Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Melanoma Res ; 21(5): 405-16, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21900792

ABSTRACT

'Cancer stem cells' (CSCs) are tumor cells with stem cell properties hypothesized to be responsible for tumorigenesis, metastatis, and resistance to treatment, and have been identified in different tumors including cutaneous melanoma, using stem cell markers such as CD133. This study explored expression of CD133 and other putative stem cell markers in uveal melanoma. Eight uveal melanoma cell lines were subjected to flow-cytometric (fluorescence-activated cell sorting) analysis of CD133 and other stem cell markers. Eight paraffin-embedded tumors were analyzed by immunohistochemistry for CD133, Pax6, Musashi, nestin, Sox2, ABCB5, and CD68 expressions. Ocular, uveal melanoma, and hematopoietic stem cell distributions of C-terminal and N-terminal CD133 mRNA splice variants were compared by reverse-transcription PCR. Fluorescence-activated cell sorting analysis revealed a population of CD133-positive/nestin-positive cells in cell lines Mel270, OMM 2.3, and OMM2.5. All cell lines studied were positive for nestin, CXCR-4, CD44, and c-kit. Immunohistochemistry identified cells positive for CD133, Pax6, Musashi, nestin, Sox2, ABCB5, and CD68 predominantly at the invading tumor front. C-terminal primers interacting with CD133 splice variant s2 detected a novel variant lacking exon 27. Differential expression of CD133 splice variants was found in iris, ciliary body, retina, and retinal pigment epithelium/choroid as well as in uveal melanoma cell lines. mRNA for nestin, Sox2, and Musashi was present in all studied cell lines. Uveal melanoma such as cutaneous melanoma may therefore contain CSCs. Further experiments are needed to isolate stem cell marker-positive cells, to evaluate their functional properties and to explore therapeutical approaches to these putative CSCs in uveal melanoma.


Subject(s)
Antigens, CD/biosynthesis , Glycoproteins/biosynthesis , Melanoma/metabolism , Neoplastic Stem Cells/metabolism , Uveal Neoplasms/metabolism , AC133 Antigen , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Female , Glycoproteins/genetics , Humans , Immunohistochemistry , Intermediate Filament Proteins/biosynthesis , Male , Melanoma/genetics , Melanoma/pathology , Mice , Middle Aged , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/biosynthesis , Nestin , Peptides/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology
2.
PLoS One ; 6(1): e14486, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21245929

ABSTRACT

BACKGROUND: Successful treatment of acute radiation syndromes relies on immediate supportive care. In patients with limited hematopoietic recovery potential, hematopoietic stem cell (HSC) transplantation is the only curative treatment option. Because of time consuming donor search and uncertain outcome we propose MSC treatment as an alternative treatment for severely radiation-affected individuals. METHODS AND FINDINGS: Mouse mesenchymal stromal cells (mMSCs) were expanded from bone marrow, retrovirally labeled with eGFP (bulk cultures) and cloned. Bulk and five selected clonal mMSCs populations were characterized in vitro for their multilineage differentiation potential and phenotype showing no contamination with hematopoietic cells. Lethally irradiated recipients were i.v. transplanted with bulk or clonal mMSCs. We found a long-term survival of recipients with fast hematopoietic recovery after the transplantation of MSCs exclusively without support by HSCs. Quantitative PCR based chimerism analysis detected eGFP-positive donor cells in peripheral blood immediately after injection and in lungs within 24 hours. However, no donor cells in any investigated tissue remained long-term. Despite the rapidly disappearing donor cells, microarray and quantitative RT-PCR gene expression analysis in the bone marrow of MSC-transplanted animals displayed enhanced regenerative features characterized by (i) decreased proinflammatory, ECM formation and adhesion properties and (ii) boosted anti-inflammation, detoxification, cell cycle and anti-oxidative stress control as compared to HSC-transplanted animals. CONCLUSIONS: Our data revealed that systemically administered MSCs provoke a protective mechanism counteracting the inflammatory events and also supporting detoxification and stress management after radiation exposure. Further our results suggest that MSCs, their release of trophic factors and their HSC-niche modulating activity rescue endogenous hematopoiesis thereby serving as fast and effective first-line treatment to combat radiation-induced hematopoietic failure.


Subject(s)
Acute Radiation Syndrome/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Radiation Injuries, Experimental/therapy , Stromal Cells/cytology , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/pathology , Animals , Cell Adhesion , Cell Cycle , Cells, Cultured , Hematopoiesis , Humans , Inflammation , Mesenchymal Stem Cells/cytology , Mice , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation Protection/methods , Stromal Cells/radiation effects , Survival Rate , Treatment Outcome
3.
World J Gastroenterol ; 16(2): 217-24, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-20066741

ABSTRACT

AIM: To test the hypothesis that liver cirrhosis is associated with mobilization of hematopoietic progenitor cells. METHODS: Peripheral blood samples from 72 patients with liver cirrhosis of varying etiology were analyzed by flow cytometry. Identified progenitor cell subsets were immunoselected and used for functional assays in vitro. Plasma levels of stromal cell-derived factor-1 (SDF-1) were measured using an enzyme linked immunosorbent assay. RESULTS: Progenitor cells with a CD133(+)/CD45(+)/CD14(+) phenotype were observed in 61% of the patients. Between 1% and 26% of the peripheral blood mononuclear cells (MNCs) displayed this phenotype. Furthermore, a distinct population of c-kit(+) progenitor cells (between 1% and 38% of the MNCs) could be detected in 91% of the patients. Additionally, 18% of the patients showed a population of progenitor cells (between 1% and 68% of the MNCs) that was characterized by expression of breast cancer resistance protein-1. Further phenotypic analysis disclosed that the circulating precursors expressed CXC chemokine receptor 4, the receptor for SDF-1. In line with this finding, elevated plasma levels of SDF-1 were present in all patients and were found to correlate with the number of mobilized CD133(+) progenitor cells. CONCLUSION: These data indicate that in humans, liver cirrhosis leads to recruitment of various populations of hematopoietic progenitor cells that display markers of intrahepatic progenitor cells.


Subject(s)
Cell Movement/physiology , Hematopoietic Stem Cells/physiology , Liver Cirrhosis/physiopathology , AC133 Antigen , Adult , Antigens, CD/metabolism , Chemokine CXCL12/metabolism , Female , Glycoproteins/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Membrane Proteins/metabolism , Middle Aged , Peptides/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Receptors, CXCR4/metabolism , Young Adult
4.
Stem Cells Dev ; 16(5): 733-45, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17999595

ABSTRACT

The transmembrane protein CD133 is expressed on somatic stem cells of various adult human tissues. To investigate whether human corneal stroma also contains CD133-expressing cells and to analyze their functional features, stromal cells were isolated by collagenase digestion, immunophenotyped, and transferred to different culture systems to determine their stem cell properties as well as their differentiation potentials. For comparison, the embryonic keratocyte cell line EK1.Br, the dermal stromal cell line NHDF, and stromal cells of diseased corneas were studied. On average, 5.3% of the normal stromal cells expressed the stem cell marker CD133 and 3.6% co-expressed CD34. Expression of CD133 but not CD34 was also demonstrated for EK1.Br cells, whereas NHDF cells were negative for both markers. Further analysis of CD133(+) normal corneal cells revealed that a significant proportion displayed a monocytic phenotype with co-expression of CD45 and CD14. In diseased corneas, up to 26.8% of the stromal cells showed expression of CD133, and virtually all CD133(+) cells co-expressed CD14 but not CD45. Moreover, using a standard clonogenic assay, normal stromal cells had the capacity to form colonies of the macrophage lineage. These colonies could be further differentiated into lumican-expressing keratocytes. Our data suggest that the human corneal stroma harbors CD133(+) monocytic progenitor cells, which possess the potential to differentiate into the fibrocytic lineage. Thus, CD133(+) /CD45(+) /CD14(+) cells might represent stromal repair cells that differentiate into keratocytes via a CD133(+)/CD45()/CD14(+) intermediate stage. The findings from our study may shed new light on regenerative processes of the human corneal stroma.


Subject(s)
Corneal Stroma/cytology , Wound Healing , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Differentiation , Cell Line , Cell Separation , Clone Cells , Collagenases/metabolism , Colony-Forming Units Assay , Corneal Diseases/pathology , Dermis/cytology , Embryo, Mammalian/cytology , Fibroblasts/cytology , Flow Cytometry , Glycoproteins/metabolism , Hematopoietic System/cytology , Humans , Immunohistochemistry , Immunophenotyping , Limbus Corneae/cytology , Neoplasm Proteins/metabolism , Peptides/metabolism , Phenotype
5.
Am J Pathol ; 171(5): 1608-18, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17916596

ABSTRACT

Cancer-associated stromal fibroblasts (CAFs) are the main cellular constituents of reactive stroma in primary and metastatic cancer. We analyzed phenotypical characteristics of CAFs from human colorectal liver metastases (CLMs) and their role in inflammation and cancer progression. CAFs displayed a vimentin(+), alpha-smooth-muscle actin(+), and Thy-1(+) phenotype similar to resident portal-located liver fibroblasts (LFs). We demonstrated that CLMs are inflammatory sites showing stromal expression of interleukin-8 (IL-8), a chemokine related to invasion and angiogenesis. In vitro analyses revealed a striking induction of IL-8 expression in CAFs and LFs by tumor necrosis factor-alpha (TNF-alpha). The effect of TNF-alpha on CAFs is inhibited by the nuclear factor-kappaB inhibitor parthenolide. Conditioned medium of CAFs and LFs similarly stimulated the migration of DLD-1, Colo-678, HuH7 carcinoma cells, and human umbilical vein endothelial cells in vitro. Pretreatment of CAFs with TNF-alpha increased the chemotaxis of Colo-678 colon carcinoma cells by conditioned medium of CAFs; however, blockage of IL-8 activity showed no inhibitory effect. In conclusion, these data raise the possibility that the majority of CAFs in CLM originate from resident LFs. TNF-alpha-induced up-regulation of IL-8 via nuclear factor-kappaB in CAFs is an inflammatory pathway, potentially permissive for cancer invasion that may represent a novel therapeutic target.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Fibroblasts/pathology , Liver Neoplasms/metabolism , Cell Line, Tumor , Cells, Cultured , Chemotaxis , Colorectal Neoplasms/immunology , Endothelial Cells/physiology , Fibroblasts/metabolism , Humans , Inflammation/pathology , Interleukin-8/biosynthesis , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , NF-kappa B/antagonists & inhibitors , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Necrosis Factor-alpha/physiology , Up-Regulation
8.
Stem Cells Dev ; 16(2): 329-38, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17521243

ABSTRACT

Recent studies have shown that in response to vascular damage or ischemia, bone marrow-derived endothelial progenitor cells (EPCs) are recruited into the circulation. To investigate whether antihypertensive treatment has an influence on the number of circulating EPCs, patients with essential hypertension were treated either with the angiotensin receptor antagonist telmisartan, the calcium channel blocker nisoldipine, or their combination for 6 weeks. At baseline and after 3 and 6 weeks of treatment, EPCs were identified and quantified by fluorescence-activated cell sorting (FACS) analysis and by their capacity to generate colony-forming units of the endothelial lineage (CFU-EC) in a methylcellulose-based assay. During treatment, patients in the nisoldipine groups, but not in the telmisartan group, showed a significant mobilization of EPCs, which in part had the capacity to generate large-sized colonies comprising more than 1,000 cells. Moreover, a remarkable correlation between the number of CFU-EC and the number of circulating CD133(+)/CD34(+)/CD146(+) cells was observed, thereby providing strong evidence that cells with this phenotype represent functional EPCs. No correlation was found between the numbers of CFU-EC and the blood pressure levels at any time point during the treatment. Hence, nisoldipine-induced mobilization of EPCs might represent a novel mechanism by which this antihypertensive compound independently of its blood pressure-lowering effect contributes to vasoprotection in patients with essential hypertension.


Subject(s)
Antihypertensive Agents/therapeutic use , Endothelial Cells/physiology , Hypertension/drug therapy , Stem Cells/physiology , Adult , Aged , Aged, 80 and over , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antigens, CD/metabolism , Benzimidazoles/therapeutic use , Benzoates/therapeutic use , Blood Pressure/physiology , Calcium Channel Blockers/therapeutic use , Cells, Cultured , Colony-Forming Units Assay , Endothelial Cells/cytology , Female , Humans , Male , Middle Aged , Nisoldipine/therapeutic use , Phenotype , Stem Cells/cytology , Telmisartan
9.
Exp Cell Res ; 313(11): 2326-35, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17475245

ABSTRACT

Recent data suggest that angiogenesis plays an important role in the pathogenesis of valvular disease. However, the cellular mechanisms underlying this process remain unknown. This study aimed at identifying and characterizing the cellular components responsible for pathological neovascularization in calcific aortic valves (CAV). Immunohistochemical analysis of uncultured CAV tissues revealed that smooth muscle alpha-actin (alpha-SMA)-positive cells, which coexpressed Tie-2 and vascular endothelial growth factor receptor-2 (VEGFR-2), can be identified prior to the initiation of capillary-like tube formation. In a second step, leaflets of CAV and non-calcific aortic valves (NCAV) were cultured and the cells involved in capillary-like tube formation were isolated. The majority of these cells displayed the same phenotype as non-cultured cells identified in CAV tissues, i.e., expression of alpha-SMA, Tie-2, and VEGFR-2. In comparison to cells isolated from cultures of NCAV leaflets, these cells showed enhanced angiogenic activity as demonstrated by migration and tube assays. The coexpression of VEGFR-2 and Tie-2 together with alpha-SMA suggests both endothelial and mesenchymal properties of the angiogenically activated cells involved in valvular neovascularization. Hence, our findings might provide new insights into the process of pathological angiogenesis in cardiac valves.


Subject(s)
Aortic Valve Stenosis/pathology , Aortic Valve/pathology , Neovascularization, Pathologic/pathology , Actins/metabolism , Antigens, CD/analysis , Aortic Valve/chemistry , Aortic Valve/metabolism , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/metabolism , Biological Assay , Cells, Cultured , Chemotaxis , Female , Flow Cytometry , Humans , Male , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Organ Culture Techniques , Phenotype , Receptor, TIE-2/metabolism , Transcription, Genetic , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Free Radic Biol Med ; 42(4): 460-5, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17275677

ABSTRACT

Activation of leukocytes, in particular polymorphonuclear neutrophils (PMN), is considered an early event in unstable coronary disease. Upon activation PMN liberate myeloperoxidase (MPO), an enzyme which binds to the vessel wall and depletes vascular NO bioavailability. Using coronary balloon angioplasty as a trigger to provoke coronary plaque injury, we assessed the time course of neutrophil activation, local and peripheral levels of myeloperoxidase, and systemic vascular NO bioavailability in patients with stable coronary artery disease. Twenty-four patients with stable CAD were enrolled prior to undergoing percutaneous interventions (PCI, n=14) and diagnostic coronary angiography (n=10), respectively. Following angioplasty arterial MPO plasma levels increased (231.5+/-67.6 to 273.8+/-80.4 pg/mg protein; P<0.01) whereas MPO levels in the coronary sinus decreased (240.8+/-74.4 vs 205.4+/-60.1 pg/mg protein; P<0.01) in the absence of elevated serum markers for myocardial necrosis. Following PCI, patients revealed impaired vascular NO bioavailability as reflected by reduced brachial flow-mediated dilation (FMD; 6.25+/-3.03 to 4.90+/-2.70%; P<0.01), whereas FMD increased in the angiography group. Coronary plaque injury provokes rapid activation of PMN in the absence of myocardial necrosis; the coronary circulation emerges as a primary site for deposition of MPO following injury of the coronary vessel wall. Activation of PMN with release of MPO is not only restricted to the target site, but can be assessed systemically and may represent a critical mechanistic link for impaired systemic vascular NO bioavailability in patients suffering unstable coronary disease.


Subject(s)
Coronary Artery Disease/immunology , Neutrophil Activation , Aged , Biomarkers/metabolism , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Female , Humans , Middle Aged , Nitric Oxide/metabolism , Peroxidase/blood
11.
Methods Enzymol ; 419: 179-93, 2006.
Article in English | MEDLINE | ID: mdl-17141056

ABSTRACT

In the developing embryo, the hemangioblast, a mesodermal precursor, gives rise to hematopoietic and endothelial cells. Recent work has shown that during postnatal life, a subset of hematopoietic progenitor cells also displays this dual differentiation capacity and can function as endothelial progenitor cells that contribute to neovascularization. Thus, this subset might be useful for therapy of various hematopoietic and vascular diseases. Here, we describe a two-step culture system that results in the generation of endothelial and hematopoietic cells from adult progenitor cells with hemangioblastic potential. We have developed growth conditions that allow retroviral gene marking of the adult hemangioblast. This culture system is amenable for single-cell analyses at distinct stages of endothelial and hematopoietic differentiation from mobilized CD133+ progenitor cells.


Subject(s)
Cell Differentiation/physiology , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Mesoderm/cytology , Cell Culture Techniques/methods , Humans
12.
Development ; 133(8): 1543-51, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16524930

ABSTRACT

Here, we report the existence of endothelial precursor (EPC) and stem cells in a distinct zone of the vascular wall that are capable to differentiate into mature endothelial cells, hematopoietic and local immune cells, such as macrophages. This zone has been identified to be localized between smooth muscle and adventitial layer of human adult vascular wall. It predominantly contains CD34-positive (+) but CD31-negative (-) cells, which also express VEGFR2 and TIE2. Only few cells in this zone of the vascular wall are positive for CD45. In a ring assay using the fragments of human internal thoracic artery (HITA), we show here that the CD34+ cells of the HITA-wall form capillary sprouts ex vivo and are apparently recruited for capillary formation by tumor cells. New vessels formed by these vascular wall resident EPCs express markers for angiogenically activated endothelial cells, such as CEACAM1, and also for mature endothelial cells, such as VE-cadherin or occludin. Vascular wall areas containing EPCs are found in large and middle sized arteries and veins of all organs studied here. These data suggest the existence of a ;vasculogenic zone' in the wall of adult human blood vessels, which may serve as a source for progenitor cells for postnatal vasculogenesis, contributing to tumor vascularization and local immune response.


Subject(s)
Cell Movement/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/growth & development , Neovascularization, Physiologic/physiology , Stem Cells/cytology , Stem Cells/physiology , Adult , Animals , Antigens, CD34/metabolism , Cell Differentiation/physiology , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , Humans , Immunohistochemistry , Macrophages/physiology , Rats , Receptors, Vascular Endothelial Growth Factor/physiology , Thoracic Arteries/cytology , Thoracic Arteries/metabolism , Thoracic Arteries/physiology
13.
J Hepatol ; 43(5): 845-53, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16139387

ABSTRACT

BACKGROUND/AIMS: Recent studies indicate that after transplantation, circulating bone marrow-derived stem cells migrate into the liver and contribute to liver regeneration. Whether such cells are actively recruited from the bone marrow for liver repair remains to be determined. In this regard, we investigated whether liver resection leads to a release of stem cell marker-positive (+) cells into the peripheral circulation. METHODS: Peripheral blood samples from 11 living liver donors were analyzed by flow cytometry one day before and 12h after partial hepatectomy (PH) using antibodies against CD133, CD34, CD45, CD14, c-kit, bcrp-1. Immunomagnetic separation was performed to select CD133+ cells for functional assays in vitro. RESULTS: A significant increase in the percentage of CD133+ cells could be demonstrated in all donors studied. Unexpectedly, virtually all CD133+ cells coexpressed CD45 and CD14, whereas only a small subset expressed CD34. No significant staining was observed for c-kit and bcrp-1. In culture, immunoselected CD133+ cells displayed characteristics of myelomonocytic precursors. In addition, enriched CD133+ generated an adherent cell population that expressed CK8, alpha-fetoprotein, and human albumin. CONCLUSIONS: PH induces mobilization of a distinct population of myelomonocytic progenitor cells, which have hepatic differentiation potential in vitro, and might play a role in liver regeneration after PH in humans.


Subject(s)
Hematopoietic Stem Cells/physiology , Hepatectomy , Liver Regeneration , Liver Transplantation , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Shape , Hematopoietic Stem Cells/cytology , Hepatocytes/cytology , Humans , Immunomagnetic Separation , Living Donors
14.
J Clin Oncol ; 23(6): 1109-17, 2005 Feb 20.
Article in English | MEDLINE | ID: mdl-15718307

ABSTRACT

PURPOSE: Bone marrow neoangiogenesis plays an important pathogenetic and possible prognostic role in acute myeloid leukemia (AML). Members of the vascular endothelial growth factor (VEGF) and angiopoietin family represent the most specific inducers of angiogenesis secreted by AML blasts. We therefore correlated expression of angiogenic factors with clinical variables. PATIENTS AND METHODS: We investigated the expression of VEGF-A, VEGF-C, angiopoietin-1 (Ang1), angiopoietin-2 (Ang2), and the receptor Tie2 by quantitative polymerase chain reaction in a cohort of 90 patients younger than 61 years with de novo AML entered into the German AML Süddeutsche Hämoblastose Gruppe Hannover 95 trial. Uni- and multivariate analyses were performed using clinical and gene expression variables. RESULTS: Univariate analysis of overall survival indicated the following variables as prognostic factors: good response on a day-15 bone marrow examination after initiation of induction chemotherapy, karyotype, and high Ang2 expression. In multivariate analysis, only bad response and log Ang2 expression remained of statistical significance, with a hazard ratio of 3.51 (95% CI, 1.91 to 6.47) and 0.75 (95% CI, 0.61 to 0.91), respectively. Subgroup analysis suggested that the prognostic impact of Ang2 expression was especially evident in cohorts with low VEGF-C and Ang1 mRNA levels. CONCLUSION: These results show that expression of Ang2 represents an independent prognostic factor in AML. Additional research into interactions of angiogenic cytokines in the pathogenesis of bone marrow angiogenesis in AML is warranted.


Subject(s)
Angiopoietin-2/metabolism , Leukemia, Myeloid/metabolism , Vascular Endothelial Growth Factors/metabolism , Acute Disease , Angiopoietin-1/metabolism , Cohort Studies , Female , Humans , Male , Middle Aged , Neovascularization, Pathologic , Polymerase Chain Reaction , Prognosis , Receptor, TIE-2/metabolism , Survival Analysis , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism
15.
Proc Natl Acad Sci U S A ; 102(2): 431-6, 2005 Jan 11.
Article in English | MEDLINE | ID: mdl-15625114

ABSTRACT

Recruitment and activation of polymorphonuclear neutrophils (PMNs) reflects a primary immunological response to invading pathogens and has also emerged as a hallmark of vascular inflammation. One of the principal enzymes released upon PMN activation is myeloperoxidase (MPO), a heme protein that not only generates cytotoxic oxidants but also impacts deleteriously on nitric oxide-dependent signaling cascades within the vasculature. Because MPO also associates with the membrane of PMN, we evaluated whether MPO could also function as an autocrine modulator of PMN activation. The extent of PMN membrane-associated MPO was elevated in patients with acute inflammatory vascular disease compared with healthy individuals. Isolated PMNs bound free MPO by a CD11b/CD18 integrin-dependent mechanism. PMNs exposed to MPO were characterized by increased tyrosine phosphorylation and p38 mitogen-activated protein kinase activation. Also, nuclear translocation of NFkappaBin PMN was enhanced after incubation with MPO, as was surface expression of CD11b. Binding of PMN to MPO-coated fibronectin surfaces amplified PMN degranulation, as evidenced by increased release of MPO and elastase. MPO also augmented PMN-dependent superoxide (O(2)(*-)) production, which was prevented by anti-CD11b antibodies, but not MPO inhibitors. Collectively, these results reveal that binding of MPO to CD11b/CD18 integrins stimulates PMN signaling pathways to induce PMN activation in a mechanism independent of MPO catalytic activity. These cytokine-like properties of MPO thus represent an additional dimension of the proinflammatory actions of MPO in vascular disease.


Subject(s)
CD11b Antigen/physiology , CD18 Antigens/physiology , Neutrophil Activation , Peroxidase/physiology , Cell Degranulation , Humans , NF-kappa B/metabolism , Phosphorylation , Superoxides/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Exp Cell Res ; 300(1): 65-71, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15383315

ABSTRACT

Bone-marrow-derived, circulating endothelial precursor cells contribute to neoangiogenesis in various diseases. Rapamycin has recently been shown to have anti-angiogenic effects in an experimental tumor model. Our group has developed a culture system that allows expansion and endothelial differentiation of human CD133(+) precursor cells. We could show by PCR analysis that mTOR, the rapamycin-binding protein, was expressed in fresh CD133(+) cells, in expanded cells after 28 days, and in differentiated endothelial cells. Rapamycin inhibited proliferation of CD133(+) cells dose dependently at similar concentrations as hematopoietic Jurkat or HL-60 cells. Apoptosis was induced by rapamycin after 48 h of treatment, which could be reduced by preincubation with FK 506. Furthermore, the development of adherent endothelial cells from expanded CD133(+) cells was dose dependently inhibited. Expression of endothelial antigens CD144 and von Willebrand factor on differentiating endothelial precursors was reduced by rapamycin. In summary, rapamycin inhibits proliferation and differentiation of human endothelial precursor cells underlining its anti-angiogenic effects.


Subject(s)
Cell Differentiation/drug effects , Endothelial Cells/drug effects , Neovascularization, Pathologic/drug therapy , Sirolimus/pharmacology , Stem Cells/drug effects , AC133 Antigen , Antigens/drug effects , Antigens/immunology , Antigens, CD , Apoptosis/drug effects , Apoptosis/physiology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Culture Techniques/methods , Cell Differentiation/immunology , Cell Division/drug effects , Cell Division/immunology , Cell Separation , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/immunology , Glycoproteins/immunology , HL-60 Cells , Humans , Immunosuppressive Agents , Jurkat Cells , Neovascularization, Pathologic/physiopathology , Peptides/immunology , Protein Kinases/drug effects , Protein Kinases/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Sirolimus/therapeutic use , Stem Cells/cytology , Stem Cells/immunology , TOR Serine-Threonine Kinases , Tacrolimus/pharmacology , von Willebrand Factor/immunology
17.
Stem Cells Dev ; 13(3): 229-42, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15186719

ABSTRACT

Recent studies show that human CD133(+) (previously known as AC133(+)) cells from mobilized peripheral blood consist of stem cells with either hematopoietic or endothelial potential. To test whether this population also contains individual precursors with both capacities, the defining characteristics of the elusive adult hemangioblast, we developed a culture system that allows single-cell analyses of differentiation. In the presence of vascular endothelial growth factor (VEGF), stem cell growth factor (SCGF), and FLT-3 ligand, CD133(+)-enriched cells were first expanded and the amplified cells were transduced with a vector encoding an enhanced green fluorescent protein (EGFP) marker gene. Single EGFP(+) cells were then cocultured with corresponding non-transduced cells from the same experiment, yielding 50-100 marked cells in 8% of the wells after 2 weeks. The resultant cells were divided and differentiated with either granulocyte colony-stimulating factor (G-CSF) or with SCGF and VEGF. These culture conditions resulted in the formation of neutrophil or endothelial cells, respectively, as identified morphologically and by phenotypic staining. Dual differentiation of EGFP(+) cells could be observed in one-quarter of clones from single-seeded cells, suggesting that 2% of EGFP(+) cells were in fact human hemangioblasts. These cells could be expanded for at least 28 days without losing this dual capacity. Hence, this culture system may be of clinical relevance in the development of cellular therapies for disorders involving hematopoiesis and the vascular system. In addition, our results provide important information related to the development of the vasculature and the potential role of hemangioblasts in vasculogenesis in adult humans.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Neovascularization, Physiologic , Peptides/metabolism , Stem Cells/physiology , AC133 Antigen , Adult , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Cells, Cultured , Cytokines/metabolism , Genes, Reporter , Hematopoietic Cell Growth Factors/metabolism , Humans , Lectins, C-Type/metabolism , Phenotype , Stem Cells/cytology , Transduction, Genetic , Vascular Endothelial Growth Factor A/metabolism , fms-Like Tyrosine Kinase 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...