The angiogenic factor CCN1 promotes adhesion and migration of circulating CD34+ progenitor cells: potential role in angiogenesis and endothelial regeneration

Blood ◽  
2007 ◽  
Vol 110 (3) ◽  
pp. 877-885 ◽  
Author(s):  
Karsten Grote ◽  
Gustavo Salguero ◽  
Matthias Ballmaier ◽  
Marc Dangers ◽  
Helmut Drexler ◽  
...  

Abstract Tissue regeneration involves the formation of new blood vessels regulated by angiogenic factors. We reported recently that the expression of the angiogenic factor CCN1 is up-regulated under various pathophysiologic conditions within the cardiovascular system. Because CD34+ progenitor cells participate in cardiovascular tissue regeneration, we investigated whether CCN1—detected for the first time in human plasma—promotes the recruitment of CD34+ progenitor cells to endothelial cells, thereby enhancing endothelial proliferation and neovascularization. In this study, we demonstrated that CCN1 and supernatants from CCN1-stimulated human CD34+ progenitor cells promoted proliferation of endothelial cells and angiogenesis in vitro and in vivo. In addition, CCN1 induced migration and transendothelial migration of CD34+ cells and the release of multiple growth factors, chemokines, and matrix metalloproteinase-9 (MMP-9) from these cells. Moreover, the CCN1-specific integrins αMβ2 and αVβ3 are expressed on CD34+ cells and CCN1 stimulated integrin-dependent signaling. Furthermore, integrin antagonists (RGD-peptides) suppressed both binding of CCN1 to CD34+ cells and CCN1-induced adhesion of CD34+ cells to endothelial cells. These data suggest that CCN1 promotes integrin-dependent recruitment of CD34+ progenitor cells to endothelial cells, which may contribute to paracrine effects on angiogenesis and tissue regeneration.

Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Karsten Grote ◽  
Gustavo Salguero ◽  
Marc Dangers ◽  
Matthias Ballmaier ◽  
Bernhard Schieffer

Introduction: CCN1 is so far known as a protein of the extracellular matrix mediating proliferation, adhesion, migration and angiogenesis in an integrin-dependent manner. We recently reported that CCN1 is up-regulated under pathophysiological conditions in the cardiovascular system. Since blood CD34 + progenitor cells have been shown to promote tissue regeneration, we investigated if CCN1 induces angiogenesis by stimulating blood CD34 + cells. Methods and Results: Circulating CCN1 was detected for the first time in human serum samples (Western blot). Rekombinant CCN1 (100 ng/mL) and supernatants from CCN1-stimulated human CD34 + progenitor cells promoted tube-formation (Matrigel-Assay; P < 0.05; n = 4) and proliferation of endothelial cells (BrdU-incorporation; P < 0.05; n = 4). Moreover, CCN1 significantly induced angiogenesis in the context of blood CD34 + cells in vivo determined in Matrigel implanted into C57/BL6 mice as measured by the hemoglobin content (Drabkin’s reagent) and by CD31 staining (immunohistochemistry). In addition, CCN1 induced migration (Transwell cell culture inserts; 7.3 ± 1.3; P < 0.05; n = 4), transendothelial invasion (Transwell cell culture inserts; 2.3 ± 0.7; P < 0.05; n = 3) of CD34 + progenitor cells and release of various growth factors, chemokines (protein array; e.g. G-CSF, GM-CSF, CCL2, TGF-β1) and matrix metalloproteinase-9 (zymography; 2.5 ± 0.5-fold; P < 0.05; n = 6) from these cells. CD34 + progenitor cells expressed the CCN1-specific integrins α M β 2 and α V β 3 (flow cytometry) and binding of CCN1 to CD34 + progenitor cells was diminished by integrin antagonizing RGD peptides. Furthermore, CCN1 stimulated integrin-dependent signaling in CD34 + progenitor cells as shown by enhanced phosphorylation of focal adhesion kinase (immunofluorescence) and promoted their adhesion to endothelial cells which could be inhibited with RGD peptides (P < 0.01; n = 4). Conclusion: Circulating CCN1 binds integrin-specific to CD34 + blood cells and thereby induces migration, adhesion and the release of chemokines and MMPs. Moreover, CCN1-dependent stimulation of blood CD34 + cells promotes endothelial cell proliferation and angiogenesis known to be important for tissue regeneration.


Cancers ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 855
Author(s):  
Paola Serrano Martinez ◽  
Lorena Giuranno ◽  
Marc Vooijs ◽  
Robert P. Coppes

Radiotherapy is involved in the treatment of many cancers, but damage induced to the surrounding normal tissue is often inevitable. Evidence suggests that the maintenance of homeostasis and regeneration of the normal tissue is driven by specific adult tissue stem/progenitor cells. These tasks involve the input from several signaling pathways. Irradiation also targets these stem/progenitor cells, triggering a cellular response aimed at achieving tissue regeneration. Here we discuss the currently used in vitro and in vivo models and the involved specific tissue stem/progenitor cell signaling pathways to study the response to irradiation. The combination of the use of complex in vitro models that offer high in vivo resemblance and lineage tracing models, which address organ complexity constitute potential tools for the study of the stem/progenitor cellular response post-irradiation. The Notch, Wnt, Hippo, Hedgehog, and autophagy signaling pathways have been found as crucial for driving stem/progenitor radiation-induced tissue regeneration. We review how these signaling pathways drive the response of solid tissue-specific stem/progenitor cells to radiotherapy and the used models to address this.


2019 ◽  
Vol 317 (4) ◽  
pp. H765-H776 ◽  
Author(s):  
Takerra K. Johnson ◽  
Lina Zhao ◽  
Dihan Zhu ◽  
Yang Wang ◽  
Yan Xiao ◽  
...  

Induced vascular progenitor cells (iVPCs) were created as an ideal cell type for regenerative medicine and have been reported to positively promote collateral blood flow and improve cardiac function in a rat model of myocardial ischemia. Exosomes have emerged as a novel biomedicine that mimics the function of the donor cells. We investigated the angiogenic activity of exosomes from iPVCs (iVPC-Exo) as a cell-free therapeutic approach for ischemia. Exosomes from iVPCs and rat aortic endothelial cells (RAECs) were isolated using a combination of ultrafiltration and size-exclusion chromatography. Nanoparticle tracking analysis revealed that exosome isolates fell within the exosomal diameter (<150 nm). These exosomes contained known markers Alix and TSG101, and their morphology was validated using transmission electron microscopy. When compared with RAECs, iVPCs significantly increased the secretion of exosomes. Cardiac microvascular endothelial cells and aortic ring explants were pretreated with RAEC-Exo or iVPC-Exo, and basal medium was used as a control. iVPC-Exo exerted an in vitro angiogenic effect on the proliferation, tube formation, and migration of endothelial cells and stimulated microvessel sprouting in an ex vivo aortic ring assay. Additionally, iVPC-Exo increased blood perfusion in a hindlimb ischemia model. Proangiogenic proteins (pentraxin-3 and insulin-like growth factor-binding protein-3) and microRNAs (-143-3p, -291b, and -20b-5p) were found to be enriched in iVPC-Exo, which may mediate iVPC-Exo induced vascular growth. Our findings demonstrate that treatment with iVPC-Exo promotes angiogenesis in vitro, ex vivo, and in vivo. Collectively, these findings indicate a novel cell-free approach for therapeutic angiogenesis. NEW & NOTEWORTHY The results of this work demonstrate exosomes as a novel physiological mechanism by which induced vascular progenitor cells exert their angiogenic effect. Moreover, angiogenic cargo of proteins and microRNAs may define the biological contributors in activating endothelial cells to form a new capillary plexus for ischemic vascular diseases. Listen to this article's corresponding podcast at https://ajpheart.podbean.com/e/angiogenic-exosomes-from-vascular-progenitor-cells/ .


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 83-83
Author(s):  
Alex J. Tipping ◽  
Cristina Pina ◽  
Anders Castor ◽  
Ann Atzberger ◽  
Dengli Hong ◽  
...  

Abstract Hematopoietic stem cells (HSCs) in adults are largely quiescent, periodically entering and exiting cell cycle to replenish the progenitor pool or to self-renew, without exhausting their number. Expression profiling of quiescent HSCs in our and other laboratories suggests that high expression of the zinc finger transcription factor GATA-2 correlates with quiescence. We show here that TGFβ1-induced quiescence of wild-type human cord blood CD34+ cells in vitro correlated with induction of endogenous GATA-2 expression. To directly test if GATA-2 has a causative role in HSC quiescence we constitutively expressed GATA-2 in human cord blood stem and progenitor cells using lentiviral vectors, and assessed the functional output from these cells. In both CD34+ and CD34+ CD38− populations, enforced GATA-2 expression conferred increased quiescence as assessed by Hoechst/Pyronin Y staining. CD34+ cells with enforced GATA-2 expression showed reductions in both colony number and size when assessed in multipotential CFC assays. In CFC assays conducted with more primitive CD34+ CD38− cells, colony number and size were also reduced, with myeloid and mixed colony number more reduced than erythroid colonies. Reduced CFC activity was not due to increased apoptosis, as judged by Annexin V staining of GATA-2-transduced CD34+ or CD34+ CD38− cells. To the contrary, in vitro cultures from GATA-2-transduced CD34+ CD38− cells showed increased protection from apoptosis. In vitro, proliferation of CD34+ CD38− cells was severely impaired by constitutive expression of GATA-2. Real-time PCR analysis showed no upregulation of classic cell cycle inhibitors such as p21, p57 or p16INK4A. However GATA-2 expression did cause repression of cyclin D3, EGR2, E2F4, ANGPT1 and C/EBPα. In stem cell assays, CD34+ CD38− cells constitutively expressing GATA-2 showed little or no LTC-IC activity. In xenografted NOD/SCID mice, transduced CD34+ CD38−cells expressing high levels of GATA-2 did not contribute to hematopoiesis, although cells expressing lower levels of GATA-2 did. This threshold effect is presumably due to DNA binding by GATA-2, as a zinc-finger deletion variant of GATA-2 shows contribution to hematopoiesis from cells irrespective of expression level. These NOD/SCID data suggest that levels of GATA-2 may play a part in the in vivo control of stem and progenitor cell proliferation. Taken together, our data demonstrate that GATA-2 enforces a transcriptional program on stem and progenitor cells which suppresses their responses to proliferative stimuli with the result that they remain quiescent in vitro and in vivo.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 261-261
Author(s):  
Selcuk Sozer ◽  
Takefumi Ishii ◽  
Wei Zhang ◽  
Jiapeng Wang ◽  
Mingjiang Xu ◽  
...  

Abstract Patients with myeloproliferative disorders (MPD) are at a high risk of developing thrombotic events. We hypothesize that one of the contributory factors to this thrombotic tendency is the involvement of vascular endothelial cells (EC) by the malignant process. In vitro and in vivo assays were used to determine the involvement of EC in patients with MPD. Endothelial progenitor cells (EPC) were assayed from the peripheral blood (PB) mononuclear cells (MNCs) of 3 normal controls (NC) and 16 patients with MPD (12 polycythemia vera (PV), 4 primary myelofibrosis (PMF). MNC were cultured for 2 days in EC growth media on fibronectin(FN)-coated plates. The non-adherent cells were then harvested and transferred to a secondary FN-coated plate for additional 5–14 days. EC colonies were identified by their morphological appearance. The colonies were plucked and analyzed for PECAM-1(CD31), VE-Cadherin(CD144), VEGFR-2, vWF, Endoglin(CD105), ULEX-1, CD45, CD14 by flow cytometry and acetylated LDL(Ac-ADL) uptake. EC colonies were CD31+CD144+VEGFR2+ULEX-1+vWF+CD105+CD45+CD14+ and capable of taking up Ac-LDL and when exposed to TNF-α and IL-1β, expressing ICAM(CD54) and E-selectin(CD62e). MPD MNC formed fewer numbers of EC colonies than normal MNC (31.1±34.2 vs 78.8±28.9; p&lt;0.01) and required more prolonged periods of culture (14 vs 5days). MPD EC colonies were also analyzed for JAK2V617F(JAK2VF) by nested-PCR. 74.6% of MPD EC colonies were homozygous(homo) JAK2VF, 14.9% were heterozygous(hetero) JAK2VF and 10.4% were wild type(wt) JAK2. Interestingly, MNCs from JAK2VF−MPD(148±47) formed greater numbers of EC colonies than NC MNC (78.8±28.9; p≤0.01). MNC from patients with a high burden of JAK2VF alleles (10.3±18.5; p&lt;0.01) formed fewer EC colonies than NC or patients with a low burden of JAK2VF (65.9±28.15; p≤0.01). These EPC assayed in vitro which produced cells with both myeloid and endothelial markers are likely due either to contamination with JAK2VF myeloid cells or the result of the transdifferentiation of myeloid progenitor cells into EC (Bailey A, et al. PNAS.2006,103:13156). The inverse correlation between the JAK2VF burden and the ability of MPD MNC to form EC colonies is possibly a consequence of the increased sensitivity of EC to apoptosis due to the constitutive activation of JAK2 (Neria F, et al. Am J Physiol Cell Physiol.2007, 292:1123). In order to assay for more primitive EPC, 2 cord blood, and 16 JAK2VF+ MPD CD34+ (10 PV, 6 PMF) cells were transplanted into sublethally irradiated NOD/SCID mice. After 8 weeks, EC-rich organs (heart, lung, liver, vessels) were harvested, single cell suspensions were positively selected for either human(h) CD31+or hCD144+ cells by immunomagnetic cell sorting and analyzed for hVEGFR2, CD144, vWF, CD45, CD14 mRNA expression and JAK2VF. These CD31+or CD144+ cells contained transcripts for CD144, vWF, VEGFR2 but not CD45 and CD14. In 77.7% of the cases the hCD31+ or hCD144+ cells were homo JAK2VF, 5.5% were hetero JAK2VF and 16.6% were wt JAK2 and these CD31+or CD144+ cells composed ≤1% of the cells within the respective tissues. hCD144+ cells were also cultured with EC growth media for 7 days and displayed EC morphology and were shown to contain JAK2VF+ cells. These CD31+CD144+JAK2VF+CD14−CD45−cells likely represent the progeny of a malignant EPC which is distinct from an HSC. The involvement of EC by the malignant process in MPD might contribute to the development of thrombosis in MPD.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2322-2322
Author(s):  
Takashi Yoshikubo ◽  
Yoshihiro Matsumoto ◽  
Masahiko Nanami ◽  
Takayuki Sakurai ◽  
Hiroyuki Tsunoda ◽  
...  

Abstract Thrombopoietin (TPO, the ligand for c-mpl) is a key factor for megakaryopoiesis. Several clinical trials of TPO have been conducted for thrombocytopenia without much success due to, in part, the production of neutralized antibodies against endogenous TPO, which causes thrombocytopenia. To overcome this problem, we previously demonstrated that mouse type minibody against c-mpl, with an amino acid sequence totally different from TPO, showed megakaryopoiesis and increased platelet numbers in monkey. This time, using CDR grafting, we generated a humanized sc(Fv)2VB22B minibody (huVB22B) against c-mpl for therapeutic use. The new minibody showed almost the same activity in vitro as TPO and the mouse type minibody, confirmed by both a human megakaryocyte cell (CD41+) differentiation assay and a proliferation assay with TPO-dependent cell line, M-07e. Single sc or iv administration of huVB22B to cynomolgus monkeys showed a dose-dependent increase in platelet numbers. Pharmacokinetic analysis showed that the plasma half-life (T1/2) of huVB22B at iv and sc administration to cynomolgus monkeys was 7–8 h and 11–16 h, respectively. After administration of huVB22B, the platelets of these monkeys increased and showed functional aggregation in response to ADP in vitro. Repeated administration of huVB22B (0.2, 2 and 20mg/kg/week) revealed that the increase in platelet level in cynomolgus monkeys was maintained for a month. Very slight reticular fibers in bone marrow were detected in a high dose group (20mg/ kg). No overt changes were detected by toxicity evaluations including clinical pathology and histopathology in 0.2 and 2mg/kg groups. No neutralized activities in plasma were observed during these experiments. Next, we examined the activities of huVB22B on human bone marrow-derived CD34-positive cells (BM-CD34+) and umbilical cord blood-derived CD34-positive cells (UCB-CD34+) in vitro. BM-CD34+ and UCB-CD34+ cells were cultured with huVB22B in serum free medium. HuVB22B induced differentiation of CD41+ cells from BM-CD34+ or UCB-CD34+ cells in a similar dose-dependent manner. However, UCB-CD34+ cells showed greater proliferation in response to huVB22B compared to BM-CD34+ cells. We then examined the in vivo activities of huVB22B on UCB CD34+ cells by treating NOD/SCID mice transplanted with human UCB-CD34+ cells with huVB22B and examining the bone marrow cells of the mice. The results showed that, compared with the control, administration of huVB22B showed an increase in the number of human hematopoietic progenitor cells (CD34+), lymphoid lineage cells (CD19+), and myeloid lineage cells (CD33+) in addition to human CFU-Meg cells (CD41+). These results suggest that c-mpl stimulation in vivo after transplantation might increase engraftment of progenitor cells in the bone marrow microenvironment and subsequently induce differentiation to multilineage cells. Umbilical cord blood transplantation faces frequent complications including a low-level stem/progenitor cell engraftment and delayed platelet recovery. Our results suggest that c-mpl stimulation might be used to increase the engraftment of UCB stem/progenitor cells and shorten the time of platelet recovery following UCB transplantation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 693-693
Author(s):  
Katherine L. Hill ◽  
Petra Obrtlikova ◽  
Diego F Alvarez ◽  
Judy A King ◽  
Qinglu Li ◽  
...  

Abstract The field of vascular regenerative medicine is rapidly growing and the demand for cell-based therapy is high. In our studies, human embryonic stem cells (hESCs) were differentiated via coculture with M2-10B4 mouse bone marrow derived stromal cells for 13–15 days. At this time, CD34+ were isolated using an immunomagnetic separation technique and further phenotyped. As shown by flow cytometric analysis, the population co-expressed typical endothelial cell surface antigens such as CD31 and Flk. Upon culture of these CD34+ cells in endothelial culture medium containing VEGF, bFGF, IGF-1, EGF, and heparin, the cells assumed a endothelial cell morphology, formed vascular like networks when placed in Matrigel, and expressed CD31, Flk1, CD146, Tie2, eNOS, vWF, and VE-cadherin (each confirmed by quantitative real time PCR, immunohistochemistry, and flow cytometry). Transmission electron micrograph images of these cells, termed hESC-ECs, showed a defined cortical filamentous rim as seen in other endothelial cells and a significant number of micro-particles being released from the cell surface. Additionally, permeability studies revealed these cells exhibit trans-electrical resistance of 1200Ω, consistent with basal barrier properties exhibited by conduit endothelial cells. These hESC-ECs also proved capable of further differentiation into smooth muscle cells, hESCSMCs. When culture conditions were changed to support SMC growth (DMEM + PDGFBB and TGF-β1), cells assumed SMC morphology including intracellular fibrils, down regulated endothelial gene transcript and protein expression, and began to express α-SMC actin, calponin, SM22, smoothelin, myocardin. Also, concomitant increases in expression of APEG-1 and CRP2/SmLIM, expressed preferentially by arterial SMCs, was found. In contrast, HUVECs placed under these SMC conditions did not display SMC characteristics. Additional studies evaluated intracellular calcium release in hESC-ECs and hESC-SMCs subjected to various pharmacological agonists. The hESC-SMC population preferentially responded to bradykinin, oxytocin, endothelin-1, histamine, and ATP, while hESC-ECs responsed to endothelin-1, histamine, bradykinin, and carbachol. Functional studies were initially done by in vitro culture of these cell populations in Matrigel. hESC-SMCs placed in Matrigel alone did not form a vascular like network. However, an improved vascular structure was seen when hESC-ECs were placed in Matrigel along with hESC-SMCs. Together, these cells formed a dense, more robust vascular network composed of thicker tube structures, indicating a more physiologically relevant model of vasculogenesis. Next in vivo studies have been initiated utilizing a mouse myocardial infarct model. NOD/SCID mice were anesthetized and subjected to ligation of the left anterior descending artery. By assessing cardiac function 3 weeks post infarction, we found that mice receiving an hESC-EC injection (1×106 cells directly into infarction sight) showed greater vascular repair and increased ejection fraction when compared to mice that did not receive an hESCEC injection [untreated control ejection fraction= 14.3% vs hESC-EC treated= 21.3%]. Currently, studies are underway evaluating combined use of hESC-ECs and hESC-SMCs in this infarct model, as we hypothesize that combined use of these cells will be more beneficial for vascular development and repair than either one population alone. Together, the phenotypic and functional studies of these hESC-derived CD34+ cells suggest these cells can act as pericytes with dual endothelial cell and SMC developmental potential and these hESC-derived pericytes can provide an important resource for developing novel cellular therapies for vascular repair.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 739-739
Author(s):  
Vijay P. S. Rawat ◽  
Natalia Arseni ◽  
Farid Ahmed ◽  
Medhanie A. Mulaw ◽  
Silvia Thoene ◽  
...  

Abstract Abstract 739 Recent studies suggest that a variety of regulatory molecules active in embryonic development such as clustered and non-clustered homeobox genes play an important role in normal and malignant hematopoiesis. Since it was shown that the Xvent-2 homeobox gene is part of the BMP-4 signalling pathway in Xenopus, it is of particular interest to examine the expression profile and function of its only recently discovered human homologue VENTX in hematopoietic development. Expression of the VENTX gene was analyzed in normal human hematopoiesis and AML patients samples by microarray and qPCR. To test the impact of the constitutive expression of VENTX on human progenitor cells, CD34+ cord blood (CB) cells were retrovirally transduced with VENTX or the empty control vector and analyzed using in vitro and in vivo assays. So far we and others have not been able to identify a murine Xenopus xvent gene homologue. However, we were able to document the expression of this gene by qPCR in human lineage positive hematopoietic subpopulations. Amongst committed progenitors VENTX was significantly 13-fold higher expressed in CD33+ BM myeloid cells (4/4 positive) compared to CD19+ BM lymphoid cells (5/7 positive, p=0.01). Of note, expression of VENTX was negligible in normal CD34+/CD38− but detectable in CD34+ BM human progenitor cells. In contrast to this, leukemic CD34+/CD38− from AML patients (n=3) with translocation t(8,21) showed significantly elevated expression levels compared to normal CD34+ BM cells (n=5) (50-fold higher; p≤0.0001). Furthermore, patients with normal karyotype NPM1c+/FLT3-LM− (n=9), NPM1c−/FLT3-LM+ (n=8) or patients with t(8;21) (n=9) had an >100-fold higher expression of VENTX compared to normal CD34+ BM cells and a 5- to 7.8-fold higher expression compared to BM MNCs. Importantly, lentivirus-mediated long-term silencing of VENTX in human AML cell lines (mRNA knockdown between 58% and 75%) led to a significant, reduction in cell number compared to the non-silencing control construct (>79% after 120h). Suggesting that growth of human leukemic cell lines depends on VENTX expression in vitro. As we observed that VENTX is aberrantly expressed in leukemic CD34+ cells with negligible expression in normal counterparts, we assessed the impact of forced VENTX gene expression in normal CD34+ human progenitor cells on the transcription program. Gene expression and pathway analysis demonstrated that in normal CD34+ cells enforced expression of VENTX initiates genes associated with myeloid development (CD11b, CD125, CD9,CD14 and M-CSF), and downregulates genes involved in early lymphoid development (IL-7, IL-9R, LEF1/TCF and C-JUN) and erythroid development such as EPOR, CD35 and CD36. We then tested whether enforced expression of VENTX in CD34+ cells is able to alter the hematopoietic development of early human progenitors as indicated by gene expression and pathway analyses. Functional analyses confirmed that aberrant expression of VENTX in normal CD34+ human progenitor cells induced a significant increase in the number of myeloid colonies compared to the GFP control with 48 ± 6.5 compared to 28.9 ± 4.8 CFU-G per 1000 initially plated CD34+ cells (n=11; p=0.03) and complete block in erythroid colony formation with an 81% reduction of the number of BFU-E compared to the control (n=11; p<0.003). In a feeder dependent co-culture system, VENTX impaired the development of B-lymphoid cells. In the NOD/SCID xenograft model, VENTX expression in CD34+ CB cells promoted generation of myeloid cells with an over 5-fold and 2.5-fold increase in the proportion of human CD15+ and CD33+ primitive myeloid cells compared to the GFP control (n=5, p=0.01). Summary: Overexpression of VENTX perturbs normal hematopoietic development, promotes generation of myeloid cells and impairs generation of lymphoid cells in vitro and in vivo. Whereas VENTX depletion in human AML cell lines impaired their growth.Taken together, these data extend our insights into the function of human embryonic mesodermal factors in human hematopoiesis and indicate a role of VENTX in normal and malignant myelopoiesis. Disclosures: No relevant conflicts of interest to declare.


2007 ◽  
Vol 293 (2) ◽  
pp. F624-F630 ◽  
Author(s):  
Maarten B. Rookmaaker ◽  
Marianne C. Verhaar ◽  
Hetty C. de Boer ◽  
Roel Goldschmeding ◽  
Jaap A. Joles ◽  
...  

The chemokine RANTES (regulated upon activation normal T-cell expressed and secreted) is involved in the formation of an inflammatory infiltrate during glomerulonephritis. However, RANTES receptor inhibition, although reducing glomerular leukocyte infiltration, can also increase damage. We hypothesized that RANTES does not only promote the influx and activation of inflammatory leukocytes but also mediates glomerular microvascular repair by stimulating the homing of bone marrow (BM)-derived endothelial progenitor cells. To investigate the role of RANTES in the participation of BM-derived cells in glomerular vascular repair, we used a rat BM transplantation model in combination with reversible anti-Thy-1.1 glomerulonephritis. Twenty-four hours after the induction of glomerulonephritis, BM-transplanted rats were treated for 7 days with either the RANTES receptor antagonist Met-RANTES or saline. The participation of BM-derived endothelial cells in glomerular repair, glomerular monocyte infiltration, and proteinuria was evaluated at days 7 and 28. Furthermore, we used an in vitro perfusion chamber assay to study the role of RANTES receptors in shear-resistant adhesion of the CD34+ stem cells to activated endothelium under flow. In our reversible glomerulonephritis model, RANTES receptor inhibition specifically reduced the participation of BM-derived cells in glomerular vascular repair by more than 40% at day 7 without impairing monocyte influx. However, no obvious change in recovery from proteinuria or morphological damage was observed. Blockade of RANTES receptors on CD34+ cells in vitro partially inhibited platelet-enhanced, shear-resistant firm adhesion of the CD34+ cells to activated endothelium. In conclusion, our data suggest that RANTES is involved in the homing and participation of BM-derived endothelial cells in glomerular repair.


2019 ◽  
Vol 34 (2) ◽  
pp. 129-137
Author(s):  
E. O. Krivkina ◽  
V. N. Silnikov ◽  
A. V. Mironov ◽  
E. A. Velikanova ◽  
E. A. Senokosova ◽  
...  

Research goals. To study the effectiveness of RGD-peptide modification of the small-diameter biodegradable vascular grafts depending on the type of a linker and RGD configuration.Material and Methods. Tubular scaffolds with a diameter of 1.5 and 4.0 mm were produced by electrospinning from polyhydroxybutyrate/valerate (PHBV) and polycaprolactone (PCL). The PHBV/PCL grafts were modified with RGD peptides. In vitro experiments showed the degree of erythrocyte hemolysis and adhesion of the platelets and endothelial cells when in contact with a modified surface. The physico-mechanical properties and the structure of graft surface were studied before and after modification. The PHBV/PCL and PHBV/PCL/RGD vascular grafts were implanted into the abdominal aorta of rats for the periods of 1 and 3 months. Explant samples were studied using confocal microscopy and histological methods.Results. The results of physical and mechanical tests showed a significant decrease in the strength properties of the PHBV/PCL/RGD grafts relative to the unmodified analogs. A significant increase in platelet aggregation was found in the modified grafts. The level of adhesion of the endothelial cells on the modified surfaces was higher than that on the unmodified surfaces. Shortterm implantation of the grafts for 1 and 3 months showed that the modified grafts had higher patency and a less tendency to calcification compared with the unmodified grafts. Immunofluorescence study demonstrated the significant superiority of the modified vascular grafts in terms of stimulating the formation of a mature endothelial monolayer. A longer linker of 4,7,10-trioxa-1,13-tridecane diamine was found to increase the bioavailability of RGD peptides; the use of RGDK and c[RGDFK] for surface modification of the grafts stimulated early endothelialization of the internal surface of the implants and reduced the prosthetic wall calcification tendency, which together increased the patency of the implanted grafts.Conclusion. In short-term implantation of biodegradable vascular grafts modified with RGD peptides, the grafts with RGDK and c[RGDFK], attached to the surface of the prostheses through the 4,7,10-triox-1,13-tridecane diamine linker, showed the best results in terms of endothelial adhesion and maintenance of the viability of the endothelial cells in vitro and endothelialization in vivo; these grafts had high patency after implantation into the bloodstream of small laboratory animals and a less tendency to calcification.


Sign in / Sign up

Export Citation Format

Share Document