Dr Paul Potter
BSc, ARCS, PhD, FRSB
Senior Lecturer and Group Leader: Disease Mechanisms and Ageing
School of Biological and Medical Sciences
Role
I am an experienced group leader and my research focuses on the genetics and pathways underlying chronic and age-related disease. I have specific interests in extracellular matrix disorders. In addition to leading my own research I collaborate with a range of groups, including the Centre for Osteoarthritis Pathogenesis at the Kennedy Institute. I was a member of the management group and a work package leader for the MouseAGE EU COST action. I continue to work on novel models of chronic and age-related disease to identify novel genes and pathways associated with disease, and to use these to search for interventions to improve health.
Areas of expertise
- Genetics and Disease,
- Ageing,
- Disease Models,
- Osteoarthritis,
- Renal Disease,
- Extracellular matrix
Teaching and supervision
Courses
Modules taught
I am Module Leader for:
- Clinical Biochemistry
- Scientific Skills
I also lecture on:
- Professional and Experimental Skills,
- Research Methods for Healthcare Scientists,
- Evidence Based Medicine,
- Independent Studies
I am also an MSc Project Supervisor.
Supervision
I have supervised a range of PhD, MSc, and undergraduate project students and will be happy to discuss opportunities to develop new projects or contribute to existing ones.
- 2014-2021, Andy Blease, Collagen I dysfunction resulting in chronic osteoarthritis (PhD).
- 2014-2018, Sara Falcone. Novel Models of Chronic Renal Disease (PhD).
- 2014-2018, Thomas Nicol. 2014. A novel model of mitochondrial dysfunction resulting in hypertrophic cardiomyopathy (PhD).
- 2017 - Pedro Romao (Diploma in Veterinary Medicine)
- 2012 - Hayley Tyrer. The role of HIF and hypoxia in acute and chronic mouse models of Otitis Media (PhD).
- 2011 - Sulzhan Bali. In vitro modelling of cellular processes in OM-BMDM studies in Junbo mice reveal defects in HIF and TGF-𝞫 signalling (PhD).
Research
My interests lie in the genetic basis of disease, with a specific focus on age-related and chronic disease. I instigated, developed, and lead, the Harwell Ageing Screen, a large-scale phenotype-driven screen to identify genes and pathways associated with age-related disease. I also collaborate on multiple individual projects around the themes of ageing and the genetic basis of disease. A key area of interest are is ECM dysfunction and current projects include novel mutations resulting in Alport syndrome, Nephrotic Syndrome or Osteoarthritis. I also have projects on sudden cardiac death, hypertrophic cardiomyopathy arising from mitochondrial dysfunction, and colitis.
Key research interest include:
- Collagen I dysfunction resulting in late-onset osteoarthritis
- Abnormal branched chain amino acid metabolism is associated with sudden cardiac death
- A novel model of Nephrotic Syndrome caused by a point mutation in Lama5
- Extracellular matrix trunover in the glomerular basement membrane
- Characterisation of a novel model of colitis
- The association of a mutation in the C lectin binding domain of aggrecan with osteoarthritis and obesity
Research grants and awards
- INFRAFRONTIER H2020, Transnational access package
Research impact
Ageing is a major risk factor for most diseases and it is essential that we understnad the pathways resulting in age-related disease, and improve our understanding of these diseases
Groups
Projects
- The role of aggrecan in adipogenesis
- A novel model of Nephrotic Syndrome
Publications
Journal articles
-
Maddirevula S, Shagrani M, Ji A-R, Horne CR, Young SN, Mather LJ, Alqahtani M, McKerlie C, Wood G, Potter PK, Abdulwahab F, AlSheddi T, van der Woerd WL, van Gassen KLI, AlBogami D, Kumar K, Mohammad HAS, Binomar H, Almanea H, Faqeih E, Fuchs SA, Scott JW, Murphy JM, Alkuraya FS., 'Large Scale Genomic Investigation of Pediatric Cholestasis Reveals a Novel Hepatorenal Ciliopathy Caused by PSKH1 Mutations'
Genetics in Medicine 26 (11) (2024)
ISSN: 1098-3600 eISSN: 1530-0366AbstractPublished here Open Access on RADARPurpose
Pediatric cholestasis is the phenotypic expression of clinically and genetically heterogeneous disorders of bile acid synthesis and flow. Although a growing number of monogenic causes of pediatric cholestasis have been identified, the majority of cases remain undiagnosed molecularly.
Methods
In a large cohort of 299 pediatric participants (279 families) with intrahepatic cholestasis, we performed exome sequencing as a first-tier diagnostic test.
Results
A likely causal variant was identified in 135 families (48.56%). These comprise 135 families that harbor variants spanning 37 genes with established or tentative links to cholestasis. In addition, we propose a novel candidate gene (PSKH1) in 4 families. PSKH1 was particularly compelling because of strong linkage in three consanguineous families who shared a novel hepatorenal ciliopathy phenotype. Two of the four families shared a founder homozygous variant while the third had a different homozygous variant in PSKH1. PSKH1 encodes a putative protein serine kinase of unknown function. Patient fibroblasts displayed abnormal cilia that are long and show abnormal transport. A homozygous Pskh1 mutant mouse faithfully recapitulated the human phenotype and displayed abnormally long cilia. The phenotype could be rationalized by the loss of catalytic activity observed for each recombinant PSKH1 variant using in vitro kinase assays.
Conclusion
Our results support the use of genomics in the workup of pediatric cholestasis and reveal PSKH1-related hepatorenal ciliopathy as a novel candidate monogenic form.
-
Aguilar C, WilliamsD, Kurapati R, Bains RS, Mburu P, Parker A, Williams J, Concas D, Tateossian H, Haynes AR, Banks G, Vikhe P, Heise I, Hutchison M, Atkins G, Gillard S, Starbuck B, Oliveri S, Blake A, Sethi S, Kumar S, Bardhan T, Jeng JY, Johnson SL, Corns LF, Marcotti M, Simon M, Wells S, Potter PK, Lad HV, 'Pleiotropic brain function of whirlin identified by a novel mutation '
iScience 27 (7) (2024)
ISSN: 2589-0042 eISSN: 2589-0042AbstractPublished here Open Access on RADARDespite some evidence indicating diverse roles of whirlin in neurons, the functional corollary of whirlin gene function and behaviour has not been investigated or broadly characterised. A single nucleotide variant was identified from our recessive ENU-mutagenesis screen at a donor-splice site in whirlin, a protein critical for proper sensorineural hearing function. The mutation (head-bob, hb) led to partial intron-retention causing a frameshift and introducing a premature termination codon. Mutant mice had a head-bobbing phenotype and significant hyperactivity across several phenotyping tests. Lack of complementation of head-bob with whirler mutant mice confirmed the head-bob mutation as functionally distinct with compound mutants having a mild-moderate hearing defect. Utilising transgenics, we demonstrate rescue of the hyperactive phenotype and combined with the expression profiling data conclude whirlin plays an essential role in activity-related behaviours. These results highlight a pleiotropic role of whirlin within the brain and implicates alternative, central mediated pathways in its function.
-
Nicol T, Falcone S, Blease A, Vikhe P, Civiletto G, Salman Omairi S, Viscomi C, Patel K, Potter PK, 'Tissue-specific differences in the assembly of mitochondrial complex I is revealed by a novel ENU mutation in ECSIT'
Cardiovascular Research 119 (12) (2023) pp.2213-2229
ISSN: 0008-6363 eISSN: 1755-3245AbstractPublished here Open Access on RADARHere we describe a mutation in the mitochondrial complex I assembly factor ECSIT which reveals tissue specific requirements for ECSIT in complex I assembly. Mitochondrial complex I assembly is a multi-step process dependant on assembly factors that organise and arrange the individual subunits, allowing for their incorporation into the complete enzyme complex. We have identified an ENU induced mutation in ECSIT (N209I) that exhibits a profound effect on complex I assembly in heart tissue resulting in hypertrophic cardiomyopathy in the absence of other phenotypes. We hypothesised that this was due to a specific loss of ECSIT’s ability to function in complex I assembly without any effect on its other characterised functions. . ThisOur data suggests the mechanisms underlying complex I assembly is tissue specific and has implications in understanding the pathogenesis of cardiomyopathy.
-
Kooblall KG, Stevenson M, Stewart M, Harris L, Zalucki O, Dewhurst H, Butterfield N, Leng H, Hough TA, Ma D, Siow B, Potter P, Cox RD, Brown SDM, Horwood N, Wright B, Lockstone H, Buck D, Vincent TL, Hannan FM, Bassett JHD, Williams GR, Lines KE, Piper M, Wells S, Teboul L, Hennekam RC, Thakker RV, 'A Mouse Model with a Frameshift Mutation in the Nuclear Factor I/X (NFIX) Gene Has Phenotypic Features of Marshall‐Smith Syndrome'
JBMR Plus 7 (6) (2023)
ISSN: 2473-4039 eISSN: 2473-4039AbstractPublished here Open Access on RADARThe nuclear factor I/X (NFIX) gene encodes a ubiquitously expressed transcription factor whose mutations lead to two allelic disorders characterized by developmental, skeletal, and neural abnormalities, namely, Malan syndrome (MAL) and Marshall–Smith syndrome (MSS). NFIX mutations associated with MAL mainly cluster in exon 2 and are cleared by nonsense-mediated decay (NMD) leading to NFIX haploinsufficiency, whereas NFIX mutations associated with MSS are clustered in exons 6–10 and escape NMD and result in the production of dominant-negative mutant NFIX proteins. Thus, different NFIX mutations have distinct consequences on NFIX expression. To elucidate the in vivo effects of MSS-associated NFIX exon 7 mutations, we used CRISPR-Cas9 to generate mouse models with exon 7 deletions that comprised: a frameshift deletion of two nucleotides (Nfix Del2); in-frame deletion of 24 nucleotides (Nfix Del24); and deletion of 140 nucleotides (Nfix Del140). Nfix+/Del2, Nfix+/Del24, Nfix+/Del140, NfixDel24/Del24, and NfixDel140/Del140 mice were viable, normal, and fertile, with no skeletal abnormalities, but NfixDel2/Del2 mice had significantly reduced viability (p Nfix Del2 was not cleared by NMD, and NfixDel2/Del2 mice, when compared to Nfix+/+ and Nfix+/Del2 mice, had: growth retardation; short stature with kyphosis; reduced skull length; marked porosity of the vertebrae with decreased vertebral and femoral bone mineral content; and reduced caudal vertebrae height and femur length. Plasma biochemistry analysis revealed NfixDel2/Del2 mice to have increased total alkaline phosphatase activity but decreased C-terminal telopeptide and procollagen-type-1-N-terminal propeptide concentrations compared to Nfix+/+ and Nfix+/Del2 mice. NfixDel2/Del2 mice were also found to have enlarged cerebral cortices and ventricular areas but smaller dentate gyrus compared to Nfix+/+ mice. Thus, NfixDel2/Del2 mice provide a model for studying the in vivo effects of NFIX mutants that escape NMD and result in developmental abnormalities of the skeletal and neural tissues that are associated with MSS.
-
Khalil Y, Carrino S, Lin F, Ferlin A, Lad HV, Mazzacuva F, Falcone S, Rivers N, Banks G, Concas D, Aguilar C, Haynes AR, Blease A, Nicol T, Al-Shawi R, Heywood W, Potter PK, Mills K, Gale DP, Peter T, Clayton PT, 'Tissue Proteome of 2-hydroxyacyl-CoA lyase Deficient Mice Reveals Peroxisome Proliferation and Activation of ω-oxidation'
International Journal of Molecular Sciences 23 (2) (2022)
ISSN: 1661-6596 eISSN: 1422-0067AbstractPublished herePeroxisomal fatty acid α-oxidation is an essential pathway for the degradation of β-carbon methylated fatty acids such as phytanic acid. One enzyme in this pathway is 2-hydroxyacyl CoA lyase (HACL1) which is responsible for the cleavage of 2-hydroxyphytanoyl-CoA into pristanal and formyl CoA. Hacl1 deficient mice do not present with a severe phenotype, unlike mice deficient in other α-oxidation enzymes such as phytanoyl-CoA hydroxylase deficiency (Refsum disease) in which neuropathy and ataxia are present. Tissues from wild-type and Hacl1-/-mice fed a high phytol diet were obtained for proteomic and lipidomic analysis. There was no phenotype observed in these mice. Liver, brain, and kidney tissues underwent trypsin digestion for untargeted proteomic liquid chromatography-mass spectrometry analysis while liver tissues also underwent fatty acid hydrolysis, extraction, and derivatisation for fatty acid gas chromatography-mass spectrometry analysis. The liver fatty acid profile demonstrated an accumulation of phytanic and 2-hydroxyphytanic acid in the Hacl1-/- liver and significant decrease in heptadecanoic acid. The liver proteome showed a significant decrease in the abundance of Hacl1 and a significant increase in the abundance of proteins involved in PPAR signaling, peroxisome proliferation, and omega oxidation, particularly Cyp4a10 and Cyp4a14. In addition, the pathway associated with arachidonic acid metabolism was affected; Cyp2c55 was upregulated and Cyp4f14 and Cyp2b9 were downregulated. The kidney proteome revealed fewer significantly upregulated peroxisomal proteins and the brain proteome was not significantly different in Hacl1-/- mice. This study demonstrates the powerful insight brought by proteomic and metabolomic profiling of Hacl1-/- mice in better understanding disease mechanism in fatty acid α-oxidation disorders.
-
Falcone S, Nicol T, Blease A, Randles MJ, Angus E, Page A, Tam FWK, Pusey CD, Lennon R, Potter PK, 'A novel model of nephrotic syndrome results from a point mutation in Lama5 and is modified by genetic background'
Kidney International 101 (3) (2021) pp.527-540
ISSN: 0085-2538 eISSN: 1523-1755AbstractPublished here Open Access on RADARNephrotic syndrome is characterised by severe proteinuria, hypoalbuminaemia, oedema and hyperlipidaemia. Genetic studies of nephrotic syndrome have led to the identification of proteins playing a crucial role in slit diaphragm signalling, regulation of actin cytoskeleton dynamics and cell-matrix interactions. The laminin α5 chain is essential for embryonic development and, in association with laminin β2 and laminin γ1, it is a major component of the glomerular basement membrane. Mutations in LAMA5 were recently identified in children with nephrotic syndrome. We have identified a novel missense mutation (E884G) in the uncharacterised L4a domain of LAMA5 where homozygous mice develop nephrotic syndrome with severe proteinuria that precedes histological and ultrastructural changes in the glomerulus. The levels of LAMA5 are reduced in vivo and the assembly of the laminin 521 heterotrimer significantly reduced in vitro. Proteomic analysis of the glomerular extracellular fraction revealed changes in the matrix composition. Importantly, the genetic background had a significant effect on aspects of disease progression from proteinuria to changes in podocyte morphology. This novel model will provide insights into patho-mechanisms of nephrotic syndrome and pathways that influence the response to a dysfunctional glomerular basement membrane.
-
Portero Vincent, Nicol Thomas, Podliesna Svitlana, Marchal Gerard A, Baartscheer Antonius, Casini Simona, Tadros Rafik, Treur Jorien L, Tanck Michael W T, Cox I Jane, Probert Fay, Hough Tertius A, Falcone Sara, Beekman Leander, Müller-Nurasyid Martina, Kastenmüller Gabi, Gieger Christian, Peters Annette, Kääb Stefan, Sinner Moritz F, Blease Andrew, Verkerk Arie O, Bezzina Connie R, Potter Paul K, Remme Carol Ann, 'Chronically elevated branched chain amino acid levels are pro-arrhythmic'
Cardiovascular Research 118 (7) (2021) pp.1742-1757
ISSN: 0008-6363 eISSN: 1755-3245AbstractPublished here Open Access on RADARAims. Cardiac arrhythmias comprise a major health and economic burden and are associated with significant morbidity and mortality, including cardiac failure, stroke, and sudden cardiac death (SCD). Development of efficient preventive and therapeutic strategies is hampered by incomplete knowledge of disease mechanisms and pathways. Our aim is to identify novel mechanisms underlying cardiac arrhythmia and SCD using an unbiased approach. Methods and results. We employed a phenotype-driven N-ethyl-N-nitrosourea mutagenesis screen and identified a mouse line with a high incidence of sudden death at young age (6–9 weeks) in the absence of prior symptoms. Affected mice were found to be homozygous for the nonsense mutation Bcat2p.Q300*/p.Q300* in the Bcat2 gene encoding branched chain amino acid transaminase 2. At the age of 4–5 weeks, Bcat2p.Q300*/p.Q300* mice displayed drastic increase of plasma levels of branch chain amino acids (BCAAs—leucine, isoleucine, valine) due to the incomplete catabolism of BCAAs, in addition to inducible arrhythmias ex vivo as well as cardiac conduction and repolarization disturbances. In line with these findings, plasma BCAA levels were positively correlated to electrocardiogram indices of conduction and repolarization in the German community-based KORA F4 Study. Isolated cardiomyocytes from Bcat2p.Q300*/p.Q300* mice revealed action potential (AP) prolongation, pro-arrhythmic events (early and late afterdepolarizations, triggered APs), and dysregulated calcium homeostasis. Incubation of human pluripotent stem cell-derived cardiomyocytes with elevated concentration of BCAAs induced similar calcium dysregulation and pro-arrhythmic events which were prevented by rapamycin, demonstrating the crucial involvement of mTOR pathway activation. Conclusions. Our findings identify for the first time a causative link between elevated BCAAs and arrhythmia, which has implications for arrhythmogenesis in conditions associated with BCAA metabolism dysregulation such as diabetes, metabolic syndrome, and heart failure.
-
Randles MJ, Lausecker F, Kong Q, Suleiman H, Kolatsi-Joannou M, Davenport B, Tian P, Falcone S, Potter P, Van Agtmael T, Norman JT, Long DA, Humphries MJ, Miner JH, Lennon R, 'Identification of an Altered Matrix Signature in Kidney Aging and Disease'
Journal of the American Society of Nephrology 32 (7) (2021) pp.1713-1732
ISSN: 1046-6673 eISSN: 1533-3450AbstractPublished here Open Access on RADARBackground: Accumulation of extracellular matrix in organs and tissues is a feature of both ageing and disease. In the kidney, glomerulosclerosis and tubulointerstitial fibrosis accompany the decline in organ function, which is irretrievable with current therapies leading to inevitable kidney failure. Whilst histological and ultrastructural patterns of excess matrix form the basis of human disease classifications, the comprehensive molecular resolution of abnormal matrix is lacking.
Methods: Using mass spectrometry-based proteomics we resolved matrix composition over age in mouse models of kidney disease. We compared the
changes in mice with a global characterization of human kidney matrix during ageing and to existing kidney disease datasets to identify common molecular features.Results: We found that ultrastructural changes in basement membranes are associated with altered cell adhesion and metabolic processes and with distinct matrix proteomes during ageing and kidney disease progression in mice. Within the altered matrix, we observed a reduction in basement membrane components (laminins, type IV collagen, type XVIII collagen) and a corresponding increase in interstitial matrix proteins (collagens I, III, VI, XV, fibrinogens and nephronectin) and this pattern was also seen in human kidney ageing. Indeed, this signature of matrix proteins was consistently modulated across all age and disease comparisons and the increase in interstitial matrix was also observed in human kidney disease datasets.
Conclusions: This study provides deep molecular resolution of matrix accumulation in kidney ageing and disease and identifies a common signature of proteins that provides insight into mechanisms of response to kidney injury and repair.
-
Bellantuono I, de Cabo R, D Ehninger, Di Germanio C, Lawrie A, Miller J,Mitchell SJ, Navas-Enamorado I, Potter P, Tchkonia T, Trejo JL, Lamming DW, 'A toolbox for the longitudinal assessment of healthspan in ageing mice'
Nature Protocols 15 (2020) pp.540-574
ISSN: 1754-2189 eISSN: 1750-2799AbstractPublished here Open Access on RADARThe number of people aged over 65 is expected to double in the next 30 years. For many, living longer will mean spending more years with the burdens of chronic diseases such as Alzheimer’s, cardiovascular disease, and diabetes. Although researchers have made rapid progress in developing geroprotective interventions that target mechanisms of ageing and delay or prevent the onset of multiple concurrent age-related diseases, a lack of standardized techniques to assess healthspan in preclinical murine studies has resulted in reduced reproducibility and slowed progress. To overcome this, major centres in Europe and the USA skilled in healthspan analysis came together to agree upon a toolbox of techniques which can be used to consistently assess the healthspan of mice. Here, we describe the agreed toolbox which contains protocols for echocardiography, novel object recognition, grip strength, rotarod, glucose and insulin tolerance tests, body composition, and energy expenditure. They can be performed longitudinally in the same mouse over a period of 4-6 weeks to test how candidate geroprotectors affect cardiac, cognitive, neuromuscular and metabolic health.
-
Falcone S, Wisby L, Nicol T , Blease A, Starbuck B, Parker A, Sanderson J, Brown SDM, Scudamore CL, Pusey CD, Tam FWK, Potter PK, 'Modification of an aggressive model of Alport Syndrome reveals early differences in disease pathogenesis due to genetic background'
Scientific Reports 9 (2019)
ISSN: 2045-2322AbstractPublished here Open Access on RADARThe link between mutations in collagen genes and the development of Alport Syndrome has been clearly established and a number of animal models, including knock-out mouse lines, have been developed that mirror disease observed in patients. However, it is clear from both patients and animal models that the progression of disease can vary greatly and can be modifed genetically. We have identifed a point mutation in Col4a4 in mice where disease is modifed by strain background, providing further evidence of the genetic modifcation of disease symptoms. Our results indicate that C57BL/6J is a protective background and postpones end stage renal failure from 7 weeks, as seen on a C3H background, to several months. We have identifed early diferences in disease progression, including expression of podocyte-specifc genes and podocyte morphology. In C57BL/6J mice podocyte efacement is delayed, prolonging normal renal function. The slower disease progression has allowed us to begin dissecting the pathogenesis of murine Alport Syndrome in detail. We fnd that there is evidence of diferential gene expression during disease on the two genetic backgrounds, and that disease diverges by 4 weeks of age. We also show that an infammatory response with increasing MCP-1 and KIM-1 levels precedes loss of renal function.
-
Christopher J, Thorsen A-S, Abujudeh S, Lourenço FC, Kemp R, Potter PK, Morrissey E, Hazelwood L, Winton DJ, 'Quantifying Microsatellite Mutation Rates from Intestinal Stem Cell Dynamics in Msh2-Deficient Murine Epithelium'
Genetics 212 (3) (2019) pp.655-665
ISSN: 0016-6731 eISSN: 1943-2631AbstractPublished hereMicrosatellite sequences have an enhanced susceptibility to mutation, and can act as sentinels indicating elevated mutation rates and increased risk of cancer. The probability of mutant fixation within the intestinal epithelium is dictated by a combination of stem cell dynamics and mutation rate. Here, we exploit this relationship to infer microsatellite mutation rates. First a sensitive, multiplexed, and quantitative method for detecting somatic changes in microsatellite length was developed that allowed the parallel detection of mutant [CA]n sequences from hundreds of low-input tissue samples at up to 14 loci. The method was applied to colonic crypts in Mus musculus, and enabled detection of mutant subclones down to 20% of the cellularity of the crypt (∼50 of 250 cells). By quantifying age-related increases in clone frequencies for multiple loci, microsatellite mutation rates in wild-type and Msh2-deficient epithelium were established. An average 388-fold increase in mutation per mitosis rate was observed in Msh2-deficient epithelium (2.4 × 10−2) compared to wild-type epithelium (6.2 × 10−5).
-
Trendelenburg AU, Scheuren AC, Potter PK, Müller R, Bellantuono I, 'Geroprotectors: A role in the treatment of frailty.'
Mechanisms of Ageing and Development 180 (2019) pp.11-20
ISSN: 0047-6374 eISSN: 1872-6216AbstractPublished here Open Access on RADARThe proportion of the population over the age of 65 is growing the most rapidly due to the longevity revolution. Frailty is prevalent in this age group and strongly associated with disability and hospitalization, having a significant impact on the costs of health and social care. New effective interventions to delay or reverse frailty are urgently required. Geroprotectors are a new class of drugs, which target fundamental mechanisms of ageing and show promise in delaying the onset of or boosting resilience in frail older people. However, there are challenges to their clinical translation. Here we review the literature for evidence that frailty can be delayed or reversed and geroprotectors can improve frailty in murine models and in patients. We will then discuss the challenges, which make their clinical testing complex and propose potential options for moving forward.
-
Paul K Potter, 'A primer on ageing studies in mice: Considerations, opportunities and limitations'
Drug Discovery Today: Disease Models 27 (2019) pp.23-29
ISSN: 1740-6757 eISSN: 1740-6757AbstractPublished here Open Access on RADAROne of the major challenges currently facing healthcare providers is an ageing population that is spending more time in ill-health. Many ageing individuals have multiple and complex needs which affect the ability to treat them effectively, which also has a significant impact on their own independence and quality of life. There are many aspects of testing interventions to improve health in old age in pre-clinical models; from breeding strategies to measurements of outcomes. Here we provide a brief overview of the major considerations to take into account in such studies and the limitations or challenges we face in these studies.
-
Curley M, Milne L, Smith S, Jørgensen A, Frederiksen H, Hadoke P, Potter P, Smith LB, 'A young testicular microenvironment protects Leydig cells against age-related dysfunction in a mouse model of premature aging.'
The FASEB Journal 33 (1) (2018) pp.978-995
ISSN: 0892-6638 eISSN: 1530-6860AbstractPublished here Open Access on RADARTesticular Leydig cells (LCs) are the primary source of circulating androgen in men. As men age, circulating androgen levels decline. However, whether reduced LC steroidogenesis results from specific effects of aging within LCs or reflects degenerative alterations to the wider supporting microenvironment is unclear; inability to separate intrinsic LC aging from that of the testicular microenvironment in vivo has made this question difficult to address. To resolve this, we generated novel mouse models of premature aging, driven by CDGSH iron sulfur domain 2 ( Cisd2) deletion, to separate the effects of cell intrinsic aging from extrinsic effects of aging on LC function. At 6 mo of age, constitutive Cisd2-deficient mice display signs of premature aging, including testicular atrophy, reduced LC and Sertoli cell (SC) number, decreased circulating testosterone, increased luteinizing hormone/testosterone ratio, and decreased expression of steroidogenic mRNAs, appropriately modeling primary testicular dysfunction observed in aging men. However, mice with Cisd2 deletion (and thus premature aging) restricted to either LCs or SCs were protected against testicular degeneration, demonstrating that age-related LCs dysfunction cannot be explained by intrinsic aging within either the LC or SC lineages alone. We conclude that age-related LC dysfunction is largely driven by aging of the supporting testicular microenvironment.-Curley, M., Milne, L., Smith, S., Jørgensen, A., Frederiksen, H., Hadoke, P., Potter, P., Smith, L. B. A Young testicular microenvironment protects Leydig cells against age-related dysfunction in a mouse model of premature aging.
-
Findlay AS, Carter RN, Starbuck B, McKie L, Nováková K, Budd PS, Keighren MA, Marsh JA, Cross SH, Simon MM, Potter PK, Morton NM, Jackson IJ, 'Mouse Idh3a mutations cause retinal degeneration and reduced mitochondrial function'
Disease Models & Mechanisms 11 (12) (2018)
ISSN: 1754-8403 eISSN: 1754-8411AbstractPublished hereIsocitrate dehydrogenase (IDH) is an enzyme required for the production of α-ketoglutarate from isocitrate. IDH3 generates the NADH used in the mitochondria for ATP production, and is a tetramer made up of two α, one β and one γ subunit. Loss-of-function and missense mutations in both IDH3A and IDH3B have previously been implicated in families exhibiting retinal degeneration. Using mouse models, we investigated the role of IDH3 in retinal disease and mitochondrial function. We identified mice with late-onset retinal degeneration in a screen of ageing mice carrying an ENU-induced mutation, E229K, in Idh3a. Mice homozygous for this mutation exhibit signs of retinal stress, indicated by GFAP staining, as early as 3 months, but no other tissues appear to be affected. We produced a knockout of Idh3a and found that homozygous mice do not survive
past early embryogenesis. Idh3a−/E229K compound heterozygous mutants exhibit a more severe retinal degeneration compared with Idh3aE229K/E229K homozygous mutants. Analysis of mitochondrial
function in mutant cell lines highlighted a reduction in mitochondrial maximal respiration and reserve capacity levels in both Idh3aE229K/E229K and Idh3a−/E229K cells. Loss-of-function Idh3b mutants do not exhibit the same retinal degeneration phenotype, with no signs of retinal stress or reduction in mitochondrial respiration.
It has previously been reported that the retina operates with a limited mitochondrial reserve capacity and we suggest that this, in combination with the reduced reserve capacity in mutants, explains the degenerative phenotype observed in Idh3a mutant mice. -
Blease A, Das Neves Borges P, Curtinha m, Javaheri IS, von Loga, Parisi I, Zarebska J, Pitsillides A, Vincent TL, Potter PK, 'Studying Osteoarthritis Pathogenesis in Mice.'
Current Protocols in Mouse Biology 8 (4) (2018)
ISSN: 2161-2617 eISSN: 2161-2617AbstractWith the increasing availability and complexity of mouse models of disease, either spontaneous or induced, there is a concomitant increase in their use in the analysis of pathogenesis. Among such diseases is osteoarthritis, a debilitating disease with few treatment options. While advances in our understanding of the pathogenesis of osteoarthritis has advanced through clinical investigations and genome-wide association studies, there is still a large gap in our knowledge, hindering advances in therapy. Patient samples are available ex vivo, but these are generally in the very late stages of disease. However, with mice, we are able to induce disease at a defined time and track the progression in vivo and ex vivo, from inception to end stage, to delineate the processes involved in disease development. © 2018 by John Wiley & Sons, Inc.Published here -
Blease A, Nicol T, Falcone S, Starbuck B, Greenaway S, Hutchinson M, Potter PK, 'Generation and Identification of Mutations Resulting in Chronic and Age-Related Phenotypes in Mice.'
Current Protocols in Mouse Biology 8 (2) (2018)
ISSN: 2161-2617 eISSN: 2161-2617AbstractAging is inevitable, and our society must deal with the consequences: namely, an increased incidence of disease and ill health. Many mouse models of disease are acute or early onset or are induced in young mice, despite the fact that aging is a significant risk factor for a range of significant diseases. To improve modeling of such diseases, we should incorporate aging into our models. Many systems are affected by aging, with a decline in mitochondrial function, an increase in senescence, a loss of resilience, telomere shortening, and a decline in immune function being key factors in the increased susceptibility to disease that is associated with aging. To develop novel models of age-related disease, we undertook a phenotype-driven screen of a pipeline of mutagenized mice. Here, we describe some of the underlying protocols and outline important aspects to consider when studying aged mice. © 2018 by John Wiley & Sons, Inc.Published here -
Potter PK, 'Deconstructing Gene Function through ENU Mutagenesis'
eLS Online first (2018)
AbstractPublished hereA great majority of genes present in the human genome are also present in the mouse, thus making it an attractive mammalian model organism to study gene function and dysfunction. Over the past few decades, the ability to manipulate the mouse genome has been developed in a variety of ways. A complementary methodology to create mutations in the mouse is to use chemical mutagenesis. N‐ethyl‐N‐Nitrosourea (ENU) is the mutagen of choice for creating random point mutations model organisms. Advances in sequencing technologies have resulted in a rapid identification of the causative mutation. ENU mutagenesis is a powerful hypothesis‐generating approach to create new mouse models through both forward and reverse genetics approaches. Furthermore, the addition of challenges can identify mutations affecting specific pathways, and specific mutant lines or strains can be used to identify modifiers.
-
Bellantuono I, Van Den Brouke R, Muller R, Van Os RP, Trendelenburg AU, Potter P, Tchkonia T, Ehninger D, Fernanades A, Trejo Perez, JL, deCabo R, van Riel N, Howlett S, 'Find drugs that delay many diseases of old age'
Nature 554 (7692) (2018) pp.293-295
ISSN: 0028-0836 eISSN: 1476-4687Published here -
Esapa CT, Piret SE, Nesbit MA, Thomas GP, Coulton LA, Gallagher OM, Simon M, Kumar S, Mallon AM, Bellantuono I, Brown MA, Croucher PI, Potter PK, Brown SDM, Cox RD, Thakker RV, 'An N-ethyl-N-nitrosourea (ENU) mutagenized mouse model for autosomal dominant non-syndromic kyphoscoliosis due to vertebral fusion'
JBMR Plus 2 (3) (2018) pp.154-163
ISSN: 2473-4039AbstractPublished hereKyphosis and scoliosis are common spinal disorders that occur as part of complex syndromes or as nonsyndromic, idiopathic diseases. Familial and twin studies implicate genetic involvement, although the causative genes for idiopathic kyphoscoliosis remain to be identified. To facilitate these studies, we investigated progeny of mice treated with the chemical mutagen N‐ethyl‐N‐nitrosourea (ENU) and assessed them for morphological and radiographic abnormalities. This identified a mouse with kyphoscoliosis due to fused lumbar vertebrae, which was inherited as an autosomal dominant trait; the phenotype was designated as hereditary vertebral fusion (HVF) and the locus as Hvf. Micro–computed tomography (μCT) analysis confirmed the occurrence of nonsyndromic kyphoscoliosis due to fusion of lumbar vertebrae in HVF mice, consistent with a pattern of blocked vertebrae due to failure of segmentation. μCT scans also showed the lumbar vertebral column of HVF mice to have generalized disc narrowing, displacement with compression of the neural spine, and distorted transverse processes. Histology of lumbar vertebrae revealed HVF mice to have irregularly shaped vertebral bodies and displacement of intervertebral discs and ossification centers. Genetic mapping using a panel of single nucleotide polymorphic (SNP) loci arranged in chromosome sets and DNA samples from 23 HVF (eight males and 15 females) mice, localized Hvf to chromosome 4A3 and within a 5‐megabase (Mb) region containing nine protein coding genes, two processed transcripts, three microRNAs, five small nuclear RNAs, three large intergenic noncoding RNAs, and 24 pseudogenes. However, genome sequence analysis in this interval did not identify any abnormalities in the coding exons, or exon‐intron boundaries of any of these genes. Thus, our studies have established a mouse model for a monogenic form of nonsyndromic kyphoscoliosis due to fusion of lumbar vertebrae, and further identification of the underlying genetic defect will help elucidate the molecular mechanisms involved in kyphoscoliosis.
-
Jackson SJ, Andrews N, Ball D, Bellantuono I, Gray J, Hachoumi , Holmes A, Latcham J, Petrie A, Potter PK, Rice ASC, Ritchie AA, Stewart M, Strepka C, Yeoman M, Chapman K, 'Does age matter? The impact of rodent age on study outcomes.'
Laboratory Animals 51 (2) (2017) pp.160-169
ISSN: 0023-6772 eISSN: 1758-1117AbstractRodent 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.Published here -
Bellantuono I, Potter PK, 'Modelling Ageing and Age-Related Disease'
Drug Discovery Today: Disease Models 20 (2016) pp.27-32
ISSN: 1740-6757 eISSN: 1740-6757AbstractPublished hereAn increased lifespan comes with an associated increase in disease incidence, and is the major risk factor for age-related diseases. To face this societal challenge search for new treatments has intensified requiring good preclinical models, whose complexity and accuracy is increasing. However, the influence of ageing is often overlooked. Furthermore, phenotypic assessment of ageing models is in need of standardisation to enable the accurate evaluation of pre-clinical intervention studies in line with clinical translation.
-
Potter PK, Bowl MR, Jeyarajan P, Wisby L, Blease A, Goldsworthy ME, Simon MM, Greenaway S, Michel V, Barnard A, Aguilar C, Agnew T, Banks G, Blake A, Chessum L, Dorning J, Falcone S, Goosey L, Harris S, Haynes A, Heise I, Hillier R, Hough T, Hoslin A, Hutchison M, King R, Kumar S, Lad HV, Law G, MacLaren RE, Morse S, Nicol T, Parker A, Pickford K, Sethi S, Starbuck B, Stelma F, Cheeseman M, Cross SH, Foster RG, Jackson IJ, Peirson SN, Thakker RV, Vincent T, Scudamore C, Wells S, El-Amraoui A, Petit C, Acevedo-Arozena A, Nolan PM, Cox R, Mallon AM, Brown SDM, 'Novel gene function revealed by mouse mutagenesis screens for models of age-related disease.'
Nature Communications 7 (2016)
ISSN: 2041-1723AbstractDetermining 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.Published here -
Nicod J, Davies RW, Cai N, Hassett C, Goodstadt L, Cosgrove C, Yee BK, Lionikaite V, McIntyre RE, Remme CA, Lodder EM, Gregory JS, Hough T, Joynson R, Phelps H, Nell B, Rowe C, Wood J, Walling A, Bopp N, Bhomra A, Hernandez-Pliego P, Callebert J, Aspden RM, Talbot NP, Robbins PA, Harrison M, Fray M, Launay JM, Pinto YM, Blizard DA, Bezzina CR, Adams DJ, Franken P, Weaver T, Wells S, Brown SD, Potter PK, Klenerman P, Lionikas A, Mott R, Flint J., 'Genome-wide association of multiple complex traits in outbred mice by ultra-low-coverage sequencing.'
Nature Genetics 48 (8) (2016) pp.912-918
ISSN: 1061-4036AbstractTwo 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.Published here -
Potter PK, 'The Mammalian Genome special issue on ageing.'
Mammalian Genome 27 (7/8) (2016) pp.257-258
ISSN: 0938-8990 eISSN: 1432-1777Published here -
Matsakas A, Prosdocimo DA, Mitchell R, Collins-Hooper H, Giallourou N, Swann JR, Potter PK, Epting T, Jain MK, Patel K, 'Investigating mechanisms underpinning the detrimental impact of a high-fat diet in the developing and adult hypermuscular myostatin null mouse.'
Skeletal Muscle 5 (2015)
ISSN: 2044-5040 eISSN: 2044-5040AbstractObese adults are prone to develop metabolic and cardiovascular diseases. Furthermore, over-weight expectant mothers give birth to large babies who also have increased likelihood of developing metabolic and cardiovascular diseases. Fundamental advancements to better understand the pathophysiology of obesity are critical in the development of anti-obesity therapies not only for this but also future generations. Skeletal muscle plays a major role in fat metabolism and much work has focused in promoting this activity in order to control the development of obesity. Research has evaluated myostatin inhibition as a strategy to prevent the development of obesity and concluded in some cases that it offers a protective mechanism against a high-fat diet.\nPregnant as well as virgin myostatin null mice and age matched wild type animals were raised on a high fat diet for up to 10 weeks. The effect of the diet was tested on skeletal muscle, liver and fat. Quantitate PCR, Western blotting, immunohistochemistry, in-vivo and ex-vivo muscle characterisation, metabonomic and lipidomic measurements were from the four major cohorts.\nWe hypothesised that myostatin inhibition should protect not only the mother but also its developing foetus from the detrimental effects of a high-fat diet. Unexpectedly, we found muscle development was attenuated in the foetus of myostatin null mice raised on a high-fat diet. We therefore re-examined the effect of the high-fat diet on adults and found myostatin null mice were more susceptible to diet-induced obesity through a mechanism involving impairment of inter-organ fat utilization.\nLoss of myostatin alters fatty acid uptake and oxidation in skeletal muscle and liver. We show that abnormally high metabolic activity of fat in myostatin null mice is decreased by a high-fat diet resulting in excessive adipose deposition and lipotoxicity. Collectively, our genetic loss-of-function studies offer an explanation of the lean phenotype displayed by a host of animals lacking myostatin signalling.\nBACKGROUND\nMETHODS\nRESULTS\nCONCLUSIONSPublished here -
Simon MM, Moresco EM, Bull KR, Kumar S, Mallon AM, Beutler B, Potter PK, 'Current strategies for mutation detection in phenotype-driven screens utilising next generation sequencing.'
Mammalian Genome 26 (9/10) (2015) pp.486-500
ISSN: 0938-8990 eISSN: 1432-1777AbstractMutagenesis-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.Published here -
de Angelis MH, Nicholson G, Selloum M, White J, Morgan H, Ramirez-Solis R, Sorg T, Wells S, Fuchs , Fray M, Adams DJ, Adams NC, Adler T, Aguilar-Pimentel A, Ali-Hadji D, Amann G, André P, Atkins S, Auburtin A, Ayadi A, Becker J, Becker L, Bedu E, Bekeredjian R, Birling MC, Blake A, Bottomley J, Bowl M, Brault V, Busch DH, Bussell JN, Calzada-Wack J, Cater H, Champy MF, Charles P, Chevalier C, Chiani F, Codner GF, Combe R, Cox R, Dalloneau E1, Dierich A, Di Fenza A10, Doe B17, Duchon A, Eickelberg O18, Esapa CT, El Fertak L, Feigel T10, Emelyanova I, Estabel J, Favor J, Flenniken A, Gambadoro A, Garrett L, Gates H, Gerdin AK, Gkoutos G, Greenaway S, Glasl L, Goetz P, Da Cruz IG, Götz A, Graw J, Guimond A, Hans W, Hicks G, Hölter SM, Höfler H, Hancock JM, Hoehndorf R, Hough T, Houghton R, Hurt A, Ivandic B, Jacobs H, Jacquot S, Jones N, Karp NA, Katus HA, Kitchen S, Klein-Rodewald T, Klingenspor M, Klopstock T, Lalanne V5, Leblanc S, Lengger C, le Marchand E, Ludwig T, Lux A, McKerlie C, Maier H, Mandel JL, Marschall S, Mark M, Melvin DG, Meziane H, Micklich K, Mittelhauser C, Monassier L, Moulaert D5, Muller S, Naton B, Neff F, Nolan PM, Nutter LM, Ollert M, Pavlovic G, Pellegata NS, Peter E, Petit-Demoulière B, Pickard A, Podrini C, Potter PK, Pouilly L, Puk O, Richardson D, Rousseau S, Quintanilla-Fend L, Quwailid MM, Racz I, Rathkolb B, Riet F, Rossant J, Roux M, Rozman J, Ryder E, Salisbury J, Santos L, Schäble KH, Schiller E, Schrewe A, Schulz H, Steinkamp R, Simon M, Stewart M, Stöger C, Stöger T, Sun M, Sunter D, Teboul L, Tilly I, Tocchini-Valentini GP, Tost M, Treise I, Vasseur L, Velot E, Vogt-Weisenhorn D, Wagner C, Walling A, Weber B, Wendling O, Westerberg H, Willershäuser M, Wolf E, Wolter A, Wood J, Wurst W, Yildirim AÖ, Zeh , Zimmer A, Zimprich A; EUMODIC Consortium, Holmes C, Steel KP, Herault Y, Gailus-Durner V, Mallon AM, Brown SD, 'Analysis of mammalian gene function through broad-based phenotypic screens across a consortium of mouse clinics.'
Nature Genetics 47 (9) (2015) pp.969-978
ISSN: 1061-4036AbstractThe function of the majority of genes in the mouse and human genomes remains unknown. The mouse embryonic stem cell knockout resource provides a basis for the characterization of relationships between genes and phenotypes. The EUMODIC consortium developed and validated robust methodologies for the broad-based phenotyping of knockouts through a pipeline comprising 20 disease-oriented platforms. We developed new statistical methods for pipeline design and data analysis aimed at detecting reproducible phenotypes with high power. We acquired phenotype data from 449 mutant alleles, representing 320 unique genes, of which half had no previous functional annotation. We captured data from over 27,000 mice, finding that 83% of the mutant lines are phenodeviant, with 65% demonstrating pleiotropy. Surprisingly, we found significant differences in phenotype annotation according to zygosity. New phenotypes were uncovered for many genes with previously unknown function, providing a powerful basis for hypothesis generation and further investigation in diverse systems.Published here -
Potter PK, 'The Interrelationship between Disease and Ageing and the Implications for Longevity.'
Current Aging Science 8 (1) (2015) pp.89-94
ISSN: 1874-6098AbstractAgeing is generally viewed as a detrimental phenotype; with age comes increasing susceptibility to disease and frailty. Recent data also suggests that disease can result in an increase in ageing phenotypes suggesting a positive feedback loop. It is clear that lifespan can be modified genetically and by interventions in certain organisms but the mechanisms by which this is achieved have not yet been fully elucidated, as indeed is the case for the ageing process itself. Because of the intimate relationship between disease, ageing and ultimately lifespan it is difficult to dissect the effects of individual changes. As we learn more about individual pathways and allelic variants influencing ageing and disease we can begin to unravel the influence of natural selection on these processes.Published here -
Esapa CT, Hannan FM, Babinsky VN, Potter PK, Thomas GP, Croucher PI, Brown MA, Brown SDM, Cox RD, Thakker RV, 'N-ethyl-N-Nitrosourea (ENU) induced mutations within the klotho gene lead to ectopic calcification and reduced lifespan in mouse models.'
PLoS ONE 10 (4) (2015)
ISSN: 1932-6203AbstractEctopic 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.Published here -
Luhmann, Carvalho, Holthaus, Cowing, Greenaway, Chu, Herrmann, Smith, Munro, Potter, Bainbridge, Ali, 'The severity of retinal pathology in homozygous Crb1rd8/rd8 mice is dependent on additional genetic factors.'
Human Molecular Genetics 24 (1) (2015) pp.128-141
ISSN: 0964-6906AbstractUnderstanding 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 Crb1(rd8/rd8) mutation, we compared the retinal pathology of Crb1(rd8/rd8)/J inbred mice with that of two Crb1(rd8/rd8) lines backcrossed with C57BL/6JOlaHsd mice. Topical endoscopic fundal imaging and scanning laser ophthalmoscopy fundus images of all three Crb1(rd8/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 Crb1(rd8/rd8) line, even at 12 months of age. This suggests that the Crb1(rd8/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.Published here -
Banks G, Heise I, Starbuck B, Osborne T, Wisby L, Potter P, Jackson IJ, Foster RG, Peirson SN, Nolan PM, 'Genetic background influences age-related decline in visual and nonvisual retinal responses, circadian rhythms, and sleep'
Neurobiology of Aging 36 (1) (2015) pp.380-393
ISSN: 0197-4580 eISSN: 1558-1497AbstractPublished hereThe 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.
-
Goldsworthy ME, Potter PK, 'Modelling age-related metabolic disorders in the mouse.'
Mammalian Genome 25 (9/10) (2014) pp.487-496
ISSN: 0938-8990 eISSN: 1432-1777AbstractAgeing can be characterised by a general decline in cellular function, which affects whole-body homoeostasis with metabolic dysfunction-a common hallmark of ageing. The identification and characterisation of the genetic pathways involved are paramount to the understanding of how we age and the development of therapeutic strategies for combating age-related disease. Furthermore, in addition to understanding the ageing process itself, we must understand the interactions ageing has with genetic variation that results in disease phenotypes. The use of model systems such as the mouse, which has a relatively short lifespan, rapid reproduction (resulting in a large number of offspring), well-characterised biology, a fully sequenced genome, and the availability of tools for genetic manipulation is essential for such studies. Here we review the relationship between ageing and metabolism and highlight the need for modelling these processes.Published here -
Copier J, Potter P, Sacks SH, Kelly AP, 'Multiple signals regulate the intracellular trafficking of HLA-DM in B-lymphoblastoid cells'
Immunology 93 (4) (1998) pp.505-510
ISSN: 0019-2805 eISSN: 1365-2567AbstractPublished herePeptide loading by major histocompatibility complex (MHC) class II molecules occurs in the endocytic pathway and is critically dependent upon the function of the class II-related molecule human leucocyte antigen-DM (HLA-DM). We have previously shown that a tyrosine-based lysosomal targeting signal present in the cytoplasmic tail of DMB has the capacity to target HLA-DM to peptide-loading compartments in HeLa cells. Here we investigate the importance of this signal in directing HLA-DM to processing compartments in professional antigen-presenting cells. We reconstituted a DMB-negative B-lymphoblastoid cell line with native or targeting-deficient DMB and show that in the absence of its tyrosine signal, DMB-Y230A is as efficient as the wild-type molecule in inducing MHC class II SDS stable dimer formation; restoring expression of the conformation-dependent DR3 epitope 16:23; the removal of CLIP; and accessing lysosomal peptide-loading compartments. By transient transfection in HeLa cells we show that Ii is able to compensate for loss of DMB-encoded targeting information. These data imply that in cells expressing physiological levels of class II, Ii and DM, there is sufficient association with Ii to direct the majority of DM into the endocytic pathway. Thus MHC class II and HLA-DM may follow similar intracellular trafficking pathways on route to antigen-processing compartments.
Book chapters
-
González-Navarro,H, Dogan S, Tuna BG, Koks G, Koks S, 'Aging in Rodents' in Suresh Rattan (ed.), Encyclopedia of Biomedical Gerontology, Elsevier (2019)
ISBN: 9780128160756AbstractPublished hereThis article gives an overview about the aging in rodents and how rodents can be used for aging modelling. The article starts with more general consideration about the modelling and some basic background. It is followed by the review of the most common progeroid syndromes along with the molecular mechanisms of aging. Then the effect of caloric restriction is described in deeper details. And finally, the role of transposable elements and the role of their activation during aging is described. Therefore, present article covers broadly the modelling of aging in the rodents with some more detailed overviews for the mechanisms explaining the potential interventions to modify the aging and aging related problems.
Conference papers
-
Nicol T, Falcone S, Blease A, Scudamore C, Hirst J, Viscomi C, Zeviani M, Brown SDM, Potter PK, 'P12 A novel model of cardiomyopathy reveals a tissue specific role for the complex i assembly factor ecsit'
104 (2018) pp.A6-
AbstractPublished hereHere we present a mouse model with a missense mutation in the gene Ecsit that shows a progressive cardiomyopathy from 4 weeks of age with no other overt phenotypes. ECSIT is known to play a role in development and immune signalling but is also thought to function as an assembly factor of complex I.
Western blot analysis of tissue lysates revealed a significant reduction in complex I proteins in heart tissue, whereas all other complexes were unaffected. In addition, Seahorse analysis of isolated mitochondria shows a significant reduction in the respiration rates of cardiac mitochondria, whilst no differences could be seen in mitochondria isolated from brain tissue.
In-gel activity demonstrated a significant drop in complex I activity of cardiac mitochondria, whilst brain mitochondria are maintained at close to normal levels. Blue native PAGE performed on cardiac mitochondria shows that this mutation affects ECSIT’s role in a limited number of complex I sub-assemblies. However, this is unique to the heart and mitochondria from brain tissue show no changes in any of the same sub-assemblies, supporting the initial findings that there is normal complex I assembly in the brain.
A potential mechanism lies in the discovery of a previously undescribed 16 kDa fragment of ECSIT that is present in WT cardiac mitochondria but not in mutant. This fragment is also undetectable in mitochondria isolated from brain tissue, indicating a tissue specific cleavage of ECSIT protein as a method of action.
Other publications
-
Schosserer M, Banks G, Dogan S, Dungel P, Fernandes A, Marolt Presen D, Matheu A, Osuchowski M, Potter P, Sanfeliu C, Tuna BG, Varela-Nieto I, Bellantuono I, 'Modelling physical resilience in ageing mice.', (2019)
AbstractGeroprotectors, a class of drugs targeting multiple deficits occurring with age, necessitate the development of new animal models to test their efficacy. The COST Action MouseAGE is a European network whose aim is to reach consensus on the translational path required for geroprotectors, interventions targeting the biology of ageing. In our previous work we identified frailty and loss of resilience as a potential target for geroprotectors. Frailty is the result of an accumulation of deficits, which occurs with age and reduces the ability to respond to adverse events (physical resilience). Modelling frailty and physical resilience in mice is challenging for many reasons. There is no consensus on the precise definition of frailty and resilience in patients or on how best to measure it. This makes it difficult to evaluate available mouse models. In addition, the characterization of those models is poor. Here we review potential models of physical resilience, focusing on those where there is some evidence that the administration of acute stressors requires integrative responses involving multiple tissues and where aged mice showed a delayed recovery or a worse outcome then young mice in response to the stressor. These models include sepsis, trauma, drug- and radiation exposure, kidney and brain ischemia, exposure to noise, heat and cold shock.Published here Open Access on RADAR -
Drechsler S, Lynch MA, Novella S, González-Navarro H, Hecimovic S, Barini E, Tucci V, Castro RE, Vandenbroucke RE, Osuchowski M, Potter PK, 'With mouse age comes wisdom: A review and suggestions of relevant mouse models for age-related conditions.', (2017)
AbstractAgeing is a complex multifactorial process that results in many changes in physiological changes processes that ultimately increase susceptibility to a wide range of diseases. As such an ageing population is resulting in a pressing need for more and improved treatments across an assortment of diseases. Such treatments can come from a better understanding of the pathogenic pathways which, in turn, can be derived from models of disease. Therefore the more closely the model resembles the disease situation the more likely relevant the data will be that is generated from them. Here we review the state of knowledge of mouse models of a range of diseases and aspects of an ageing physiology that are all germane to ageing. We also give recommendations on the most common mouse models on their relevance to the clinical situations occurring in aged patients and look forward as to how research in ageing models can be carried out. As we continue to elucidate the pathophysiology of disease, often through mouse models, we also learn what is needed to refine these models. Such factors can include better models, reflecting the ageing patient population, or a better phenotypic understanding of existing models.Published here -
Kõks S, Dogan S, Tuna B, González-Navarro H, Potter PK, Vandenbroucke R, 'Mouse models of ageing and their relevance to disease.', (2016)
AbstractAgeing is a process that gradually increases the organism's vulnerability to death. It affects different biological pathways, and the underlying cellular mechanisms are complex. In view of the growing disease burden of ageing populations, increasing efforts are being invested in understanding the pathways and mechanisms of ageing. We review some mouse models commonly used in studies on ageing, highlight the advantages and disadvantages of the different strategies, and discuss their relevance to disease susceptibility. In addition to addressing the genetics and phenotypic analysis of mice, we discuss examples of models of delayed or accelerated ageing and their modulation by caloric restriction.Published here
Professional information
Memberships of professional bodies
- Fellow of the Royal Society of Biology
- Member of the Centre for Osteoarthritis Pathogenesis funded by Versus Arthritis, Kennedy Centre, Oxford
- Member of the Society for Experimental Biology and Medicine
- Editorial Board; Experimental Biology and Medicine
Further details
Other experience
- Head of Disease Model Discovery, Mammalian Genetics Unit, Medical Research Council, Harwell (Sept 2010 to April 2018), Honorary Senior Lecturer (September 2009-Oct 2011, Kennedy Institute for Rheumatology, Imperial College)
- London Technology Network Business Fellow (March 2010-Oct 2011)
- Head of Mutagenesis (Nov 2007 – Aug 2010), Deputy Scientific Manager (Nov 2006 – Oct 2007), Mary Lyon Centre, Medical Research Council, Harwell
- Research Associate (reporting to Prof M. Botto, Prof Mark Walport)Faculty of Medicine, Imperial College (Hammersmith Hospital), London: Rheumatology Section (Nov 1999 – October 2006)
- Research Associate (reporting to Dr. K. Gould) ICSM (St. Mary’s Hospital), London: Department of Immunology (Jan 1998 – Oct 1999)
- Research Associate (reporting to Dr. A. Kelly) Guy’s Hospital, London: Renal Laboratory, Department of Medicine (July 1994- Jan 1998)
- Research Assistant (reporting to Dr. A.K. So) RPMS, Hammersmith Hospital, London: Rheumatology Unit (Jan 1988- July 1994)
- Research Assistant (reporting to Dr. John Harfield) Coulter Electronics, Luton (1983-1988) Particle sizing lab