• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 27
  • 22
  • 10
  • 4
  • 3
  • 2
  • 1
  • Tagged with
  • 78
  • 78
  • 78
  • 22
  • 21
  • 20
  • 18
  • 17
  • 16
  • 13
  • 12
  • 11
  • 10
  • 10
  • 10
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Role of endothelial progenitor cells in acute vascular injury in man

Padfield, Gareth John January 2013 (has links)
Percutaneous coronary intervention (PCI) acutely improves coronary blood flow and myocardial perfusion but at the expense of endovascular laceration and endothelial denudation. PCI associated vascular injury is associated with intense inflammation and a loss of vascular function that may lead to significant in-stent restenosis (ISR), and the potentially catastrophic, acute stent thrombosis. Reendothelialisation is essential to the restoration of normal homeostasis and facilitating vascular healing. Attention has recently focused on a novel mechanism of reendothelialisation mediated by bone marrow-derived precursor or stem cells: endothelial progenitor cells (EPC). EPC are thought to home to, and reendothelialise sites of endothelial denudation, and therefore offer the potential to provide exciting new developments in the management of cardiovascular disease. Understanding the role of EPC following vascular injury may help us to enhance vascular repair following PCI. The following studies were performed to clarify the relationships between putative EPC and vascular injury associated with PCI. In studies of patients undergoing elective PCI for stable anginal symptoms I found that concentrations of traditional circulating phenotypic EPC expressing CD34+VEGFR-2+ were unaffected, unlike CD34+CD45- cell concentrations, which were transiently increased six hours following PCI, subsequently returning to normal by 24 hours, notably without an increase in CD34+ adhesion molecule expression or VEGF-A production. However, the purported progeny of CD34+VEGFR-2+ cells, endothelial cell-colony forming units (EC-CFU), were mobilised at 24 hours, commensurate with a systemic inflammatory response. Interestingly the concentration of circulating CD34+VEGFR-2+ cells and EC-CFU were unrelated to each other, emphasising the distinction between these two cell populations. Although EC-CFU contained proliferating cells and exhibited some endothelial characteristics, EC-CFU predominantly expressed the leukocyte antigen CD45 in addition to the lymphocyte markers CD4 and CD8, and most intensely, the surface markers CD68 and CD105, epitopes commonly expressed on macrophages. Notably, EC-CFU were a potent stimulus for the migration of mononuclear cells. However, despite being mobilised in the context of an acute systemic inflammatory response and being composed of leukocytes, isolated systemic inflammation in healthy volunteers (induced by Salmonella Typhus vaccination) in the absence of vascular injury did not cause selective mobilisation of EC-CFU or indeed of putative phenotypic EPC. It is therefore likely that EC-CFU mobilisation is a relatively specific inflammatory response to cardiovascular injury. In a cohort of 201 patients undergoing coronary angiography, traditional circulating phenotypic EPC (CD34+VEGFR-2+ and CD34+VEGFR-2+CD133+) were very rare indeed and were not increased in response to an acute coronary syndrome (ACS). Furthermore traditional EPC concentrations bore no relation to atheroma burden or clinical outcome. In contrast, concentrations of CD34+CD45- cells were increased in patients with coronary artery disease compared to those with normal coronary arteries and were increased in association with more severe coronary disease. Increased concentrations of circulating CD34+CD45- cells were also associated with a shorter cumulative event-free survival. Both EC-CFU and angiogenic monocytes expressing Tie-2 and VEGFR-2 were increased following acute myocardial infarction but did not relate to coronary atheroma or clinical outcome. These studies examine the behavior of putative EPC in response to both discrete vascular injury and myocardial infarction, and isolated inflammation in the absence of vascular injury. I have identified novel characteristics of the EC-CFU assay and determined that specific factors associated with cardiovascular injury likely trigger EC-CFU mobilisation. The clinical relevance of the traditional phenotypic EPC population is uncertain, but a novel CD34+CD45- population is mobilised acutely following discrete vascular injury and is significantly associated with coronary atheroma and clinical events. It is probable that the circulating CD34+CD45- concentration reflects vascular injury and atheroma burden, and I suggest that CD34+CD45- cells are released directly from the vessel wall following PCI, and do not reflect a reparatory response. In order to determine the impact of EPC populations on vascular healing, prospective studies examining the impact of periprocedural EPC concentrations on vascular healing following PCI are required.
2

The Role of MicroRNAs in Endothelial Progenitor Cell Function

Behbahani, John January 2016 (has links)
Cultures of peripheral blood mononuclear cells (MNCs) give rise to at least two different variants of endothelial progenitor cells (EPCs), early and late outgrowth EPCs. We investigated whether microRNAs in early and late EPCs could serve as markers of internal processes that can be exploited to distinguish cell identity and functional capacity. We hypothesized that as MNCs give rise to early and late EPCs, there is a gradual change in total microRNA profile, reflecting a total change in processes within the predominant cell population. Using a candidate microRNA array, early and late EPCs showed vastly different microRNA expression profiles. MiR-146a expression increased progressively as early EPCs emerged around 5-7 days (p<0.05). Through targeting TRAF6 and IRAK1, miR-146a conferred inflammatory tolerance in early EPCs, likely contributing to their purported ability to suppress inflammation. MiR-146a knock down (KD) in endotoxin-stimulated early EPCs reduced anti-inflammatory cytokine IL-1RA (p<0.001), and increased expression of pro-inflammatory cytokines IL-1 (p<0.001) and IL-8 (p<0.01). Interestingly, the microRNA expression profile of late EPCs was highly congruent to mature endothelial cells, with 100-fold greater miR-126 expression than monocytes and early EPCs (p<0.01). MiR-126KD in late EPCs abolished matrigel-network formation (p<0.05); while overexpression (OE) in early EPC augmented network formation (p<0.05) and chemotactic migration (p<0.001). We also found that the melanoma cell adhesion molecule or MCAM (CD146) identified late EPC precursors. Only MCAM+MNCs from adult blood (<5% of total MNCs) yielded late EPC-like colonies. Robust miR-126 expression in these cells predicted the generation of late EPCs. Overall, our results suggest that miR-146a in early EPCs likely contributes to repair by suppressing inflammation during cardiovascular injury; while in late EPCs, miR-126 directly promotes angiogenesis and vascular repair. Finally, we highlight a unique method for the efficient generation of late EPCs by using MCAM selection and screening for miR-126.
3

Characterization and Assessment of Lung and Bone Marrow Derived Endothelial Cells and their Bone Regenerative Potential

Valuch, Conner R. 12 1900 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Fracture repair is costly and difficult to treat. One of the main causations of nonunion is a lack of essential blood supply. The needed blood is supplied by the growth of new blood vessels, a process known as angiogenesis, that invade the damaged tissue early in the healing process. We proposed using bone tissue engineering as an effective therapy. This therapy uses stem cells to aid in tissue regeneration. Endothelial progenitor cells (EPCs) were selected due to their ability to form tube-like networks in vitro. EPCs were isolated from murine bone marrow and lung tissue. We tested EPC’s tube forming, proliferative, and wound migration ability in vitro. To test their ability in vivo we created a femoral fracture in young and old mice. EPCs were seeded to the fracture site upon a collagen scaffold. The in vitro studies displayed that the bone marrow and lung-derived endothelial cells presented EPC traits. In the mouse fracture model bone marrow, endothelial cells did not significantly improve the healing process. In the future, we want to improve our cell extraction and purification method, as well as test a new stem cell delivery biomaterial. We also want to select and use a growth factor (GF) that can help to promote bone regeneration in tandem with the EPCs.
4

Biological Effects of Osteopontin on Endothelial Progenitor Cells

Altalhi, Wafa 03 October 2011 (has links)
Endothelial Progenitor Cells (EPCs) are thought to participate in the healing of injured vascular endothelium by incorporating into the defect sites to mediate endothelial recovery. Recently, osteopontin (OPN) was shown to be fundamental in accelerating estrogen-dependent healing of injured blood vessels. Here, we are investigating the effect OPN has on EPC behavior. Late outgrowth human EPCs (LEPCs) were derived from circulating monocytes isolated by leukophoresis, and grown in culture until passage six. L-EPCs were then assayed for adhesion, spreading, chemotaxis, and haptotaxis, as well as resistance to detachment by flow electric cellsubstrate impedance sensing (ECIS). The results of standard and ECIS methods showed both dose and time dependent responses in cell adhesion and spreading. In addition, OPN promoted haptotactic migration of EPCs in Boyden chamber assays. LEPCs seeded onto 10μM OPN substrates and exposed to laminar flow had grater survival and higher resistance to detachment than OPN/static and flow only conditions. CD44 and !1 integrins were only responsible for approximately 50% of LEPCs adhesion to OPN compared to the unblocked condition. Western blots showed that Rho GTPases were activated in L-EPCs seeded on OPN. However, this activation could not be completely blocked by either CD44 or !1 integrin antagonists. These data confirm the direct effects of OPN on EPCs adhesion, and suggest that OPN works by mediating cell adhesion during vascular injury.
5

The Use of Endothelial Progenitor Cells to Promote Bone Healing in a Defect Model in the Rat Femur

Atesok, Kivanc 01 December 2011 (has links)
The objective of this project was to evaluate the effects of local endothelial progenitor cell (EPC) therapy on bone regeneration in a segmental defect in the rat femur. Animals from the EPC-treated (N=28) and control (N=28) groups were sacrificed at 1, 2, 3, and 10 weeks post-operatively. Bone healing was evaluated with radiographic, histological, and micro computed tomography (micro-CT) scans. Radiographically; mean scores of the EPC group at 1, 2, and 3 weeks were significantly higher compared to control group. At 10 weeks, all the animals in the EPC group had complete union (7/7), but in the control group none achieved union (0/7). Histologically, specimens from EPC-treated animals had abundant new bone formation compared to controls. Micro-CT assessment showed significantly improved parameters of bone healing for the EPC group compared to control group. In conclusion, local EPC therapy significantly enhanced bone regeneration in a segmental bone defect in rat femur.
6

The Use of Endothelial Progenitor Cells to Promote Bone Healing in a Defect Model in the Rat Femur

Atesok, Kivanc 01 December 2011 (has links)
The objective of this project was to evaluate the effects of local endothelial progenitor cell (EPC) therapy on bone regeneration in a segmental defect in the rat femur. Animals from the EPC-treated (N=28) and control (N=28) groups were sacrificed at 1, 2, 3, and 10 weeks post-operatively. Bone healing was evaluated with radiographic, histological, and micro computed tomography (micro-CT) scans. Radiographically; mean scores of the EPC group at 1, 2, and 3 weeks were significantly higher compared to control group. At 10 weeks, all the animals in the EPC group had complete union (7/7), but in the control group none achieved union (0/7). Histologically, specimens from EPC-treated animals had abundant new bone formation compared to controls. Micro-CT assessment showed significantly improved parameters of bone healing for the EPC group compared to control group. In conclusion, local EPC therapy significantly enhanced bone regeneration in a segmental bone defect in rat femur.
7

Biological Effects of Osteopontin on Endothelial Progenitor Cells

Altalhi, Wafa 03 October 2011 (has links)
Endothelial Progenitor Cells (EPCs) are thought to participate in the healing of injured vascular endothelium by incorporating into the defect sites to mediate endothelial recovery. Recently, osteopontin (OPN) was shown to be fundamental in accelerating estrogen-dependent healing of injured blood vessels. Here, we are investigating the effect OPN has on EPC behavior. Late outgrowth human EPCs (LEPCs) were derived from circulating monocytes isolated by leukophoresis, and grown in culture until passage six. L-EPCs were then assayed for adhesion, spreading, chemotaxis, and haptotaxis, as well as resistance to detachment by flow electric cellsubstrate impedance sensing (ECIS). The results of standard and ECIS methods showed both dose and time dependent responses in cell adhesion and spreading. In addition, OPN promoted haptotactic migration of EPCs in Boyden chamber assays. LEPCs seeded onto 10μM OPN substrates and exposed to laminar flow had grater survival and higher resistance to detachment than OPN/static and flow only conditions. CD44 and !1 integrins were only responsible for approximately 50% of LEPCs adhesion to OPN compared to the unblocked condition. Western blots showed that Rho GTPases were activated in L-EPCs seeded on OPN. However, this activation could not be completely blocked by either CD44 or !1 integrin antagonists. These data confirm the direct effects of OPN on EPCs adhesion, and suggest that OPN works by mediating cell adhesion during vascular injury.
8

Biological Effects of Osteopontin on Endothelial Progenitor Cells

Altalhi, Wafa 03 October 2011 (has links)
Endothelial Progenitor Cells (EPCs) are thought to participate in the healing of injured vascular endothelium by incorporating into the defect sites to mediate endothelial recovery. Recently, osteopontin (OPN) was shown to be fundamental in accelerating estrogen-dependent healing of injured blood vessels. Here, we are investigating the effect OPN has on EPC behavior. Late outgrowth human EPCs (LEPCs) were derived from circulating monocytes isolated by leukophoresis, and grown in culture until passage six. L-EPCs were then assayed for adhesion, spreading, chemotaxis, and haptotaxis, as well as resistance to detachment by flow electric cellsubstrate impedance sensing (ECIS). The results of standard and ECIS methods showed both dose and time dependent responses in cell adhesion and spreading. In addition, OPN promoted haptotactic migration of EPCs in Boyden chamber assays. LEPCs seeded onto 10μM OPN substrates and exposed to laminar flow had grater survival and higher resistance to detachment than OPN/static and flow only conditions. CD44 and !1 integrins were only responsible for approximately 50% of LEPCs adhesion to OPN compared to the unblocked condition. Western blots showed that Rho GTPases were activated in L-EPCs seeded on OPN. However, this activation could not be completely blocked by either CD44 or !1 integrin antagonists. These data confirm the direct effects of OPN on EPCs adhesion, and suggest that OPN works by mediating cell adhesion during vascular injury.
9

The role of endothelial progenitor cells in the utero-placental vasculature

Sipos, Peter January 2013 (has links)
Fetal growth in utero depends on nutrient and oxygen reaching the fetus through the uterine and placental microcirculations, both undergoing massive expansion during pregnancy. Aberrations of the placental vasculature are associated with Intrauterine Growth Restriction (IUGR), a common pathological outcome of pregnancy; however, the cellular components responsible for vessel formation in the placenta and the uterus remain unknown. Endothelial Progenitor Cells (EPC) are a group of morphologically and functionally varied bone marrow derived vasculogenic cell types, divided into two major subsets: (i) Circulating Angiogenic Cells (CACs), which promote vessel formation by interfering with the extracellular matrix and (ii) Endothelial Colony Forming Cells (ECFCs), which provide the source for new endothelium. This role has been demonstrated in pathophysiological studies, but not in normal physiological events in vivo. Fetal ECFCs are more proficient than their adult counterparts, but it is unclear in what specific fetal or maternal physiological situations fetal ECFCs are involved. Based upon these considerations, it was hypothesised that: (i) fetal-derived ECFCs play a role in placental vasculogenesis, (ii) these cells transmigrate the placenta and home to loci of vessel formation in the pregnant uterus, and that (iii) intrinsic alterations in their capabilities are associated with fetal growth restriction during intrauterine life. To support these hypotheses the following experiments were performed;(i) EPCs in blood from pairs of human umbilical arteries and veins were counted by flow cytometry. Numbers of EPCs in these samples showed an arterio-venous gradient suggesting their placental sequestration. Furthermore, ECFCs were isolated from human umbilical blood using established culture techniques. Labelled human fetal ECFCs were transplanted into the circulation of murine fetuses using an ultrasound-guided intra-cardiac injection. Using a fluorescent imager and microscopy these cells were shown to home to the murine placenta and participate in vasculogenesis.(ii) Male mice ubiquitously expressing eGFP were crossbred with native females, and fetal (eGFP-positive) endothelial-like cells integrated into the uterine microvasculature. Human fetal ECFCs injected into murine fetuses were shown to migrate to the maternal uterus and became functionally involved with the microvasculature. In humans, microvessels were isolated from uterine biopsies of mothers with male offspring. Copies of the male specific SRY gene (quantified by RT-QPCR) indicated that cells of fetal origin constituted 12% of the endothelium in these vessels. In cross-sections, hybridisation of the Y-chromosome demonstrated the presence of fetal cells in the maternal endothelium of the human uterus. (iii) Using flow cytometry, fewer EPCs were defined within the peripheral circulation of growth-restricted babies. Functional assays showed that ECFCs derived from these growth-restricted cases had intrinsically impaired proliferation, migration, matrix-metalloproteinase (MMP-2) production, and generated fewer blood vessels in a murine vasculogenic bioassay. These results demonstrated the vasculogenic capacity of human fetal ECFCs in vivo and established them as key players in human placental vasculogenesis and uterine vessel expansion. Notably, these results also showed a link between impaired function of fetal ECFCs and IUGR, which is associated with increased cardiovascular risk of both the fetus as an adult, and mother in later life. From these findings it could be speculated, that intrinsic changes in ECFC-biology may be the causative link between IUGR and fetal and maternal cardiovascular susceptibility. Insight into these processes may contribute to early diagnosis, prevention and treatment of IUGR and associated conditions.
10

Loss of vascular homeostasis with age : correlation of structural changes in endothelial glycosaminoglycans with endothelial progenitor cell function

Williamson, Kate January 2012 (has links)
Ageing poses one of the largest risk factors for the development of cardiovascular disease (CVD). The increased propensity towards vascular pathology with advancing age maybe explained, in part, by a reduction in the ability of circulating endothelial progenitor cells (EPCs) to contribute to vascular repair and regeneration. Among all current putative EPC populations, outgrowth endothelial cells (OECs) display the most features consistent with a human postnatal vasculogenic cell. Cell-surface heparan sulfate (HS) proteoglycans, by virtue of specific sulfated domains within the glycosaminoglycan chain, are able to bind and modulate the activities of a variety of proteins important for EPC mobilisation, homing and function at sites requiring neovascularization. This study aimed to determine if human OEC function is impaired with age, and to ascertain whether this is accompanied by changes in the fine structure of OEC HS.Using in vitro cell culture methods, OECs were isolated from healthy subjects across an age range and cell phenotype was verified by the demonstration of numerous endothelial, but not hematopoietic, cell characteristics. The functional capacity of peripheral blood derived OECs from young and old subjects, and comparative cord blood derived OECs, was assessed in terms of their susceptibility to apoptosis, proliferative, migratory and tube-forming capabilities. In vitro scratch and transwell migration assays revealed that the migratory capacity of peripheral blood derived OECs isolated from old subjects was impaired in comparison to those from young subjects and cord blood derived OECs. Structural analysis of HS by high performance liquid chromatography (HPLC) demonstrated a significant reduction in the relative percentage of the trisulfated disaccharide, 2-O-sulfated-uronic acid, N, 6-O-sulfated-glucosamine (UA[2S]-GlcNS[6S]), within OEC HS with age (r = -0.847, p=<0.01). Moreover, a decline in the migratory response of OECs towards a gradient of VEGF significantly correlated with the percentage expression of this disaccharide (r = 0.840, p<0.01). Disruption of cell surface HS by pre-treatment with heparinase I and III was found to significantly reduce the VEGF-induced migratory response of peripheral blood derived OECs isolated from young subjects to levels similar to that observed for OECs from older individuals. Understanding the role of HS in regulating the directional migration of EPCs to sites requiring neovascularization and developing approaches to facilitate EPC migration may aid in the design of more successful strategies to optimise the regenerative capacity of these cells in the ageing vasculature.

Page generated in 0.1072 seconds