoid Receptor Modulators/biosynthesis , Endocannabinoids , Endothelium, Vascular/cytology , Glycerides/biosynthesis , Stem Cells/metabolism , Arachidonic Acids/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Glycerides/pharmacology , Humans , Polyunsaturated Alkamides/pharmacology , Stem Cells/classification , Stem Cells/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
11.
Exp Hematol ; 35(7): 1109-18, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17588480

ABSTRACT

OBJECTIVE: Endothelial progenitor cells (EPCs) are used for angiogenic therapies or as biomarkers to assess cardiovascular disease risk. However, there is no uniform definition of an EPC, which confounds EPC studies. EPCs are widely described as cells that coexpress the cell-surface antigens CD34, AC133, and vascular endothelial growth factor receptor-2 (VEGFR-2). These antigens are also expressed on primitive hematopoietic progenitor cells (HPCs). Remarkably, despite their original identification, CD34+AC133+VEGFR-2+ cells have never been isolated and simultaneously plated in hematopoietic and endothelial cell (EC) clonogenic assays to assess the identity of their clonal progeny, which are presumably the cellular participants in vascular regeneration. METHODS: CD34+AC133+VEGFR-2+ cells were isolated from human umbilical cord blood (CB) or granulocyte colony-stimulating factor-mobilized peripheral blood and assayed for either EPCs or HPCs. RESULTS: CD34+AC133+VEGFR-2+ cells did not form EPCs and were devoid of vessel forming activity. However, CD34+AC133+VEGFR-2+ cells formed HPCs and expressed the hematopoietic lineage-specific antigen, CD45. We next tested whether EPCs could be separated from HPCs by immunoselection for CD34 and CD45. CD34+CD45+ cells formed HPCs but not EPCs, while CD34+CD45- cells formed EPCs but not HPCs. CONCLUSIONS: Therefore, CD34+AC133+VEGFR-2+ cells are HPCs that do not yield EC progeny, and the biological mechanism for their correlation with cardiovascular disease needs to be reexamined.


Subject(s)
Antigens, CD34/analysis , Antigens, CD/analysis , Endothelial Cells/cytology , Glycoproteins/analysis , Hematopoietic Stem Cells/cytology , Peptides/analysis , Stem Cells/cytology , Vascular Endothelial Growth Factor Receptor-2/analysis , AC133 Antigen , Adult , Cell Separation , Cells, Cultured , Female , Humans , Leukocyte Common Antigens/analysis , Male , Middle Aged
12.
Stem Cells ; 25(2): 297-304, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17023514

ABSTRACT

Endothelial progenitor cells (EPCs) circulate in the peripheral blood and reside in blood vessel walls. A hierarchy of EPCs exists where progenitors can be discriminated based on their clonogenic potential. EPCs are exposed to oxidative stress during vascular injury as residents of blood vessel walls or as circulating cells homing to sites of neovascularization. Given the links between oxidative injury, endothelial cell dysfunction, and vascular disease, we tested whether EPCs were sensitive to oxidative stress using newly developed clonogenic assays. Strikingly, in contrast to previous reports, we demonstrate that the most proliferative EPCs (high proliferative potential-endothelial colony-forming cells and low proliferative potential-endothelial colony-forming cells) had decreased clonogenic capacity after oxidant treatment. In addition, EPCs exhibited increased apoptosis and diminished tube-forming ability in vitro and in vivo in response to oxidative stress, which was directly linked to activation of a redox-dependent stress-induced kinase pathway. Thus, this study provides novel insights into the effect of oxidative stress on EPCs. Furthermore, this report outlines a framework for understanding how oxidative injury leads to vascular disease and potentially limits the efficacy of transplantation of EPCs into ischemic tissues enriched for reactive oxygen species and oxidized metabolites.


Subject(s)
Clone Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Oxidative Stress , Stem Cells/cytology , Stem Cells/metabolism , Adult , Animals , Apoptosis/drug effects , Catalysis/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Colony-Forming Units Assay , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Female , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/enzymology , Humans , Hydrogen Peroxide/pharmacology , Infant, Newborn , MAP Kinase Kinase Kinase 5/metabolism , Male , Mice , Mice, SCID , Neovascularization, Physiologic/drug effects , Oxidants/pharmacology , Oxidative Stress/drug effects , Rats , Stem Cells/drug effects , Stem Cells/enzymology
13.
Blood ; 109(5): 1801-9, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17053059

ABSTRACT

The limited vessel-forming capacity of infused endothelial progenitor cells (EPCs) into patients with cardiovascular dysfunction may be related to a misunderstanding of the biologic potential of the cells. EPCs are generally identified by cell surface antigen expression or counting in a commercially available kit that identifies "endothelial cell colony-forming units" (CFU-ECs). However, the origin, proliferative potential, and differentiation capacity of CFU-ECs is controversial. In contrast, other EPCs with blood vessel-forming ability, termed endothelial colony-forming cells (ECFCs), have been isolated from human peripheral blood. We compared the function of CFU-ECs and ECFCs and determined that CFU-ECs are derived from the hematopoietic system using progenitor assays, and analysis of donor cells from polycythemia vera patients harboring a Janus kinase 2 V617F mutation in hematopoietic stem cell clones. Further, CFU-ECs possess myeloid progenitor cell activity, differentiate into phagocytic macrophages, and fail to form perfused vessels in vivo. In contrast, ECFCs are clonally distinct from CFU-ECs, display robust proliferative potential, and form perfused vessels in vivo. Thus, these studies establish that CFU-ECs are not EPCs and the role of these cells in angiogenesis must be re-examined prior to further clinical trials, whereas ECFCs may serve as a potential therapy for vascular regeneration.


Subject(s)
Cell Differentiation , Clone Cells/cytology , Endothelial Cells/cytology , Hematopoietic System/cytology , Stem Cells/cytology , Adult , Animals , Antigens/metabolism , Biomarkers , Cell Transplantation , Cells, Cultured , Colony-Forming Units Assay , Endothelial Cells/metabolism , Female , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Macrophages/cytology , Male , Mice , Middle Aged , Monocytes/cytology , Mutation/genetics
14.
Methods Enzymol ; 419: 439-73, 2006.
Article in English | MEDLINE | ID: mdl-17141066

ABSTRACT

Cord blood has served as a source of hematopoietic stem and progenitor cells for successful repopulation of the blood cell system in patients with malignant and nonmalignant disorders. It was information on these rare immature cells in cord blood that led to the first use of cord blood for transplantation. Further information on these cells and how they can be manipulated both in vitro and in vivo will likely enhance the utility and broadness of applicability of cord blood for treatment of human disease. This chapter reviews information on the clinical and biological properties of hematopoietic stem and progenitor cells, as well as the biology of endothelial progenitor cells, and serves as a source for the methods used to detect and quantitate these important functional cells. Specifically, methods are presented for enumerating human cord blood myeloid progenitor cells, including granulocyte-macrophage (CFU-GM), erythroid (BFU-E), and multipotential (CFU-GEMM or CFU-Mix) progenitors, and their replating potential; hematopoietic stem cells, as assessed in vitro for long-term culture-initiating cells (LTC-ICs), cobblestone area-forming cells (CAFCs), and myeloid-lymphoid-initiating cells (ML-ICs), and as assessed in vivo for nonobese diabetic (NOD)/severe combined immunodeficient (SCID) mouse repopulating cells (SRCs); and high and low proliferative potential endothelial progenitor cells (EPCs).


Subject(s)
Fetal Blood/cytology , Fetal Stem Cells , Adult Stem Cells , Animals , Cell Culture Techniques/methods , Humans
15.
Hum Mol Genet ; 15(11): 1858-69, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16648142

ABSTRACT

Genetic inactivation of tumor suppressor genes initiates human cancers. However, interaction of accessory cells with the tumor-initiating cell within the microenvironment is often required for tumor progression. This paradigm is relevant to understanding neurofibroma development in neurofibromatosis type I patients. Somatic inactivation of the Nf1 tumor suppressor gene, which encodes neurofibromin, is necessary but not sufficient to initiate neurofibroma development. In contrast, neurofibromas occur with high penetrance in mice in which Nf1 is ablated in Schwann cells in the context of a heterozygous mutant (Nf1+/-) microenvironment. Neurofibromas are highly vascularized, and recent studies suggest that Nf1+/- mice have increased angiogenesis in vivo. However, the function of neurofibromin in human endothelial cells (ECs) and the biochemical mechanism by which neurofibromin regulates neoangiogenesis are not known. Utilizing Nf1+/- mice, primary human ECs and endothelial progenitor cells harvested from NF1 patients, we identified a discrete Ras effector pathway, which alters the proliferation and migration of neurofibromin-deficient ECs in response to neurofibroma-derived growth factors both in vitro and in vivo. Thus, these studies identify a unique biochemical pathway in Nf1+/- ECs as a potential therapeutic target in the neurofibroma microenvironment.


Subject(s)
Neurofibroma/pathology , Neurofibromin 1/genetics , Alleles , Animals , Collagen/chemistry , Drug Combinations , Endothelium, Vascular/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Growth Substances/metabolism , Humans , Laminin/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurofibromin 1/physiology , Proteoglycans/chemistry , Schwann Cells/metabolism , Signal Transduction
16.
Hum Mol Genet ; 15(11): 1921-30, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16644864

ABSTRACT

Neurofibromatosis type I (NF1) is a genetic disorder caused by mutations in the NF1 tumor suppressor gene. Neurofibromin is encoded by NF1 and functions as a negative regulator of Ras activity. NF1 patients develop renal artery stenosis and arterial occlusions resulting in cerebral and visceral infarcts. Further, NF1 patients develop vascular neurofibromas where tumor vessels are invested in a dense pericyte sheath. Although it is well established that aberrations in Ras signaling lead to human malignancies, emerging data generated in genetically engineered mouse models now implicate perturbations in the Ras signaling axis in vascular smooth muscular cells (VSMCs) as central to the initiation and progression of neointimal hyperplasia and arterial stenosis. Despite these observations, the function of neurofibromin in regulating VSMC function and how Ras signals are terminated in VSMCs is virtually unknown. Utilizing VSMCs harvested from Nf1+/- mice and primary human neurofibromin-deficient VSMCs, we identify a discrete Ras effector pathway, which is tightly regulated by neurofibromin to limit VSMC proliferation and migration. Thus, these studies identify neurofibromin as a novel regulator of Ras activity in VSMCs and provide a framework for understanding cardiovascular disease in NF1 patients and a mechanism by which Ras signals are attenuated for maintaining VSMC homeostasis in blood vessel walls.


Subject(s)
Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Neurofibromin 1/metabolism , Neurofibromin 1/physiology , ras Proteins/metabolism , Animals , Becaplermin , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis
17.
Blood ; 105(7): 2783-6, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15585655

ABSTRACT

Endothelial progenitor cells (EPCs) can be isolated from adult peripheral and umbilical cord blood and expanded exponentially ex vivo. In contrast, human umbilical vein endothelial cells (HUVECs) or human aortic endothelial cells (HAECs) derived from vessel walls are widely considered to be differentiated, mature endothelial cells (ECs). However, similar to adult- and cord blood-derived EPCs, HUVECs and HAECs derived from vessel walls can be passaged for at least 40 population doublings in vitro. Based on this paradox, we tested whether EPCs reside in HUVECs or HAECs utilizing a novel single cell deposition assay that discriminates EPCs based on their proliferative and clonogenic potential. We demonstrate that a complete hierarchy of EPCs can be identified in HUVECs and HAECs derived from vessel walls and discriminated by their clonogenic and proliferative potential. This study provides evidence that a diversity of EPCs exists in human vessels and provides a conceptual framework for determining both the origin and function of EPCs in maintaining vessel integrity.


Subject(s)
Aorta/cytology , Endothelium, Vascular/cytology , Stem Cells/cytology , Umbilical Veins/cytology , Age Factors , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured , Fetal Blood/cytology , Humans
18.
Blood ; 104(9): 2752-60, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15226175

ABSTRACT

Emerging evidence to support the use of endothelial progenitor cells (EPCs) for angiogenic therapies or as biomarkers to assess cardiovascular disease risk and progression is compelling. However, there is no uniform definition of an EPC, which makes interpretation of these studies difficult. Although hallmarks of stem and progenitor cells are their ability to proliferate and to give rise to functional progeny, EPCs are primarily defined by the expression of cell-surface antigens. Here, using adult peripheral and umbilical cord blood, we describe an approach that identifies a novel hierarchy of EPCs based on their clonogenic and proliferative potential, analogous to the hematopoietic cell system. In fact, some EPCs form replatable colonies when deposited at the single-cell level. Using this approach, we also identify a previously unrecognized population of EPCs in cord blood that can achieve at least 100 population doublings, replate into at least secondary and tertiary colonies, and retain high levels of telomerase activity. Thus, these studies describe a clonogenic method to define a hierarchy of EPCs based on their proliferative potential, and they identify a unique population of high proliferative potential-endothelial colony-forming cells (HPP-ECFCs) in human umbilical cord blood.


Subject(s)
Blood Cells/cytology , Endothelium, Vascular/cytology , Stem Cells/cytology , Cell Culture Techniques , Cell Proliferation , Clone Cells , Colony-Forming Units Assay , Endothelial Cells/cytology , Fetal Blood/cytology , Humans , Immunophenotyping , Stem Cells/classification , Telomerase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...