Network


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

Hotspot


Dive into the research topics where Russell Hughes is active.

Publication


Featured researches published by Russell Hughes.


International Journal of Experimental Pathology | 2004

Current methods for assaying angiogenesis in vitro and in vivo

Carolyn A. Staton; Stephen M. Stribbling; Simon Tazzyman; Russell Hughes; Nicola J. Brown; Claire E. Lewis

Angiogenesis, the development of new blood vessels from an existing vasculature, is essential in normal developmental processes and in numerous pathologies, including diabetic retinopathy, psoriasis and tumour growth and metastases. One of the problems faced by angiogenesis researchers has been the difficulty of finding suitable methods for assessing the effects of regulators of the angiogenic response. The ideal assay would be reliable, technically straightforward, easily quantifiable and, most importantly, physiologically relevant. Here, we review the advantages and limitations of the principal assays in use, including those for the proliferation, migration and differentiation of endothelial cells in vitro, vessel outgrowth from organ cultures and in vivo assays such as sponge implantation, corneal, chamber, zebrafish, chick chorioallantoic membrane (CAM) and tumour angiogenesis models.


Biochimica et Biophysica Acta | 2009

Tumor-associated macrophages: Effectors of angiogenesis and tumor progression

Seth B. Coffelt; Russell Hughes; Claire E. Lewis

Tumor-associated macrophages (TAMs) are a prominent inflammatory cell population in many tumor types residing in both perivascular and avascular, hypoxic regions of these tissues. Analysis of TAMs in human tumor biopsies has shown that they express a variety of tumor-promoting factors and evidence from transgenic murine tumor models has provided unequivocal evidence for the importance of these cells in driving angiogenesis, lymphangiogenesis, immunosuppression, and metastasis. This review will summarize the mechanisms by which monocytes are recruited into tumors, their myriad, tumor-promoting functions within tumors, and the influence of the tumor microenvironment in driving these activities. We also discuss recent attempts to both target/destroy TAMs and exploit them as delivery vehicles for anti-cancer gene therapy.


Blood | 2009

Hypoxia inducible factors 1 and 2 are important transcriptional effectors in primary macrophages experiencing hypoxia

Hsin-Yu Fang; Russell Hughes; Craig Murdoch; Seth B. Coffelt; Subhra K. Biswas; Adrian Llewllyn Harris; Randall S. Johnson; Hongxia Z. Imityaz; M. Celeste Simon; Erik Fredlund; Florian R. Greten; Jordi Rius; Claire E. Lewis

Ischemia exists in many diseased tissues, including arthritic joints, atherosclerotic plaques, and malignant tumors. Macrophages accumulate in these sites and up-regulate hypoxia-inducible transcription factors (HIFs) 1 and 2 in response to the hypoxia present. Here we show that the gene expression profile in primary human and murine macrophages changes markedly when they are exposed to hypoxia for 18 hours. For example, they were seen to up-regulate the cell surface receptors, CXCR4 and GLUT1, and the potent, tumor-promoting cytokines, vascular endothelial growth factor A, interleukin (IL)-1beta and IL-8, adrenomedullin, CXCR4, and angiopoietin-2. Hypoxia also stimulated their expression and/or phosphorylation of various proteins in the nuclear factor-kappaB (NF-kappaB) signaling pathway. We then used both genetic and pharmacologic methods to manipulate the levels of HIFs-1alpha and 2alpha or NF-kappaB in primary macrophages to elucidate their role in the hypoxic induction of many of these key genes. These studies showed that both HIF-1 and -2, but not NF-kappaB, are important transcriptional effectors regulating the responses of macrophages to such a period of hypoxia. Further studies using experimental mouse models are now warranted to investigate the role of such macrophage responses in the progression of various diseased tissues, such as malignant tumors.


Cancer Research | 2015

Perivascular M2 Macrophages Stimulate Tumor Relapse after Chemotherapy

Russell Hughes; Bin-Zhi Qian; Charlotte Rowan; Munitta Muthana; Ioanna Keklikoglou; Oakley C. Olson; Simon Tazzyman; Sarah Danson; Christina L. Addison; Mark Clemons; Ana M. Gonzalez-Angulo; Johanna A. Joyce; Michele De Palma; Jeffrey W. Pollard; Claire E. Lewis

Tumor relapse after chemotherapy-induced regression is a major clinical problem, because it often involves inoperable metastatic disease. Tumor-associated macrophages (TAM) are known to limit the cytotoxic effects of chemotherapy in preclinical models of cancer. Here, we report that an alternatively activated (M2) subpopulation of TAMs (MRC1(+)TIE2(Hi)CXCR4(Hi)) accumulate around blood vessels in tumors after chemotherapy, where they promote tumor revascularization and relapse, in part, via VEGF-A release. A similar perivascular, M2-related TAM subset was present in human breast carcinomas and bone metastases after chemotherapy. Although a small proportion of M2 TAMs were also present in hypoxic tumor areas, when we genetically ablated their ability to respond to hypoxia via hypoxia-inducible factors 1 and 2, tumor relapse was unaffected. TAMs were the predominant cells expressing immunoreactive CXCR4 in chemotherapy-treated mouse tumors, with the highest levels expressed by MRC1(+) TAMs clustering around the tumor vasculature. Furthermore, the primary CXCR4 ligand, CXCL12, was upregulated in these perivascular sites after chemotherapy, where it was selectively chemotactic for MRC1(+) TAMs. Interestingly, HMOX-1, a marker of oxidative stress, was also upregulated in perivascular areas after chemotherapy. This enzyme generates carbon monoxide from the breakdown of heme, a gas known to upregulate CXCL12. Finally, pharmacologic blockade of CXCR4 selectively reduced M2-related TAMs after chemotherapy, especially those in direct contact with blood vessels, thereby reducing tumor revascularization and regrowth. Our studies rationalize a strategy to leverage chemotherapeutic efficacy by selectively targeting this perivascular, relapse-promoting M2-related TAM cell population.


Breast Cancer Research | 2007

Inflammation and breast cancer. Microenvironmental factors regulating macrophage function in breast tumours: hypoxia and angiopoietin-2

Claire E. Lewis; Russell Hughes

Considerable evidence has now accumulated for tumour-associated macrophages stimulating key aspects of tumour progression, including the proliferation, survival and metastasis of tumour cells, tumour angiogenesis and suppression of the anti-tumour functions of other immune effectors at the tumour site. Tumour micro-environmental factors such as hypoxia have profound, direct effects on these cells, stimulating many of their pro-tumour functions. Hypoxia also does so indirectly by stimulating the release of the cytokine angiopoietin-2 from tumour cells and tumour blood vessels. This in turn then recruits Tie-2-expressing monocytes into tumours from the bloodstream and inhibits their production of anti-apoptotic and anti-angiogenic cytokines.


Cancer Research | 2013

Macrophage Delivery of an Oncolytic Virus Abolishes Tumor Regrowth and Metastasis After Chemotherapy or Irradiation

Munitta Muthana; Samuel Rodrigues; Yung-Yi Chen; Abigail F. Welford; Russell Hughes; Simon Tazzyman; Magnus Essand; Fiona Morrow; Claire E. Lewis

Frontline anticancer therapies such as chemotherapy and irradiation often slow tumor growth, but tumor regrowth and spread to distant sites usually occurs after the conclusion of treatment. We recently showed that macrophages could be used to deliver large quantities of a hypoxia-regulated, prostate-specific oncolytic virus (OV) to prostate tumors. In the current study, we show that administration of such OV-armed macrophages 48 hours after chemotherapy (docetaxel) or tumor irradiation abolished the posttreatment regrowth of primary prostate tumors in mice and their spread to the lungs for up to 27 or 40 days, respectively. It also significantly increased the lifespan of tumor-bearing mice compared with those given docetaxel or irradiation alone. These new findings suggest that such a novel, macrophage-based virotherapy could be used to markedly increase the efficacy of chemotherapy and irradiation in patients with prostate cancer.


Nature Communications | 2015

Directing cell therapy to anatomic target sites in vivo with magnetic resonance targeting

Munitta Muthana; Aneurin J. Kennerley; Russell Hughes; Ester Fagnano; Jay Richardson; Melanie Paul; Craig Murdoch; Fiona Wright; Christopher Payne; Mark F. Lythgoe; Neil Farrow; Jon Dobson; Joe Conner; Jim M. Wild; Claire E. Lewis

Cell-based therapy exploits modified human cells to treat diseases but its targeted application in specific tissues, particularly those lying deep in the body where direct injection is not possible, has been problematic. Here we use a magnetic resonance imaging (MRI) system to direct macrophages carrying an oncolytic virus, Seprehvir, into primary and metastatic tumour sites in mice. To achieve this, we magnetically label macrophages with super-paramagnetic iron oxide nanoparticles and apply pulsed magnetic field gradients in the direction of the tumour sites. Magnetic resonance targeting guides macrophages from the bloodstream into tumours, resulting in increased tumour macrophage infiltration and reduction in tumour burden and metastasis. Our study indicates that clinical MRI scanners can not only track the location of magnetically labelled cells but also have the potential to steer them into one or more target tissues.


Bone | 2015

Rapid modification of the bone microenvironment following short-term treatment with Cabozantinib in vivo

Marie Therese Haider; Keith D. Hunter; Simon P. Robinson; Timothy J. Graham; Eva Corey; T. Neil Dear; Russell Hughes; Nicola J. Brown; Ingunn Holen

Introduction Bone metastasis remains incurable with treatment restricted to palliative care. Cabozantinib (CBZ) is targeted against multiple receptor tyrosine kinases involved in tumour pathobiology, including hepatocyte growth factor receptor (MET) and vascular endothelial growth factor receptor 2 (VEGFR-2). CBZ has demonstrated clinical activity in advanced prostate cancer with resolution of lesions visible on bone scans, implicating a potential role of the bone microenvironment as a mediator of CBZ effects. We characterised the effects of short-term administration of CBZ on bone in a range of in vivo models to determine how CBZ affects bone in the absence of tumour. Methods Studies were performed in a variety of in vivo models including male and female BALB/c nude mice (age 6–17-weeks). Animals received CBZ (30 mg/kg, 5 × weekly) or sterile H2O control for 5 or 10 days. Effects on bone integrity (μCT), bone cell activity (PINP, TRAP ELISA), osteoblast and osteoclast number/mm trabecular bone surface, area of epiphyseal growth plate cartilage, megakaryocyte numbers and bone marrow composition were assessed. Effects of longer-term treatment (15-day & 6-week administration) were assessed in male NOD/SCID and beige SCID mice. Results CBZ treatment had significant effects on the bone microenvironment, including reduced osteoclast and increased osteoblast numbers compared to control. Trabecular bone structure was altered after 8 administrations. A significant elongation of the epiphyseal growth plate, in particular the hypertrophic chondrocyte zone, was observed in all CBZ treated animals irrespective of administration schedule. Both male and female BALB/c nude mice had increased megakaryocyte numbers/mm2 tissue after 10-day CBZ treatment, in addition to vascular ectasia, reduced bone marrow cellularity and extravasation of red blood cells into the extra-vascular bone marrow. All CBZ-induced effects were transient and rapidly lost following cessation of treatment. Conclusion Short-term administration of CBZ induces rapid, reversible effects on the bone microenvironment in vivo highlighting a potential role in mediating treatment responses.


Archive | 2011

Role of Tumour-Associated Macrophages in the Regulation of Angiogenesis

Russell Hughes; Hsin-Yu Fang; Munitta Muthana; Claire E. Lewis

As mentioned in previous chapters, tumours consist not only of malignant cells but also of various stromal cell types including tumour-associated macrophages (TAMs) (Sica et al. 2008). One early sign that TAMs might influence tumour angiogenesis was the finding that TAM numbers positively correlate with tumour angiogenesis in breast carcinomas (Leek et al. 1996). Several subsequent studies have confirmed such a link in a wide array of tumour types (Aharinejad et al. 2004; Bailey et al. 2007; Koide et al. 2004; Ohta et al. 2003; Saji et al. 2001) and showed that high-TAM numbers are also often linked to poor prognosis (Bingle et al. 2002; Lewis and Pollard 2006). However, definitive evidence for the pro-angiogenic effect of TAMs in tumours was provided using various murine tumour models.


bioRxiv | 2018

Non-canonical HIF-1 stabilization is essential for intestinal tumorigenesis

Nadine Rohwer; Sandra Jumpertz; Merve Erdem; Antje Egners; Klaudia Theresa Warzecha; Athanassios Fragoulis; Anja Kuehl; Rafael Kramann; Sabine Neuss; Ines Rudolph; Tobias Endermann; Christin Zasada; Ivayla Apostolova; Marco Gerling; Stefan Kempa; Russell Hughes; Claire E. Lewis; Winfried Brenner; Maciej Malinowski; Martin Stockmann; Lutz Schomburg; William J. Faller; Owen J. Sansom; Frank Tacke; Markus Morkel; Thorsten Cramer

The hypoxia-inducible transcription factor HIF-1 is appreciated as a promising target for cancer therapy. However, conditional deletion of HIF-1 and HIF-1 target genes in cells of the tumor microenvironment can result in accelerated tumor growth, calling for a detailed characterization of the cellular context to fully comprehend HIF-1’s role in tumorigenesis. We dissected cell type-specific functions of HIF-1 for intestinal tumorigenesis by lineage-restricted deletion of the Hif1a locus. Intestinal epithelial cell-specific Hif1a loss reduced activation of wnt/β-catenin, tumor-specific metabolism and inflammation, significantly inhibiting tumor growth. Deletion of Hif1a in myeloid cells reduced the expression of fibroblast-activating factors in tumor-associated macrophages resulting in decreased abundance of tumor-associated fibroblasts and robustly reduced tumor formation. Interestingly, hypoxia was detectable only sparsely and without spatial association with nuclear HIF-1α in intestinal adenomas, pointing towards a functional importance of hypoxia-independent, i.e. non-canonical HIF-1 stabilization that has not been previously appreciated. This adds a further layer of complexity to the regulation of HIF-1α and suggests that hypoxia and HIF-1α stabilization can be uncoupled in cancer. Collectively, our data show that HIF-1 is a pivotal pro-tumorigenic factor for intestinal tumor formation, controlling key oncogenic programs in both the epithelial tumor compartment and the tumor microenvironment.

Collaboration


Dive into the Russell Hughes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fiona Wright

University of Sheffield

View shared research outputs
Top Co-Authors

Avatar

Hsin-Yu Fang

University of Sheffield

View shared research outputs
Top Co-Authors

Avatar

Ingunn Holen

University of Sheffield

View shared research outputs
Top Co-Authors

Avatar

Jim M. Wild

University of Sheffield

View shared research outputs
Top Co-Authors

Avatar

Joe Conner

University of Sheffield

View shared research outputs
Researchain Logo
Decentralizing Knowledge