Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sarah Chambers is active.

Publication


Featured researches published by Sarah Chambers.


Immunobiology | 2013

The role of immune-related myeloid cells in angiogenesis.

Sarah Chambers; Christina L. O’Neill; T. Michelle O’Doherty; Reinhold Medina; Alan W. Stitt

Macrophage function is not restricted to the innate and adaptive immune responses, but also includes host defence, wound healing, angiogenesis and homeostatic processes. Within the spectrum of macrophage activation there are two extremes: M1 classically activated macrophages which have a pro-inflammatory phenotype, and M2 alternatively activated macrophages which are pro-angiogenic and anti-inflammatory. An important property of macrophages is their plasticity to switch from one phenotype to the other and they can be defined in their polarisation state at any point between the two extremes. In order to determine what stage of activation macrophages are in, it is essential to profile various phenotypic markers for their identification. This review describes the angiogenic role for myeloid cells: circulating monocytes, Tie-2 expressing monocytes (TEMs), myeloid-derived suppressor cells (MDSCs), tumour associated macrophages (TAMs), and neutrophils. Each cell type is discussed by phenotype, roles within angiogenesis and possible targets as a cell therapy. In addition, we also refer to our own research on myeloid angiogenic cells (MACs), outlining their ability to induce angiogenesis and their similarities to alternatively activated M2 macrophages. MACs significantly contribute to vascular repair through paracrine mechanisms as they lack the capacity to differentiate into endothelial cells. Since MACs also retain plasticity, phenotypic changes can occur according to disease states and the surrounding microenvironment. This pro-angiogenic potential of MACs could be harnessed as a novel cellular therapy for the treatment of ischaemic diseases, such as diabetic retinopathy, hind limb ischaemia and myocardial infarction; however, caution needs to be taken when MACs are delivered into an inflammatory milieu.


Stem Cells | 2013

Ex Vivo Expansion of Human Outgrowth Endothelial Cells Leads to IL-8-Mediated Replicative Senescence and Impaired Vasoreparative Function

Reinhold Medina; Christina O'Neill; T. Michelle O'Doherty; Sarah Chambers; Jasenka Guduric-Fuchs; Jessica Neisen; David Waugh; David Simpson; Alan W. Stitt

Harnessing outgrowth endothelial cells (OECs) for vasoreparative therapy and tissue engineering requires efficient ex vivo expansion. How such expansion impacts on OEC function is largely unknown. In this study, we show that OECs become permanently cell‐cycle arrested after ex vivo expansion, which is associated with enlarged cell size, β‐galactosidase activity, DNA damage, tumor suppressor pathway activation, and significant transcriptome changes. These senescence hallmarks were coupled with low telomerase activity and telomere shortening, indicating replicative senescence. OEC senescence limited their regenerative potential by impairing vasoreparative properties in vitro and in vivo. Integrated transcriptome‐proteome analysis identified inflammatory signaling pathways as major mechanistic components of the OEC senescence program. In particular, IL8 was an important facilitator of this senescence; depletion of IL8 in OECs significantly extended ex vivo lifespan, delayed replicative senescence, and enhanced function. While the ability to expand OEC numbers prior to autologous or allogeneic therapy remains a useful property, their replicative senescence and associated impairment of vasorepair needs to be considered. This study also suggests that modulation of the senescence‐associated secretory phenotype could be used to optimize OEC therapy. STEM Cells 2013;31:1657–1668


Cardiovascular Research | 2016

Endothelial cell-derived pentraxin 3 limits the vasoreparative therapeutic potential of circulating angiogenic cells

Christina O'Neill; Jasenka Guduric-Fuchs; Sarah Chambers; Michelle O'Doherty; Barbara Bottazzi; Alan W. Stitt; Reinhold Medina

Aims Circulating angiogenic cells (CACs) promote revascularization of ischaemic tissues although their underlying mechanism of action and the consequences of delivering varying number of these cells for therapy remain unknown. This study investigates molecular mechanisms underpinning CAC modulation of blood vessel formation. Methods and results CACs at low (2 × 105 cells/mL) and mid (2 × 106 cells/mL) cellular densities significantly enhanced endothelial cell tube formation in vitro, while high density (HD) CACs (2 × 107 cells/mL) significantly inhibited this angiogenic process. In vivo, Matrigel-based angiogenesis assays confirmed mid-density CACs as pro-angiogenic and HD CACs as anti-angiogenic. Secretome characterization of CAC-EC conditioned media identified pentraxin 3 (PTX3) as only present in the HD CAC-EC co-culture. Recombinant PTX3 inhibited endothelial tube formation in vitro and in vivo. Importantly, our data revealed that the anti-angiogenic effect observed in HD CAC-EC co-cultures was significantly abrogated when PTX3 bioactivity was blocked using neutralizing antibodies or PTX3 siRNA in endothelial cells. We show evidence for an endothelial source of PTX3, triggered by exposure to HD CACs. In addition, we confirmed that PTX3 inhibits fibroblast growth factor (FGF) 2-mediated angiogenesis, and that the PTX3 N-terminus, containing the FGF-binding site, is responsible for such anti-angiogenic effects. Conclusion Endothelium, when exposed to HD CACs, releases PTX3 which markedly impairs the vascular regenerative response in an autocrine manner. Therefore, CAC density and accompanying release of angiocrine PTX3 are critical considerations when using these cells as a cell therapy for ischaemic disease.


Stem Cells Translational Medicine | 2018

Preclinical Evaluation and Optimization of a Cell Therapy Using Human Cord Blood‐Derived Endothelial Colony‐Forming Cells for Ischemic Retinopathies

Emma Reid; Jasenka Guduric-Fuchs; Christina O'Neill; Lynsey-Dawn Allen; Sarah Chambers; Alan W. Stitt; Reinhold Medina

Cell therapy using endothelial progenitors holds promise for vascular repair in ischemic retinopathies. Using a well‐defined subpopulation of human cord blood‐derived endothelial progenitors known as endothelial colony‐forming cells (ECFCs), we have evaluated essential requirements for further development of this cell therapy targeting the ischemic retina, including dose response, delivery route, and toxicity. First, to evaluate therapeutic efficacy relating to cell dose, ECFCs were injected into the vitreous of mice with oxygen‐induced retinopathy. Using angiography and histology, we found that intravitreal delivery of low dose (1 × 103) ECFCs was as effective as higher cell doses (1 × 104, 1 × 105) in promoting vascular repair. Second, injection into the common carotid artery was tested as an alternative, systemic delivery route. Intracarotid ECFC delivery conferred therapeutic benefit which was comparable to intravitreal delivery using the same ECFC dose (1 × 105), although there were fewer human cells observed in the retinal vasculature following systemic delivery. Third, cell immunogenicity was evaluated by injecting ECFCs into the vitreous of healthy adult mice. Assessment of murine ocular tissues identified injected cells in the vitreous, while demonstrating integrity of the host retina. In addition, ECFCs did not invade into the retina, but remained in the vitreous, where they eventually underwent cell death within 3 days of delivery without evoking an inflammatory response. Human specific Alu sequences were not found in healthy mouse retinas after 3 days of ECFC delivery. These findings provide supportive preclinical evidence for the development of ECFCs as an efficacious cell product for ischemic retinopathies. Stem Cells Translational Medicine 2018;7:59–67


Stem Cells | 2018

The Vasoreparative Function of Myeloid Angiogenic Cells Is Impaired in Diabetes Through the Induction of IL1β

Sarah Chambers; Christina O'Neill; Jasenka Guduric-Fuchs; Kiran J. McLoughlin; Aaron Liew; Aoife M. Egan; Timothy O'Brien; Alan W. Stitt; Reinhold Medina

Myeloid angiogenic cells (MACs) promote revascularization through the paracrine release of angiogenic factors and have been harnessed as therapeutic cells for many ischemic diseases. However, their proangiogenic properties have been suggested to be diminished in diabetes. This study investigates how the diabetic milieu affects the immunophenotype and function of MACs. Both MACs isolated from diabetic conditions and healthy cells exposed to a diabetic environment were used to determine the potential of MACs as a cell therapy for diabetic‐related ischemia. MACs were isolated from human peripheral blood and characterized alongside proinflammatory macrophages M (LPS + IFNγ) and proangiogenic macrophages M (IL4). Functional changes in MACs in response to high‐d‐glucose were assessed using an in vitro 3D‐tubulogenesis assay. Phenotypic changes were determined by gene and protein expression analysis. Additionally, MACs from type 1 diabetic (T1D) patients and corresponding controls were isolated and characterized. Our evidence demonstrates MACs identity as a distinct macrophage subtype that shares M2 proangiogenic characteristics, but can be distinguished by CD163hi expression. High‐d‐glucose treatment significantly reduced MACs proangiogenic capacity, which was associated with a significant increase in IL1β mRNA and protein expression. Inhibition of IL1β abrogated the antiangiogenic effect induced by high‐d‐glucose. IL1β was also significantly upregulated in MACs isolated from T1D patients with microvascular complications compared to T1D patients without microvascular complications or nondiabetic volunteers. This study demonstrates that Type 1 diabetes and diabetic‐like conditions impair the proangiogenic and regenerative capacity of MACs, and this response is mediated by IL‐1β. Stem Cells 2018;36:834–843


Frontiers of Medicine in China | 2018

The vasoreparative potential of endothelial colony forming cells: A journey through pre-clinical studies

Christina O'Neill; Kiran J. McLoughlin; Sarah Chambers; Jasenka Guduric-Fuchs; Alan W. Stitt; Reinhold Medina

For over a decade various cell populations have been investigated for their vasoreparative potential. Cells with the capacity to promote blood vessel regeneration are commonly known as endothelial progenitor cells (EPCs); although such a definition is currently considered too simple for the complexity of cell populations involved in the reparative angiogenic process. A subset of EPCs called endothelial colony forming cells (ECFCs) have emerged as a suitable candidate for cytotherapy, primarily due to their clonogenic progenitor characteristics, unequivocal endothelial phenotype, and inherent ability to promote vasculogenesis. ECFCs can be readily isolated from human peripheral and cord blood, expanded ex vivo and used to revascularize ischemic tissues. These cells have demonstrated efficacy in several in vivo preclinical models such as the ischemic heart, retina, brain, limb, lung and kidney. This review will summarize the current pre-clinical evidence for ECFC cytotherapy and discuss their potential for clinical application.


Investigative Ophthalmology & Visual Science | 2016

Pro-Tissue Repair capacity of circulating angiogenic cells (CACs) is impaired when exposed to High Glucose

Jasenka Guduric-Fuchs; Sarah Chambers; Christina L. O’Neill; Reinhold Medina; Alan W. Stitt


Journal of Vascular Research | 2015

Vasgulogenic Properties of Endothelial Colony Forming Cells (ECFCS) in Hypoxia are Regulated by MIR-130

Jasenka Guduric-Fuchs; Christina O'Neill; B. James; Sarah Chambers; Reinhold Medina; Alan W. Stitt


Joint Meeting of the European-Society-for-Microcirculation (ESM) and European-Vascular-Biology-Organisation (EVBO) | 2015

The cellular density of Myeloid Angiogenic Cells (MACs) dictates their angiogenic function

Christina O'Neill; Michelle O'Doherty; Sarah Chambers; Jasenka Guduric-Fuchs; Barbara Bottazzi; Alan W. Stitt; Reinhold Medina


International Cellular Senescence Association (ICSA) conference | 2015

IL8 is a key component of the senescence-associated secretome in human endothelial colony-forming cells

Reinhold Medina; Christina O'Neill; Sarah Chambers; Jasenka Guduric-Fuchs; Alan W. Stitt

Collaboration


Dive into the Sarah Chambers's collaboration.

Top Co-Authors

Avatar

Alan W. Stitt

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Reinhold Medina

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Christina O'Neill

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emma Reid

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Simpson

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

David Waugh

Queen's University Belfast

View shared research outputs
Researchain Logo
Decentralizing Knowledge