Network


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

Hotspot


Dive into the research topics where Paul K. Potter is active.

Publication


Featured researches published by Paul K. Potter.


PLOS Genetics | 2011

HIF-VEGF Pathways Are Critical for Chronic Otitis Media in Junbo and Jeff Mouse Mutants

Michael Cheeseman; Hayley E. Tyrer; Debbie Williams; Tertius Hough; Paras Pathak; Maria R. Romero; Helen Hilton; Sulzhan Bali; Andrew E. Parker; Lucie Vizor; Tom Purnell; Kate Vowell; Sara Wells; Mahmood F. Bhutta; Paul K. Potter; Steve D.M. Brown

Otitis media with effusion (OME) is the commonest cause of hearing loss in children, yet the underlying genetic pathways and mechanisms involved are incompletely understood. Ventilation of the middle ear with tympanostomy tubes is the commonest surgical procedure in children and the best treatment for chronic OME, but the mechanism by which they work remains uncertain. As hypoxia is a common feature of inflamed microenvironments, moderation of hypoxia may be a significant contributory mechanism. We have investigated the occurrence of hypoxia and hypoxia-inducible factor (HIF) mediated responses in Junbo and Jeff mouse mutant models, which develop spontaneous chronic otitis media. We found that Jeff and Junbo mice labeled in vivo with pimonidazole showed cellular hypoxia in inflammatory cells in the bulla lumen, and in Junbo the middle ear mucosa was also hypoxic. The bulla fluid inflammatory cell numbers were greater and the upregulation of inflammatory gene networks were more pronounced in Junbo than Jeff. Hif-1α gene expression was elevated in bulla fluid inflammatory cells, and there was upregulation of its target genes including Vegfa in Junbo and Jeff. We therefore investigated the effects in Junbo of small-molecule inhibitors of VEGFR signaling (PTK787, SU-11248, and BAY 43-9006) and destabilizing HIF by inhibiting its chaperone HSP90 with 17-DMAG. We found that both classes of inhibitor significantly reduced hearing loss and the occurrence of bulla fluid and that VEGFR inhibitors moderated angiogenesis and lymphangiogenesis in the inflamed middle ear mucosa. The effectiveness of HSP90 and VEGFR signaling inhibitors in suppressing OM in the Junbo model implicates HIF–mediated VEGF as playing a pivotal role in OM pathogenesis. Our analysis of the Junbo and Jeff mutants highlights the role of hypoxia and HIF–mediated pathways, and we conclude that targeting molecules in HIF–VEGF signaling pathways has therapeutic potential in the treatment of chronic OM.


Stem Cells | 2012

Age‐Related Changes in Speed and Mechanism of Adult Skeletal Muscle Stem Cell Migration

Henry Collins-Hooper; Thomas E. Woolley; Louise Dyson; Anand Patel; Paul K. Potter; Ruth E. Baker; Eamonn A. Gaffney; Philip K. Maini; Philip R. Dash; Ketan Patel

Skeletal muscle undergoes a progressive age‐related loss in mass and function. Preservation of muscle mass depends in part on satellite cells, the resident stem cells of skeletal muscle. Reduced satellite cell function may contribute to the age‐associated decrease in muscle mass. Here, we focused on characterizing the effect of age on satellite cell migration. We report that aged satellite cells migrate at less than half the speed of young cells. In addition, aged cells show abnormal membrane extension and retraction characteristics required for amoeboid‐based cell migration. Aged satellite cells displayed low levels of integrin expression. By deploying a mathematical model approach to investigate mechanism of migration, we have found that young satellite cells move in a random “memoryless” manner, whereas old cells demonstrate superdiffusive tendencies. Most importantly, we show that nitric oxide, a key regulator of cell migration, reversed the loss in migration speed and reinstated the unbiased mechanism of movement in aged satellite cells. Finally, we found that although hepatocyte growth factor increased the rate of aged satellite cell movement, it did not restore the memoryless migration characteristics displayed in young cells. Our study shows that satellite cell migration, a key component of skeletal muscle regeneration, is compromised during aging. However, we propose clinically approved drugs could be used to overcome these detrimental changes. STEM CELLS2012;30:1182–1195


PLOS Genetics | 2013

Unlocking the Bottleneck in Forward Genetics Using Whole-Genome Sequencing and Identity by Descent to Isolate Causative Mutations

Katherine R. Bull; Andrew J. Rimmer; Owen M. Siggs; Lisa A. Miosge; Carla M. Roots; Anselm Enders; Edward M. Bertram; Tanya L. Crockford; Belinda Whittle; Paul K. Potter; Michelle Simon; Ann-Marie Mallon; Steve D.M. Brown; Bruce Beutler; Christopher C. Goodnow; Gerton Lunter; Richard J. Cornall

Forward genetics screens with N-ethyl-N-nitrosourea (ENU) provide a powerful way to illuminate gene function and generate mouse models of human disease; however, the identification of causative mutations remains a limiting step. Current strategies depend on conventional mapping, so the propagation of affected mice requires non-lethal screens; accurate tracking of phenotypes through pedigrees is complex and uncertain; out-crossing can introduce unexpected modifiers; and Sanger sequencing of candidate genes is inefficient. Here we show how these problems can be efficiently overcome using whole-genome sequencing (WGS) to detect the ENU mutations and then identify regions that are identical by descent (IBD) in multiple affected mice. In this strategy, we use a modification of the Lander-Green algorithm to isolate causative recessive and dominant mutations, even at low coverage, on a pure strain background. Analysis of the IBD regions also allows us to calculate the ENU mutation rate (1.54 mutations per Mb) and to model future strategies for genetic screens in mice. The introduction of this approach will accelerate the discovery of causal variants, permit broader and more informative lethal screens to be used, reduce animal costs, and herald a new era for ENU mutagenesis.


Nature Genetics | 2016

Genome-wide association of multiple complex traits in outbred mice by ultra-low-coverage sequencing

Jérôme Nicod; Robert W. Davies; Na Cai; Carl Hassett; Leo Goodstadt; Cormac Cosgrove; Benjamin K Yee; Vikte Lionikaite; Rebecca E McIntyre; Carol Ann Remme; Elisabeth M. Lodder; J.S. Gregory; Tertius Hough; Russell Joynson; Hayley Phelps; Barbara Nell; Clare Rowe; Joe Wood; Alison Walling; Nasrin Bopp; Amarjit Bhomra; Polinka Hernandez-Pliego; Jacques Callebert; Richard M. Aspden; Nick P. Talbot; Peter A. Robbins; Mark Harrison; Martin Fray; Jean-Marie Launay; Yigal M. Pinto

Two bottlenecks impeding the genetic analysis of complex traits in rodents are access to mapping populations able to deliver gene-level mapping resolution and the need for population-specific genotyping arrays and haplotype reference panels. Here we combine low-coverage (0.15×) sequencing with a new method to impute the ancestral haplotype space in 1,887 commercially available outbred mice. We mapped 156 unique quantitative trait loci for 92 phenotypes at a 5% false discovery rate. Gene-level mapping resolution was achieved at about one-fifth of the loci, implicating Unc13c and Pgc1a at loci for the quality of sleep, Adarb2 for home cage activity, Rtkn2 for intensity of reaction to startle, Bmp2 for wound healing, Il15 and Id2 for several T cell measures and Prkca for bone mineral content. These findings have implications for diverse areas of mammalian biology and demonstrate how genome-wide association studies can be extended via low-coverage sequencing to species with highly recombinant outbred populations.


Neurobiology of Aging | 2015

Genetic background influences age-related decline in visual and nonvisual retinal responses, circadian rhythms, and sleep

Gareth Banks; Ines Heise; Becky Starbuck; Tamzin Osborne; Laura Wisby; Paul K. Potter; Ian J. Jackson; Russell G. Foster; Stuart N. Peirson; Patrick M. Nolan

The circadian system is entrained to the environmental light/dark cycle via retinal photoreceptors and regulates numerous aspects of physiology and behavior, including sleep. These processes are all key factors in healthy aging showing a gradual decline with age. Despite their importance, the exact mechanisms underlying this decline are yet to be fully understood. One of the most effective tools we have to understand the genetic factors underlying these processes are genetically inbred mouse strains. The most commonly used reference mouse strain is C57BL/6J, but recently, resources such as the International Knockout Mouse Consortium have started producing large numbers of mouse mutant lines on a pure genetic background, C57BL/6N. Considering the substantial genetic diversity between mouse strains we expect there to be phenotypic differences, including differential effects of aging, in these and other strains. Such differences need to be characterized not only to establish how different mouse strains may model the aging process but also to understand how genetic background might modify age-related phenotypes. To ascertain the effects of aging on sleep/wake behavior, circadian rhythms, and light input and whether these effects are mouse strain-dependent, we have screened C57BL/6J, C57BL/6N, C3H-HeH, and C3H-Pde6b+ mouse strains at 5 ages throughout their life span. Our data show that sleep, circadian, and light input parameters are all disrupted by the aging process. Moreover, we have cataloged a number of strain-specific aging effects, including the rate of cataract development, decline in the pupillary light response, and changes in sleep fragmentation and the proportion of time spent asleep.


Human Molecular Genetics | 2015

The severity of retinal pathology in homozygous Crb1rd8/rd8 mice is dependent on additional genetic factors

Ulrich F.O. Luhmann; Livia S. Carvalho; Sophia-Martha kleine Holthaus; Jill A. Cowing; Simon Greenaway; Colin Chu; Philipp Herrmann; Alexander J. Smith; Peter M.G. Munro; Paul K. Potter; James W. Bainbridge; Robin R. Ali

Understanding phenotype–genotype correlations in retinal degeneration is a major challenge. Mutations in CRB1 lead to a spectrum of autosomal recessive retinal dystrophies with variable phenotypes suggesting the influence of modifying factors. To establish the contribution of the genetic background to phenotypic variability associated with the Crb1rd8/rd8 mutation, we compared the retinal pathology of Crb1rd8/rd8/J inbred mice with that of two Crb1rd8/rd8 lines backcrossed with C57BL/6JOlaHsd mice. Topical endoscopic fundal imaging and scanning laser ophthalmoscopy fundus images of all three Crb1rd8/rd8 lines showed a significant increase in the number of inferior retinal lesions that was strikingly variable between the lines. Optical coherence tomography, semithin, ultrastructural morphology and assessment of inflammatory and vascular marker by immunohistochemistry and quantitative reverse transcriptase-polymerase chain reaction revealed that the lesions were associated with photoreceptor death, Müller and microglia activation and telangiectasia-like vascular remodelling—features that were stable in the inbred, variable in the second, but virtually absent in the third Crb1rd8/rd8 line, even at 12 months of age. This suggests that the Crb1rd8/rd8 mutation is necessary, but not sufficient for the development of these degenerative features. By whole-genome SNP analysis of the genotype–phenotype correlation, a candidate region on chromosome 15 was identified. This may carry one or more genetic modifiers for the manifestation of the retinal pathology associated with mutations in Crb1. This study also provides insight into the nature of the retinal vascular lesions that likely represent a clinical correlate for the formation of retinal telangiectasia or Coats-like vasculopathy in patients with CRB1 mutations that are thought to depend on such genetic modifiers.


PLOS ONE | 2015

N-ethyl-N-Nitrosourea (ENU) induced mutations within the klotho gene lead to ectopic calcification and reduced lifespan in mouse models.

Christopher T. Esapa; Fadil M. Hannan; Valerie N. Babinsky; Paul K. Potter; Gethin P. Thomas; Peter I. Croucher; Matthew A. Brown; Steve D. M. Brown; Roger D. Cox; Rajesh V. Thakker

Ectopic calcification (EC), which is the pathological deposition of calcium and phosphate in extra-skeletal tissues, may be associated with hypercalcaemic and hyperphosphataemic disorders, or it may occur in the absence of metabolic abnormalities. In addition, EC may be inherited as part of several monogenic disorders and studies of these have provided valuable insights into the metabolic pathways regulating mineral metabolism. For example, studies of tumoural calcinosis, a disorder characterised by hyperphosphataemia and progressive EC, have revealed mutations of fibroblast growth factor 23 (FGF23), polypeptide N-acetyl galactosaminyltransferase 3 (GALNT3) and klotho (KL), which are all part of a phosphate-regulating pathway. However, such studies in humans are limited by the lack of available large families with EC, and to facilitate such studies we assessed the progeny of mice treated with the chemical mutagen N-ethyl-N-nitrosourea (ENU) for EC. This identified two mutants with autosomal recessive forms of EC, and reduced lifespan, designated Ecalc1 and Ecalc2. Genetic mapping localized the Ecalc1 and Ecalc2 loci to a 11.0 Mb region on chromosome 5 that contained the klotho gene (Kl), and DNA sequence analysis identified nonsense (Gln203Stop) and missense (Ile604Asn) Kl mutations in Ecalc1 and Ecalc2 mice, respectively. The Gln203Stop mutation, located in KL1 domain, was severely hypomorphic and led to a 17-fold reduction of renal Kl expression. The Ile604Asn mutation, located in KL2 domain, was predicted to impair klotho protein stability and in vitro expression studies in COS-7 cells revealed endoplasmic reticulum retention of the Ile604Asn mutant. Further phenotype studies undertaken in Ecalc1 (kl203X/203X) mice demonstrated elevations in plasma concentrations of phosphate, FGF23 and 1,25-dihydroxyvitamin D. Thus, two allelic variants of Kl that develop EC and represent mouse models for tumoural calcinosis have been established.


Nature Communications | 2016

Novel gene function revealed by mouse mutagenesis screens for models of age-related disease

Paul K. Potter; Michael R. Bowl; Prashanthini Jeyarajan; Laura Wisby; Andrew Blease; Michelle Goldsworthy; Michelle Simon; Simon Greenaway; Vincent Michel; Alun R. Barnard; Carlos Aguilar; Thomas Agnew; Gareth Banks; Andrew Blake; Lauren Chessum; Joanne Dorning; Sara Falcone; Laurence Goosey; Shelley Harris; Andy Haynes; Ines Heise; Rosie Hillier; Tertius Hough; Angela Hoslin; Marie Hutchison; Ruairidh King; Saumya Kumar; Heena V. Lad; Gemma Law; Robert E. MacLaren

Determining the genetic bases of age-related disease remains a major challenge requiring a spectrum of approaches from human and clinical genetics to the utilization of model organism studies. Here we report a large-scale genetic screen in mice employing a phenotype-driven discovery platform to identify mutations resulting in age-related disease, both late-onset and progressive. We have utilized N-ethyl-N-nitrosourea mutagenesis to generate pedigrees of mutagenized mice that were subject to recurrent screens for mutant phenotypes as the mice aged. In total, we identify 105 distinct mutant lines from 157 pedigrees analysed, out of which 27 are late-onset phenotypes across a range of physiological systems. Using whole-genome sequencing we uncover the underlying genes for 44 of these mutant phenotypes, including 12 late-onset phenotypes. These genes reveal a number of novel pathways involved with age-related disease. We illustrate our findings by the recovery and characterization of a novel mouse model of age-related hearing loss.


Mammalian Genome | 2015

Current strategies for mutation detection in phenotype-driven screens utilising next generation sequencing

Michelle Simon; Eva Marie Y Moresco; Katherine R. Bull; Saumya Kumar; Ann-Marie Mallon; Bruce Beutler; Paul K. Potter

Mutagenesis-based screens in mice are a powerful discovery platform to identify novel genes or gene functions associated with disease phenotypes. An N-ethyl-N-nitrosourea (ENU) mutagenesis screen induces single nucleotide variants randomly in the mouse genome. Subsequent phenotyping of mutant and wildtype mice enables the identification of mutated pathways resulting in phenotypes associated with a particular ENU lesion. This unbiased approach to gene discovery conducts the phenotyping with no prior knowledge of the functional mutations. Before the advent of affordable next generation sequencing (NGS), ENU variant identification was a limiting step in gene characterization, akin to ‘finding a needle in a haystack’. The emergence of a reliable reference genome alongside advances in NGS has propelled ENU mutation discovery from an arduous, time-consuming exercise to an effective and rapid form of mutation discovery. This has permitted large mouse facilities worldwide to use ENU for novel mutation discovery in a high-throughput manner, helping to accelerate basic science at the mechanistic level. Here, we describe three different strategies used to identify ENU variants from NGS data and some of the subsequent steps for mutation characterisation.


Laboratory Animals | 2017

Does age matter? The impact of rodent age on study outcomes

Samuel J. Jackson; Nick Andrews; Doug Ball; Ilaria Bellantuono; James R. J. Gray; Lamia Hachoumi; Alan Holmes; Judy Latcham; Anja Petrie; Paul K. Potter; Andrew S.C. Rice; Alison Ritchie; Michelle Stewart; Carol Strepka; Mark Yeoman; Kathryn Chapman

Rodent models produce data which underpin biomedical research and non-clinical drug trials, but translation from rodents into successful clinical outcomes is often lacking. There is a growing body of evidence showing that improving experimental design is key to improving the predictive nature of rodent studies and reducing the number of animals used in research. Age, one important factor in experimental design, is often poorly reported and can be overlooked. The authors conducted a survey to assess the age used for a range of models, and the reasoning for age choice. From 297 respondents providing 611 responses, researchers reported using rodents most often in the 6–20 week age range regardless of the biology being studied. The age referred to as ‘adult’ by respondents varied between six and 20 weeks. Practical reasons for the choice of rodent age were frequently given, with increased cost associated with using older animals and maintenance of historical data comparability being two important limiting factors. These results highlight that choice of age is inconsistent across the research community and often not based on the development or cellular ageing of the system being studied. This could potentially result in decreased scientific validity and increased experimental variability. In some cases the use of older animals may be beneficial. Increased scientific rigour in the choice of the age of rodent may increase the translation of rodent models to humans.

Collaboration


Dive into the Paul K. Potter's collaboration.

Top Co-Authors

Avatar

Michelle Simon

Medical Research Council

View shared research outputs
Top Co-Authors

Avatar

Andrew Blease

Medical Research Council

View shared research outputs
Top Co-Authors

Avatar

Gareth Banks

University College London

View shared research outputs
Top Co-Authors

Avatar

Sara Wells

Medical Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tertius Hough

Medical Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ann-Marie Mallon

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ines Heise

Medical Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge