Driving Discoveries 2023: Connecting our fellows and students
Abstract presentation details for our oral and poster presentations
Oral abstract presentations:

Louise McAlister
Authors and affiliations:
Louise McAlister1, Vanessa Shaw2, Rukshana Shroff1
1Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH
2UCL Great Ormond Street Institute of Child Health, London WC1N 1EH
Background:
Controlling dietary phosphate intake is integral to the management of chronic kidney disease but the practical implementation is not well described. We aimed to evaluate the quality and content of paediatric phosphate educational materials (PEMs).
Methods:
UK and international PEMs, obtained from paediatric renal dietitians, were screened. Duplicates, adult and non-phosphate focused resources were excluded. Quality was evaluated using suitability (Suitability Assessment of Materials [SAM]) and readability (Flesch Reading Ease [FRE] and Flesch-Kincaid [FK] Grade Level) instruments. Codes were assigned for format, appearance, target audience, resource type and content, and agreed by 3 dietitians (aiming for inter-coder agreement >80%). Content was compared to Paediatric Renal Nutrition Taskforce (PRNT) recommendations.
Results:
Of the 64 PEMs obtained, 37 (28 written, 2 pictorial, 1 recipe, 1 video, 1 app, 4 games) were paediatric-focused. The written PEMs’ target audience were caregivers (32%), children (25%) or unspecified (43%). 75% stated production date: 19% produced >5 years ago; 57% 2-<5 years; 24% <2 years. The median FRE test score was 68.2 (IQR 61.1-75.3) and FK reading grade level was 5.6 (4.5-7.7). 54% rated “superior” (SAM ≥70), 38% “adequate” (40-69) and 8% “not suitable” (£39). PEMs without a summary (12%), visuals (59%), cover-graphics (35%) or relevant images (50%) received the lowest scores. Inter-rater coding reliability was 87%. 50-96% included specific advice comparable to PRNT recommendations; 89% mentioned additives (Table1, Figure1).
Conclusion:
The written paediatric PEMs were pitched at an appropriate literacy level for adults (median FK grade 5.7, “aged 10-11 years”; FRE 68.2, “understood by 13-15-year-olds”). Only half rated “superior” (SAM), due to the absence of graphics and relevant illustrations with >⅓ containing inaccuracies and <⅓ including practical advice. In contrast to PRNT recommendations, >⅓ did not limit eggs, fish with bones, phosphate raising agents or seeds, and >⅓ limited wholegrains and legumes.
Lay summary
Title:
Dietary phosphate education materials for paediatric chronic kidney disease: is inconsistent messaging reducing their impact?
Background:
When the kidneys are not working well, a mineral called phosphate may accumulate in the body as the usual losses in the urine are decreased. This eventually weakens bones and stiffens blood vessels, damaging the heart. To prevent this, children and young people (CYP) with chronic kidney disease (CKD) are advised to eat less dietary phosphate but many find this difficult.
Your methodology: what have you done in your study?
We evaluated over 60 national and international phosphate educational materials (PEMs) for quality and readability. The advice was compared to international recommendations.
Results: What have you discovered?
Most PEMs were pitched at an appropriate adult reading level (10-11 years reading age) but a third contained inaccuracies. Inclusion of more relevant illustrations and practical advice could improve their quality. Over half limited the same foods as published recommendations, including avoidance of foods containing phosphate additives. However, some restricted wholegrains and legumes (eg peas and beans), which disagrees with recommendations.
What are the potential benefits that this research could bring to patients?
Low quality PEMs, including confusing messages, may make following dietary advice difficult. This work is part of a Kidney Research UK-funded study (IMPACT) which aims to improve phosphate control by hearing the real-life experiences and challenges of CYP and caregivers.
Authors and affiliations:

Anthony Onoja
Anthony Onoja1, Philip Kalra2, Maarten Taal3,4, and Nophar Geifman1
1School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey
2Dept of Renal Medicine, Salford Royal, Northern Care Alliance NHS Foundation Trust
3Department of Renal Medicine, University Hospitals of Derby and Burton NHS Foundation Trust
4Centre for Kidney Research and Innovation, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham
Background:
Chronic kidney disease (CKD), an increasing public health concern, requires biomarkers that can diagnose disease more specifically and sensitively, predict response to therapy, and predict course of disease. However, CKD is a broad term, covering a number of conditions, with many possible aetiologies, and high patient variability, all of which complicate the identification of biomarkers. Novel artificial intelligence approaches can help identify clinically meaningful groups of CKD patients, informative of biomedical profiles. Here, we aimed to identify clusters of patients with CKD, which correlate with phenotypic and clinical outcomes, and demonstrate greater association with potential biomarkers.
Methods:
Data on around 3000 adult CKD patients from the National Unified Renal Translational Research Enterprise (NURTuRE) were used to identify subgroups, based on clinical and demographic characteristics. We applied Latent Class Analysis, and derived the optimal number of classes (clusters). Differing associations between classes, in baseline clinical phenotypes and putative biomarkers, were then investigated.
Results:
Six distinct classes of CKD patients, exhibiting unique clinical characteristics, were identified. Class 1 consisted primarily of female patients; Class 5 included mainly male patients and the highest proportion of stage-2 CKD, as well as Glomerular disease. Class 2 included the highest proportion of stage-4 CKD patients while Class 6 included the highest proportion of stage-5 patients and those with ischaemic nephropathy. Class 3 included the largest proportion of those with diabetic nephropathy. Comparison of blood and urine biomarkers indicated that several exhibited significant differences between the six patient clusters.
Conclusion:
The identification of six distinct and clinically relevant classes of patients within the CKD population underscores the significant heterogeneity within the CKD population. By recognising these diverse profiles, approaches that consider the specific characteristics and risks associated with each patient class can be tailored. Further, associated biomarkers may provide mechanistic insights into treatments and personalised care.
Lay summary
Background:
Chronic Kidney Disease (CKD) is a multifaceted condition characterised by progressive kidney damage. Identifying differences, or patient diversity, within the CKD population is crucial for personalised disease management and care, taking us away from a more “one size fits all” approach.
Your methodology: what have you done in your study?
An artificial intelligence method that can identify grouping of patients from large and complex data was applied to NURTuRE clinical patient data; and the resulting identified groups were associated with blood- or urine-borne chemical differences (changes in specific proteins and other molecules) and clinical characteristics.
Results: What have you discovered?
Our approach identified 6 distinct patient subgroups, each associated with differing clinical characteristics. Further, the different groups were found to be associated with different blood and urine chemicals (biomarkers).
What are the potential benefits that this research could bring to patients?
These findings highlight the differences in CKD disease in the CKD patient population and the potential of blood or urine chemicals that we can measure for differentiating between patients and their disease characteristics. Understanding these distinct patterns aids in personalised treatment and helps in finding new targets in the body for drug treatment in CKD.
Keywords: CKD, Patient heterogeneity, Clustering, Biomarkers, Stratified medicine

Zilong Li
Authors and affiliations:
Zilong Li1, Hangfeng Zhang1, Haixue Yan1, and Lei Su1.
Matthew Bartlett, Royal Free Hospital
Ben Lindsay, Royal London Hospital
1Queen Mary University of London, 2Royal Free Hospital, 3The Royal London Hospital
The scarcity of medical resources and the increasing popularity of the Internet of Everything concept are driving innovation in the medical field, particularly in the development of sensors. Ultrasound sensors, capable of measuring blood flow and velocity using Doppler techniques, are particularly beneficial in renal monitoring and cardiovascular applications, such as assessing heart function, detecting blockages in blood vessels, or evaluating the effectiveness of treatments. The demand for flexible and wearable electronic devices has gained significant attention in recent years, with a particular focus on their use in medical applications for continuous cardiovascular monitoring. Sensors for medical applications are developing towards simplicity, flexibility, and intelligence for real-time monitoring of vital signs. To this end, the researchers are developing a flexible wearable medical device with a small size, high sensitivity and high stability. The core component of ultrasound sensors is the piezoelectric transducer, responsible for generating and detecting ultrasonic waves. Currently, lead titanate-based relaxor ferroelectric ceramics such as Pb(Mg0.33Nb0.67)xTi(1-x)O3 (PMNPT) represent the state-of-the-art materials, with superior piezoelectric performance, surpassing traditional commercial PbZrxTi1-xO3 based materials. To further improve the piezoelectric performance, we have conducted rare earth element doping in PMNPT to alter the free energy landscape of the material and reduce the energy barrier between different polar phases. Prototype 3 x 3 flexible transducer arrays were fabricated using developed ceramics and their ultrasound performance measured in water using hydrophone, revealing an operating frequency of 3.47 MHz and a bandwidth of 38.6% with an insertion loss of -6 dB. The potential of these flexible transducer arrays was demonstrated through the capturing of the flow speed variation of blood-mimicking fluid in a vessel-mimicking tube.
Lay summary
Lay title: Body-worn sensor for point-of-care monitoring
Our research is exploring the integration and miniaturisation of flexible ultrasonic sensing arrays and modules such as data processing, information transmission, and power supply. We have completed the patterning of the electrode array based on inkjet printing method. And we have completed the assembly and packaging of the ultrasonic sensor array and electrode array. Based on this flexible ultrasound array, we are able to locate blood vessels in the tissue-mimicking phantom and measure blood flow velocity. The development of a flexible wearable medical device with high sensitivity and stability could revolutionise continuous cardiovascular monitoring. It would allow patients to monitor their heart health in real-time, potentially detecting abnormalities or changes that may require medical attention earlier. The use of flexible and wearable devices could provide patients with greater comfort and convenience compared to traditional monitoring methods. It may eliminate the need for frequent hospital visits or uncomfortable procedures, allowing patients to carry out monitoring in their everyday lives.
Authors and affiliations:
Katherine Clark1, Eleanor Chilton2, Lauren Collins2, Sophie Webster2, Kathryn Dalrymple1, Kate Bramham1
1King’s College London, 2Women’s Health Research, King’s College NHS Foundation Trust
Background:
Acute Kidney Injury (AKI) is a rapid deterioration in kidney function and when it occurs in pregnancy (Pr-AKI) it is an independent risk factor for elevated maternal and fetal mortality. Optimal management of Pr-AKI is limited by lack of identification. Serial point-of-care testing to monitor changes in creatinine has been demonstrated to be successful in predicting and preventing AKI in other clinical settings but these tools have not been validated in pregnancy.
Methods:
A prospective longitudinal feasibility study (APRICOTS, IRAS 279788) is being undertaken at a tertiary London maternity unit. Pregnant people over 18-years old and able to give consent were offered participation. On intrapartum admission serial point-of-care NovaMaxCreat enzymatic creatinine testing using one drop of finger-prick capillary blood is offered at two or six-hourly intervals for participants with or without risk factors for Pr-AKI. Participants complete a postpartum acceptability survey postpartum giving scores out of 100 (0 totally unacceptable and 100 totally acceptable).
Results:
In six-months, 86 women were screened and 52 were consented to participate (60% recruitment rate). 20/52(38.5%) have completed the acceptability questionnaire to date with some outstanding. The mean score for overall acceptability of the test was 89.65(SD15.92). Mean scores for how easy, quick, convenient, and simple the test was to have done were 92.9(SD12.03), 91.55(SD15.84), 85.3(SD18.90) and 94.8(SD7.77) respectively. 55%(11/20) were happy to have the test more than 3-to-5 times in 24hours. Participants reported that it was fair to offer the test to someone who is pregnant (82.7,SD20.31) or who has given birth (82.30,SD20.31) with a lower mean score during labour (59.80,SD31.64).
Conclusion:
The feasibility progression criteria is expected to be met (75-recruits in nine months). Acceptability scores are reassuring but these findings will be confirmed on completion of the study with interviews performed to further explore aspects that are considered less acceptable.
Lay summary
Lay title: Acute kidney injury in pregnancy: can we improve diagnosis and find it earlier?
Background:
Acute kidney injury (AKI) is the sudden loss of kidney function. It can even lead to kidney and heart problems and even death. About one-third of AKI cases are preventable. The number of pregnant women with AKI has increased. Testing of creatinine (marker of kidney function) from finger-prick blood has been found to reduce AKI, but this test has not been studied in pregnancy.
Your methodology: what have you done in your study?
We are doing a study to check whether women are willing to have finger-prick tests for creatinine to predict AKI. People are asked to have finger-prick tests during their labour and birth. They are offered the test more often if they are at increased risk of AKI. After they have given birth they complete a questionnaire telling us how they feel about the test.
Results: What have you discovered?
People are happy to take part in the study. 86 people were asked to take part and 52 agreed. Participants agreed that the test was acceptable, and it was easy, quick, convenient and simple. 55% of people who were asked said that they were happy to have the test more than 3-5 times in 24 hours.
What are the potential benefits that this research could bring to patients?
When we finish this study we will design a future trial to reduce AKI in pregnancy.

Dr Simon Baker
Authors and affiliations:
Andrew S. Mason1, George Hatton1, Richard Gawne1, Sally James2, Andrew Macdonald3, Reuben S. Harris4, Matthew Wellberry-Smith5, Omar Masood5, Jennifer Southgate1 and Simon C. Baker1,*
1Jack Birch Unit for Molecular Carcinogenesis, Department of Biology and York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK.
2Bioscience Technology Facility, Department of Biology, University of York, Heslington, York YO10 5DD, UK.
3Faculty of Biological Sciences, School of Molecular and Cellular Pathology, University of Leeds, Leeds, UK.
4UT Health San Antonio, University of Texas, USA.
5Leeds Kidney Unit, St James’s University Hospital, Leeds, UK.
Background:
Whilst age and smoking are the established risk factors for urothelial carcinoma (UCa), studies of mutational signatures show the anti-viral apolipoprotein B mRNA editing enzyme catalytic polypeptide 3 (APOBEC3) enzymes, are responsible for the preponderance of mutations in bladder tumour genomes. BK polyomavirus (BKPyV) infects >80% of the UK population in childhood and infections persist into adulthood by lying dormant in the renal epithelium. Persistent BKPyV infections can be reactivated in the adult kidney at times of immune insufficiency (due to aging or immuno-suppression), leading to viruria and urothelial infection. However, UCa samples do not frequently contain BKPyV, suggesting a hit-and-run mechanism.
Methods:
We used two models of mitotically quiescent normal human urothelium to study sequalae of BKPyV-infection. Chronic infections of an in vitro tissue-engineered model were used to generate protein, RNA and DNA. DNA was used for Nanorate sequencing (NanoSeq) and mutational signature analysis. A ureteric organ culture system was assessed by immunohistology.
Results:
Ureteric organ cultures revealed that APOBEC3 protein induction occurs in “bystander” cells involved in the clearance of their infected neighbours by apical extrusion into the urine. As BKPyV-infections are lytically cytopathic, this novel observation of APOBEC3-induction in non-infected cells provides an explanation for the persistence of mutations after clearance of the infection. APOBEC3-induction during chronic BKPyV-infections of in vitro tissue-engineered urothelium was associated with the development of APOBEC3 mutational signatures, highly reminiscent of those observed in UCa genomes analysed in parallel. Analysing the wider context of APOBEC3 mutations in our models and UCa samples suggested APOBEC3A could be the enzyme responsible for initiating carcinogenesis.
Conclusion:
Using state-of-the-art human tissue models this study makes a robust case for BKPyV-infections as an important risk factor UCa by inducing the APOBEC3-mediated mutagenesis known to initiate the disease. Work is now ongoing to collect supporting data in patient samples.
Lay summary
Lay title: BK virus infections may explain the greater risk of bladder cancer in kidney transplant recipients.
BK virus infects most children and can hide in the kidneys throughout adulthood. In around one-in-five kidney transplant patients, the medication used to protect the transplant (called immunosuppression) leads to reactivation of BK infections.
When kidney cells sense that they have a BK infection, they slough off into the urine. Infected kidney cells pass down the tubes of the ureter and into the bladder, where the virus can infect the cells of the bladder lining.
The patterns of DNA damage or “mutations” that start cancers can be thought of like a fingerprint. Scientists use these patterns to identify the process causing the damage. This study used cutting-edge human bladder tissue models to show, for the first time, that BK virus infections in the lab can create similar patterns of mutations to those we observe in bladder cancers.
We know kidney transplant patients develop bladder cancers three-times more often than the general population, and commonly experience BK infections, so these results are especially relevant to kidney transplant recipients. However, as we age, BK virus is detected in urine samples from one-in-three adults, so this study has wider implications for those who develop bladder cancer in old age.

Roseanne E Billany
Authors and affiliations:
Roseanne E Billany1, Zahra Mubaarak1, Stephanie Burns2, Hannah ML Young3 Nicolette C Bishop4, Alice C Smith3, Matthew PM Graham-Brown1,2
1Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
2John Walls Renal Unit, University Hospitals of Leicester NHS Trust, UK
3Department of Population Health Sciences, University of Leicester, Leicester, UK
4School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
Background:
Kidney transplant recipients (KTRs) are prone to high rates of infection, malignancy and cardiovascular disease. Poor physical fitness and physical inactivity remain pertinent targets to improve post-transplant clinical outcomes.
Methods:
Eligible KTRs were randomised (1:1) to either a 12-week structured home-based exercise programme (INT, n=25) or 12-week best standard care control (CTR, n=25). Figure 1 outlines the home-based exercise programme. The a-priori thresholds for specific feasibility and acceptability criteria are as follows: recruitment success of 20% of eligible participants (≥2 participants per month), adherence (an average of three exercise sessions per week) and attrition (≤30%).
Results:
Ninety-four patients were approached and 50 (53%) recruited across 22 months of recruitment. Participant characteristics were: 50±14 years (INT 49±13; CTR 51±15), 23 male (INT 10; CTR 13), eGFR 59±19 ml/min/1.73 m2 (INT 60±20; CTR 61±21), 35 White British (WB),13 Asian, 1 Caribbean, and 1 Mixed ethnicity (INT 17 WB, 7 Asian, 1 Mixed; CTR 18 WB, 6 Asian, 1 Caribbean). Three participants withdrew from the intervention group (1 Covid-19 infection, 1 recurrent urine infections unrelated to the trial, 1 time/family circumstances) and one from the control group (lost to follow-up; 8% attrition). There were no adverse events reported. Intervention participants (n=22 completed) recorded an average of 4.4±1.4 exercise sessions per week (aerobic 2.8±1.1; strength 1.6±0.5).
Conclusion:
Results show that engagement with the home-based exercise programme in KTRs was excellent and the study comfortably exceeded a-priori thresholds relating to recruitment, retention and completion suggesting patients are interested in the study and the programme of exercise despite the current evidence showing physical activity levels are low. The groups are well matched and there is encouraging representation of female participants and participants from a non-white background. These initial results support further assessment and development of home-based programmes of exercise and activity for KTRs.
Lay summary
Lay title: Feasibility of home-based exercise in kidney transplant recipients
Background:
Kidney transplant recipients (KTRs) are at risk of many diseases but most importantly ones that affect the heart. Poor physical fitness and low levels of exercise are linked to more heart problems. Only 27% of KTRs are active enough to support good health. Research suggests that more support is needed.
Your methodology: what have you done in your study?
We recruited 50 KTRs and randomly assigned them to either a 12-week home-based exercise programme (Figure 1) or continuing on as they usually do for 12 weeks (25 in each). We took various health measurements at the start and after the 12 weeks.
Results: What have you discovered?
We recruited 50 of the 94 patients that we approached, suggesting patients were interested in the study. Only 4 participants withdrew from the study. And of the participants who completed the exercise programme, they recorded an average of 4 exercise sessions per week.
What are the potential benefits that this research could bring to patients?
Results suggest engagement with the home-based exercise programme in KTRs was excellent and that further research would be beneficial to develop home-based programmes to help KTRs become more active and lower the risk of heart problems.

Sarah Fawaz
Authors and affiliations:
Sarah Fawaz1, Ivan Hartling2, Rebecca Vaughan1, Rutger Ploeg1, Philip Charles2, Maria Kaisar1
1University of Oxford, Nuffield department of Surgical Sciences
2University of Oxford, Target Discovery Institute, Nuffield Department of Medicine
Background:
Organ transplantation is a life-saving treatment of end-stage diseases. Yet, shortage of donors and uncertainty for the quality of donor organs limit the full benefit of organ transplantation. Recent research suggests that biomarkers of the TNFα pathway are associated with kidney function decline. Soluble TNFα, TNFR1 and TNFR2 receptors are inflammatory markers with strong prognostic value of progression from acute injury to chronic kidney dysfunction. In this study, we evaluated donor longitudinal circulating levels of TNF-α, TNFR1 and TNFR2 during donor management and compute relevant associations with 12-month posttransplant function.
Methods:
Plasma samples from 251 deceased donors (donation after brain death [DBD] = 132 and donation after circulatory death [DCD] = 119) were obtained by the Quality in Organ Donor (QUOD) biobank. Plasma samples from living donors (n=20) obtained from the Oxford Transplant Biobank, provided a baseline reading for these analytes. Using Luminex assay, we quantified the circulating levels of TNF-α and receptors in serial donor plasma collected during donor management at three time points (time of brain stem death test; B2, start of retrieval; B3, prior to cross clamp; B4) in DBD and two corresponding time points in DCD (prior to withdrawal of life support; B2, start of retrieval; B3).
Results:
Circulating levels of TNF-α and receptors differ among donor types. In DBDs, high levels of TNF receptors following brain death reduced during donor management before reaching their highest level prior to kidney retrieval (time point B4). Notably, only in DBDs plasma levels of TNF-α (p=0.00001) and TNFR1 (p=0.012) strongly associated to 12-month suboptimal function (eGFR<40ml/min) posttransplant. All analytes had baseline levels in living donor plasma prior to kidney procurement.
Conclusion:
Our study demonstrates that circulating inflammatory levels differ in DBDs when compared to DCDs. Increased levels of TNF-α mediators in deceased donors may deem donor organs susceptible to posttransplant injury and suboptimal function at 12-m post-transplant function.
Lay summary
Lay title: Can we predict how well kidneys will work after transplant by looking at the inflammation level specifically TNF-α in the donor blood?
Background:
Kidney transplantation is the best treatment for kidney disease. However, a high number of patients die while on the transplant list. To save more lives, transplant clinicians accept more kidneys from older or sicker donors who may suffer from undetected kidney disease. Some of these donor kidneys may not function so well long term, resulting in the recipient having to return to dialysis. Here, we measured certain markers in donor plasma to predict how well the transplanted kidneys might function in the recipients. These markers include soluble TNFα, TNFR1, and TNFR2 receptors, which are indicators of inflammation.
Your methodology: what have you done in your study?
Plasma samples were collected from a total of 251 deceased donors, with 132 brain-death donors (DBD) and 119 donors classified as donation after circulatory death (DCD). Using a technique called Luminex assay, we measured the levels of TNF-α and its receptors in the donor plasma samples at different time points from the beginning of brain stem death to the retrieval of the kidney.
Results: What have you discovered?
In DBDs, the levels of TNF-α and TNFR1 in the plasma strongly correlates with declining transplant function (eGFR<40ml/min) at 12 months after transplantation. Higher levels of donor plasma TNF-α and TNFR1 levels associated with poorer kidney function in the recipient after the transplant.
Overall, these findings suggest that TNF-α and its receptors may play a role in predicting the post-transplant kidney function, particularly in the case of DBDs.
What are the potential benefits that this research could bring to patients?
These findings highlight the importance of considering the inflammatory status of donors when evaluating the quality and potential success of organ transplantation. By understanding these differences, we can develop better tools to assess donor kidneys and improve the overall outcomes for transplant recipients.

Haresh Selvaskandan
Authors and affiliations:
Haresh Selvaskandan1, Charlotte Boys2, Izabella Pawluczyk1, Jonathan Barratt1
1Mayer IgA Nephropathy Group, University of Leicester
2Heidelberg University
Background:
Many living with IgA nephropathy (IgAN) progress to kidney failure (KF). Endocapillary hypercellularity (E1, a histopathological lesion) confers an increased KF risk, which appears reversible with systemic immunosuppression. This improves outcomes but confers severe side effects. E1 is defined by the obliteration of glomerular capillary lumens by inflammatory cells; delineating transcriptome changes in glomerular endothelial cells (GEnCs) associated with E1 may highlight safer therapeutic targets. GEnC transcriptomes have never been profiled in diseased kidneys before. We used digital spatial profiling (DSP) to achieve this.
Methods:
DSP was performed using a Nanostring GeoMx. 5micrometre sections were collected from formalin-fixed paraffin-embedded (FFPE) biopsies (four E1 and five E0). Tissues were deparaffinized, antigen retrieved, and incubated with whole transcriptome probes. GEnCs and macrophages were stained (CD31, CD68) with primary conjugated antibodies. A custom JavaScript function was used to select GEnCs and macrophages (segmentation) within glomeruli (Fig-1). Photocleaved nucleotide barcodes from individual cell types were sequenced (Illumina).
Single-cell enrichment was assessed using single-cell deconvolution, differential gene expression (DGE) explored using a linear mixed effects model, and pathway analysis was performed using Reactome.
Results:
The JavaScript function allowed good segmentation (Fig-1). Single cell deconvolution performed against the human kidney cell atlas showed significant enrichment of GEnCs signals (Fig-2). TRIM23, IL27RA, TMEM139, P14K2B and PSMD9, were associated with E1 on DGE analysis, after p-value adjustment (Fig-3). Pathway analysis based on DGE revealed the complement cascade was enhanced in E1 GEnCs (Fig-4).
Conclusion:
DSP with the GeoMx was effective at enriching for GEnC transcript signals from FFPE tissue. This preliminary data also shows that E1 GEnCs display an inflammatory phenotype, likely related complement activity. With several trials investigating complement-targeting therapeutics for IgAN, validation of these findings may highlight a cohort of patients with IgAN likely to respond to complement pathway treatments.
Lay summary :
Discovering safer treatments for those with a kidney disease called IgA nephropathy.
Background:
IgA nephropathy (IgAN) is a common disease which affects the kidney’s filters; many affected develop kidney failure (KF). ‘Endocapillary hypercellularity (E1) is a type of microscopic kidney injury that occurs in IgAN. It increases the risk of KF, but seems treatable with medications that suppress the body’s immune system. These seem beneficial but have severe side effects. This project used cutting-edge technology to study E1 to find safer treatments for it.
Your methodology: what have you done in your study?
We used digital spatial profiling (DSP) to study E1. This involves marking kidney cells affected by E1 with different colours, and then shining a precise beam of light on these cells to study which genes are switched on and off in them, by triggering a controlled chemical reaction. We then used computer programs to study active genes in E1, to identify new ways of treating it.
Results: What have you discovered?
We found overactivity of an aggressive biological system called the complement system in E1. This usually protects us from infections. Overactivity of the complement system causes many kidney diseases, and already treatments are being developed to temper it. If we can confirm these results, it can highlight a group of patients with IgAN who would be ideal for these types of treatment.

Ahmed Ahmed
Authors and affiliations:
Ahmed Ahmed1,2, Anna Winterbottom1,2, Shenaz Ahmed3, John Stoves 4, Sunil Daga1,5
1 Department of Renal Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
2 Leeds Institute of Health Sciences, Faculty of Medicine and Health, University of Leeds, Leeds, England, United Kingdom
3 Division of Psychological and Social Medicine, Leeds Institute of Health Sciences, University of Leeds, United Kingdom
4 Department of Renal Medicine, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, United Kingdom,
5 Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, England, United Kingdom
Background:
Decision-making about living donor kidney transplant (LDKT) is challenging for people with advanced kidney disease (AKD). Fear of risks, insufficient knowledge, religious beliefs, and complex family dynamics are cited as reasons for not choosing LDKT. With the aim of improving informed decision-making and experience of care, our study examined the decisional needs of people with AKD in a diverse population in the United Kingdom.
Methods:
Semi-structured interviews with people with AKD from two kidney units in West Yorkshire were conducted. Purposive sampling (N=30) ensured ethnic diversity (19 minority ethnic groups), gender balance (16 females), and socially disadvantaged (19). Participants from the waiting list (13) and those who had a kidney (only) transplant within the last two years (7 received LDKT and 9 deceased donor kidney transplant) participated. Thematic analysis was conducted.
Results:
Three themes were generated:
- Knowledge: about benefits of LDKT, donor work-up process, financial implication, and long-term outcomes. Knowledge gaps were predominantly identified among those with low education and socially deprived areas irrespective of their ethnic groups.
- Family and social matters: post-transplant relationship concerns, guilt around potential transplant failure, community perception of organ donation, fear of isolation and compromised opportunities for marriage and starting a family for female.
- Deliberation and validation: need to clarify religious views, deliberation around fate and religion as well as cultural norms, desire to meet others in receipt of transplant of a similar background. Needs related to religion and culture were only identified among older minority ethnic groups.
Conclusion:
While ethnicity has been identified as a significant factor associated with transplantation decision preference, it appears to intersect with other factors such as age, gender, social deprivation, and education levels. These findings and those of a parallel study exploring kidney health professionals’ views, will inform the development of a decision support intervention for people with AKD from diverse ethnic backgrounds in UK kidney units.
Lay summary
Lay title:
What do people with kidney disease from various backgrounds need when they decide about pursuing a kidney transplant from a living donor?
Background:
People with kidney disease often struggle to decide about living-donor kidney transplant. Some worry about the health of their loved ones, while others may not have enough information. Our study examined the decisional needs of a diverse UK population to help more kidney disease patients make better care decisions.
Your methodology: what have you done in your study?
We conducted interviews with people with kidney disease in West Yorkshire ensuring diversity in gender, ethnicity, education, and socioeconomic status.
Results: What have you discovered?
Decisional needs identified:
- Knowledge: of living-donor kidney transplantation benefits, donor assessment and preparation, health and financial impact of donation. Low-educated, socially deprived people had the most knowledge gaps, regardless of ethnicity.
- Deliberation and validation: Including the need to clarify religious beliefs around transplantation, fate, and culture. Kidney transplant recipients felt the need to meet other transplant recipients from similar backgrounds. Only older minority ethnic groups had religious and cultural needs around transplantation.
- Family and social matters: Relationship concerns, guilt over a possible transplant failure, how the community views organ donation, fear of being alone, and concerns over women's chances of marrying and having children after transplantation.
What are the potential benefits that this research could bring to patients?
These results and a separate study on kidney health professionals' opinions will be used to create transplant decision-support interventions for UK kidney disease patients of different ethnicities.

Dr Jennifer Chandler
Authors and affiliations:
Jennifer C. Chandler1, Daniyal Jafree1, Saif Malik1, Gideon Pomeranz1, Maria Kolatsi-Joannou1, Andrew Mason2, Aoife M Waters1, David A Long1
1 Developmental Biology and Cancer, UCL Institute of Child Health, London, UK
2 Department of Biology, University of York, UK
Background:
Cellular communication within the kidney glomerulus is critical for healthy filtration and its disruption is associated with adult diseases such as diabetic nephropathy. However, little is known about glomerular cell communication in childhood glomerulopathies, many of which have no effective treatments. We chose to explore this in the context of WT1 glomerulopathy, mutations in which are one of the most common causes of childhood glomerular disease.
Methods:
Disease progression was characterised in a murine model (Wt1R394W/+) of childhood WT1 glomerulopathy, using biochemical and histological approaches. Single-cell RNA sequencing (scRNA-seq) and bioinformatic analyses were conducted on glomeruli isolated from wildtype (Wt1+/+) and mutant (Wt1R394W/+) mice. Validation was performed on murine and human tissue using immunofluorescence, RT-qPCR, ELISA and in vitro assays.
Results:
Wt1R394W/+ mice have elevated urinary albumin (p<0.0001) from 4 weeks of age, prior to overt histological features of glomerulosclerosis which occur by 8 weeks. Using ligand-receptor analysis on scRNA-seq data from glomeruli isolated at 4 weeks, we identified vascular signaling to be the cellular interaction most disrupted in Wt1R394W/+ glomeruli, with CD31+ staining demonstrating a loss of the glomerular endothelium by 8 weeks of age (p<0.0001). Patient tissue carrying the same WT1p.R394W mutation also showed a loss of the glomerular endothelium (p<0.0001). Amongst a milieu of disrupted molecules, we found that adrenomedullin (AM) was upregulated in Wt1R394W/+ murine podocytes and serum, a finding confirmed in Nephroseq data from human glomerular diseases. Finally, we conducted an in vitro angiogenesis assay with glomerular endothelial cells exposed to media conditioned by Wt1+/+ and Wt1R394W/+ primary podocytes, demonstrating a correlation between AM concentration in the conditioned media and endothelial cell tubular formation.
Conclusion:
This work has generated a scRNA-seq dataset for childhood WT1 glomerulopathy. Wt1R394W/+ podocytes show disruption to several pro-vascular molecules in early disease, driving endothelial loss. Amongst these molecules, AM warrants further investigation as a therapeutic candidate to support vascular health in glomerulopathies.
Lay summary
Lay title: Supporting kidney blood vessels may slow the progression of childhood glomerular disease
Background:
Many children with kidney disease do not respond to commonly used drugs; this is particularly true of genetic disorders that affect the kidney glomerulus, the filtration units of the kidney. Therefore, there is a critical need to establish new treatment options for these young patients.
Your methodology: what have you done in your study?
We analysed a pre-clinical model of childhood glomerular disease, using a technique that measures the level of all the genes produced in each glomerular cell. We then counted the number of glomerular blood vessels in the disease-model and patient tissue. Finally, we measured the levels of a protein called adrenomedullin in glomerular cells and blood serum.
Results: What have you discovered?
The production of several genes important for blood vessel health was disrupted during early disease in the glomerulus. This was associated with a loss of blood vessels in later disease in our model and in patients. Amongst the disrupted factors, we identified adrenomedullin, a protein that can support the blood vessels and potentially prevent their loss.
What are the potential benefits that this research could bring to patients?
Adrenomedullin is a potential therapeutic candidate which could slow damage to glomerular blood vessels and thus the onset of kidney failure. In the long term, this could provide an alternative therapy for patients, delaying the need for dialysis or transplantation.

Dr Tanya Smith
Authors and affiliations:
Dr Tanya Smith1,2, Dr Yueh-An Lu3, Dr Sumukh Deshpande2, Dr Chia-Te Liao3, Dr Bnar Talabani2, Dr Robert Andrews2, Dr Timothy Bowen1,2, Professor Philip Taylor4 , Professor Donald Fraser1,2
1Wales Kidney Research Unit, School of Medicine, Cardiff
2Division of Infection and Immunity, School of Medicine, Cardiff
3Division of Nephrology, Kidney Research Center, Linkou Chang Gung Memorial Hospital, Taipei
4Dementia Research Institute, Cardiff
Background:
The cellular states exhibited by renal proximal tubular cells (PTCs) during dedifferentiation, proliferation, and reacquisition of a fully differentiated phenotype have been poorly characterised. This critical knowledge gap is likely to be of importance for PTC proliferation, physiological growth of the kidney, as well as in recovery from kidney injury. In addition, PTCs exhibit sex differences in propensity for recovery, but sex-specific differences in PTC phenotype are not well characterised.
Methods:
Kidneys were harvested from naïve female and male mice at 1, 2, 4, and 12 weeks of age (n=2 per group, 16 mice in total). Libraries were prepared on the 10x platform, and single nuclear RNA sequencing (snRNAseq) was completed using the Illumina NextSeq 550 System.
Results:
Unbiased clustering analysis was performed on 68,775 nuclei obtained from whole kidney. All expected cell types were identified in the primary analysis. High levels of proliferation were evident at early time points in some clusters (e.g. tubular cells) but were absent in others (e.g. podocytes). Proliferation was especially evident in PTCs which are the most abundant cell type in the adult kidney. Uniquely, when compared to other kidney cell types, PTCs demonstrated sex-specific expression profiles at 4 and 12 weeks. Mapping of PTC differentiation pathways using techniques including trajectory and RNA Velocity analyses delineated increasing PTC specialization and sex-specific phenotype specification. Trajectory analysis has identified the differentiation pathway for tubular cells. Ongoing work is focussed on ligand-receptor analysis, pathways underpinning stromal-epithelial interaction, and localisation studies employing fluorescence microscopy.
Conclusion:
For the first time, we have characterised sex-specific phenotypical subtypes of PTCs in the growing kidney and differentiation pathways leading to these phenotypes. This new appreciation may enable us to develop ways to protect patients from kidney disease and more effectively treat them.
Lay summary
Lay title: What’s so special about the female kidney? Understanding the differences in the healthy male and female kidney
Background:
Acute kidney injury (AKI) remains a serious problem. Even for those who recover, AKI may have a lasting impact, with a risk of progression to chronic kidney disease. For reasons unknown, the female kidney appears to be protected from some forms of kidney injury and is more likely to recover than the male kidney. My project aims to understand these differences which may help us to develop ways to protect patients from kidney disease and more effectively treat them.
Your methodology: what have you done in your study?
I applied a new technique, called ‘single cell sequencing’, which allows me to identify the different cells (the individual building blocks of the kidney) in the kidney.
Results: What have you discovered?
For the first time, we have discovered sex-specific subtypes of kidney cells in the growing kidney.
What are the potential benefits that this research could bring to patients?
Knowing which cells are different within the male and female kidney may enable us to understand the genes responsible for protecting the kidney from damage. In the future, I hope that this will allow us to personalise therapy and to better inform patients of their risk of kidney damage prior to surgery.

Samin Chowdhury
Authors and affiliations:
Samin Chowdhury1
1University of Lancaster
Background:
Multimorbidity is the concurrent presence of two or more long-term health conditions in the same individual. It fragments the healthcare delivery and affects the quality of life. Chronic kidney disease (CKD) often occurs with multimorbidity. The prevalence of CKD is rising. However, there is a lack of evidence on prevalence, patterns, and the impacts of multimorbidity on adverse clinical outcomes in patients with CKD.
Methods:
Systematically conducted literature review. A search was conducted on EMBASE, MEDLINE, CINAHL, and SCOPUS (2019-2023). The main search terms were “chronic kidney disease” and “multimorbidity.” Eligibility criteria were observational studies with adult participants with all stages of CKD (CKD stage 1-5 including those on renal replacement therapy). The exposure was multimorbidity quantified by measures. All-cause mortality, kidney disease progression, hospitalisation, and cardiovascular events were outcomes. Joanna Briggs Institute (JBI) checklist was used for the risk of bias assessment. Due to heterogeneity in design and methods, Jennie Popay’s narrative synthesis was used for data synthesis.
Results:
Of 6879 papers, nine papers met the inclusion criteria. Most studies included participants with all stages of CKD (CKD stage 1-5).
The prevalence of multimorbidity ranged from 86.6% to 99.1%. Hypertension was the most prevalent comorbidity.
The combination of concordant multimorbidity (hypertension, diabetes, and cardiovascular diseases) was highly prevalent.
Multimorbidity was significantly associated with mortality, cardiovascular events, kidney disease progression, and hospitalisation. While older people had more multimorbidity burdens, younger patients with CKD were at a higher risk of death from multimorbidity (HR 1.96 [95% CI: 1.61-2.37]). Severe CKD with clusters of cardiovascular diseases, diabetes, chronic pain, and depression were significantly associated with all-cause mortality.
Conclusion:
There are associations between multimorbidity and adverse clinical outcomes in patients with CKD. However, there is a lack of data on Black, Asian, and Minority Ethnic participants from low and middle-income countries.
Lay summary
Lay title: Multiple health conditions in people with chronic kidney disease: frequency, trends, and associated health results.
Background:
Multiple health conditions describes when a person simultaneously has two or more long-term illnesses. It affects how long a person lives and how well they live. Chronic kidney disease (CKD) is progressive damage to the kidneys. It affects 13% of people around the world and this number is rising. It often takes place with multiple health conditions.
There is not enough data to tell us how multiple health conditions affect someone with chronic kidney disease. So, this study aims to discover the common multiple health conditions and how they affect someone with kidney disease.
Your methodology: what have you done in your study?
This study looked at a large amount of research already done in this area. People with CKD and multiple health conditions were included. The effects of multiple health conditions on the risk of hospital admission, kidney disease progression, and death were studied.
Results: What have you discovered?
Multiple health conditions are highly common in people with CKD (86.6%-99.1%). They are associated with a higher risk of hospital admission, worsening of kidney function, and death.
What are the potential benefits that this research could bring to patients?
In the near future, this study will help healthcare workers identify people with kidney disease who have multiple health conditions earlier. This will reduce their risk of hospital admission, worsening kidney function, and death.

Sarah Gleeson
Authors and affiliations:
Sarah Gleeson1,2, Jack Beadle1,2, Linda Moran2, Liz Lightstone1,2, Candice Roufosse1,2, Michelle Willicombe1,2
1 Imperial College London
2 Imperial College Healthcare NHS Trust
Background:
Tubuloreticular inclusions(TRIs) seen on electron microscopy(EM) are classically associated with lupus nephritis(LN) and systemic viral infections in native biopsies. Traditionally a marker for enhanced type I interferon expression, little is known about their significance post-transplant. We aimed to look at a large cohort of transplant biopsies showing TRIs to investigate associations and outcomes.
Methods:
A retrospective analysis was performed on two prospective databases; an in-centre transplant registry and a histopathology database holding data on all kidney biopsies performed at our centre. All patients biopsied since 2015, who had EM examination were included. Where patients had more than one biopsy showing a TRI the earliest one was included. Demographic, clinical and transplant data was collected from the laboratory records. ABO incompatible transplants were excluded.
Results:
7998 kidney transplant biopsies were performed between December 2005 and December 2022; 1740 (21.7%) had EM performed. Of 1740 with EM, 256 (14.7%) had evidence of TRIs on at least one biopsy. Of 256 patients, 34% were female, the median age was 52.3 years (38.9-59.4), 28% had underlying glomerulonephritis as their cause of end-stage kidney disease, 61% were deceased donors and 68% were of non-white ethnicity.
TRIs were associated with serological evidence of autoimmunity (17%), viral infections (28%) and donor specific antibodies (35 %), with no association found in 34%. Rejection occurred in 127 (50%), including 38(44%) of patients with no recognised association with TRIs.
Allograft outcomes were poor, with all-cause allograft survival and death-censored allograft survival of 54% and 70% after a follow up of 5.7 +/- 3.8 years post index biopsy. A comparison with a matched control group is underway.
Conclusion:
In extension to previous work, we show that TRIs appear to be associated with alloimmunity. In this regard they may be a useful biomarker especially in cases where the diagnosis is unclear or biopsy findings are ‘subthreshold’. Irrespective of aetiology, TRIs are associated with poor outcomes and warrant further consideration.
Lay summary
Lay title: Tubuloreticular inclusions and kidney transplant
Background:
When someone’s kidney transplant isn’t working, we often do a biopsy to get some more information about why it’s not working. Sometimes we see structures on electron microscopy (EM), a special part of the biopsy called tubuloreticular inclusions (TRIs). The importance of these structures isn’t understood. In non-transplant kidney biopsies TRIs are often seen when patients have a kidney disease called lupus or when they have a viral infection. A previous study suggested that, in kidney transplants TRIs may be associated with rejection.
Your methodology: what have you done in your study?
I have looked back at all patients transplanted in my centre who have had a TRI on a transplant biopsy. I have also looked at a control group of patients with EM who didn’t have TRIs. I looked at the findings on the biopsy, the clinical situation at the time of the biopsy and how the patients did long term. This was compared between the groups.
Results: What have you discovered?
7998 kidney transplant biopsies were performed between December 2005 and December 2022; 1740 (21.7%) had EM performed. Of 1740 with EM, 256 (14.7%) had evidence of TRIs on at least one biopsy. Of 256 patients, 34% were female, the median age was 52.3 years (38.9-59.4), 28% had underlying glomerulonephritis (immune kidney disease) as their cause of ESKD. 61% of the transplants were from deceased donors.
What could this mean for patients?
When we do biopsies we want to get as much information as possible about the underlying cause of kidney function. The presence of a TRI could give us more information which may allow us to start treatment earlier or ensure we do closer monitoring to hopefully allow us to keep the kidney transplants working for longer.

Nancy Smart
Authors and affiliations:
Nancy Smart1, Babarinde Omolola Adefunke, Caroline Bull, Emma E. Vincent1, Gavin Welsh1, Kaitlin Wade1 and Abigail Lay2
1University of Bristol
2University of Manchester
Background:
Despite being the leading cause of kidney disease, diabetic kidney disease (DKD) has few treatment options, and its incidence is continually increasing. The microbiome impacts many aspects of host health, such as physiology and immunity, making it a potential avenue to discover and develop novel therapeutic targets and biomarkers for DKD. The metabolome has also been explored as a potential mechanism for the action of the microbiome. Through the use of Mendelian randomization (MR), this study aims to identify if the microbiome has a causal role in the development of DKD that can be exploited for therapeutic treatments.
Methods:
Bi-directional two-sample Mendelian randomization analyses using inverse variance weighted models were performed to assess the role of the microbiome and metabolome on DKD. Both the estimated glomerular filtration rate (eGFR) and the urine albumin-creatinine ratio (uACR) have been used as proxies to measure DKD. Single nucleotide polymorphisms (SNPs) associated with these proxies, the metabolome, or microbiome, have been taken from publicly available Genome-Wide-Association Studies (GWAS) of European populations. This includes data from the CKDGen Consortium, Nightingale Health Laboratories and UKBiobank amongst others. Sensitivity analyses including Radial MR and colocalization are being performed to check for pleiotropy and heterogeneity.
Results:
Current analysis suggests that the metabolites glycine and glycoprotein acetyls could both promote kidney health, with glycine associated with lower uACR levels and glycoprotein acetyls promoting eGFR levels. Additional results for the microbiome MR analyses will be obtained before the conference, alongside further sensitivity analysis data.
Conclusion:
Certain metabolites do appear to have protective effects on the kidney, which could be exploited to create therapeutic treatments. The microbiome may not play a causal role in DKD but could still be used to create diagnostic tests allowing more precise tracking of DKD incidence and progression.
Lay summary
Lay title: Microorganisms within the human gut could be causing diabetic kidney disease
Background:
Diabetic kidney disease (DKD) is the leading cause of kidney disease globally. Despite this there are few treatment options and those available, such as dialysis or transplantation, can have a significant impact on a patient’s mental and physical health. There is a desperate need for more treatments and preventative strategies.
Your methodology: what have you done in your study?
Data from large European populations has been analysed to see if there are relationships between DKD and either metabolites in blood, or the bacteria in the gut.
Results: What have you discovered?
We have identified several metabolites that are associated with DKD that have potentially protective effects.
Our research could help the development of non-invasive diagnostic tests, allowing early detection of DKD and thus early interventions. Metabolites or bacteria identified as significant could also be exploited to create new treatments that prevent the development of DKD. It might even be possible to create a diet that promotes gut health and subsequently kidney health.

Jess Ivy
Authors and affiliations:
Georgios Krilis1, Frances Turner1, Hannah M Costello1, Matthew A Bailey1, Jessica R Ivy1
1University of Edinburgh
Background:
Major physiological parameters have a circadian rhythm including blood pressure (BP), which is elevated during the day and dips at night. Loss of this BP rhythm (termed “non-dipping”) is common in kidney disease and is an independent risk factor for adverse cardiovascular outcomes. Restoring normal circadian BP rhythm is important clinical goal but underlying mechanisms remain obscure. Molecular clocks, present in all cells, set the rhythm of gene expression and allow predictive temporal control of physiological function. This rhythm is set by the master clock in the brain and is communicated to the body via glucocorticoid signaling, allowing the synchronization of peripheral clocks to the external world. Attenuation of the glucocorticoid rhythm robustly induces non-dipping BP. The renal arteries and arterioles influence renal haemodynamics and contribute to long-term BP stability. We hypothesise that glucocorticoid treatment disrupts the rhythm of vascular function and gene expression in the renal artery and contributes to non-dipping BP.
Methods:
Male C57BL6 mice kept on a 12:12 light:dark schedule, were treated chronically with corticosterone to flatten the normal circadian rhythm of plasma glucocorticoids. Mice were culled either at 7am/7pm and renal arteries dissected for functional assessment using wire myography or every 2 hours for 48 hours and renal arteries collected for RNA sequencing.
Results:
Renal arteries in control mice exhibited diurnal variation in their dilatory response to sodium nitroprusside with increased dilation during the active period. This variation was absent in the corticosterone-treated mice. In control renal arteries 3% (465/14 425) of protein-coding genes were rhythmic. 156 of these genes lost their rhythm following treatment, including components of the molecular clock but surprisingly 465 genes gained a rhythm.
Conclusion:
In the mouse renal artery, vasodilatory function exhibits a diurnal rhythm. Arrhythmic glucocorticoid signaling abolishes this rhythm and causes dampening of genes involved in normal circadian rhythms and induces de novo rhythms.
Lay summary
Lay title: Stress hormones dampen the daily rhythm of kidney artery.
Body clocks allow us to adapt to our rhythmic environment of light and dark. They control our physiology, including blood pressure. Blood pressure has a daily rhythm, being 10% lower at night than during the day. When this rhythm is blunted, which happens in most chronic kidney disease patients, this is called non-dipping. Non-dipping is associated with an increased risk of cardiovascular disease over and above the risk associated with high blood pressure. Blood vessels in the kidney contribute to blood pressure regulation but we don’t know this works over a 24-hour day. Stress hormones cause non-dipping and change our body clocks. In this study we measured the function and gene changes of kidney arteries at different time points in mice treated with stress hormones. We found that kidney artery dilation has a rhythm with better dilation during wakefulness, but this rhythm was absent in stress-hormone treated mice. We found stress hormone treatment caused 156 genes to lose their rhythm but surprisingly 465 genes gained a rhythm. Discovering how these genes change kidney artery function over the course of the day could unveil new ways to improve blood pressure rhythmicity and the associated cardiovascular disease risk in people with non-dipping blood pressure.

Amy J. Osborne
Authors and affiliations:
Amy J. Osborne,1 Agnieszka Bierzynska,2 Elizabeth Colby,2 Philip A. Kalra,3 Maarten W. Taal,4 Gavin I. Welsh,2 Moin A. Saleem,2 Colin Campbell1*
1Intelligent Systems Laboratory, University of Bristol, Bristol, BS8 1TW, United Kingdom.
2Bristol Renal, University of Bristol and Bristol Royal Hospital for Children, Bristol, BS1 3NY, United Kingdom.
3Department of Renal Medicine, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Stott Lane, Salford, M6 8HD, UK.
4Centre for Kidney Research and Innovation, University of Nottingham, Derby, UK.
Background:
Chronic kidney disease (CKD) affects approximately 10% of the population and is a heterogeneous group of multiple renal pathologies, some with unknown disease mechanisms. Nephrotic syndrome (NS) is a major cause of kidney disease that can be due to a range of genetic and non-genetic causes. Several genetic risk loci have been associated with overall CKD susceptibility by statistical analyses of common single nucleotide polymorphisms (SNPs). However, it is currently unknown whether multiple SNPs show associations with each of the CKD and idiopathic NS (INS) sub-types, progression rates and proteinuria levels.
Methods:
We analysed the correlation between SNP genotypes and CKD subtype, estimated glomerular filtration rate (eGFR) slope and time-average proteinuria, jointly, in each of the NURTuRE-CKD and Salford Kidney Study datasets, by using a multivariate statistical technique called canonical correlation analysis (CCA). We also used CCA to analyse SNPs and INS subtypes in NURTuRE-INS. We tested whether neural network supervised machine learning could help to predict disease subtypes and progression rates based on the CCA-identified SNPs.
Results:
We identified 101 replicated SNPs that showed significant correlation with CKD-subtype, proteinuria and eGFR slope, jointly, and 161 SNPs with CKD subtype only. Furthermore, we identified 943 SNPs that showed significant correlation with membranous nephropathy. These replicated SNPs were mainly located in the HLA region and also showed associations with immune gene expression quantitative trait loci, including C4A. Of the identified SNPs, six had previous disease associations including with nephrotic syndrome and membranous glomerulonephritis. In addition, using combination-SNP CCA followed by neural network supervised machine learning of INS subtype data, we identified several SNPs suggestive of predicting INS subtype with good accuracy (83%), however further testing in additional cohorts would be needed.
Conclusion:
In summary, our analyses suggest that multiple immune genetic markers are associated with CKD and INS phenotypes and progression rates.
Lay summary
Lay title: Using machine learning to improve understanding of genetic modifiers of different types of kidney disease.
Background:
Chronic kidney disease (CKD) is a global problem affecting 9% of the population. Up to 50% of CKD patients suffer from CKD of unknown origin and in some cases there is a poor understanding of the underlying disease mechanisms. Nephrotic syndrome is a major cause of kidney disease however it is a clinical presentation that can be due to a range of genetic and non-genetic causes.
Your methodology: what have you done in your study?
We analysed the correlation between genetic variants and known kidney disease types. We tested whether a type of supervised machine learning called “neural networks” could predict the type of kidney disease and rate of progression by using genetic variant data.
Results: What have you discovered?
We found several genetic variants in immune system genes that may be useful for predicting membranous nephropathy and different types of nephrotic syndrome (NS).
What are the potential benefits that this research could bring to patients?
Applying machine learning to genetic data will help to refine the diagnosis of CKD and idiopathic nephrotic syndrome (INS) and improve understanding of their causes. This would help to manage clinical decisions for CKD and INS patients.

Lauren G Russell
Authors and affiliations:
Lauren G Russell1, Jennifer C Chandler1, Maria Kolasti-Joannou1, Daniyal J Jafree1, Christopher J Rowan2, Norman D Rosenblum2, Paul JD Winyard1, David A Long1
1University College London, London, UK
2The Hospital for Sick Children, Toronto, Canada
Background:
Hedgehog (Hh) signalling is critical in kidney development, with genetic alterations of pathway components leading to severe defects in renal formation. However, the localisation and functional role of the Hh pathway activator Indian hedgehog (Ihh) in kidney development is not yet understood.
Methods:
We utilised a cutting-edge 3D imaging technique, combining wholemount fluorescence in situ hybridisation and immunolabelling to visualise Ihh mRNA localisation in the mouse embryonic kidney. To functionally interrogate the role of Ihh in the developing mouse kidney, we utilised both a global Ihh knockout model and an inducible Osr1CreERT2 model to conditionally delete Ihh in the intermediate mesoderm.
Results:
Ihh mRNA was detected in the developing kidney, primarily localised to proximal tubules from embryonic day (E)16.5 through to the first day postnatally. Ihh global knockout kidneys were smaller with reduced total glomerular number at E16.5, indicating renal hypoplasia in these embryos. A similar phenotype was observed with conditional deletion of Ihh in the Osr1+ intermediate mesoderm, leading to significantly decreased kidney/bodyweight ratio at E18.5. Additionally, we identified a significant reduction in the number of SIX2+ nephron progenitor cells, intermediate nephrogenic structures, and decreased total glomerular number in Osr1Cre;IhhloxP/loxP mice. These mice had normal stromal formation and no defects in nephron segmentation.
Conclusion:
Using a novel imaging technique, we identified the onset of Ihh expression in maturing proximal tubules of the mouse embryonic kidney. We observed renal developmental defects in both Ihh total knockout and a conditional Ihh deletion models. Deletion of Ihh results in smaller kidneys with decreased number of developing nephrons, providing evidence of a role for Ihh in the nephron development in the kidney. This gives us insight into molecular pathways potentially involved in the kidneys of patients with renal developmental defects.
Lay summary
Lay title: Investigating the function of the gene ‘Indian hedgehog’ in the development of the kidney.
Background:
Changes in our genes which lead to defects in the kidneys and urinary system are the most common reason for severe childhood kidney problems. Examining the processes involved in kidney development is key to understanding why many children are born with kidney developmental defects. This project involves looking at a molecule called ‘Indian hedgehog’ which we propose is needed for the kidney to develop properly.
Your methodology: what have you done in your study?
Using new 3D imaging techniques, we identified Indian hedgehog in the developing nephron, the kidneys functional unit. We have used two experimental models to investigate the function of this gene in kidney development: mice that completely lack the Indian hedgehog gene and mice without Indian hedgehog gene specifically in the kidney.
Results: What have you discovered?
We have found that kidneys without the gene Indian hedgehog are smaller, have less nephrons, and lower numbers of the cells and structures that form the functional nephron. Therefore, this study indicates an importance for Indian hedgehog in kidney development, specifically in nephron formation.
What are the potential benefits that this research could bring to patients?
Advancing our understanding of kidney development will provide key information on why problems occur in patients with childhood kidney problems and aid research to try and prevent this in the future.

Laura Wilson
Authors and affiliations:
Laura Wilson1, Judith Allen2, Adrian Woolf2, David Long1
1Great Ormond Street Institute of Child Health, University College London.
2The University of Manchester.
Background:
Recent studies show that macrophages modulate progression of polycystic kidney disease (PKD). However, there is no information regarding the role of macrophages in the initiation of cystogenesis. We hypothesised that a specific population of macrophages may be needed to initiate kidney cystogenesis.
Methods:
We characterised macrophage subpopulations in cystic Cpk and Pkd1RC mouse kidneys from the embryonic period. Additionally, we specifically depleted Lyve1+ macrophages by crossing Lyve1-Cre with Csf1r-floxed mice in the context of wild type and Cpk mice.
Results:
Using macrophage-specific markers F4/80 and CD11b, we identified two populations of macrophages (F4/80hiCD11blo and F4/80loCD11bhi) by flow cytometry. CD206, Lyve1, TIMD4 and CCR2 markers were used to further characterise the subpopulations. Renal F4/80hiCD11blo macrophages were CD206+, some expressed Lyve1 and some were TIMD4+. F4/80loCD11bhi macrophages were CCR2+ and negative for the other markers. At embryonic day 18.5, when homozygous Cpk and Pkd1RC mutant mice have dilated tubules, there was a significant increase in the proportion of Lyve1+ macrophages in mutant compared with wild-type kidneys, with no changes in other populations. By studying a detailed time course of Cpk disease progression, we observed that the initial accumulation of Lyve1+ macrophages was followed by an increase in F4/80hiCD11blo macrophages in kidneys as cysts initiate. All macrophage populations were significantly increased in severely cystic kidneys. Depleting Lyve1+ macrophages did not compromise kidney development in non-PKD mutant mice, with no significant change in glomerular numbers in Lyve1-Cre Csf1rfl/fl embryos. There were no differences in PKD severity between Cpk-/- and Lyve1+ macrophage-depleted Cpk-/- mice based on kidney/body weight, renal cystic index and blood urea nitrogen two weeks after birth.
Conclusion:
Lyve1+ macrophages accumulate before PKD cysts initiate during embryonic development. Genetic depletion of Lyve1+ macrophages from this period, however, does not affect the severity of PKD.
Lay summary
Lay title: Investigating the immune system in polycystic kidney disease.
Background:
Polycystic kidney disease (PKD) is a condition people inherit if their parents pass on a faulty PKD gene. Large, fluid-filled sacs (called cysts) form in the kidney that cause damage over time, leading to kidney failure. There is no cure for PKD and more treatments are needed.
Your methodology: what have you done in your study?
We looked at different types of macrophages in kidneys of mice with a faulty PKD gene that develop cystic kidneys similar to the human disease. Macrophages are a type of white blood cell that play an important part in the immune system. We studied a type of macrophage that expresses a molecule called Lyve1 to see if they influence PKD progression.
Results: What have you discovered?
We found the number of Lyve1+ macrophages in kidneys of mice that have a faulty PKD gene was increased compared to normal mouse kidneys. We deleted Lyve1+ macrophages in mice and found this did not affect kidney development or influence PKD severity.
What are the potential benefits that this research could bring to patients?
Previous studies have shown macrophages regulate PKD severity, but it is unknown which types of macrophages are involved. Further studies investigating different types of macrophages could identify the important macrophages, which could provide an alternative therapy for PKD patients in the future.
Abstract poster presentations:

Professor Maria Grazia De Angelis
Authors and affiliations:
Thomas Fabiani1, Maria Grazia De Angelis1, Simone Dimartino1, Cristiana Boi2, Eleonora Ricci3
1University of Edinburgh
2University of Bologna
3 National Center for Scientific Research Demokritos, Athens
Background:
A wearable artificial kidney (WAK) is the holy grail of hemodialysis. WAKs would improve the lifestyle and health of patients, especially those with comorbidities, as dialysis would be performed more gently and even at night, reducing restrictions on patients' lives. Furthermore, it would enable huge water savings in a water-intensive process. The biggest technological obstacle to making a WAK is the purification and recirculation of the dialysate, which is the wastewater containing the molecules removed from the blood. Previous authors have used a biochemical reaction to deplete the more abundant uremic toxins, e.g. urea, but those clinical trials failed because the byproducts formed from the reaction are toxic to patients.
Methods:
Our goal is to eliminate the bottleneck of WAK by synthesizing a material that can safely and completely deplete uremic toxins from dialysate without releasing byproducts. To this end, we performed molecular simulations and experimental tests to screen a large database of materials capable of removing uremic toxins, particularly urea, from water.
Results:
Commercially available materials do not provide the purification capacity needed for a miniaturized system. After examining hundreds of existing and new materials, we realized that some porous materials with ordered nanopores (Covalent Organic Framework) could get good removal of urea from water, if they were heavily loaded with fluorine atoms. Two materials were studied in more detail.
Conclusion:
Our calculations allowed us to sift through a large database of porous materials and pre-select those suitable for WAK. We were also able to understand the physical and chemical mechanism behind their performance.
Lay summary
Lay title: Redefining hemodialysis with materials science and data-driven innovation: towards the wearable artificial kidney
Background:
Our goal is to create a wearable artificial kidney, i.e., a self-supporting dialysis system that can function in the absence of a water supply. Ideally these would allow for more continuous and less invasive treatment for patients, nocturnal dialysis and would be less life-shattering than home dialysis systems, even the most advanced ones.
Your methodology - what have done in your study?
From the study of the previous works carried out and from our engineering expertise, we have identified the main bottleneck in the realization of the WAK in the materials for the purification of water. Therefore, we are focusing on this section of the machine, looking for materials that can safely and permanently capture uremic toxins from the water circulating in the device. We have carried out experimental tests on some commercial materials and calculations on a family of about 500 candidate materials suitable for our process.
Results: What have you discovered?
Commercial materials do not have the ability to remove all uremic toxins. Some new materials have the potential to capture urea from water in large quantities. Their chemical nature appears to play a large part in this, especially their fluorine atom content.
What are the potential benefits that this research could bring to patients?
Patients could perform hemodialysis overnight, travel easily, and perform some light physical activity during treatment. They may be far from a water source. The treatment itself would be much less invasive, require less water and be more accessible to populations with limited access to water or medical facilities.

Thandi Ferris
Authors and affiliations:
Thandi Ferris 1, Katarzyna Szymanska1, Colin A Johnson1
1University of Leeds
Background:
Autosomal recessive polycystic kidney disease (ARPKD) is a rare, severe disorder characterized by polycystic kidneys during the perinatal period. Mutations in the PKHD1 gene, encoding the ciliary-associated protein fibrocystin, are responsible for ARPKD. Several studies have presented compelling evidence that mutations in the PKHD1 gene, resulting in reduced or absent fibrocystin, correlated to dysregulated cilia and the cystic phenotype. However, a definitive pathomechanism establishing a relationship between these factors remains unknown.
Methods:
Here, using an eSpCas9GFP/gRNA system, three (SCTi003-A) human iPSC lines were generated, two carrying compound heterozygous mutations; c.[106_107insC];[106_112delACGTGGA], c.[99_100insA];[98_106delCAGGGGGAA], and one with the heterozygous mutation, c.106_113insTCACTGTG all in exon 2 of PKHD1. These cell lines were characterized and validated for pluripotency cell markers Oct-4 and SSEA-3/4 using immunocytochemistry/immunofluorescence and confocal microscopy.
Mutation analysis was done with Sanger sequencing. The mutational consequences on the PKHD1 gene product, fibrocystin, were analyzed with Western blot. RT-PCR analysis is to be done to further assess the mutation consequences on cDNA. To characterize the impact of these mutations on fibrocystin localization to primary cilia, iPSCs will be stained with antibodies specific for cilia (ARL13B) and fibrocystin (binding to residues 143-235 on the 2nd TIG domain) and immunofluorescence assessed via confocal microscopy. Karyotyping of the cell lines will be completed in due course.
Results:
iPSCs expressed normal immunofluorescence for SSEA-3/4 and Oct-4. Mutation analysis revealed no off-target events, and the targeted mutations were present in each cell line.
Western blot analysis indicated the presence of potential fibrocystin isoforms within the 75- 150kDa range.
Conclusion:
These iPSCs mutant PKHD1 cell lines expressed pluripotency cell markers and therefore markers and, therefore, have the potential to be differentiated into suitable ARPKD kidney organoid models.
Lay summary
Lay title: Modelling ARPKD Compound Heterozygous Mutations in Human Induced Pluripotent Stem Cell Line
Autosomal recessive polycystic kidney disease (ARPKD) is a rare form of polycystic kidney disease that is typically diagnosed during the perinatal period and early childhood. This inherited disease causes cysts to grow in the kidneys. This often leads to kidney failure, where patients require dialysis or kidney transplants since there are no specific drugs for ARPKD.
ARPKD is caused by mutations in the PKHD1 gene encoding for the ciliary-associated protein fibrocystin. Cilia are hair-like structures that protrude from the apical surface of cells and are actively involved in several cellular processes, including kidney organogenesis.
Studies have demonstrated a link between PKHD1 mutations and dysfunctional cilia in animal models that implicates their role in cystic kidney disease. However, the exact mechanism causing cyst formation is poorly understood. Although animal models have provided substantial insights into ARPKD disease mechanisms, they fail to effectively recapitulate human phenotypes.
This study characterizes and validates human induced pluripotent stem cells (iPSCs) with induced compound mutations comparable to ARPKD mutations. These cell lines can potentially be differentiated into 3D kidney organoid ARPKD models to further investigate the molecular mechanisms of cyst formation and develop therapeutic strategies to abrogate the cystic kidney phenotype.

Magdalena Karwatka
Authors and affiliations:
Magdalena Karwatka1, Joe Cockburn1, Steve Evans1, John Ladbury1, Colin A Johnson1, Michelle Peckham1; Rowan Taylor2
1University of Leeds
2Human Centric Drug Discovery, Oxford
Background:
Primary cilia are microtubule-based organelles protruding from the surface of most mammalian cells. They are important signaling hubs with essential roles in vertebrate development. Defects in cilia cause ciliopathies that range from non-syndromic inherited retinal diseases to severe multi-organ disorders, invariably presenting with renal cystic dysplasia.
CEP290 (Centrosomal protein of 290kDa) is a key regulator of ciliary structure and ciliary vesicular trafficking. CEP290 mutations are a major cause of ciliopathies. Nearly half of CEP290 coding exons are in-frame “skippable” exons that can be spliced from the transcript without altering the reading frame. This can remove the frame-disrupting segment of mRNA, restoring the reading frame and producing a truncated protein that retains some or all function.
Methods:
Using CRISPR-Cas9, we knocked-in frame-disrupting pathogenic mutations in exon 7, 25 and 36 of CEP290 in induced pluripotent stem cells (iPSC). Kidney organoids derived from iPSCs will be imaged and assessed for key cellular phenotypes. We will test splice-switching antisense oligonucleotides (ssAONs) targeting exons 7, 25 and 36 for exon-skipping efficacy in cells by qRT-PCR and western blotting. The ssAONs will then be encapsulated in liposomes attached to microbubbles to assess deliverability and accessibility in organoids following bubble rupture using ultrasound.
Results:
We have generated CRISPR-edited iPSC lines with mutations in exon 36 of CEP290 and have used these to generate organoid models of CEP290-related disease. We show that exon 36 is skipped in kidney but not retinal organoids, exon-skipping does not affect primary ciliogenesis, but a frameshift mutant markedly reduces cilia size in iPSCs.
Conclusion:
We have developed models and a workflow for assessing exon-skipping as a potential therapy for CEP290-related forms of renal cystic dysplasia. We will use ssAONs in kidney cells to trigger skipping of exons carrying frame-disrupting mutations and assess the suitability of microbubbles as a carrier for targeted ssAON delivery.
Lay summary
Lay title: Early testing of oligonucleotide therapies as potential treatments for cystic kidney diseases
Background:
Cilia are antennae-like structures on cells that receive and transduce signals from their surroundings during human development. Defects in cilia cause a range of human developmental disorders called “ciliopathies”. CEP290 (Centrosomal Protein 290kDa) is an essential molecule in the cilium, and mutations are the most frequent cause of ciliopathies, invariably associated with cystic kidney disease. This project aims to identify and develop therapeutics for patients with these “untreatable” conditions. CEP290 contains many “in-frame” exons. If a mutation occurs within an in-frame exon, cellular processes (“splicing”) can remove this segment (“exon-skipping”), avoiding the detrimental effects of the mutation.
Your methodology - what have done in your study?
We have edited different types of CEP290 mutations into induced pluripotent stem cells (iPSCs) from which we can grow 3D human cell models (“organoids”) of kidney disease. We will assess if new therapeutic agents called antisense oligonucleotides will force cellular processes to perform exon-skipping on an exon containing a mutation, which has the potential to relieve disease symptoms.
Results: What have you discovered?
We have shown that exon-skipping occurs for only some types of CEP290 mutations in kidney but not retina cells, providing the basis for development of new disease models.
What are the potential benefits that this research could bring to patients?
Using antisense splice-switching oligonucleotides to induce exon-skipping is a potential pre-clinical therapeutic approach that targets CEP290 mutations causing renal ciliopathies.

Cheuk Yan Man
Authors and affiliations:
Cheuk Yan Man1, Hortensja Ł Brzóska2, Owen Williams2, Michael G Robson3, Jonathan Elliott1, David A Long2, Elisavet Vasilopoulou1
1Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
2Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
3 MRC Centre for Transplantation, King’s College London, London
Background:
Glomerular disease involves both injury of glomerular cells and inflammatory responses that drive disease progression. Thymosin β4 (Tβ4) is a peptide that is expressed in podocytes and macrophages in the mouse kidney and has a protective role in glomerular disease. Tβ4-knockout mice develop exacerbated glomerular disease compared to wild-type controls, with impaired renal function, reduced podocyte number and increased macrophage accumulation. However, the precise contribution of leukocyte and resident kidney cell-produced Tβ4 to glomerular disease progression is unknown. We aimed to answer this question in our study.
Methods:
Transplantation of bone marrow cells from wild-type (WT) or Tβ4-knockout (KO) mice into irradiated recipient mice was carried out to generate four groups of chimeric mice; WT mice with WT bone marrow (WT/WT, n=9), WT mice with KO bone marrow (WT/KO, n=6), KO mice with WT bone marrow (KO/WT, n=6) and KO mice with KO bone marrow (KO/KO, n=7). Glomerulonephritis was induced ten weeks later by administration of nephrotoxic serum (NTS). Bone marrow reconstitution (flow cytometry), macrophage accumulation, podocyte density (immunohistochemistry) and albuminuria (ELISA) were assessed 21 days post-NTS.
Results:
Bone marrow transplantation was efficient with >75% of circulating cells being donor-derived. Macrophage accumulation in the periglomerular area was significantly lower in WT/WT mice (10.18±0.34 macrophages/glomerulus) compared with WT/KO (13.73±0.49, p<0.0001), KO/WT (15.24±0.46, p<0.0001), and KO/KO mice (18.26±0.47, p<0.0001). Podocyte density was highest in WT/WT mice (6.64±0.18 podocytes/100μm2) compared with WT/KO (5.54±0.19, p<0.0001), KO/WT (5.98±0.22, p=0.0083), and KO/KO mice (4.94±0.14, p<0.0001). Albuminuria was significantly lower in WT/WT (9,947±3,403 μg/24hours) compared with KO/KO mice (29,177±4,838, p=0.0070), but there were no statistical differences with WT/KO (13,468±3,056) and KO/WT (14,635±3,729) mice.
Conclusion:
Our findings show that Tβ4 expression by both bone marrow-derived and kidney-resident cells has a protective role in glomerular disease.
Lay summary
Lay title: Investigating the protective effect of thymosin β4 in chronic kidney disease
Background:
One of the leading causes of end-stage kidney disease is damage to the glomeruli, the components of the kidney where the blood is filtered to make urine. This damage is caused by disruption of the structure of the glomeruli and by the build-up of white blood cells, which causes inflammation. Thymosin β4 is a naturally occurring protein that can reduce inflammation and the severity of glomerular disease in mice. The aim of this study is to understand how thymosin β4 exerts its protective effect in the kidney.
Your methodology - what have done in your study?
Mice with selective deletion of thymosin β4 either in circulating white blood cells, or in the kidney, were generated followed by the initiation of glomerular disease.
Results: What have you discovered?
The results show that lack of thymosin β4 in either white blood cells or kidney cells, exacerbates glomerular damage and inflammation. Loss of thymosin β4 in both white blood cells and kidney cells had a cumulative effect.
What are the potential benefits that this research could bring to patients?
These results show that thymosin β4 regulates both white blood cells and kidney cells to limit the effects of kidney injury. This work provides the rationale to harness this protective pathway to develop new therapies to slow the progression of glomerular disease.

Katarzyna Szymanska
Authors and affiliations:
Katarzyna Szymanska1, Claire Smith1, Alice Lake1, Andrew Streets2, Albert Ong2, Colin A Johnson1
1Leeds Institute of Medical Research, University of Leeds
2 University of Sheffield Medical School
Background:
Polycystic kidney disease (PKD) and other syndromic forms of cystic kidney disease (CyKD) are a major cause of childhood morbidity and mortality in the UK, with approximately 5,000 patients in the UK needing either dialysis or transplantation every year due to kidney failure. Tolvaptan is a drug that slows the decline in renal function for autosomal dominant polycystic kidney disease (ADPKD). However, common (11 to 13%) side-effects of Tolvaptan mean that many patients do not tolerate treatment. In this work we aim to model a series of CyKD patient mutations (from severe syndromic ciliopathies such as Meckel-Gruber syndrome to non-syndromic PKD) in kidney organoids (KiO) and treat cystic phenotypes with ROCK inhibitors (fasudil and hydroxyfasudil), recently identified to rescue normal ciliogenesis in cell-based models and diminish cystogenesis in mouse model.
Methods:
Patient mutations were introduced in MKS1, PKD2, BICC1 and PKHD1 in induced pluripotent stem cells (iPSCs) line SCTi003-A using CRISPR-Cas9 genetic engineering. Mini KiO, grown in non-adherent 96-well plates, were optimised for drug treatment to induce cellular cystic phenotypes using forskolin, blebbistatin and 8Br-cAMP.
Results:
SCTi003-A iPSCs were successfully differentiated into KiO with nephron structures (expressing markers NPHS1 for podocytes, LTL for proximal tubules, and CDH1 for distal tubules) following an adaptation of existing protocols. We are currently optimising a protocol that differentiates KiO with both nephron and collecting duct structures. We have successfully developed iPSC lines with biallelic MKS1, PKD2, BICC1 and PKHD1 mutations. These will be characterised, differentiated into KiO and assessed for rescue of cellular cystic phenotypes by treatment with ROCK inhibitors.
Conclusion:
We have developed pre-clinical iPSC-derived models of CyKD with a series of patient mutations for use in potential pre-clinical testing of ROCK inhibitors for this disease indication.
Lay summary
Lay title: New treatments for cystic kidney disease.
Tolvaptan is a drug that slows the decline in renal function for autosomal dominant polycystic kidney disease (ADPKD). However, common (11 to 13%) side-effects of Tolvaptan mean that many patients do not tolerate treatment. Furthermore, there are no preventative treatments or new therapeutic interventions that may modify disease progression of patients with ADPKD or other types of cystic kidney disease. This has created an acute clinical need for the potential repurposing of existing drugs or the development of novel lead compounds that could treat cystic kidney disease. We identified and validated two approved drugs (fasudil and hydroxyfasudil) and now want to test these in cell-based disease models of kidney cysts. We will use kidney organoids (mini kidneys grown in a dish) that we can artificially produce from special “induced pluripotent stem cells” (iPSCs). We will model renal tubule formation and cyst formation in response to drug treatment in a physiologically relevant tissue, an essential step in drug efficacy testing. The overall aim is to re-purpose and translate a known drug into Phase II clinical trials in less than 5 years, significantly reducing the time and investment required to make the move from laboratory to the clinic.

Mannie Sher
Authors and affiliations:
Mannie Sher1, Francis Clay1
1The Tavistock Institute of Human Relations
Background:
There is a relatively low take-up of home dialysis in the UK. This project will investigate the psycho-social, socio-technical and socio-ecological dynamics (the emotional, behavioural, technical and environmental properties) of home dialysis of patients with chronic kidney disease (CKD), their families, carers and hospital renal staff.
Methods:
The method of investigation will focus on how patients relate to and feel about their illness, their patient role, and the impact of these on their identities. The method will consist of an investigation into personal, conscious, and unconscious factors and environmental contextual factors in determining individual and systemic behaviours in relation to CKD and treatments; it will seek to understand various influences in decision-making and patient resistance to facing and dealing with their illness and treatment.
In-depth interviews of up to 20 CKD patients will allow for the collection of rich, qualitative data. The patient, treatment, and context (environment, culture, educational, social and economic circumstances) will lie at the heart of this project.
The core research activity will comprise individual and group interviews. Attitudes and beliefs towards CKD and its treatment will be examined in-depth in patients and families, renal ward staff and policy makers. The questions will aim to answer practical questions like diet, the onset of the disease, symptoms, attitudes concerning health-promoting and health-diminishing behaviours. Patient life-cycle histories will be investigated, with reference to early childhood relationships and milestone events, cultural food habits, education and work histories, sport, accidents, inter-personal relationships, personality and identity-formation, sexual development, behaviour and relationships. Patients and their families will be asked about their in-the-moment experiences of dialysis and their expectations of home dialysis.
Results:
The resulting material will be subject to rigorous content analysis to produce trends and detailed pictures of patient attitudes and experiences of hospital and home dialysis.
Conclusion:
The research project is in the application-for-funding stage; therefore, no conclusions are available at present.
Lay summary
Lay title: An investigation into the emotions (feelings), attitudes and behaviours towards home dialysis of CKD patients and their families, hospital renal staff, policy makers. A primary aim of the research is to increase the take-up of home dialysis.
Background:
The take up of home dialysis is about 1% of patients with kidney disease. This is a startling figure, given that home dialysis offers patients improved health, increased chances of survival, greater longevity, greater freedom and flexibility of lifestyle and treatment frequencies. For the NHS too, there are advantages of home dialysis since hospital dialysis costs over 2.5 times more than home dialysis and produces a healthier population. Some obstacles to having dialysis at home are practical, e.g. lack of space, ageing and frailty, poor hand-eye coordination, clinically unsuitable; others are psychological and emotional, e.g. fear of needling oneself, fear of things going wrong. Health inequalities in society also factors regarding home dialysis, e.g. poverty, poor education levels, the rural/urban divide, disadvantaged families, immigrant families, transient populations.
Your methodology - what have you done in your study?
The methodology of this study will involve new approaches to investigating decisions about home dialysis.
Results: What have you discovered?
The research project is at the application for funding stage; therefore, no results are available at present.
What are the potential benefits that this research could bring to patients?
Home dialysis offers patients better health outcomes, increased chances of survival, greater longevity, greater freedom and flexibility of lifestyle and treatment frequencies.
Authors and affiliations:
Praveen D Sudhindar1, Elisa Molinari1, Seamus M McLafferty1, Colin A Johnson3, Colin Miles1, John A Sayer1
1Newcastle University
3University of Leeds
Renal ciliopathies are a heterogeneous class of disorders caused by dysfunctions of the primary cilia. They are often multisystem disorders characterized by extensive genetic heterogeneity and clinical variability with high levels of lethality and there is marked phenotypic overlap among distinct ciliopathy syndromes. Nephronophthisis (NPHP) is a typical renal ciliopathy phenotype that causes kidney failure often within early childhood, for which there are no curative treatments beyond dialysis and transplantation.
To identify novel therapeutics for NPHP, we designed a high-throughput ciliary phenotype-driven screening strategy to interrogate the Tocris® library of 1120 biologically active compounds, making use of the Operetta high-content imaging system with Harmony/Columbus software. Initially, 33 compounds were identified that restored ciliary phenotype in renal epithelial cells derived from Cep290 mutant mice. These compounds were subjected to a secondary screen using NPHP patient fibroblasts (P-BB), carrying compound heterozygous CEP290 mutations, including an allele that we had previously shown to be amenable to Anti-sense Oligonucleotide (ASO) mediated exon skipping. In this screen, 12 compounds either restored ciliogenesis or corrected cilia length defects.
A tertiary phenotypic screen of the 12 TOCRIS compounds was then carried out in human urine-derived renal epithelial cells (hURECs) from a NPHP patient (P-HB) carrying CEP290 mutation (p. (Thr832Asnfs*12) and p.(Gly1890*)) along with control hURECs from an unaffected sibling. The initial hUREC phenotypic screen of P-HB cells displayed a ciliogenesis defect with elongated cilia typically found in CEP290 patients, compared to the unaffected sibling who had normal cilia. Two of the Tocris® chemical compounds rescued the ciliary phenotype in patient (P-HB). We will employ RNAseq to identify the underlying molecular pathways using the Tocris® compounds which may reveal novel insights into the mechanisms underlying NPHP secondary to CEP290 mutations and explore this in additional genetic causes.
Lay summary
Lay title: Small molecule drug treatments for inherited kidney disease: Nephronophthisis
Background:
Cilia are slender hair like projections that extend from almost all types of eukaryotic cells. They are involved in cellular signalling and sensory roles which are important for maintaining the local cellular environment. Dysfunction of cilia leads to number of human diseases which are termed as ‘ciliopathies’. Nephronophthisis (NPHP) is a kidney ciliopathy, which is a rare genetic disorder with an estimated occurrence of 1 in 922,000 in the US, that causes kidney failure often within early childhood, for which there are no curative treatments beyond dialysis and transplantation.
Your methodology - what have done in your study?
To identify novel therapeutics for NPHP we employed a screening strategy to interrogate a selection of biologically active chemical compounds using high resolution microscopic imaging. Primary, secondary, and tertiary screening were conducted in kidney epithelial cells derived from mouse models, patient fibroblast cells and patient kidney epithelial cells respectively.
Results: What have you discovered?
The patient kidney epithelial cells had longer cilia and dramatically reduced ciliation rates compared to a healthy sibling. Treatment with selected compounds either restored the ciliogenesis or corrected the cilia length defects.
What are the potential benefits that this research could bring to patients?
We will employ deep sequencing methodologies in the future to identify molecular mechanisms underlying NPHP. This study will provide valuable insights for future treatment of NPHP in affected patients.

Amarpreet Thind
Authors and affiliations:
Dr Amarpreet K Thind*1,2, Dr Michelle Willicombe1,2 and Professor Edwina A Brown1,2 on behalf of the KTOP Study Investigator Group3-6.
1Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London,
2Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust,
3Central London Community Healthcare NHS Trust,
4Department of Surgery and Cancer, Imperial College London,
5Institute of Health and Social Care, London South Bank University,
6The Centre for Health Services and Clinical Research, The University of Hertfordshire.
Background:
Older people with kidney failure are vulnerable to developing frailty, which affects all aspects of kidney transplantation (KT). To better inform, guide, and support older people considering KT a holistic understanding of experiences is required.
Methods:
The Kidney Transplantation in Older People (KTOP) study recruited KT candidates aged >60, assessed frailty (Edmonton Frail Scale), quality of life (QoL) (Short Form-12 V2), and clinical outcomes on the waitlist (WL) and post-KT from October 2019-June 2023. It was powered for QoL differences. Comparative and mixed-effect analysis determined variations by frailty.
Results:
210 patients were recruited, with 118 transplanted. At recruitment 63.4% (118) were not frail, 19.4% (36) were vulnerable, and 17.2% (32) were frail. A trend towards poorer WL and KT outcomes in the vulnerable/frail was observed. Vulnerable/frail WL candidates were more likely to experience infection events (60% vulnerable/frail c.f. 24.4% not frail, p=0.001) and spend longer suspended (mean 434 days vulnerable/frail c.f. 307 not frail, p=0.025). Vulnerable/frail KT recipients were more likely to be hospitalised (73% vulnerable/frail c.f. 52% not frail, p=0.028), have longer admissions (mean 36.6 days vulnerable/frail c.f. 24.8 not frail, p=0.024), with a trend towards higher graft loss (29.4% vulnerable/frail c.f. 9.9% not frail, p=0.059) and mortality (18.2% vulnerable/frail c.f. 7% not frail, p=0.078).
WL physical component scores (PCS) remained stable in not frail candidates but declined in vulnerable/frail. Post-KT, not frail recipients PCS declined initially and then recovered, whilst PCS stabilised in vulnerable/frail. WL mental component scores (MCS) improved in both groups. Post-KT MCS initially worsened and then improved in not frail recipients but declined in the vulnerable/frail.
Conclusion:
WL and KT clinical outcomes were poorer in vulnerable/frail older people, however QoL experiences were mixed. KT did not drastically change QoL in either group. Assessing frailty is crucial to older peoples’ care, enabling tailored understanding, risk assessment, counselling, and interventions to be implemented.
Lay summary
Lay title: Waitlist and Kidney Transplant Experiences in Older People Living with Frailty
The number of older people waiting for and receiving transplants is increasing. Older people with kidney failure are at risk of frailty, which can affect their ability to cope and manage after transplantation. The Kidney Transplantation in Older People study has used questionnaires to measure frailty and quality of life in older people on the waitlist and after a transplant. Medical events that occurred were also counted to see how these varied with different levels of frailty.
210 older people have been included in the study, 118 of whom have been transplanted. Vulnerable/frail older people had worse medical events both on the waitlist (major infection events and longer suspended from the waitlist) and after a transplant (more and longer hospital admissions). In the vulnerable/frail older people physical quality of life stabilised after a transplant but mental quality of life worsened. Across all older people however, transplantation did not lead to drastic changes in quality of life. This research provides more detailed information on how experiences vary in older people and why these variations exist (frailty). Consequently, more accurate, realistic information can be given to older people and now we can find specific ways to support older people during this time.

Priscilla Smith
Authors and affiliations:
P Smith1, K Dalrymple1, K Clarke1, Y Wang1, T Harris2, A Webb1, L Chappell1, K Bramham1
1King’s College London
2PKD Charity
Background: 46% of women with moderate-severe chronic kidney disease (CKD) will require dialysis or lose at least 25% of kidney function within six months of delivery with no development of preventative treatments.
Methods: Biological samples, longitudinal and outcome data will be collected in a prospective cohort and eligible women identified to participate in ORCHARD-BEET randomised controlled feasibility trial. Inclusion criteria: singleton pregnancies;<25 wks gestation; CKD (pre-pregnancy eGFR<90mls/min/m2 or pregnancy Cr>70µmol/l). Randomisation:1 to 1. Standard care or daily beetroot juice supplement (nitrate 400mg). Primary outcome: recruitment rate; secondary outcomes: tolerability, acceptability, eGFR change 6 months postpartum.
Results: 118 pregnant persons consented to participate to the cohort and 104 were randomised for ORCHARD-BEET trial. Cohort maternal baseline characteristics are presented in Table 1.
Conclusion: To our knowledge this is the largest prospective cohort study with embedded pragmatic feasibility trial with concurrent biobanking of pregnant participants with CKD (close April 2024), which is representative of women with moderate and severe CKD. Findings will be used to inform future intervention trials to prevent pregnancy-associated progression of kidney disease.
Lay summary
Lay title: ORCHARD study of kidney disease in pregnancy
Background
Kidney disease affects 3% of women of childbearing age. Many women will lose kidney function or need to start dialysis during or after pregnancy. We have no treatments to prevent this and there have been no improvements in the last 20 years.
We hope to find out whether drinking a small amount of beetroot juice each day can help to protect kidney function in pregnancy. Beetroot juice contains ‘dietary nitrate’, which has been shown in non-pregnant and pregnant individuals to lower blood pressure and improve blood flow by relaxing the blood vessels.
Your methodology - what have done in your study?
Women with kidney disease pre-pregnancy were asked to participate if under 25 weeks pregnant and not on dialysis. If randomly allocated to intervention arm, they were asked to drink a 70ml Beetroot Juice concentrate “shot” daily along with usual clinical care. They were seen up to 3 times during pregnancy and twice post-partum; biological samples were collected, blood pressure checked and those taking the beet shot were asked about any missed doses or problems with the drink.
Results: What have you discovered?
We have been able to recruit women with moderate to severe CKD to an interventional trial during pregnancy and meet our recruitment target overall (See Table 1). We will report full results once all follow-ups have been completed.
What are the potential benefits that this research could bring to patients?
Potential therapy to prevent worsening of kidney function during or following pregnancy.

Maryline Fresquet
Authors and affiliations:
Maryline Fresquet1-Patrick Hamilton1, Bernard Davenport1, Emily Williams1, Alex Mironov1, Wayne Lim1, Ronan o’cualain1, Craig Lawless1, Alex Nyström2, Paul Brenchley3, Rachel Lennon1
1Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health Sciences, School of Biological Sciences, University of Manchester, Manchester, UK.
2Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.
3Central Manchester University Hospitals NHS Foundation Trust and Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK.
Scientific summary
Background:
Membranous nephropathy (MN) is an autoimmune disease affecting the kidney. 80% of MN patients have autoantibodies to the phospholipase A2 receptor found on podocytes and IgG-PLA2R immune complexes deposit in the glomerular basement membrane (GBM). We currently have limited understanding about the consequences of the antibody deposition in the filter. In this study, we defined the ultrastructural and compositional changes of the disorganised GBM in patients with MN.
Methods:
A MN human biopsy was stained and analysed by volume electron microscopy. The acquired images were used to model segment containing the immune deposits in the GBM using the IMOD image processing software.
Glomerular sections from 5 MN patient biopsies and 5 control samples were collected. ~75 glomeruli/patient were isolated by laser-microdissection and prepared for proteomic analysis.
Results:
The 3D model of the glomerulus highlighted novel ultrastructural features of the disorganised filtration barrier in MN patients. We observed a thickened appearance of the overall GBM due to the presence of large immune complex deposits, formation of spikes between the deposits and visualised vesicles in the immune complex. We found adhesions sites formed between the podocyte foot process and the basement membrane, possibly to resist mechanical force caused by the deposits.
Proteomic analysis identified enriched pathways; regulation of immune response and complement activation, hallmarks of MN pathology. Additional analysis showed an increased level of proteins in the MN glomeruli, namely PLA2R, COL7A1, COL12A1, FBLN5 and EFEMP1, all basement membrane proteins. These proteins are involved in collagen fibril organisation, extracellular matrix assembly and dysregulation of the GBM and remodelling. We validated these proteins using immunofluorescence and found PLA2R, COL7A1 and COL12A1 specifically enriched within the MN patient glomeruli.
Conclusion:
This study improves our understanding about the pathobiology associated with GBM defects. These findings have now the potential to identify new therapies that target GBM repair.
Lay summary
Lay title: Investigating the basement membrane in the autoimmune kidney disease, membranous nephropathy.
Background:
Membranous nephropathy (MN) is an auto-immune disease that affects the filters (glomeruli) of the kidney and can cause protein in the urine. Patients have circulating antibodies in their blood which accumulate in the filter, then become inflamed. We currently have limited understanding about the consequences of the antibody deposition in the filter.
Your methodology - what have done in your study?
In this study we sought to define the changes in structure and composition of healthy and diseased glomeruli. We used MN patient kidney biopsies and analysed them using electron microscopy and mass spectrometry.
Results: What have you discovered?
We described novel ultrastructural features of the glomerular filtration barrier, which appeared very abnormal in MN, thickened, with immune complex deposits. We found key proteins present in patient samples potentially involved in the dysregulation of the filter.
What are the potential benefits that this research could bring to patients?
Improving the understanding of the disease mechanism will help in finding treatments targeting the removal of autoantibodies from the kidney.

Richard Naylor
Authors and affiliations:
Richard W. Naylor1, Rachel Lennon1
1University of Manchester
Scientific summary
Background:
>90% of cases of Autosomal Dominant Polycystic Kidney Disease (ADPKD) are caused by mutations in the PKD1 and PKD2 genes, which encode for polycystin-1 (PC1) and PC2, respectively. The polycystins form an ion channel that is important for proper ciliary function. Much effort has been made to investigate the molecular mechanisms driving pathology in ADPKD. However, a detailed analysis of the proteome, using up-to-date cutting edge mass spectrometers, in ADPKD has not been performed.
Methods:
Using the latest mass spectrometry approaches, proteomics analysis was performed on kidneys taken from P28 wild-type and Pkd1nl/nl (a Pkd1 hypomorphic mouse model of ADPKD). 4 biological replicates were assessed, and data processing was performed using MaxQuant and msqrob2 differential analysis.
Results:
Differential protein intensity analysis showed a number of previously identified markers of PKD are up-regulated in the Pkd1nl/nl model. This includes Lcn2, Acta2, multiple claudins, markers of leukocytes (such as Itgam), downstream effectors of the Yap/Taz Hippo pathway, and multiple markers of fibrosis (TgfB, Col1a1, Postn). Novel proteins were also detected that are likely to be important in pathogenesis, including multiple RhoGEFs, Thy1, Grb2 and CD44. We have a particular interest in cell-matrix interactions and how these underpin disease. Of note, we find multiple integrins are more highly expressed in ADPKD, with integrin alpha1, alpha3, beta1 and beta5 particularly up-regulated. Given loss of integrin beta1 is known to preclude cystogenesis, we are currently investigating further the roles of integrin alpha subunits in ADPKD, with the specific aim to generate therapeutics against these more desirable targets.
Conclusion:
Improved proteomics analyses have enabled greater understanding of ADPKD kidneys and highlights the power of the modern and cutting-edge mass spectrometers available to researchers. We have found multiple molecules that are likely to be important in disease progression in ADPKD and are developing ways to disrupt the function of these proteins with the aim of slowing down disease progression.
Lay summary
Lay title: Understanding the changes in protein content of PKD kidneys highlights new insights into the disease
Background:
Many researchers have looked at how kidneys change in terms of their mRNA transcripts. However, mRNA is not the functional readout of the tissue. In all cells, mRNA is converted into protein, which performs a specific function in the cell. How quickly the protein is degraded or altered (such as by interactions with other proteins or by being modified) is more important with respect to how it is able to change cell behavior. What this ultimately means is that the level of mRNA does not always correspond to the level/activity of the protein. Given this, we have directly analyzed all the proteins in the kidneys of a mouse model of ADPKD and compared them to normal kidneys to get a more refined understanding of how the kidney changes in ADPKD.
Your methodology - what have done in your study?
We used a machine that detects proteins, called a mass spectrometer, and compared non-ADPKD to ADPKD kidneys to see which proteins are changed in terms of their amounts.
Results: What have you discovered?
We have discovered a number of proteins that are likely to be important in the progression of ADPKD and are currently developing methods to stop these changes in order to develop new therapies.
What are the potential benefits that this research could bring to patients?
The main aim of this research is to discover new targets that I will use to apply for further funding to develop as potential novel therapies. This includes commercialization of new technologies targeting candidates in order to more speedily impact patients in a clinical setting.
Authors and affiliations:
Ivan Hartling1, Sarah Fawaz2, Rebecca Vaughan2,3, Ioannis Michelakis2,4, Rutger Ploeg2,3, Philip Charles1, Cecilia Lindgren1, and Maria Kaisar2,3
1 Big Data Institute, Nuffield Department of Medicine, University of Oxford, United Kingdom
2 Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
3 Research and Development, NHS Blood and Transplant, Bristol, United Kingdom
4 Onassis Cardiac Surgery Center, Athens, Greece
Background:
Kidney transplantation is a life-saving treatment for end stage kidney disease, but every year hundreds of patients with kidney failure die while waiting for a transplant. Donor organ acceptance criteria have been expanded in an effort to decrease organ shortage; however, current acceptance criteria depend heavily on immune matching and donor age and lack granularity. This study aimed to identify donor circulating protein signatures to improve granularity of assessing donor kidneys, predicting kidney transplant outcomes.
Methods:
We analysed deceased donor plasma samples that were linked to complete donor and recipient metadata obtained from the QUOD biobank. A selection of 49 analytes were measured at three timepoints in the plasma of 132 brain death donors (DBD) and at two timepoints in the plasma of 119 circulatory death (DCD) donors. These measurements, along with donor age, height, and sex, were used to construct separate DBD and DCD 10-fold cross-validated lasso regression models using 12-m recipient averaged eGFR as outcome end point. Models were compared using root mean squared error (RMSE) which is a measure of prediction accuracy.
Results:
Lasso regression identified unique protein signatures in both DBD and DCD donors consisting of 20 and 22 proteins respectively with 8 proteins common to both models. The DBD model achieved a RMSE of 18.7 mL/min/1.73 m2 while the DCD model RMSE was 19.0 mL/min/1.73 m2. Both models performed considerably better than models containing clinical variables alone which had RMSEs of 21.7 mL/min/1.73 m2 and 22.7 mL/min/1.73 m2 for DBD and DCD respectively.
Conclusion:
This study identified protein signatures in DBD and DCD kidney donor plasma that could improve prediction of post-transplant outcome compared to using clinical variables alone. Identified protein signatures will be validated in a follow-up study on 1000 donors to develop a predictive score which could be used to improve transplant outcome prediction.
Lay summary
Lay title: Improving assessment of donated kidneys
Background:
Kidney transplantation is a life-saving treatment of end stage kidney disease, but every year hundreds of patients die waiting for a transplantation. To save more lives, transplant clinicians accept kidneys from higher risk donors; however, these organs may have poor function following transplantation. To improve assessment of donor organs, this study used blood markers and machine learning techniques to improve methods for selecting donor organs that will function well in the recipients.
Your methodology - what have done in your study?
We analysed blood samples collected during donor management to identify markers that can predict how well grafts will function after transplantation. We combined blood markers with clinical variables and used machine learning models to select the most useful markers and variables. We then determined how much improvement was provided by the blood markers.
Results: What have you discovered?
We identified unique markers that can improve the selection of donor kidneys. We found that using these markers along with donor age, height, and sex had better accuracy than using age, height, and sex alone.
What are the potential benefits that this research could bring to patients, whether that be in the near or distant future ?
These markers will now be used in a large cohort study to develop a predictive score to help clinicians to better select organs for transplantation. This will help to both increase the number of donor organs and improve the outcomes of kidney transplantation.
Authors and affiliations:
Griffiths, JD1; Streets, AJ1; Johnson, CA2; Smythe, E3; Ong, ACM1
1 Kidney Genetics Group, Academic Unit of Nephrology, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
2 The School of Medicine, University of Leeds, Leeds LS2 9JT, UK
3 Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK.
Background:
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic kidney disease in man and a leading cause of end-stage renal failure. In the majority of cases, germline mutations in PKD1 or PKD2, encoding Polycystin-1 (PC1) and Polycystin-2 (PC2) are causative. Whilst the mechanisms underlying cyst development remain unclear, recent work demonstrates abnormal actin organisation modulated by Rho GTPases. We have identified the RhoGAP protein, SH3BP1, as a novel interactor of PC1 through a mammalian-2-hybrid screen. SH3BP1 is a RhoGAP with selectivity for the Rho-GTPases Cdc42 and Rac1, converting them to their inactive GDP-bound states. SH3BP1 can modulate cell-cell junction formation, directional cell migration and endocytosis. In ADPKD cyst development, a number of these processes are known to be defective, suggesting that a functional SH3BP1-PC1 interaction could be a major regulator of actin cytoskeletal dynamics.
Methods:
Analysis of protein-protein interactions was performed using immunoprecipitation techniques and recombinant tagged protein expression. Renal epithelial mutant cell lines were generated using the CRISPR-CAS9 system. Endocytosis was evaluated through incubation in 3kDa Fluorescein-conjugated dextran before analysis via fluorescence-activated cell sorting (FACS).
Results:
An interaction between SH3BP1 and the C terminus of PC1 has been confirmed in PC1 and SH3BP1. Knockout SH3BP1 renal epithelial cell lines have been generated and are currently undergoing phenotypic analysis alongside PKD1 knockout cells and wild type controls.
Conclusion:
This work identifies a novel interaction between PC1 and SH3BP1 which may have a functional relevance in cyst development. The potential link between SH3BP1 and cyst development will be explored through analysis of SH3BP1 and PKD1 mutant cell lines.
Lay summary
Lay title: Investigating how cell structure and behavior might contribute to cyst growth in autosomal dominant polycystic kidney disease
Background:
Autosomal dominant polycystic kidney disease (ADPKD) is the most common kidney disease which is passed down through families. Patients with ADPKD develop fluid-filled cysts in their kidneys leading to kidney failure in some patients. ADPKD is usually caused by a fault in the proteins Polycystin-1 (PC1) or Polycystin-2 (PC2). We do not know why problems with these genes cause cysts, but recent work shows that PC1 can affect the internal scaffolding of cells causing changes in their shape and behaviour.
Your methodology - what have done in your study?
We have used kidney cells in the lab to look at how PC1 interacts with other proteins that might affect cyst growth. We have also been able to look at how PKD cells take in material from its surrounding environment.
Results: What have you discovered?
We have discovered that PC1 can interact with another protein, SH3BP1, which also regulates how cells behave. By making kidney cells without SH3BP1 we can explore if it affects cyst growth. We have also seen changes in how PKD cells take up material from around the cell which may relate to how cysts develop.
What are the potential benefits that this research could bring to patients?
By exploring these processes further, we hope to better understand how cysts develop and identify new drug targets that can be used for future treatments.
Authors and affiliations:
John P Stone1,2, William R Cowey1,2, Corban JT Bowers1,2, Amy F Stewart1,2, Erin R Armstrong1,2, Marc Clancy5 Timothy R Entwistle1,2, Kavit Amin1,2,3,4 and James E Fildes
1The Ex-Vivo Research Centre, Alderley Park, Macclesfield, UK
2Pebble Biotechnology Laboratories, Building 3, Alderley Park, Macclesfield, UK
3 Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.
4 Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
5Department of Renal Transplantation, NHS Greater Glasgow and Clyde-Queen Elizabeth University Hospital, Glasgow, UK.
6The Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom.
Background:
Normothermic machine perfusion (NMP) offers a superior alternative to existing hypothermic preservation strategies but is currently limited to 1-3 hours. Extending the time a kidney can be sustained using this technology could electivise transplantation, and enable physiological assessments of renal function. We aimed to develop a protocol that allows the safe preservation of donor kidneys for 12 hours using this technique.
Methods:
Porcine kidneys (n=20) were retrieved and flushed with 1L preservation solution before being stored on ice. Following a cold ischaemic time of 3.5 hours, kidneys were placed onto a NMP circuit and perfused for 12 hours. Renal haemodynamics, biochemistry and urine output were recorded and analysed. At the end of perfusion, kidneys were scored based on the clinical assessment score and their suitability for transplant determined. Biopsies were collected at the end for histological assessment.
Results:
All kidneys were successfully reperfused with immediate recordable renal blood flow (RBF). RBF continually improved over the course of the perfusions, peaking at 12 hours, and negatively correlated with intra-renal resistance. Perfusate sodium concentrations remained stable and within physiological parameters. Sodium bicarbonate increased over time with a corresponding decrease in lactate concentrations, demonstrating active renal gluconeogenesis. Urine production began immediately in all kidneys and was sustained, indicating a maintenance of functionality. Under the clinical perfusion assessment score, all kidneys received a score of 1 and would be considered suitable for transplantation. Histological assessment revealed kidneys were injury-free with a REMUZZI score of 0.
Conclusion:
We have developed a NMP protocol that safely preserves donor kidneys for 15.5 hours. Successful perfusion was achieved with stable haemodynamics, blood-perfusate biochemistry, and maintained urine output. Importantly, kidneys remained in optimal health, with no evidence of injury. This protocol may enable the electivisation of transplantation, while reducing ischaemic injury associated with static cold storage.
Lay summary
Lay title: Keeping Kidneys Healthy: Safely Preserving Donor Kidneys for 12 Hours with New Technique
Background:
Maintaining donor kidney health before transplantation is crucial. Presently, kidneys are preserved on ice which can harm them. An alternative technique, "normothermic machine perfusion," involves circulating blood through kidneys to warm them and provide nutrients. However, it's limited to 1-3 hours. Our goal was to extend this to 12 hours, offering flexibility for transplants and enhancing post-transplant organ performance.
Your methodology - what have done in your study?
Kidneys were collected from abattoir pigs and stored on ice for a short period. They were then connected to a machine that copies the body's conditions, keeping them healthy and functioning for 12 hours. Blood flow, chemicals, and urine production were monitored continuously to see how well the kidneys were doing. Kidney tissue was collected after 12 hours to look at how healthy it was.
Results: What have you discovered?
Kidneys quickly received sufficient blood flow upon connection, improving over 12 hours, indicating better function. Blood chemistry remained stable. All kidneys consistently produced urine, showing maintained function. After 12 hours, kidneys were suitable for transplant.
What are the potential benefits that this research could bring to patients?
A 12-hour normothermic machine perfusion protocol was developed, preserving kidney health. This promising technique could improve organ transplant safety and success by extending the time kidneys remain viable before surgery.

Katie Mylonas
Authors and affiliations:
Ross A Campbell1, Marie-Helena Docherty1, David P. Baird1, David A Ferenbach1, Jeremy Hughes1 and Katie J Mylonas1.
1Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh EH16 4UU, Scotland, United Kingdom.
Scientific summary
Background:
Senescent cells (SCs) accumulate in the kidney with age/injury. They are metabolically active, promoting inflammation/fibrosis via release of senescence associated secretory phenotype (SASP) cytokines. We have shown ageing induces renal epithelial senescence, with administration of the senolytic drug ABT- 263 prior to renal ischaemia reperfusion injury (IRI) being protective; reducing fibrosis and inflammation, whilst promoting preserved function. ABT-263 also reduces macrophage numbers. I hypothesise that SCs compromise renal repair/regeneration after IRI in mice by driving excessive monocyte/macrophage recruitment and negatively influencing macrophage phenotype.
Methods:
In vitro: human proximal tubular epithelial cells (PTECs) were irradiated with 10 gy radiation, to induce senescence. Chemokine CCL2, phospatidylserine (PS), a pro-phagocytic ‘eat me’ signal, and CD47, an anti-phagocytic ‘don’t eat me’ signal, were measured by various methods.
Macrophages were cultured from healthy human volunteer blood and exposed to the supernatant (conditioned medium; CM) of senescent or control PTECs. The phagocytic receptor, MerTK, was measured by flow cytometry (FC).
In vivo: macrophages were interrogated in old, young and young mice with inducible renal senescence (TG) by FC and immunofluorescence. IRI was carried out on WT and CD47-/- mice, with nephrectomy of the CLK at 5 weeks. Kidney function was measured via serum cystatin c 6 weeks post-IRI.
Results:
In vitro: senescent PTECs produced significantly increased levels of the chemokine CCL2 (p<0.01) and PS (p<0.01), but also CD47 (p<0.001). Macrophage MerTK was reduced after exposure to senescent CM vs control (p<0.05).
In vivo: old and TG mice had more renal inflammatory macrophages than young WTs (p<0.05).
CD47-/- mice had better kidney function after 6 weeks of IRI than WTs(p<0.01) indicating that SC clearance by macrophage can be enhanced with positive results in the kidney.
Conclusion:
SC negatively affect macrophage phenotype/function. These studies will establish the foundation for immune-enhancing therapies in disease to reduce renal fibrosis.
Lay summary
Lay title: Exploring how harmful senescent cells interact with the immune system in order to promote kidney repair.
Background:
Harmful senescent cells accumulate with age/injury. Removing them increases healthy lifespan in mice. Ageing mice had increased kidney cell senescence which was reversed with the novel drug ABT-263 that destroys senescent cells. ABT-263-treatment protected kidneys from scarring after injury.
White blood cells of the immune system called macrophages appear in the kidney after injury to aid healing. However, excessive macrophages can also be damaging.
I propose that the ability of these macrophages to promote kidney repair is negatively affected by the presence of senescent cells. I will test whether I can enhance kidney repair by changing how these cells interact with each other.
Your methodology: what have you done in your study?
We have measured:
- factors produced by senescent cells, and macrophages that have interacted with senescent cells
- levels of macrophages in the kidneys of mice that have lots of senescent cells
- kidney function in mice where macrophages can clear senescent cells more easily.
Results: what have you discovered?
Senescent cells change the way macrophages work, affecting how they help heal injured kidneys.
What are the potential benefits that this research could bring to patients?
I hope that these studies will establish the foundation for therapies to improve how macrophages help heal the injured kidney.

Winnie Chan
Authors and affiliations:
Winnie Chan1,2, Carolyn Greig1, Richard Borrows2,3
1School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham
2Department of Nephrology, University Hospitals Birmingham NHS Foundation Trust
3School of Immunity and Infection, University of Birmingham
Scientific summary
Background: Kidney transplantation is the treatment of choice for patients reaching end-stage renal disease. In addition to rectifying uraemia and metabolic complications contributing to an overall sense of well-being, kidney transplantation improves mortality, morbidity and quality of life. After successful transplantation, many patients regain some physical capacity, but frailty represents a prevalent phenomenon, affecting >50% of this population.
Frailty is a state of increased vulnerability to physical stressors. It is characterised by progressive and sustained degeneration in multiple physiological systems, and is associated with worsened mortality, morbidity and quality of life in kidney transplant recipients. To date, no studies have evaluated the effects of resistance exercise training (RET) combined with protein supplementation (RETPS) in ameliorating frailty in these patients. This randomised controlled feasibility study explores 1] the feasibility of implementing a novel low-cost home-based RET programme with/without protein supplementation; 2] the effects of RET with/without protein supplementation on frailty and functional status, muscle strength, and quality of life; 3] the cost-effectiveness of RETPS compared to RET alone; and 4] the potential efficacy measures.
Methods and Results: A total of 40 clinically stable kidney transplant recipients will be enrolled and randomly assigned to either a RET group (n=20) or a RETPS group (n=20), each with a 12-week intervention period. Measurements will be undertaken pre- and post- intervention, including assessments of study feasibility, frailty and functional status, muscle strength, muscle thickness, body composition, kidney function, inflammation, cardiometabolic status and patient-reported outcome measures.
Conclusion: This study sets out to introduce a holistic approach, incorporating optimum and safe level of protein supplementation with achievable and effective home-based exercises, thereby improving strength and combating frailty in kidney transplant recipients. Ultimately, this study will inform the design of a full-scale randomised controlled trial to investigate the impact of RET and RETPS in kidney transplantation.
Lay summary
Lay Title: Can home-based exercise and protein supplements improve frailty in kidney transplant recipients?
Background:
Kidney transplantation is the preferred treatment for advanced kidney disease. It restores the ability to eliminate body toxins, prolongs longevity and improves quality of life. However, frailty remains a common problem after transplantation, accounting for recurrent hospitalisation and death.
No studies have evaluated the effects of resistance exercise training with or without protein supplementation on frailty status in kidney transplant patients. This study aims to answer 4 research questions: 1. Is it feasible to implement a home-based resistance exercise training programme? 2. What is the impact of resistance exercise training with or without protein supplementation on frailty status? 3. Is combining protein supplementation with exercise training cost-effective? 4. What are the potential efficacy measurements?
Your methodology: what have you done in your study?
Forty kidney transplant patients will be randomly assigned to either resistance exercise training only, or resistance exercise training in combination with protein supplementation for 12 weeks. Measurements will be undertaken before and after the intervention including assessments of feasibility, frailty status, functional status, muscle strength, muscle thickness, body composition, kidney function, inflammation, cardiometabolic status, and patient-reported outcome measures.
Potential Benefits: This study has the potential to introduce a holistic approach, combining the correct and safe level of protein supplementation with achievable home-based exercises to combat frailty and inform future larger-scale studies.

Dr Jessica Kepple
Authors and affiliations:
Jessica D. Kepple12, Phalguni Rath1, Katherine R Bull123
Wellcome Centre for Human Genetics1, Nuffield Department of Medicine2, MRC Human Immunology Unit, Radcliffe Department of Medicine3; University of Oxford, Oxford, UK.
Scientific summary
Background: SHDRA or “structural heart defects and renal anomalies syndrome” is a rare genetic disorder arising from mutations in the TMEM260 gene, encoding a broadly expressed endoplasmic reticulum-bound O-mannosyltransferase protein. Infants with biallelic mutations have severe multiorgan abnormalities, including early-onset renal impairments and defects, with high paediatric mortality rates. Given the patient renal phenotypes, we hypothesize that TMEM260 is a novel gene required for early renal development.
Methods: To examine the renal function of human TMEM260 variants and isoform-specific deletions, we are generating TMEM260 deficient renal organoids from induced pluripotent stem cells (iPSC) using CRISPR-Cas9 gene editing. We will assess morphology, gene expression, and proteomic changes in control, full deletion, and TMEM260 variant organoids. Immunofluorescent imaging will elucidate cellular changes in tubular and glomeruli structures at serial developmental timepoints. Single-cell RNA Sequencing (ScRNA-Seq) will reveal cell type specific transcriptional changes, supported by mass spectrometry for 4000+ proteins.
Results: We have generated RNA sequencing data from human renal transplant donors demonstrating broad renal TMEM260 expression. We performed analysis mapping known human mutations to the protein structure, revealing most mutations map to the ER luminal domain involved in substrate binding. We have established a complete TMEM260 deficient renal organoid model, with the deletion confirmed by Sanger sequencing and qPCR. Assessment of gross organoid morphology revealed variable structures and sizes between mutants and controls. Organoids lacking TMEM260 have reduced expression in developmental genes including DAPL1, SIX1, and GATA3. Further assessment is being done to determine cell specific changes upon TMEM260 loss in the developing organoid.
Conclusion: Overall, this project will provide novel insights into renal TMEM260 function, with possible implications for alternatively impacted organ systems and gene discovery. Having established an organoid model for complete TMEM260 loss, we seek to develop human variants and isoform-specific deletion models to measure cellular interactions and identify altered regulatory pathways.
Lay summary
Lay Title:
Exploring TMEM260 as a novel protein required for kidney development.
Background:
The rare disease SHDRA or “structural heart defects and renal anomalies syndrome” is caused by loss of the gene TMEM260, leading to severe developmental defects in the heart, brain, and kidney and death in early childhood. Currently, we know very little about how TMEM260 loss causes disease. We predict that TMEM260 is important for normal kidney development, with its loss leading to significant structural and functional changes.
Your methodology: what have you done in your study?
To determine TMEM260 function, we have deleted it in human cells that we then programmed into kidney organoids, miniaturised kidneys in a dish. We are measuring how TMEM260 loss affects kidney structures, genes, and proteins in the different cell types.
Results: what have you discovered?
We found that TMEM260 is present in many human kidney cells. Organoids that lack TMEM260 grow visibly different structures and sizes to controls and have lower amounts of important developmental genes. We are working to further explore cell changes in these kidney organoids.
What are the potential benefits that this research could bring to patients?
Given the unknown function of TMEM260 in human kidneys, this project will advance our understanding of its role in kidney development, expand the use of organoids as a tool to investigate kidney disease, and may lead to uncovering new targets for SHDRA treatments.

Louise Oni
Authors and affiliations:
Alexandra Weightman1, Andrew J. Chetwynd1, Louise Oni1
1University of Liverpool & Alder Hey Children’s Hospital, Liverpool
Scientific summary
Background:
A limitation to understanding rare diseases is capturing every patient. In high throughput discovery science (‘omics’) large amounts of data can be collected from small biosample volumes (<100µL). Micro-sampling is therefore an interesting approach to mechanistic studies. Dried blood spots (DBS) are collected via a finger prick. This technique allows patients to collect samples from home and return by post, saving time and money. A growing theme is the expansion to other biofluids such as urine and saliva. This work aimed to evaluate the perception of the use of at home dried biofluid collection.
Methods: We devised a 23-item questionnaire distributed to members of Kidney Research UK’s`s ‘Kidney Voices for Research’.
Results: In total 86 people responded, 95% expressed that they would provide a dried biofluid from home. In addition, 80% of patients replied stating that it would increase their likelihood to take part in medical research with the remaining 20% saying they would have volunteered regardless. Greater than 99% reported use of at home testing ranging from Covid-19 screening to DNA testing for ancestry research. The majority of patients reported willingness to provide dried blood (90.7%), urine (95.3%) and saliva (95.3%) with an overall preference in collecting samples monthly (35%-36%).
Patients were more willing to provide urine and saliva samples (weekly if needed) compared to blood sampling, this may be a result of the more invasive nature of a finger prick test. Patients cited that this method was ‘convenient’, ‘easy to do’ ‘no blood taken from vein’ hence ‘saving’ them for future.
Conclusion:
Patients are supportive of at home biofluid micro-sampling. This enhances equity in involvement in clinical research and increases opportunities to monitor diseases.
Lay summary
Lay title: What do people with kidney disease think of taking ‘microsamples’?
Background:
A lot of people are unable to help with kidney research because they live a long way from a research hospital or cannot afford the time or money on travelling to give samples. This makes it hard to do research that is reflective of the true kidney patient populations.
Your methodology: what have you done in your study?
To fix this we are interested in patients collecting small amounts of blood, urine and saliva from home and posting them back to us once they have dried. To see if this idea is popular with patients, we asked the Kidney Research UK Kidney Voices for Research members to complete a questionnaire looking at different aspects of this work.
Results: what have you discovered?
Over 95% of patients are happy to provide samples this way with 80% saying they are more likely to give samples for research this way. They were happy to provide blood (91%), urine (95%) and saliva (95%) with around 80% of patients willing to provide a sample at least once a month.
What are the potential benefits that this research could bring to patients?
These findings show that kidney patients are happy to provide samples from home and post them back. This makes it easier for patients and researchers to collect samples more frequently to track disease. It also reduces barriers to taking part in research and includes people from all locations and backgrounds.

Manjo Valluru
Authors and affiliations:
Valluru MK1, Genomics England Research Consortium, and Ong ACM1
1Division of Clinical Medicine, University of Sheffield
Scientific summary
Background: ADPKD is generally an adult-onset disease caused by germline variants in PKD1 and PKD2. However, ADPKD can rarely present in infancy or early childhood and occasionally in late childhood or early teenage years. We recently reported that 70% of cases with very-early onset ADPKD (age<18 months) had biallelic PKD1 variants, most commonly a pathogenic variant in combination with a hypomorphic variant (Durkie et al, 2021; 2023). To investigate the potential of other known or novel genes to contribute to an early-onset-ADPKD phenotype (2-16 Years), we hypothesised that an early-onset clinical presentation was more likely to be associated with a digenic or oligogenic inheritance. To address this question, whole genome sequencing data from young ADPKD patients (age<16yr) recruited to the UK 100,000 Genomes Project (100K GE), found to carry a PKD1 variant, was analysed alongside that of one or both parents.
Methodology: The inclusion and genotyping of participants in the 100K GE were managed by Genomics England Limited (GEL). Initially, affected PKD1 cystic kidney disease Trio or Duo probands were filtered based on AGE<16 and analysed in GEL Research Environment. Recorded data were subjected to downstream bioinformatics analysis.
Findings: 21 PKD1 positive cases under 16 years were identified in 100K GE. Ten cases had a negative family history of PKD, and no cases of very early-onset-ADPKD were found. Of these, 6 were monoallelic, 2 were biallelic and 13 were digenic or oligogenic. Unlike the very early-onset-ADPKD cohort, only two children with early-onset-ADPKD in the 100K GE cohort had biallelic PKD1 variants. We identified 24 PKD1 variants inherited in trans with a second gene variant derived from 21 other genes (REVEL >0.4). Of these, 10 were known to be associated with PKD or congenital anomalies of the kidney and urinary tract (CAKUT) phenotypes and 11 were associated with other renal syndromes. These digenic variants could be strong candidates as potential genetic modifiers of the human ADPKD phenotype.
Lay summary
Lay title: Exploring Genetic Factors in Childhood-onset Autosomal Dominant Polycystic Kidney Disease
Background: ADPKD most commonly presents in adults but can rarely occur in infancy, childhood, or teenage years. Our recent research found that ADPKD presenting in infancy (<18 months) is commonly due to two PKD1 gene faults, often a combination of a harmful and a milder change.
To better understand whether this observation extended to children, we investigated whether faults in other genes might contribute to ADPKD presenting in childhood. We suspected that when ADPKD starts early in life, other genes could be involved.
Your methodology: what have you done in your study?
We obtained genetic data from young ADPKD patients and their parents recruited in the UK's 100,000 Genomes Project (age<16years) and analysed children with a PKD1-related gene variation alongside their parents' genes.
Results: what have you discovered?
Among the participants, 21 with PKD1 gene changes were identified. Of significance, changes in 24 other kidney genes were present in the majority of these cases, often inherited from the unaffected parent. Of interest, these genes had been linked previously to other kidney diseases.
What are the potential benefits that this research could bring to patients?
Our study uncovers a strong genetic basis for childhood-onset ADPKD. This insight could lead to better ways of identifying, managing and treating the disease in children.

Andrew Streets
Authors and affiliations:
Streets AJ1, Valluru MK1, Chang L1, Simms RJ1, Pei Y2 and Ong ACM1
1Division of Clinical Medicine, University of Sheffield
2Division of Nephrology, University of Toronto
Scientific summary
Background: ADPKD is the most common inherited cause of kidney failure, caused by germline mutations affecting PKD1 and PKD2 in >90% of patients. The approval of tolvaptan as a disease-modifying therapy in ADPKD patients at risk of rapid disease progression has highlighted the need to develop more accurate prognostic models. We recently showed that global urine extracellular vesicles (UEV) miRNA profiling was highly informative in this regard (Magayr TA, et al. Kidney Int. 2020). In combination, five miRNAs improved the diagnostic performance (AUC) of ultrasound mean kidney length (US-MKL) to discriminate patients with >3ml/min/yr and <3ml/min/yr annualised historical estimated glomerular filtration rate (eGFR) slopes over 5 years. In this study, we report the refinement of differentially expressed (DE) UEV miRNA between healthy controls and ADPKD patients in an independent validation cohort enriched for patients with early disease as the next step to validating its use in prognostic models.
Methodology: In silico bioinformatics analysis was performed to identify a panel of DE UEV miRNAs that discriminate between healthy controls and ADPKD patients. Panel miRNA screening by TaqMan qPCR was conducted in UEV microRNAs isolated from an independent validation cohort (n=70) comprising healthy volunteers (n=25), ADPKD patients with early (eGFR>60ml/min, n=35) or late (eGFR<60ml/min, n=10) disease. Based on this focused panel, bioinformatics analysis was performed to identify the major predicted miRNA-mRNA pathways altered in ADPKD datasets.
Findings: Of 21 DE miRNAs (Panel, AUC>0.8), seven miRNAs were found to be altered in the validation cohort. Furthermore, the combination of five of these improved the classification of early (AUC=0.76) and late (AUC=0.93) disease by ROC analysis. From miRNA-mRNA network analysis, we identified 90 targets altered in ADPKD. Over-representation analysis revealed enrichment for altered Cell-migration/adhesion/matrix and Rho-GTPase-signalling Genesets. Our results have validated a focused panel of five UEV miRNAs that can discriminate ADPKD patients from controls.
Lay summary
Lay title: Spotting Kidney Disease Early: A Urine Test for Rapid ADPKD progression
Background:
A common genetic kidney disease called ADPKD can lead to kidney failure. Scientists are working to develop better ways to predict the progression of this disease. One promising approach involves studying tiny particles called extracellular vesicles (UEVs) in urine. These UEVs contain molecules called microRNAs (miRNAs) that can provide important information about the disease. Our previous study found that analysing specific miRNAs in UEVs from urine samples could help predict the disease's severity.
Your methodology: what have you done in your study?
To improve prediction, we tested urine samples from a new group using a selected panel of miRNAs.
Results: what have you discovered?
We identified seven miRNAs that are altered between healthy people and ADPKD patients. By combining five of these miRNAs, it may be possible tobetter classify patients with early and late stages of the disease. We also looked at the connections between miRNAs and the genes they influence. We found that target genes are involved in cell movement, adhesion, and communication.
What are the potential benefits that this research could bring to patients, whether that be in the near or distant future?
We have found a way to analyse specific miRNAs in urine samples to predict the severity of ADPKD. This could help doctors identify patients at risk of rapid disease progression and provide them with more targeted care. The miRNAs we identified offer insights into the underlying processes involved in the disease.