Driving Discoveries 2022: Connecting our fellows and students
Abstract presentation details for our oral and poster presentations
Oral abstract presentations:

Fiona Chapman
Authors and affiliations (if published):
Fiona A. Chapman, David E. Newby, Neeraj Dhaun (Bean)
Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh
Background:
Chronic kidney disease (CKD) affects one in ten people and cardiovascular disease (CVD) is its commonest complication. Despite standard-of-care, many patients with CKD progress to kidney failure and/or die of CVD. Apelin is an endothelium-dependent vasodilator and an attractive therapeutic target for CKD. We examined, for the first time, the local vascular actions of apelin in CKD.
Methods:
We recruited 15 patients with non-diabetic CKD and 15 age, and sex-matched healthy volunteers. We assessed blood pressure (BP) and arterial stiffness (pulse wave velocity, PWV), and examined vasodilatory responses to acetylcholine (ACh, endothelium-dependent vasodilatation), sodium nitroprusside (SNP, endothelium-independent vasodilatation), and pyroglutamated apelin-13 ([Pyr1]apelin-13) using gold-standard forearm plethysmography. To assess the effect of apelin on endogenous fibrinolysis, we measured tissue plasminogen activator (tPA) and plasminogen activator inhibitor-1 (PAI-1).
Results:
CKD patients had a mean age of 55±4 years and 53% were male. Patients with CKD had a higher BP (mean arterial pressure: 102mmHg versus 93mmHg, p<0.05) and increased PWV (7.5±2.2m/s versus 6.0±0.9 m/s, p<0.01) compared to healthy volunteers. Similar dose-dependent vasodilatation to ACh and SNP was seen in both groups. [Pyr1]apelin-13 increased forearm blood flow by ~30% in both healthy patients and CKD patients (p<0.01 compared to baseline for both). Net tPA antigen release increased 20-70 fold in response to apelin in both healthy patients and CKD patients, with a trend to a greater release in CKD patients. There was no association between apelin response and kidney function.
Conclusion:
Apelin causes vasodilatation in optimally managed patients with CKD. Systemic studies are needed to further examine the actions of apelin in CKD.
Lay summary:
Kidney disease is common and affects one in 10 people. Many people with kidney disease have high blood pressure and go on to have heart attacks or strokes, and some die from this. Current treatment for kidney disease is limited and unfortunately many people still develop kidney failure. We want to find new treatments for kidney disease. Apelin is a small protein found in the body. It lowers blood pressure, helps the heart pump better and may help the kidneys get rid of salt and water. We want to see if it might be a new treatment for kidney patients that could also protect them from heart disease. We explored what apelin did to blood vessels in people with kidney disease. We gave apelin into the arms of 15 people with kidney disease and 15 healthy people and measured how well their blood vessels relaxed. Apelin caused the blood vessels in people with kidney disease to relax the same amount as blood vessels in healthy people. This is very promising. We now need to give a longer treatment with apelin throughout the body and see what happens to blood pressure, heart function and the kidneys.
Biography:
Chronic kidney disease is a potentially devastating, life-changing condition with very few treatment options. As a renal registrar I have been fortunate to work in the renal units in both Edinburgh and Glasgow. I really enjoy working with patients and wanted to move into clinical research to try and have a greater impact for all patients with kidney disease.
Knowledge of the apelin system has increased enormously in the last decade. Excitingly, animal studies suggest it is a new potential target for treating kidney disease. My study is the first to explore the actions of the apelin system on the kidneys in humans. This is not only essential to better understand the system but will also provide the first insight into whether it is a viable target for treating chronic kidney disease. Ultimately, my study could pave the way for larger clinical trials and the development of a new treatment for CKD.
Being awarded a Kidney Research UK training fellowship has transformed my career. I have been incredibly privileged to work with international experts who have helped me ensure my studies are well planned and successful. I am so grateful to them, and to the patients who have taken part.

Richard Naylor
Authors and affiliations (if published):
Richard W. Naylor, Rachel Lennon
Wellcome Centre for Cell-Matrix Research, University of Manchester
Background:
Autosomal dominant polycystic kidney disease (ADPKD) is the most common, potentially life-threatening monogenic disease. A major disease feature of ADPKD is the formation of large fluid-filled sacs, called cysts, in the kidney. Over time, these cysts induce fibrosis and preclude kidney function. Half of patients with ADPKD require renal replacement therapy by the age of 60, and one in eight kidney transplants in the UK are performed on patients with ADPKD. Therefore, ADPKD places significant burden on both patients and the NHS. ~93% of ADPKD cases are caused by mutations in either PKD1 or PKD2, which encode for polycystin-1 (PC1) and PC2, respectively. ADPKD is considered a ciliopathy as PC1/PC2 form a calcium ion channel that is located at the cilium. Many studies have confirmed the role of PC1/PC2 in regulating calcium signalling and other downstream pathways, such as cAMP signalling. However, the contribution of the extracellular matrix in the early stages of ADPKD disease progression has not been assessed. Given the extracellular matrix is important in many processes and signalling pathways that are implicated in ADPKD progression, it is vital we understand how it contributes to cystogenesis.
Methods:
Mass spectrometry-based proteomic approaches were utilised on micro-dissected zebrafish embryonic kidney tubules to determine the changes in the extracellular matrix that occur in a zebrafish model of ADPKD. qRT-PCR and immunostaining approaches were also employed to develop novel ways of determining rates of cell proliferation in this tissue.
Results:
It was found that altering the composition of the extracellular matrix in the zebrafish embryonic kidney is sufficient to reduce cell proliferation in control and ADPKD zebrafish embryos. However, analysis of the embryonic kidney proteome in ADPKD zebrafish embryos shows little change is observed in the overall make-up of the extracellular matrix.
Conclusion:
These results suggest that the extracellular matrix can play an important role in regulating cell proliferation in normal and ADPKD kidney tissue. However, in the earliest stages of the disease, there appears to be little change in the composition of the extracellular matrix, supporting the conclusion that matrix dysregulation is not required for the hyper-proliferation phenotype associated with cystogenesis in ADPKD.
Lay summary:
Autosomal dominant polycystic kidney disease (ADPKD) is caused by variants in the PKD1 and PKD2 genes. The major feature of the disease is the formation of fluid-filled sacs, cysts, in the kidneys. These cysts can grow very large, to the size of golf balls in extreme cases. Half of patients with ADPKD will need renal replacement therapy by the age of 60, which places a huge burden on patient mental health and the NHS. Cysts are able to form because cells in the kidney tubules begin proliferating, enabling cyst growth. Targeting this increased proliferation is a potential therapeutic approach, and this is one of the ways that tolvaptan works to reduce cyst formation. The protein mesh that surrounds cells, called the extracellular matrix is of greatest interest. This matrix is extremely important as it enables cells to proliferate and grow, therefore I hypothesise that changes in the extracellular matrix in the early stages of ADPKD is important for cyst growth and progression of the disease.
Biography:
I am intrigued by the complex structure of the kidney and how important it is to human health, which often is not fully realised by the wider population. My career started looking at how the kidney forms, but now I am more invested in understanding how the kidney changes during disease, which often involves many developmental pathways being re-initiated.
My research is aiming to determine new paradigms in kidney disease, especially in ADPKD. I have developed new tools to aid ADPKD research and I believe understanding the under-appreciated role of the extracellular matrix is fundamental to many kidney diseases.
Kidney Research UK has offered me the amazing opportunity to work independently and guide my own research. Attending conferences such as Driving Discoveries enables me to network with people who will help my research now and in the future. The charity's patient-focus also gives me access to the people that we are doing the research for, the patients. I hope in the future such access will guide me on better understanding the areas of research that are most essential in kidney disease.

Amarpreet Thind
Authors and affiliations (if published):
Amarpreet K Thind1,2,Annabel Rule2,3, Dawn Goodall2, Shuli Levy2, Sarah Brice2, Frank JMF Dor2,4, Nicola Evans5, David Ospalla2, Nicola Thomas6, David Wellsted7, Lina Johansson1,2, Michelle Willicombe1,2, Edwina A Brown1,2
1Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London,
2Imperial College Healthcare NHS Trust, Hammersmith Hospital,
3Central London Community Healthcare NHS Trust,
4Department of Surgery and Cancer, Imperial College London,
5Guy’s and St Thomas’ NHS Foundation Trust,
6Institute of Health and Social Care, London South Bank University,
7The Centre for Health Services and Clinical Research, The University of Hertfordshire.
Background:
Older end-stage kidney disease (ESKD) patients are particularly vulnerable to developing frailty, and the presence of frailty is well recognised as impacting on clinical and experiential outcomes. With kidney transplantation rates in older people increasing, achieving a more detailed understanding of patient experiences is necessary for guiding appropriate treatment decision making.
The “Kidney Transplantation in Older People (KTOP): impact of frailty on outcomes” study is an active study exploring the experiences of older people with ESKD awaiting and undergoing transplantation. This manuscript presents a secondary analysis of the Edmonton Frail Scale (EFS) and its relationship with patient experience scores, at recruitment into the KTOP study.
Methods:
The KTOP study is a single centre, prospective, mixed methods study, which began in October 2019. All ESKD patients aged ≥60 who were being considered for transplantation at Imperial College Renal and Transplant Centre were eligible. Recruited participants had their frailty assessed by the EFS and completed 5 questionnaires to assess patient experience and quality of life (Short Form- 12 (v2), Palliative Care Outcome Scale – Symptoms Renal, Depression Patient Health Questionnaire-9, Illness Intrusiveness Ratings Scale, and Renal Treatment Satisfaction Questionnaire). Categories of components within the EFS were then created to support the analysis; psychosocial, physical function, medical status, and general health status.
Results:
210 patients have been recruited (aged 60-78), 186 of whom had completed EFS assessments. 118 (63.4%) participants were identified as not frail, 36 (19.4%) vulnerable, and 32 (17.2%) participants were frail. Across all study questionnaires poorer patient experiences and quality of life were reported as the identified frailty status worsened. Severe deficits in the psychosocial category of the EFS showed a significant association with higher depression screen scores, lower physical and mental function scores from the Short Form-12(v2), and lower renal treatment satisfaction scores. Deficits in the physical function and medical status categories of the EFS showed some association with the patient experiences scores reported.
Conclusion:
In the KTOP study cohort at recruitment, vulnerable and frail candidates reported worse quality of life and patient experiences. Severe deficits in the psychosocial domains of the EFS show a strong association with patient experience and quality of life, whilst deficits in the physical function and medical status domains showed a lesser association. This study has highlighted specific domains within the EFS that may be suitable for targeted interventions to improve patient experience and optimise outcomes.
Lay summary:
The quality of life for older people with kidney disease is highly variable. Older people with kidney disease are at a higher risk of becoming frail, and frailty has been shown to affect their quality of life. The Kidney Transplantation in Older People study uses questionnaires to better understand the experiences of older people as they wait for and receive a kidney transplant. The study is running in West London and so far, 210 older people are involved. Early results have shown that older people who are vulnerable to frailty or already frail, do describe a poorer quality of life and experiences of living with kidney disease, compared to those who are not frail. We have also found that specific aspects of an older person’s life play a bigger role in their experiences of living with kidney disease. These include difficulties with mood, memory, taking medications and the social support they have around them. Low levels of physical activity also seem to contribute negatively. These early results have suggested areas where we may be able to better support older people as they wait for a kidney transplant, to help improve their quality of life and their experiences of kidney disease.
Biography:
Amarpreet Thind is a clinical research fellow at Imperial College London and the Imperial College Renal and Transplant Centre. Having graduated from Newcastle University in 2012, with an intercalated MRes degree, she is currently completing her PhD in geriatric nephrology. She is part of a multi-disciplinary team conducting the Kidney Transplantation in Older People (KTOP) clinical study. The study is exploring the impact of frailty on clinical and experiential outcomes in older people, accelerating our understanding of the unique clinical factors, influences, and priorities that drive care and decision making in this dynamic population.
During her early registrar training Amarpreet developed a realisation for the varied and significant impact that living with kidney disease has on the everyday lives of patients, families and their carers. A desire to better understand these perspectives and find practical ways to improve clinical care drove Amarpreet to conduct and continue with renal research. Only through working with Kidney Research UK has this valuable research been possible. In addition to the essential financial support, the charity has created a collaborative, multi-professional environment, to bring the project into fruition, promote the findings, and ultimately bring us closer to providing better patient-centred care for older people.

Dr Ben Reynolds
Authors and affiliations (if published):
Ben C Reynolds1, Claire Hagerty1 Vassilis Charissis2, Soheeb Khan2, Lyall Campbell2,
1Royal Hospital for Children, Glasgow
2Glasgow Caledonian University
Background:
Undertaking training of patients/families in peritoneal dialysis (PD) can be stressful for families and requires face to face availability of educators. In paediatrics, children are usually admitted for this training to occur. The rarity of paediatric kidney disease limits opportunities for families to meet peers/others, so the decision to undertake PD may not be truly informed or shared between clinicians and families.
We hypothesized that virtual reality (VR) could offer an alternative supplemental route for training of patients, families, and staff in dialysis. We aimed to develop a prototype as proof of concept that VR is suitable for this.
Methods:
Qualitative interviews were conducted with families, nursing staff, and nurse educators to determine the key elements of the VR program, and main objectives that VR education could fulfil. A multimedia library was composed to permit 3D model creation of all aspects of PD. A VR application was coded and developed with stages of preparation/machine set-up, connection, and disconnection of a dialysis session. Two brief troubleshooting scenarios were also developed. Following feedback from medical and nursing staff, a tutorial was added and several adjustments made to the program. Several iterations were completed with feedback from staff members at each stage.
Results:
A prototype VR PD educational tool has been developed for use with the MetaQuest 2 VR headset. Informal feedback sought from patients and families familiar with PD has been generally very positive. One parent had significant motion sickness and could not tolerate use of the headset. One family with experience of VR and PD reported the application was ‘epic’. Two families have trialled the tool prior to undertaking any education in PD – both reported that the tool alleviated anxiety, and that they could relate the VR experience to their training or nurses setting up the machine on the ward. For the one family who have since completed face to face training, this took three days (usually five to seven in our unit).
Conclusion:
VR offers a potentially very useful supplement to training in medical technologies such as dialysis. Training duration for families may be shortened. Formal evaluation of the application’s utility as an educational tool is needed, as is objective evidence of its benefit to patients/families. Multiple potential alternative uses of VR in medical education also exist.
(One aspect of evaluation planned for August 2022 but not available at time of abstract submission)
Lay summary:
Learning how to do dialysis at home can be stressful for patients and families. For children, families may not have the opportunity to meet with dialysis-experienced patients as end stage kidney disease is rare. Parents may have to decide on the type of dialysis, or agree to undertake dialysis training, without a full understanding of what this entails.
We have developed a prototype virtual reality application which provides a realistic simulation of setting up, connecting, and disconnecting a peritoneal dialysis session for a child. Interviews with families, nursing staff, and nurse specialists, helped identify the key areas that training should cover. A multimedia library was created to allow recreation of all aspects of a dialysis session. The program was developed, with several versions tested by medical and nursing staff to identify any areas that needed to change.
A small number of families have tried out the application – all were impressed with it and reported that they could see a real place for it in training patients in the future. Two families used the application before any kind of training – both said it was very helpful and made them less stressed before starting. A formal assessment of the application is planned.
Biography
Ben is a consultant paediatric nephrologist based at Royal Hospital for Children, Glasgow, and the clinical lead for transplantation. His interests include the immunology of transplantation, managing the challenges associated with adolescence and transition to adult care, and the patient experience/journey.
The pandemic brought challenges and isolation for many. Ben staved off some of the isolation by immersing himself in the virtual reality world and realised that there was real potential for these tools to be used for patient and family education. Similar enthusiasm from Kidney Research UK has provided funding for the development of these tools.
Being directly involved in kidney research is always exciting. Being able to witness the direct impact of research findings on patients and families is a privilege for all clinicians; being able to also contribute to that research is even more powerful.

Simon Baker
Authors and affiliations (if published):
S.C. Baker1,*, A.S. Mason1, R.G. Slip1, K.T. Skinner1, A. Macdonald2, M. Wellberry-Smith3, O. Masood3, R.S. Harris4, T.R. Fenton5, M. Periyasamy7, S. Ali7 and J. Southgate1
1Jack Birch Unit for Molecular Carcinogenesis, Department of Biology and York Biomedical Research Institute, University of York, UK.
2Faculty of Biological Sciences, School of Molecular and Cellular Pathology, University of Leeds, UK.
3Leeds Kidney Unit, St James’s University Hospital, UK.
4College of Biological Sciences, University of Minnesota, USA.
5School of Cancer Sciences, University of Southampton, UK.
6Department of Surgery & Cancer, Imperial College London, UK.
*Presenting author
Background:
Age and smoking are the main risk factors for bladder cancer. However, studies of mutational signatures show the anti-viral apolipoprotein B mRNA editing enzyme catalytic polypeptide (APOBEC) enzymes, and not smoke-derived carcinogens, are responsible for the preponderance of mutations in bladder tumour genomes. BK polyomavirus (BKPyV) infects >80% of the 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.
Methods:
We used two models of mitotically-quiescent normal human urothelium to study sequalae of BKPyV infection; an in vitro tissue-engineered model and a ureteric organ culture system. Post-infection urothelia were evaluated using RNA-sequencing, western blotting, immunolabelling and APOBEC-activity assays. Host genome damage was assessed by quantifying apurinic/apyrimidinic sites.
Results:
BKPyV infection significantly elevated APOBEC3A/B proteins, deaminase activity and numbers of apurinic/apyrimidinic sites in the host urothelial genome. Large T antigen (LT-Ag) pushed mitotically-quiescent urothelial cells into an arrested G2 cell cycle state where Rad51-mediated DNA-displacement loops formed during homologous recombination potentially provide a single-stranded DNA substrate for APOBECs. Proximity ligation assays indicate LT-Ag interacts with both Rad51 and APOBEC3 proteins at DNA-displacement loops. In vitro findings were validated in organ cultures, where infected cells were extruded apically from the urothelium in a manner synonymous with production of decoy cells in patients.
Conclusion:
These initial results support reactivated BKPyV infections in adults as a risk factor for bladder cancer in immune-insufficient populations, including transplant patients and the elderly. As further validation, nanorate sequencing will compare mutational signatures from in vitro infections to those of bladder cancers.
Lay summary:
BK virus infects most children without obvious symptoms and can hide, dormant in the kidneys during adulthood. We don’t understand what triggers the sleeping virus to awaken, but this reactivation can happen in around one-in-five kidney transplant patients due to the medication used to prevent transplant rejection called immunosuppression).
When kidney cells sense that they have a BK virus infection, they slough off into the urine. Infected kidney cells also release virus into the urine. Once it has left the kidneys, urine travels down the tubes of the ureter and into the bladder, where it is stored until a person urinates.
This study found BK virus infects human bladder cells leaving changes in the DNA that could lead to cancer. This study is especially relevant to kidney transplant recipients, but has wider implications for others who develop bladder cancer in old age.
We know kidney transplant patients develop ureter and bladder cancers much more frequently than the general population, but currently it is not clear how this happens. We are working with kidney and bladder clinical specialists to understand whether BK virus infections lead to the increased risk of urinary tract cancers in kidney transplant patients.
Biography:
My research seeks to understand the causes of bladder cancer. Following the evidence led me to persistent polyomavirus infections of the kidney as a potential risk factor. Our building evidence suggests understanding how persistence and reactivation of polyomaviruses in the kidney are regulated may hold the key to preventing bladder cancer as well as nephropathy and graft-rejection in transplant recipients.
My Kidney Research UK fellowship is acting as a focal point to reignite old initiatives and start new activities in the BK polyomavirus field. We are bringing together diverse experts in the field and there is a real buzz around BK virus research, both in the UK and internationally.
My Kidney Research UK fellowship has given me the independence and resources to ask the biggest questions in my field and the charity’s relationship with patients is helping us drive change in clinical practice.

Claudia Bruno
Authors and affiliations (if published):
Claudia Bruno1, Rayan Moumneh1, Lynsey Stronach2, Emilie Sauvage1, Ian Simcock2, Silvia Schievano1,2, Rukshana Shroff1,2, Claudio Capelli1,2.
1University College London, Institute of Cardiovascular Science (London, UK)
2Great Ormond Street Hospital for Children, NHS Foundation Trust (London, UK)
Background:
Central venous lines (CVLs) used for haemodialysis (HD) in children are associated with a high complication rate leading to inadequate HD and line replacement in nearly half of the patients. Computational modelling and simulations are powerful engineering tools that use computers to study complex systems by means of mathematics, physics and computer science. This project aimed at providing a computational fluid dynamics characterization of CVLs commonly used in children to understand the causes of their complications, and to improve their performance by optimizing their design.
Methods:
Four models of CVLs of varying design and sizes (6.5Fr, 8Fr, 10Fr and 14Fr) were recreated starting from medical images. Each design presented multiple ports for blood exchange (i.e. tip and side holes). Computational fluid dynamics (CFD) analyses were set up to simulate the CVLs’ performance under both ideal and realistic models of superior vena cava. A variety of flow rates, routinely used in real hospital settings, were applied to study the whole range of working conditions. Haemodynamics features were analysed together with the effect of the catheter insertion inside the vein. CFD findings were then compared to clinical CVLs outcomes from patients (n=26 with 57 lines) over the past two years. Fluid dynamics characteristics of the CVLs studied were used to set up a preliminary design optimisation process.
Results:
In all the simulated CVLs, the arterial lumens, where blood is withdrawn from the body, showed the highest stagnation and recirculation areas. The role of arterial tips appeared to be negligible for the blood exchange and the proximal side holes played a major role for blood aspiration being also subject to the highest levels of shear stress (Figure 1a), regardless of the design. In all the anatomical models, blood velocity increased after catheter insertion (Figure 1b) together with wall shear stresses. CFD results were in accordance with the clinical data which showed a higher recurrence rate of thrombosis for the 8Fr and 10Fr CVLs. Also, microCT images of the explanted lines confirmed presence of thrombosis at the level of the catheter tips. Results suggested that the shear stress is the parameter more strongly related to clinical complications. Preliminarily optimised designs showed a better haemodynamic performance, reducing stagnation areas (Figure 1c).
Conclusion:
In this project, we carried out the fluid dynamics characterisation of commercially available CVLs for paediatric use to try to explain the associated clinical outcomes. This process will guide the optimisation of their design.

Figure 1: a) Shear stress levels contours plotted for the 10Fr model. Velocity magnitude contours: b) inside the vein after CVL insertion; c) inside a 6.5Fr model before and after optimisation to reduce stagnation areas.
Lay summary:
Haemodialysis is a life-saving treatment for children with kidney failure. Central venous lines (CVLs) are the tubes most commonly used for dialysis access. Although ‘lifelines’ for dialysis patients, CVLs are fraught with complications like poor blood flow, clots and infections. In an international study 45% of all CVLs needed replacement due to complications. This results in hospitalisations and multiple operations to replace failing lines, damages the child’s vessels and limits their quality of life. Currently available CVLs are simply miniaturised versions of adult CVLs, and not optimised for the child’s anatomy or blood flow characteristics. In this project we have used a bioengineering approach to better comprehend the fluid dynamics in CVLs and to address their high failure rate. A bioengineering approach can provide data on the fluid dynamics inside the line and can be used to address the high failure rate of paediatric CVLs. We have evaluated different CVL designs currently in use in children and correlated clinical data with engineering results. These results will allow us to set up the optimisation process which will lead to new designs with better flow distribution and low level of damage to blood cells.
Biography:
During the final year of my MSc in biomedical engineering, I had the opportunity to research central venous line performance for paediatric dialysis in collaboration with clinicians at Great Ormond Street Hospital and engineers from University College London. In those six months I came to understand the severity of kidney disease and the importance of the dialysis treatment for paediatric patients. What surprised me the most was that, despite the high rate of complications that paediatric patients encounter due to the poor performances of central venous lines, there was a lack of engineering studies which sought to uncover the underlying causes and address them. The potential impact that advanced engineering studies can have on improving the quality of patient’s treatment keep me in renal research and is the focus of my current research.
Kidney Research UK is a vibrant community that gave me the opportunity to conduct my research and present my findings to other researchers, clinicians and, most importantly, to patients. Lastly, the MedTech Academy Programme offered by Kidney Research UK has been extremely useful and helped me gain skills to successfully translate research into the market and build a network of like-minded people in the renal community.
Authors and affiliations (if published):

Katia Nazmutdinova
Katia Nazmutdinova1,4, Cheuk Yan Man1, Philip Beales2, Karen Price1, Stephen Walsh3 and David Long1
1Kidney Development and Disease Group, Developmental Biology and Cancer Department, University College London, London, UK
2Genetics & Genomic Medicine Department, University College London, London, UK
3Department of Renal Medicine, University College London, London, UK
4Encelo Laboratories, UK
Background:
Urine has emerged as a non-invasive source of patient-specific renal cells, however reliable cell extraction has not been demonstrated yet, largely due to the short shelf life of samples and poor and inconsistent recovery rates, reported in the literature. This project is set to optimise the current centrifugation method for cell isolation and preservation from urine samples.
Methods:
We custom-built a unique benchtop filtration unit, a Cell Catcher, for processing urine samples in clinical settings, without the need for any equipment and compared its efficiency to the current gold standard, centrifugation. Sixty-three fresh urine samples were collected from healthy individuals and patients affected by rare genetic conditions, with or without kidney involvement (Bardet-Biedl Syndrome patients and various tubulopathies patients). Samples were processed either using the Cell Catcher (Clinic Edition) (Encelo, UK) within 30 minutes of collection, on-site, or transported to the lab on ice to be centrifuged within 4 hours. Urine-derived pellets were plated and maintained in culture for 4-6 weeks. Colonies were quantified manually during the second week of culturing, while RNA was extracted from cells at passage 1 for gene expression analysis.
Results:
We report that cell cultures can be established in up to 90% of samples, if they are processed in the Cell Catcher, an increase of 26%, compared to centrifugation. In addition, we report higher cell yield in Cell Catcher-processed samples, with a high degree of variation in colonies formed per sample among patients (1-200), but not in healthy controls (1-12). We have successfully expanded cells from patients and report achievable cell yields of 0.5-2.2 million within 2 weeks. Our study has confirmed that the majority of the urine-derived cells express proximal tubule cell markers.
Conclusion:
Cell Catcher standardises urine processing method and captures more viable cells from more samples, compared to conventional centrifugation method. It solves logistical issues associated with immediate access to laboratory equipment currently needed to retrieve viable cells. This advancement aims to help facilitate research in personalised medicine in the renal field and beyond, by turning biowaste into valuable patient-specific cells more efficiently.
Lay summary:
Every day, dozens of cells, originating from the kidney are shed in urine, as part of normal physiological processes. Studying these cells in a dish from patients, affected by or predisposed to renal disease, can help researchers to understand the mechanisms underlying kidney disease and identify novel diagnostic tools and personalised treatments. Current methods of cell extraction from urine samples have a low probability of success and require almost immediate processing of samples in laboratory settings, which means that patients must travel to the hospital to provide a sample and participate in research. However, an alternative method is currently under development. It is a novel point of care device Cell Catcher, which allows patients to collect their samples in the comfort of their home, and then simply mail them to the laboratory, with live cells preserved. We have demonstrated up to 90% efficiency of the beta version of Cell Catcher as part of a validation study using over 60 urine samples, as part of the feasibility study conducted by biotech start-up Encelo and UCL. By turning biowaste into valuable patient-specific cells we believe this will help advance scientific knowledge in the renal field, leading to faster diagnoses and treatments.
Biography:
Dr Katia Nazmutdinova is a geneticist with a passion to make patient-specific kidney cells more accessible for research, improving the translatability of studies. She obtained her PhD in Genetics of Rare Diseases at University College London in 2018. Katia was studying urine-derived cells of renal origin, extracted from patients affected by a rare genetic condition, Bardet-Biedl Syndrome. A major limitation of the study was a low success rate of cell extraction, reliance on rapid lab-based centrifugation and infrequent patient visits. Katia decided to develop a user-friendly pee cup that can be sent to patients, for live cells to be collected by post.
She launched the company to develop the device Cell Catcher in 2018, securing pre-seed funding from the private sector and an innovation grant in paediatrics from Kidney Research UK in 2021. The grant enabled the first functional prototype to be built and tested, in collaboration with a nephrology group led by David Long at the UCL Great Ormond Street Hospital Institute of Child Health. Turning biowaste into valuable patient-specific cells more efficiently will help advance scientific knowledge in the renal field, leading to faster diagnoses and treatments, as well as opening new routes in biobanking.

Christina Pearce
Authors and affiliations (if published):
Christina Joanne Pearce* and Natalie Hall
*Kings College, London
Background:
Depression is common within patients with chronic kidney disease (CKD) and is correlated to poorer quality of life, worse clinical outcomes and greater healthcare costs. Little is known about how depression is identified and managed in practice within the UK. Prior to the coronavirus-19 pandemic Seekles and colleagues mapped the UK renal psychosocial workforce and found that none of the renal centres met the recommended benchmark of social worker to patient ratios as set out in the 2002 workforce report and only four units met the recommended psychologist to patient ratios. The aim of this study was to assess the variability in psychosocial care for patients with CKD across UK renal centres.
Methods:
All 71 adult renal care centres across the UK were invited to take part in the online survey. The survey was comprised of three sections: 1) general kidney treatment and patient profile, to be completed by the clinical director or lead nurse, (2) psychological support, to be completed by psychological support staff, as identified in section 1, and (3) social work support, to be completed by social work staff, as identified in section 1. The survey was circulated on the 4 December 2021 and closed on the 31 April. Due to the small number descriptive statistics are predominant utilised for this analysis conducted in STATA.
Results:
Of the 71 centres invited to take part 56 centres (79%) opened the survey. Valid responses were given by 41 (58% response rate) Clinical Directors/lead nurses, 19 responses for the psychology module (where applicable) and 14 responses from the social work module (where applicable). Centres from England, Scotland, Wales and Northern Ireland responded to the survey. Of the centres that responded, acute transplant surgery was the group with the least provision for psychosocial care (n=15, 37%). Cognitive behavioural therapy is the most common psychological intervention provided (79% of respondents). The majority of respondents had a dedicated in-centre social worker (83% of respondents to the social work module). Psychologists reported that 84% of their services had formal referral pathways into their services.
Conclusion:
Less than half of all adult renal centre respondents had psychology and/or social work provision. Where the psychosocial modules were completed by relevant staff, the renal services often have dedicated social workers and a formal pathway for the identification and management of depression and/or anxiety. This survey highlights the psychosocial provision environment and areas for improvement.
Lay summary:
Depression is common within patients with chronic kidney disease (CKD) and is related to a lower quality of life and worse health outcomes. Little is known about how depression is identified in patients and managed in UK kidney centres. Before the coronavirus-19 pandemic research that mapped the UK kidney psychological and social workforce identified low levels of social workers and psychologists compared to the recommended numbers.
The aim of this study was to assess the variability in psychosocial care for patients with CKD across UK renal centres. The study survey, sent to 71 adult kidney centres across the UK, was made up of 3 sections: general questions for the medical team, one section for the social worker and one for the psychologist.
Forty-one centres (58% of the UK centres) responded to the survey with usable information. Responses came from centres from all countries in the UK. Psychologists reported cognitive behavioural therapy to be the most common talk therapy for depression (79%) and that formal referral routes were available in 84% of centres with psychological care providers. Less than half of all adult kidney centres who responded to our survey had psychology and/or social workers as part of their team.
Biography:
Christina is a chartered psychologist who joined King’s College London in 2020 as a postdoctoral research associate in the health psychology section. Her current research focuses on evaluating services for the identification and management of depression in people with chronic kidney disease across the UK.
Christina gained her PhD from University College London, Centre for Behavioural Medicine in 2020 which explored the reasons for and patterns of non-adherence to inhaled medication in paediatric patients with severe asthma. Christina’s research focuses on living with long-term conditions in terms of health behaviour, self-management and accompanying mental health issues. Christina’s work in health psychology has spanned several conditions including medically unexplained symptoms, psoriasis, psoriatic arthritis, cardiovascular disease, asthma, polypharmacy, and chronic kidney disease.

Monica Gamez
Authors and affiliations (if published):
Monica Gamez1, Hesham El Hegni1, Sarah Fawaz1, Matthew Butler1, Elizabeth Wasson1, Michael Crompton1, Raina Ramnath1, Yan Qiu1, Jerry Turnbull2, Olga Zubkova3, Gavin Welsh1, Simon Satchell1, Rebecca Foster.1
1Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Whitson St, Bristol, UK. BS1 4NE. Corresponding author: [email protected]
2Department of Biochemistry, University of Liverpool, Liverpool, UK. L69 3BX
3Ferrier Research Institute, 69 Gracefield Rd, Lower Hutt, Victoria University of Wellington, New Zealand.
Background:
An estimated 647 million people worldwide will have diabetes mellitus (DM) by 2040, which causes life altering microvascular complications, such as diabetic nephropathy (DN). The endothelial glycocalyx (eGlx) is a protective layer that lines the luminal side of blood vessels and contains proteoglycans (core proteins with glycosaminoglycan (GAG) sidechains) that help maintain vascular permeability, and are damaged during DM. Heparanase degrades the GAG, heparan sulphate (HS), and is upregulated in DN. Heparanase inhibition and knock-down both prevent the development of DN. The objective of this study was to show that HS, specifically within the endothelial glycocalyx, is important in glomerular barrier function and that prevention of its shedding, by a novel class of heparanase inhibitor (HI), is protective in DM.
Methods:
Endothelial glycocalyx HS was removed in mice by intravenous injection of heparinase III, or by knock-down of Ext-1 (an HS biosynthesis enzyme) in endothelial cells using Tie2rtTA, tet-O-Cre, Ext-1fl/fl (Ext-1fl/fl) mice. A mouse model of type 2 diabetes (db/db) was used whereby HI or vehicle was given, i.p. daily, from 9-11wk of age. Mice were ringer perfused for glomerular permeability studies or Alcian blue/ glutaraldehyde perfused for electron microscopy (EM). Glomeruli were isolated from ringer perfused kidneys and apparent albumin permeability was measured in single capillaries from individual glomeruli. Alcian blue perfused kidneys were processed for EM, imaged, and eGlx depth and percent coverage were measured using ImageJ. Urine albumin creatinine ratios (uACR) were measured at endpoint.
Results:
A significant reduction in glomerular endothelial glycocalyx depth and coverage was seen with heparinase III treatment and Ext-1fl/fl mice. These were associated with significant increases in glomerular albumin permeability. In diabetic mice, eGlx depth and coverage was significantly reduced and uACR was significantly increased. Diabetic mice treated with HI no longer had a significant increase in uACR and eGlx depth/coverage and glomerular albumin permeability was significantly restored when HI was given.
Conclusion:
We confirm that endothelial glycocalyx HS plays a direct role in the glomerular filtration barrier demonstrated by targeted HS removal. We also demonstrate that heparanase inhibition in DM, using a novel and clinically relevant inhibitor, directly enhances the glomerular endothelial glycocalyx, resulting in normalised glomerular albumin permeability.
Lay summary:
The glycocalyx is a sugar layer which lines all of the blood vessels in the body. In diabetes, this layer is damaged and is associated with blood vessel leakiness. In the kidney, this leads to diabetic kidney disease. Heparan sulphate is one of the main types of sugar in this layer, and it has been shown that heparanase, a protein which is able to break down heparan sulphate, is increased in diabetes. In two mouse models of heparan sulphate loss from this layer, we found that there was increased blood vessel leakiness in the kidney. This suggests that heparan sulphate plays an important role in the glycocalyx. We then aimed to protect heparan sulphate in the glycocalyx in diabetic mice, using a new drug to block heparanase. We found that protection of the glycocalyx through use of this drug resulted in protected blood vessels and prevention of diabetic kidney disease. This work is evidence that heparan sulphate is an important component of the glycocalyx, and that preventing loss of this sugar molecule in diabetes can help to prevent the development of diabetic kidney disease.
Biography:
My initial attraction to renal research began in the first lab I worked in as an undergraduate at the University of Michigan. Our work focused on BK polyomavirus, a ubiquitous virus which can result in polyomavirus-associated nephropathy in kidney transplant patients, increasing chances of transplant loss. I later worked as a technician in the Bristol Renal group at the University of Bristol. Here, I went on to complete my PhD focusing on therapeutically targeting the glycocalyx in the kidney, an important layer in the vasculature. I came to fully appreciate how fascinating the kidney is, and just how many people are impacted by kidney disease globally. This urgent need for renal research has kept me interested. I aim to accelerate research by understanding mechanistically how to prevent vascular dysfunction in kidney disease and applying this to other vascular beds. Ultimately, our goal is to prevent progression of kidney disease and related cardiovascular complications for patients. Thanks to Kidney Research UK, who has funded much of the work in Bristol Renal, we are able to carry out important research which will go on to help kidney patients. Without organisations like Kidney Research UK, advancements in kidney treatments would not be possible.

Eoin O'Sullivan
Authors and affiliations (if published):
Eoin D O’Sullivan. David A Ferenbach.
University of Edinburgh
Background:
Progressive fibrosis and maladaptive organ repair results in significant morbidity and millions of premature deaths annually. Senescent cells accumulate with ageing and after injury and are implicated in organ fibrosis, but the mechanisms by which senescence influences repair are poorly understood.
Methods:
Two models of murine renal injury were analysed using bulk and single cell RNA sequencing to assess pathways augmented in senescent cells and the impact of the senolytic agent ABT263. Further analysis explored conserved pathways present in senescent epithelia across mice, humans, and other organ systems. Candidate molecules were identified and validated using in vitro and in vivo models of fibrotic disease.
Results:
We show that obstructive injury generates senescent epithelia which persist after resolution of the original injury, promote ongoing fibrosis and impede adaptive repair. Depletion of senescent cells with ABT263 reduces fibrosis in reversed ureteric obstruction and after renal ischaemia-reperfusion injury. We validate these findings in humans, showing that senescence and fibrosis persist after relieved renal obstruction. We next characterise senescent epithelia in murine renal injury using single cell RNA-Seq. We extend our classification to human kidney and liver disease and identify conserved pro-fibrotic proteins which we validate in vitro and in human disease. We demonstrate that one such molecule, Protein Disulfide Isomerase Family A Member 3 (PDIA3), is essential for TGF-beta mediated fibroblast activation. Inhibition of PDIA3 in vivo significantly reduces kidney fibrosis during ongoing renal injury.
Conclusion:
Inhibition of PDIA3 represents a new potential therapeutic pathway to reduce organ fibrosis after injury. Analysis of the signalling pathways of senescent epithelia connects senescence to organ fibrosis, permitting rational design of anti-fibrotic therapies.
Lay summary:
Our understanding of why injured and aged kidneys are more susceptible to developing progressive scarring (fibrosis) and working less well is incomplete.
In this project we used the latest advances in cell analysis to look for novel pathways which are switched on in the injured kidney – seeking ways to treat these pathways to prevent fibrosis.
We uncovered a range of candidate ‘bad molecules’ – and narrowed our list by looking in mice, in humans, in kidneys and in injured livers. We identified a molecule called PDIA3 which was raised in all the settings described – and in human kidney disease. When we explored how PDIA3 works we discovered that it is an important driver of kidney fibrosis and that an experimental drug can block its activity and protect the aged and injured kidney.
This work opens up a new avenue to prevent kidney scarring and we are currently working in collaboration with academic and pharmaceutical teams to design novel agents which can be used to move from mouse studies back to human disease.
Biography:
Kidney diseases tend to be complex and nuanced and people living with kidney disease often have several interacting medical issues while they live with their conditions for their entire lives. These challenges mean that providing care and performing research is both intellectually rewarding but also highly impactful as you can potentially greatly enhance the quality and length of people’s lives.
Our group has identified new molecules which can lead to renal scarring. These molecules likely drive renal fibrosis and cause progression of CKD/scarring after AKI. We have shown these pathways can be targeted successfully in mice, reducing scarring in their kidneys after injury. This could be extremely useful to patients after AKI to protect their kidneys.
Kidney Research UK has been central to my research. The charity supported my scientific training, funded and guided the research, and built a network of passionate researchers and advocates which has allowed me to build new collaborations, enhancing research quality and generating new ideas and partnerships.

Lucia Marinas del Rey
Authors and affiliations (if published):
Lucia Marinas del Rey1,2,3, Krista Rombouts3, Giuseppe Mazza3,4, Jill Norman2, Reza Motallebzadeh1
1UCL Division of Medicine and Interventional Science
2UCL Department of Renal Medicine
3UCL Institute for Liver and Digestive Health
4Engitix Ltd
Background:
Chronic kidney disease (CKD), which describes an irreversible alteration of kidney function, represents a significant worldwide healthcare burden. It can result from many different aetiologies and is one of the leading causes of mortality worldwide. Irrespective of the cause, a common feature of CKD is fibrosis in the tubulointerstitium, where persistent injury causes glomerulosclerosis, tubular atrophy, and fibrosis. The lack of effective antifibrotic therapies to stop the progression of CKD evidences the need for a better understanding of tubulointerstitial fibrosis.
Methods:
Tissue from normal human kidney cortex (from deceased donors) and fibrotic human kidney cortex (from explants of kidney transplants with chronic rejection) was dissected into cubes and decellularised with sodium dodecyl sulphate. Decellularised cubes were assessed by DNA content (DNeasy Kit, Qiagen), histology (haematoxylin and eosin (H&E) and picro-sirius red (PSR)) and immunohistochemistry. To develop human kidney extracellular matrix (hkECM) hydrogels, decellularised cubes were homogenised and lyophilised into a fibrous powder, which was then dissolved in 0.01M HCl to a concentration of 13 mg/ml and digested with 1 mg/ml pepsin (Sigma). hkECM (3, 4.5, and 6 mg/ml) was mixed with a nanocellulose-based material (30%) and human proximal tubular epithelial cells (HK-2 cell line) and cultured for up to 21 days.
Results:
Kidney tissue was successfully decellularised as shown by DNA content reduction of 97% after decellularisation. Additionally, histological analysis confirmed the elimination of nuclei by H&E staining, and lack of cellular material (yellow in PSR staining); and preservation of collagen (red; PSR). Immunohistochemistry confirmed maintenance of key ECM proteins (collagens I and IV, fibronectin, laminin α5). By decellularisation of normal kidney cortical tissue, we were able to produce a specific biomaterial derived from the tubulointerstitial extracellular matrix (hkECM). Combination of the hkECM with nanocellulose produced stable hydrogels which supported growth and differentiation of HK-2 cells. Additionally, a method for generating hydrogels from fibrotic tissue is being developed in the same manner to create models that reproduce fibrotic tissue.
Conclusion:
Using combinations of different renal cell types and normal and fibrotic ECM to create kidney organoids, this system has the potential to recapitulate the biomechanical and biochemical properties of the tissue and the cell-ECM interactions. Comparison of the normal and fibrotic organoids will provide insights into the role of the ECM in regulating cell differentiation and function, model tubulointerstitial fibrosis, and potentially establish a platform for drug testing.
Lay summary:
Chronic kidney disease (CKD), which describes an irreversible alteration of kidney function, represents a significant worldwide healthcare burden, being one of the leading causes of mortality worldwide. Irrespective of the cause, a common feature of CKD is fibrosis in the tubulointerstitium, where persistent injury causes glomerulosclerosis, tubular atrophy, and fibrosis. The lack of effective antifibrotic therapies to stop the progression of CKD evidences the need for a better understanding of tubulointerstitial fibrosis.
The aim is to use tissue engineering to develop a 3D hydrogel model of the tubulointerstitium and study renal fibrosis. By decellularisation of normal and fibrotic kidney cortical tissue, we produced a specific biomaterial derived from the tubulointerstitial extracellular matrix (ECM) - hkECM. Combination of the hkECM with nanocellulose generated stable hydrogels which supported growth and differentiation of human proximal tubular epithelial cells. Combining different renal cell types to create kidney organoids, this system can recapitulate the biomechanical and biochemical properties of the tissue and the cell-ECM interactions. Comparison of the normal and fibrotic organoids will provide insights into the role of the ECM in regulating cell differentiation and function, model fibrosis, and potentially establish a platform for drug testing.
Biography:
I am dedicating my PhD at UCL to developing models of the kidney tubulointerstitium in normal and fibrotic conditions. Having graduated in biomedical engineering and done an MSc in regenerative medicine, working in tissue engineering has always been my goal. This is what drove me to move to London from Madrid, to be a part of the exciting research that organisations such as Kidney Research UK and UCL are involved in. In my two years of PhD, I have developed an understanding and appreciation for this field.
I am attracted to the complexity of the kidney and kidney disease, and its many implications for our overall health. Since the beginning, I was very surprised that there was no effective cure, and I believe my work in establishing accurate in-vitro kidney models will enable future progress in the field. I hope that joining Kidney Research UK will give me the chance to meet like-minded people working in kidney research and establishing collaborations to contribute to the field.

Holly Stowell-Connolly
Authors and affiliations (if published):
Holly Stowell-Connolly1, Abigail C. Lay1,2, Jennifer A Hurcombe1, Mark Graham1, and Richard J. Coward1
1Bristol Renal, University of Bristol
2Division of Cardiovascular Sciences, University of Manchester
Background:
Glomerular podocytes are structurally, morphologically, and biochemically similar to neurons, despite different bodily and organ location and germ line origin. Additionally, diseases causing neuronal degeneration show associations with chronic kidney disease (CKD), as evidenced by a three-fold increase in the likelihood of developing dementia in patients with renal failure (RF). Due to such associations, we hypothesise common mechanistic pathways contributing to disease progression in both cell types, specifically in diabetes. This work aims to understand differential podocyte expression and activation of TBK1, a protein abnormally deactivated in neurons in Amyotrophic Lateral Sclerosis (ALS, aka Motor Neuron Disease).
Methods:
Transcriptomic and proteomic analysis determined differential renal expression of genes known to be involved in neuronal degeneration, specifically in conditions related to diabetes. Conditionally immortalized human podocytes (cihPods) were grown in basal or diabetic conditions (DC) (physiologically high concentrations of insulin, glucose, TNF-α, and IL-6), and validation of aberrant expression of TBK1 was confirmed by qRT-PCR, western blotting (WB), and immunofluorescence (IF). Importance of podocyte TBK1 expression and signalling was investigated by generating a constitutive TBK1 knock-down (KD) in using lentiviral transduction of shRNA. KD was confirmed by qRT-PCR and IF. Cell line characterization was achieved via WB and scratch assays.
Results:
Transcript- and proteomic analysis determined TBK1 mRNA and protein expression to be increased in human and mouse podocytes grown in DC (p<0.05). Expression is unchanged in glomerular endothelial or proximal tubular cells. IF demonstrates TBK1 expression is upregulated in DC, as well as activation indicated by at S172 phosphorylation. pTBK1 S172 translocates to the nucleus where it colocalizes with SUMO and PML, suggesting sequestration at PML-nuclear bodies in response to an insulin- and IL-6-rich environment. KD of TBK1 within cihPods results in aberrant cell growth, as demonstrated by increased cellular and nuclear area, and abnormal actin architecture. Early work indicates this is not a result of re-entry of the cell cycle. Current work will determine a role of Hippo signalling and initiation of cell death mechanisms. Additional early work indicates increased cell motility in KD cells. Current work aims to understand the response of cihPods to DC with a TBK1 KD.
Conclusion:
Proteins involved in neuronal degeneration can be abnormally regulated in cihPods in DC. TBK1 expression and activation is upregulated in cihPods grown in DC, with a novel nuclear sequestration in podocyte nuclei. TBK1 KD causes aberrant actin fibre growth, leading to increases in cellular and nuclear area.
Lay summary:
Neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and Motor Neuron Disease (ALS), occur when a specific set of cells within the brain, neurons, become injured and eventually die. Patients with dementias including Alzheimer’s disease often develop other diseases, including diabetes and chronic kidney disease (CKD). Interestingly, patients with dementia are three times as likely as healthy individuals to also develop CKD. The causes of this association are currently unknown. Injury to neurons is often a response to abnormal regulation of specific proteins. This work aims to understand how proteins involved in such injury are regulated within a type of kidney cell, the podocyte, and how diabetes can affect its regulation and function. We demonstrate podocyte presence of TBK1, known to be deactivated in some ALS patients, and show increased expression and activation in diabetes. We aim to assess the importance of TBK1 in kidney health, by removing TBK1 from podocytes and measuring changes in cell growth and function. Understanding how proteins involved in both neuron and podocyte injury may lead to the repurposing of current treatments, or the development of new therapies for both neurodegenerative and CKD patients.
Biography:
I first found myself interested in the renal field whilst studying the kidney during my A-Levels and my undergraduate degree, and so when applying for a laboratory experience it was my first choice! During this time, my interest in the field grew further, initiating my desire to continue my renal research with generous funding from Kidney Research UK. My current work is quite novel, and I hope that it can provide advances in patient treatments for, not just kidney disease, but also neurodegenerative diseases. Not only has Kidney Research UK facilitated this research, but their funding has also allowed me to continue in a field I greatly enjoy.

David Ferenbach
Authors and affiliations (if published):
David Ferenbach
Background:
Progressive fibrosis is a feature of aging and chronic tissue injury in multiple organs including the kidney and heart. Gli1+ mesenchymal stem cells are a major source of activated fibroblasts in multiple organs but the links between injury, inflammation and Gli1+ stem cell expansion and tissue fibrosis remain incompletely understood. Here we show that leukocyte derived tumour necrosis factor alpha (TNFa) promotes Gli1+ stem cell proliferation and cardio-renal fibrosis via downstream induction and release of Indian Hedgehog (IHH) from renal epithelia.
Methods:
Single cell RNA sequencing was performed on healthy young, healthy aged and young fibrotic kidneys from mice. Pharmacological and conditional genetic blockade was used to determine the key molecules generated from injured and aged kidneys, and their impact on cardiac and renal fibrosis assessed in experimental mice and in human kidney disease.
Results:
Using single cell RNA sequencing of aged and fibrotic kidneys, we identify an ‘inflammatory’ proximal tubular epithelia (iPT) population expressing high levels of ubiquitin D (UBD) responsible for TNFa/NFkB induced IHH production in vivo. TNFa induces UBD expression in human proximal tubular cells in vitro and in vivo in experimental murine and human renal disease and ageing. Ubd+ iPT derived IHH activates canonical Hedgehog signalling in Gli1+ progenitors, inducing their activation and proliferation with resultant fibrosis in the injured and ageing kidney and heart. Of note, this occurs independent of blood pressure and renal functional change and is inhibited in mice by selective genetic Ihh deletion from Pax8 expressing renal epithelia, and by pharmacological blockade of TNFa, NFkB, Hedgehog or Gli signalling. Increased levels of circulating IHH associate with more rapid loss of renal function and higher rates of cardiovascular disease in patients with chronic kidney disease.
Conclusion:
Indian Hedgehog is the upstream ligand connecting leukocyte activation to Gli1+ mesenchymal stem cell expansion and fibrosis. These studies provide a mechanistic basis for translational studies in humans to inhibit the early components of signalling (via TNFa inhibitors), and for the design of novel agents to selectively block the fibrotic effects of IHH upon the kidney and heart.
Lay summary:
Our understanding of why injured and aged kidneys are more susceptible to developing progressive scarring (fibrosis) and why kidney disease promotes heart problems remains poor.
In this project we use the latest advances in cell analysis to identify a novel sub-type of kidney cell present in injured and aged kidneys. We show that these cells are the result of activation by white blood cells within the kidney (as is known to occur after injury and ageing). Of key importance – these cells also release a molecule called Indian Hedgehog (IHH) which is an important driver of fibrosis in the kidney and heart. Testing human samples from another Kidney Research UK funded study, we show that human patients with higher levels of IHH develop further kidney scarring and cardiac problems more rapidly than those with low levels. By blocking the release of this molecule, we demonstrate that we can protect against these processes.
On the basis of this work (and published evidence from other conditions that blocking TNFa is safe and helps prevent deteriorating kidney function) we propose that human trials should be undertaken to test the effects of TNFa blockers in chronic kidney disease (to prevent kidney and cardiac disease) and in dialysis patients (seeking to prevent cardiac problems).
Biography:
David Ferenbach graduated in medicine from the University of Edinburgh and, after training posts in Edinburgh and Glasgow, completed his PhD in the Centre for Inflammation Research in Edinburgh. Here he developed a research interest in the mechanisms driving accelerated fibrosis in injured and aged kidneys funded by a clinical training fellowship from Kidney Research UK. He pursued this goal further during his Wellcome Trust intermediate fellowship in the Bonventre Laboratory of Harvard Medical School. Today, his laboratory in Edinburgh is focused on understanding the role played by senescent epithelia in driving kidney scarring – with the aim of developing new treatments for human fibrotic disease.
As a clinically practicing nephrologist I am constantly reminded of both the huge advances we have made in treating kidney disease – but also by the major problems that remain unsolved:
- Why are elderly people at increased risk of acute kidney injury and longer-term chronic kidney disease?
- How can we prevent kidney fibrosis from progressing?
- How could we best prevent our patients from suffering from hugely increased rates of cardiovascular disease and death?
My laboratory remains focused on advancing our understanding of the questions above, with the goal of translating this to novel treatments for our patients.
Over the past two years we have published exciting work delineating how previous kidney injury and ageing leads to dysfunctional, “senescent” cells within the kidney (with two papers published/in press in Science Translational Medicine and another in JCI Insight). This work revealed novel targets for anti-fibrotic therapies – which we have recently reached agreement with pharmaceutical companies to generate and validate novel therapies designed to preserve kidney function and prevent fibrosis in the aftermath of injury.
Without the backing of Kidney Research UK I would not be in renal research today - the charity has support me throughout the last 15+ years: at the stage of my PhD, awarding my lab its first project grant and now, with two additional innovation awards. I am immensely grateful to the entire Kidney Research UK team for their support.

Serena MacMillan, PhD student at The University of Cambridge
Authors and affiliations (if published):
Serena MacMillan, Sarah A Hosgood, Michael Nicholson.
Department of Surgery, University of Cambridge, UK.
This work has been accepted for publication as a Short Report for the British Journal of Surgery.
Background:
A major restriction to transplantation is the requirement for ABO blood group compatibility between donor and recipient. In this study, we have used an a-galactosidase from Bacteroides fragilis to enzymatically remove type B blood group antigens from human kidneys using ex vivo normothermic machine perfusion (NMP) to overcome the ABO barrier in kidney transplantation.
Methods:
Human donor kidneys rejected for transplantation and offered for research were used in this study with approval by the National Research Ethics committee and Research and Development office (NRES: 15/NE/0408). An a-galactosidase (GH110B) from B. fragilis was used to remove type B blood group antigens from fixed tissue from five type B donor kidneys, quantified with immunofluorescence staining of blood group B and H antigens on the vascular endothelium. Three further human kidneys of blood group B then underwent five hours acellular NMP supplemented with 2.5mg/ml of GH110B alongside one untreated control kidney and serial biopsies were stained and quantified for antigen removal.
Results:
In all biopsies examined, B antigen expression was restricted to peritubular capillaries. In fixed tissue, GH110B removed up to 99% of endothelial blood group B antigens after 1hr of incubation. In three whole kidneys, between 67-94% of B antigens were removed after 5hrs of acellular NMP.
Conclusion:
For the first time, we have shown that blood group antigens can be successfully removed from whole human kidneys using NMP. This proof-of-principle work paves the way for future development and clinical application to increase the number of ABO-compatible grafts in transplantation.
Lay summary:
All people have one of four bloods groups: O, A, B or AB and this is called their ABO blood type. Kidney transplants cannot be performed unless the donor and the recipient have compatible ABO types. If the blood groups don’t match, then the kidney transplant can be rejected immediately. Importantly, anyone can receive a kidney that is blood type O and is therefore called the universal donor. Unfortunately, less than half of all donated kidneys are type O.
We are researching a new way of changing the blood group of a kidney. Blood type is determined by A or B markers that line kidney blood vessels. Blood group O kidneys do not have either A or B markers or ‘antigens’. Enzymes that work as ‘biological scissors’ can remove A and B antigens from cell surfaces, effectively making cells blood group O. We have now applied this idea to human kidneys that have been turned down for transplantation and donated for research. In this work, we have removed between 67-94% of blood group B antigens from three blood group B kidneys. This research could transform the way that kidney transplantation is delivered throughout the world.
Biography:
When deciding to pursue a PhD, I was looking for research that was translational and had close ties with clinical work. Everything fell into place when I discovered the amazing work being undertaken in kidney transplantation research. Being able to feel the momentum behind life-changing renal research drives my work and provides real satisfaction in my day-to-day lab work on blood group conversion of human kidneys.
My work is at an exciting stage now we have shown the first proof-of-principle work of converting human kidneys from type A or B blood groups to a universally transplantable type. The research has the chance to make a real change to organ allocation for kidney transplantation and to know that it may one day improve outcomes for those on the waiting list is an amazing boost.
Kidney Research UK has funded my PhD and so I am incredibly grateful to the charity for giving me the opportunityy to pursue this work. The charity has also provided amazing exposure for the research and the field of kidney transplantation, which is so critical for public engagement with renal research.

Joyce Popoola
Authors and affiliations (if published):
Joyce Popoola1, Suhaylah Bauhadoor1, Jonathan Bartley2
1Consultant Nephrologist, Young Adult Worker Department of Nephrology and Transplantation, St George’s University Hospitals NHS Foundation Trust, London
2Patient Support and Advocacy Worker Kidney Care UK
Background:
The population of young adults (16-29 years) with renal disease is increasing partly due to a rising number transferring from the paediatric units and increased life expectancy. Unfortunately, they are also at risk of significant co-morbidity and early mortality compared to their contemporaries. Young adults often struggle to understand their conditions and adherence is most challenging in this age group. We have carried out local surveys exploring the needs of young adults from their perspective. Overwhelmingly they highlight; lack of peer support and information on their condition not presented in a readily accessible generation friendly format.
Methods and results:
We are developing a bespoke tool “My Nephros” for young adults on renal disease and transplantation providing targeted succinct answers to their healthcare queries. Responses to their most, frequently asked questions (FAQs) are presented using interactive modern technology in a contemporary fashion. Short videos and audios as sound bites are being used for responses. This is being achieved with the direct input of representatives from our young adult population, young adult worker, health advisor and psychologist. The responses are scientifically sound and formulated in a Millennial and Generation Z accessible fashion.
Conclusion:
Patients and carers nowadays are keen to do their own research in relation to matters related to their healthcare. The usual place for the young adults to do this is via internet searches and social media platforms. More time and motivation may be required to stimulate changes in patient behaviours related to educational opportunities. We however present a tool with potential to be used in helping patients’ values guide healthcare acceptance.
Lay summary:
Here we present the use of a tool for Young Adults, “My Nephros” to help them learn more about their kidney disease or transplant. It is based on commonly asked questions by young adults in relation to understanding and managing their condition and will be accessible on the internet via search engines and social media.
Empowering young adults helping them understand their condition may help improve their self-care.
Biography:
Joyce Popoola is a renal consultant at St George’s University Hospital NHS Foundation Trust, an honorary senior lecturer and clinical subdean, St George’s University London.
Renal medicine is a relatively new specialty, and yet affects at least 10% of the world’s population including young people. In the absence of targeted intervention this can only get worse resulting in devastation of individuals, families, and communities. Self-actualisation and global economies are particularly impacted when the youth are affected.
Laboratory, clinical translational and epidemiological studies form the bulk of research. Joyce’s team are additionally interested in researching the development and embedding of relevant trusted decision-making educational tools. She believes these are essential to maximising and ensuring lasting research footprints, particularly among younger patients who tend to ‘’do their own research’’.
Joyce had interest in innovative practice from her early years in training. Kidney Research UK gave an amazing head start by awarding her a fellowship enabling research into transplantation. This opened a new world to her in research, education and cutting-edge clinical care in nephrology and transplantation. An international transplant fellowship enabled post-doctoral studies in Boston. She returned to the UK to take up the role of lead transplant physician and later of young adult care.
Abstract poster presentations:

Belinda Ameyaw
Authors and affiliations (if published):
Belinda Ameyaw, Phalguni Rath, John A. Todd, Katherine Bull
Wellcome Centre for Human Genetics, University of Oxford
Background:
Oxidative stress is thought to be a central component in the pathogenesis of DKD. Hyperphosphorylation of Tau and aggregation into neurofibrillary tangles is a contributor to Alzheimer’s disease and frontotemporal dementia. Tau has also been identified as a downstream effector of the antioxidant transcription factor NRF2 suggesting a role in oxidative stress response. Kidney biopsies from DKD patients show an increase in Tau compared to healthy controls but its role in renal disease is unknown.
Methods:
Using a dual guide CRISPR-CAS9 approach, Tau’s gene, MAPT, is first knocked out in the neuroblastoma cell line, SH-SY5Y. Flow cytometry is used to quantify basal and induced oxidative stress in these cells relative to wildtype and an existing cell line edited only in exon 1. A rescue with a Tau plasmid into the knockouts is assessed for effect on oxidative stress phenotype. Mass Spectrometry proteomics identifies differentially expressed proteins between the wildtype and knockout SH-SY5Y cells. Kidney organoids generated from hiPSCs are validated for expression of key kidney markers and Tau, before the dual guide approach is employed to knockout Tau in organoids.
Results:
In two clones obtained from the dual guide method, neither N nor C-terminal antibodies detect Tau protein on western blots in contrast to a cell line edited within exon 1 alone. MAPT-/- cells show increased basal and induced oxidative stress with decreased cell viability in response to stress induction. NRF2 and KEAP1 expression increase in knockouts alongside NRF2 effectors HMOX1 and NQO1. The stress phenotype is rescued by MAPT overexpression but not by Taxol treatment nor CDDO-imidazole activation, indicating a mechanism independent of tubulin binding or the NRF2 pathway. The proteomics data confirms this and indicates altered RNA binding and splicing in the absence of Tau. Kidney organoids express Tau in the proximal tubules, mimicking human kidney, and the dual guide approach deletes a 22 kb internal section of MAPT in hiPSCs.
Conclusion:
Multiexonal targeting of MAPT generates a robust new knockout in SH-SY5Y cells. The absence of Tau increases basal and induced oxidative stress with a concomitant induction of the NRF2-KEAP1 pathway. This work confirms a role for Tau in oxidative stress, for the first time in human cells, and indicates that rather than acting via Tau’s known tubulin binding function, this may be mediated via disruptions to the RNA spliceosome. Kidney organoids from wildtype and MAPT-/- hiPSCs provide new tools to investigate the role of Tau in DKD.
Lay summary:
Tau protein usually keeps cells working effectively but disruptions in their structure can occur leading to cell death in the brain. This has been observed in Alzheimer’s disease patients and partially explains symptoms such as memory loss. Here, we investigated the effect of completely removing this protein from a cell line similar to the ones found in the brain and observed the cells showing signs of stress. This effect is reversed when the Tau protein is brought back into the cells. Since Tau is also found in the kidney and has been shown to increase in people with diabetic kidney disease, we generated ‘mini kidney’ tissues in a dish and removed the Tau. This will allow us to test whether Tau has the same role in preventing stress in the kidney and how that relates to known indicators of diabetic damage.
Biography:
I am a final year DPhil student at the University of Oxford researching the role of oxidative stress in diabetic kidney disease. My interest in renal research began with a curiosity about how a diabetic milieu induces an oxidative stress environment and how this alters the renal structure to cause kidney disease. I believe that understanding the mechanism of disease development, especially during the very early stages, will enable us to identify and repurpose drugs to reduce the occurrence of end stage renal disease. Kidney Research UK provides a much-needed network of individuals with whom research ideas can be exchanged, provides opportunities to showcase research and financial support to aid the work I do.

David Baird
Authors and affiliations (if published):
David P. Baird, Maximilian Reck, Ross Campbell, Eoin D. O’Sullivan, Marie-Helena Docherty, Carolynn Cairns, Cyril Carvalho, Matthieu Vermeren, Jeremy Hughes, Laura Denby, Bryan Conway, Katie J. Mylonas, David A. Ferenbach.
Background:
Renal tubular senescence in response to ageing and injury is proposed as a key driver of kidney fibrosis. Senescent cell depletion in mice improves outcomes in multiple organs including the kidney. There are currently no non-invasive biomarkers for quantifying renal senescence available. A multi-omics approach will be used, where human renal proximal tubular epithelial cells(hRPTECs) in culture, and a murine model of renal senescence and human samples will be utilised, to identify urinary biomarkers of renal tubular senescence.
Methods:
A model has been optimised to induce senescence in hRPTECs in culture using irradiation and treatment with MDM2-antagonist Nutlin 3A. Transcriptomic studies using bulk RNAseq were performed to identify genes that are differentially expressed in senescent cells compared to proliferating controls. Total body irradiation (TBI) induces accelerated senescence in mice; LC-MS proteomic studies were performed on urine from mice comparing healthy controls with mice exposed to TBI with or without subsequent senolytic therapy. Additionally, tubular senescence has been quantified histologically in human biopsy samples from 55 patients with chronic kidney disease using a triple immunofluorescent stain, which included p21CIP1, Ki67 and tubular markers CD10 and CKPAN.
Results:
Irradiated and Nutlin 3A treated cells both had an increase in CDKN1A and a fall in LMNB1 and MKI67 in keeping with senescence induction. Other factors including CXCL8, TNF and IL6 rose in irradiated cells compared to controls but fell in Nutlin 3A treated cells. 1272 genes were differentially expressed in irradiated cells compared to controls; 760 of these genes were differentially expressed in the same direction in Nutlin 3A treated samples. Murine LC-MS studies identified 15 proteins that fell in mice exposed to TBI compared to healthy controls but reverted towards baseline with senolytic treatment. By combining these datasets with publicly available data, several candidate biomarkers of senescence have been identified (not named due to pending patent applications). Renal tubular senescence in human kidney biopsies correlates with eGFR and age (both p < 0.01). Further LC-MS and ELISA studies in matched urine samples are planned that will allow us to determine which molecules most closely correlate with senescence histologically in the human kidney.
Conclusion:
We have identified several candidate urinary biomarkers of senescence; ongoing studies will determine which molecules correlate with renal senescence histologically. Further planned studies in a cohort of >380 patients with >4 years follow-up will determine if the most promising biomarkers predict renal outcomes.
Lay summary:
Senescence is the term used to describe cells that have stopped functioning and regenerating normally. Senescent cells accumulate in aged and injured kidneys, and drive scarring and loss of function. In recent studies, giving medications to mice that kill senescent cells (known as senolytic medications) improved the function of multiple organs including the kidneys. At present, the only methods available for measuring senescence in the kidney require a kidney biopsy.
Several approaches have been used to find potential biomarkers of kidney senescence, that is molecules that are either increased or decreased in senescent cells in the kidney compared to healthy cells and that can be detected in urine. This has included using human kidney cells grown in the lab and experiments using mice treated with senolytic medications. Senescence in kidney biopsies from 55 patients with kidney disease has also been measured and currently, urine collected at the same time as those biopsies is being used to determine which urinary senescence biomarkers most closely match the amount of senescence found in the biopsy. Urine samples from 380 patients will then be used to see if the most promising biomarkers can predict which patients develop a more rapid decline in kidney function.
Biography:
I am a clinical research training fellow at the University of Edinburgh. Through my clinical work as a renal registrar, I have seen first-hand the incredible burden kidney disease can have on patients and their families, but also the remarkable potential there is to improve patient care through scientific innovations. My research is focused on finding urinary biomarkers of kidney senescence, which could be used to identify patients that are at higher risk of kidney disease progression and who might benefit from therapies targeting senescent cells. Through an innovation grant, Kidney Research UK funded the initial bulk RNA sequencing studies of in vitro samples in my project, comparing senescent and non-senescent cells. I have since been awarded an MRC fellowship, building on this work, and moving toward human translation.

Andrew Chetwynd
Authors and affiliations (if published):
Andrew J. Chetwynd, Julien Marro, Jenny Hawkes, Sarah Northey, Louise Oni
Background:
Chronic kidney disease is the only long-term side-effect associated with immunoglobulin A Vasculitis (IgAV). IgAV is the most common form of vasculitis in children and is typically self-limiting however, 1-2 % of children will develop end stage renal disease requiring renal replacement therapy. In order to improve renal outcomes, methods to predict or prevent IgAV-nephritis (IgAVN) such as earlier diagnostics biomarkers and therapeutic targets are required. In this work we investigate urinary complement factor B and D and MASP-1 and the renin-angiotensinogen-aldosterone-system (RAAS), known triggers of renal inflammation for children with IgAV.
Methods:
IgAV patients and healthy controls (HC) were recruited into the IgA Vasculitis (IgAV) study at Alder Hey Children’s Hospital (REC 17/NE/0390). Children with IgAV were further classified based upon renal inflammation into IgAVN and IgAV without nephritis (IgAVwoN). The complement branch of this work incorporated 49 patients: IgAVN n=14, IgAVwoN n=20 and HC n=15. The RAAS aspect 40 patients were incorporated IgAVN: 9, IgAVwoN: 19 and HC 14. Commercial ELISA kits were purchased for all analytes. All samples were normalized the urinary creatine concentration.
Results:
Urinary MASP-1 was significantly elevated in the IgAVN group compared to HC (p<0.05) the same was also the case for complement factor B. Interestingly for complement factor D the IgAVN patients returned significantly greater concentrations compared to IgAVwoN (p<0.01) and HC (p<0.01).
Urinary renin concentrations had no significant differences between HC and IgAV patients or between patients separated by renal inflammation. Urinary angiotensinogen was significantly elevated in IgAV patients compared to HC (p=0.0015). When differentiated by renal inflammation IgAVN was significantly elevated compared to HC (p=0.0006) and IgAVwoN (p=0.0492). IgAVwoN concentrations were also significantly elevated relative to HC (p=0.0233). Interestingly aldosterone was elevated in all IgAV compared to HC (p<0.0001) however when split based upon renal inflammation the IgAVwoN aldosterone concentrations were significantly greater compared to IgAVN (p=0.0035) and HC (p<0.0001).
Conclusion:
This work demonstrates complement activation through the lectin and alternative pathways in the urine of IgAVN patients. Currently, clinical trials are underway using inhibitors of complement factors B and D pathways. This data provides preliminary evidence that paediatric IgAV patients may benefit also. We also demonstrate data to support the role of the RAAS pathway and there may be a role for early RAAS inhibition
Lay summary:
Children with IgA-vasculitis have a rash on their legs and normally get better on their own. However, 2% will get kidney damage requiring dialysis or transplant. Currently there is no way to know which children will get kidney damage which is only picked up once a lot of damage has happened. To stop children getting kidney damage, we need markers that indicate kidney damage more quickly than current methods. Here we looked at two pathways that trigger kidney damage and looked at the concentration of six molecules in urine. The complement molecules bind with the IgA antibody and deposit in the kidney causing damage. Here we showed that in urine all three molecules studied were higher in children with kidney damage. Two of these are currently being tested as targets for medication and these findings provide evidence that testing these new medicines in children with IgA vasculitis may help. The other three molecules are from a pathway linked to blood pressure and kidney damage. One was at high levels in patients with kidney damage and another was at high levels in patients with vasculitis but no kidney damage. This suggests that some existing medicines that target this pathway may help prevent kidney damage.
Biography:
I am an analytical chemist with a background in metabolomics and proteomics. I currently work as a post doc at the University of Liverpool. Here I investigate the O-glycans of the hinge region of IgA1 and other protein biomarkers in paediatric patients with IgA vasculitis. This work aims to help predict which patients will develop renal inflammation and show further mechanistic insights into this.
My PhD was focussed on the development of novel highly sensitive urine metabolomics methods to study renal damage in HIV patients on combined anti-retroviral therapy. Subsequently I was involved in a clinical metabolomics facility before moving into materials science to develop proteomics skills.
In 2021, I returned to renal research to apply the methods I have developed during my career to medical research. Funding from Kidney Research UK allowed me to return to my passion for medical research. This enables me to continue to grow and develop as an analytical chemist while being able to apply this to paediatric renal disease. Furthermore, this role allows me to work between clinicians at Alder Hey and basic science researchers at Liverpool, providing a unique perspective which I have not experienced previously in my career.

Isaac Chung
Authors and affiliations (if published):
Isaac Chung1, Mukunthan Srikantharajah, Robin Ramphul, Juan Carlos Kaski, Debasish Banerjee1,2
1Renal and Transplantation Unit, St George’s University Hospitals NHS Foundation Trust;
2Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St George’s, University of London
Background:
Chronic kidney disease patients (CKD) are at a higher risk of cardiovascular disease and mortality, which partially improves with kidney transplant but remains elevated compared to general population. This study aims to investigate the progression of vascular structure and function in CKD and transplant patients.
Methods:
Vascular markers including carotid intima-medial thickness [c-IMT], ankle-brachial pressure index [ABPI], brachial artery flow mediated dilatation [ba-FMD], brachial artery nitroglycerin mediated dilation [ba-NMD], augmentation index [AIx] and carotid-femoral pulse-wave velocity [cf-PWV] were measured at baseline and at 3-year follow-up.
Results:
There was no significant difference between CKD and kidney transplant patients at baseline in ba-FMD (6.67 ± 1.47 vs 5.75 ± 0.80; p-value=0.91), ba-NMD (11.1 ± 5.52 vs 11.95 ± 5.25; p-value=0.76), c-IMT (0.67 ± 0.15 vs 0.58 ± 0.80; p-value=0.20), AIx (24.67 ± 7.58 vs 17.50 ± 14.60; p-value=0.21), and cf-PWV (9.13 ± 2.27 vs 8.50 ± 1.84; p-value=0.58).
In the CKD cohort, there was no difference between baseline and follow-up in ba-FMD (4.70 ± 2.75 vs 3.57 ± 2.41, p-value=0.41), ba-NMD (11.06 ± 5.51 vs 11.61 ± 4.37, p-value=0.94), c-IMT (0.67 ± 0.15 vs 0.63 ± 0.11, p-value=0.61), Aix (24.67 ± 7.58 vs 26.50 ± 9.59, p-value=0.42), and cf-PWV (9.13 ± 2.27 vs 9.81 ± 1.42, p-value=0.55). In the transplant cohort, there was no difference between baseline and follow-up in ba-FMD (4.90 ± 4.03 vs 5.58 ± 2.69, p- value =0.66), c-IMT (0.58 ± 0.08 vs 0.57 ± 0.10, p-value =0.97), Aix (17.50 ± 14.07 vs 23.20 ± 19.11, p-value=0.46), and cf-PWV (8.50 ± 1.84 vs 8.92 ± 2.68, p-value=0.69). Ba-NMD improved in 3-years follow-up compared to baseline in the transplant cohort (11.94 ± 5.25 vs 16.65 ± 3.21, p-value=0.03).
Conclusion:
CKD and kidney transplant patients have similar measures of vascular structure and function at baseline; after 3-years, CKD patients remain similar to baseline in vascular health, while renal transplant patients remain similar to baseline apart from increased Ba-NMD when compared to baseline.
Lay summary:
Patients with chronic kidney disease (CKD) are more likely to have heart disease. This is partly due to the toxins in their blood making arteries stiffer and less stretchy. We are aiming to study the progression of blood vessel disease in patients with CKD and kidney transplant patients. Previously, we have shown the blood vessels in patients with a kidney transplant are more stretchy when compared to patients with CKD and on haemodialysis in 3-months follow-up.
In this study, we have found at 3-year follow-up, CKD and kidney transplant patients have similar blood vessel elasticity. We also found kidney transplant patients have blood vessels that dilate more to a drug called glyceryl trinitrate when compared to patients with CKD.
Table 1 Baseline characteristics & results from PACK & PACK-E.
CKD (n=6) | CKD | Baseline vs Follow-up | Transplant (n=10) | Transplant | Baseline vs Follow-up | |
Baseline | 3 Years from Baseline | p-value | Baseline | 3 Years from Baseline | p-value | |
Age | 62.80 ± 10.94 | 65.80 ± 10.94 | NA | 50.57 ± 16.29 | 53.57 ± 16.29 | NA |
Male/Female | 3/3 | 3/3 | NA | 6/4 | 6/4 | NA |
eGFR | 28.40 ± 9.86 | 47.80 ± 33.50 | 57.58 ± 31.71 | 51.57 ± 25.01 | ||
Ba-FMD (%) | 4.70 ± 2.75 | 3.57 ± 2.41 | 0.47 | 4.90 ± 4.03 | 5.58 ± 2.69 | 0.66 |
Ba-NMD (%) | 11.06 ± 5.51 | 11.61 ± 4.37 | 0.85 | 11.94 ± 5.25 | 16.65 ± 3.21 | 0.03 |
AIx (%) | 24.67 ± 7.58 | 26.50 ± 9.59 | 0.72 | 17.50 ± 14.07 | 23.20 ± 19.11 | 0.46 |
cf-PWV (m/s) | 9.13 ± 2.27 | 9.81 ± 1.42 | 0.55 | 8.50 ± 1.84 | 8.92 ± 2.68 | 0.69 |
c-IMT (cm) | 0.67 ± 0.15 | 0.63 ± 0.11 | 0.68 | 0.58 ± 0.08 | 0.57 ± 0.10 | 0.97 |
Biography:
Renal research allowed me to speak to a variety of patients with varied experiences. CKD is such a hidden disease. I find it interesting to hear people’s stories. Furthermore, as a medical student, research in renal disease has allowed me to learn additional techniques such as ultrasound to further my clinical practice in the future.
Cardiovascular disease remains the primary cause of death within kidney disease patients. My work mainly looks at the natural progression of vascular disease within patients with chronic kidney disease. I hope my work can shed some light on the link between cardiovascular disease and kidney disease.
Kidney Research UK has given me an opportunity to undertake my intercalated BSc and learn research techniques. I hope to bring the things I learnt in this research year into my future endeavours.

Silothabo Dilso
Authors and affiliations (if published):
Silothabo Dliso1, Julien Marro2, Catherine McBurney1, Jessica Tiffin1 Andrew J. Chetwynd2, Rachael D. Wrigh2, and Louise Oni2,3*
1NIHR Alder Hey Clinical Research Facility, Clinical Research Division, Alder Hey Children’s NHS Foundation Trust, Liverpool L14 5AB, UK; [email protected] (SD); [email protected] (CM); [email protected] (JT)
2Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L12 2AP, UK; [email protected] (J.M.); [email protected] (A.J.C.); [email protected] (R.D.W.)
3Department of Paediatric Nephrology, Alder Hey Children’s NHS Foundation Trust Hospital, Liverpool L14 5AB, UK
Background:
The IgA Vasculitis Study is a longitudinal observational cohort study, aimed to improve the understanding of Immunoglobin A vasculitis (IgAV), with a focus on preventing the evolution of kidney disease. The study opened in August 2019. Like many other studies, it was affected by the Covid-19 Pandemic in a number of ways. In order to focus efforts on Covid-19 research many studies had to be paused or closed, causing significant disruption to recruitment and contributing to participants being lost to follow up; thus, making longitudinal sample collection difficult. The aim of this abstract is to illustrate some of the challenges in establishing a cohort study during this era.
Methods:
The IgA Vasculitis Study includes patients of any age or sex with a clinical diagnosis of IgAV. Additionally, healthy controls are recruited using children with no known inflammatory conditions or significant illnesses. A multidisciplinary research team have facilitated the set up and delivery of this study and include a consultant nephrologist, research nurses, a physician associate, a post-doctoral research associate and medical/research students. IgAV patients are recruited and followed up on the Medical Day Case Unit (MDU) during the course of their clinical monitoring using the Alder Hey Henoch Schonlein Purpura Pathway. Healthy control participants (HCs) were recruited from MDU and Surgical Day Care units.
Results:
To date, 91 patients with IgAV and 32 HCs have been recruited. The study was forced to pause during the first national lockdown period and was open subsequently, but recruitment was significantly impacted thereafter. Figure 1 demonstrates the impact of the pandemic on study recruitment.

Figure 1. Recruitment of IgAV patients and healthy controls between august 2019 and April 2022, with periods of UK national lockdown highlighted.
There have been several cross-sectional research projects that have come out from the IgA Vasculitis Study, these include a systematic review on urinary biomarkers, a clinical cohort data description paper, urine complement laboratory studies, urine and plasma IgA analysis, urine renin-angiotensin-aldosterone system studies and a urinary protein array study. Efforts are underway to strengthen the longitudinal cohort to explore the scientific evolution of nephritis.
Conclusion:
Despite the challenges of the past two years, the study has been relatively successful, producing several outputs, including abstracts and journal articles. The success of the recovery of this study has been due to the diverse, multidisciplinary team who have worked together to maximise recruitment and sample collection.
Lay summary:
Immunoglobin A vasculitis (IgAV) is an inflammatory disease affecting the blood vessels and can sometimes lead to kidney problems. IgAV is relatively rare, and there is much that is still not known about it. The IgA Vasculitis Study seeks to improve understanding about the condition, however over the past couple of years, like many other studies, it was significantly impacted by the Covid-19 pandemic. At times recruitment was not possible and even when it was, there were difficulties due to restrictions on a national level and apprehension from the public around people coming to hospital. By creating a team made of diverse professionals, the study was able recover and go on to successfully recruit over 130 participants. The study has also led to other interesting smaller projects, some of which have gained recognition nationally and the results of which have gone on to be published in international journals.
Biography:
I am a Physician Associate in Paediatric Clinical Research at Alder Hey Children’s Hospital, an Honorary Fellow at the University of Liverpool, and Chair of the Faculty of Physician Associates Research Committee.
I am part of the IgA vasculitis study team at Alder Hey, where we are investigating renal inflammation in IgA vasculitis patients. We are seeking to gain insight into the pathophysiology of IgA vasculitis and the renal inflammation that is associated with it, as well as identifying biomarkers to help predict disease progression and potential treatment targets.
Our Kidney Research UK grant funds our postdoctoral research associate, which in turn allows us to be able to run a lot of experiments ourselves and grow the study as we get more data.
I was first drawn to renal research by the opportunity to work with experienced researchers and learn from them. What keeps me interested in it is the potential we have within our IgA vasculitis study to make ground-breaking discoveries. Additionally, we have been using the study as a vehicle to change practice around inclusion in research, this hopefully will influence the wider research community.

Dr Sarah Gleeson, Imperial College London
Authors and affiliations (if published):
Sarah Gleeson1,2, Michelle Willicombe1,2, Liz Lightstone1,2, Candice Roufosse1,2
1 Imperial College Healthcare NHS Trust
2 Department of Immunology and Inflammation, Imperial College London
Background:
Membranous nephropathy (MN) recurs in 7-44% of cases following transplantation, with more cases identified when protocol biopsies are performed. Live donor transplant, short time on the waiting list, steroid free regimens and positive PLA2R at the time of transplantation have all been associated with recurrence, however the associations remain uncertain. Histopathological appearances are similar to those seen in native MN, however the first evidence of early recurrence may beC4d staining along the glomerular basement membrane. We sought to examine the incidence, risk factors, histopathology and outcomes of recurrent membranous nephropathy in our cohort to better characterise the natural history post-transplant and, where appropriate, allow us to intervene earlier to prolong transplant survival.
Methods:
All patients with biopsy proven membranous nephropathy listed as the cause of their end stage kidney disease (ESKD) transplanted at our centre from 2006 were included. Detailed demographic, clinical and histopathological details were gathered and analysed.
Results:
40 patients received 42 kidney transplants. The mean age at transplant was 57.5 years, 67.5% male and 47.5% were Caucasian. 59.5% received deceased donor kidneys. 80.9% received alemtuzumab induction. Mean follow up was eight years. 23.8% lost their graft, 25.7% had a rejection episode. 35/42 had at least one biopsy post-transplant. 10 patients had a biopsy proven recurrent MN. 60% received live donor transplants versus 28% without recurrence (p 0.11). 30% had a pre-emptive transplant compared to 9%. The mean time on the waiting list was 1.8 years compared with 3.4 years (p=0.9).
Membranous nephropathy recurred 2.25 years post-transplant. Proteinuria at diagnosis ranged from six to 1126 mg/mmol. Four patients required immunosuppressive treatment. All were treated with rituximab with a partial or complete response in all patients.
There was no difference in graft loss or time to loss between the groups (p 0.18).
Conclusion:
23% of patients with membranous nephropathy had a recurrence post-transplant. Although live donor transplant, pre-emptive transplant and shorter time on the waiting list are associated with recurrence, this is not statistically significant in our single centre cohort. Rituximab treatment, when indicated, appears successful. Although patients have lost their grafts due to recurrence, there is not a higher rate of graft loss in the recurrence group. A detailed study of tissue and serology is now being undertaken to identify early features of recurrence to allow early intervention and to correlate with treatment response and graft outcomes.
Lay summary:
Membranous nephropathy (MN) is a cause of kidney failure. This can sometimes come back (recur) after kidney transplant, in 7-44% of transplants. Receiving a kidney from a live donor and less time waiting for a transplant have been associated with recurrence, but it is not a definite link. As this area is relatively understudied, we planned to look at it in more detail in our patients to see how often it occurs, if there is any risk factors and how people do if it recurs.
After examining our hospital records, we found that 42 patients had MN who received a transplant. The average age at the time of transplant was 57, 67% were male and 47% Caucasian. The average follow up was 8 years. In this time 35% had at least one episode of rejection and 24% lost their transplant. MN recurred in 24% of transplants, an average of 2.3 years following transplant. Those that had a recurrence were more likely to have had a live donor kidney and had waited a shorter time for their transplant, however this difference was not statistically significant. If they needed treatment this was with rituximab and generally worked well to improve the disease.
Biography:
Sarah Gleeson is a renal trainee who has undertaken her training in Ireland, New Zealand and the UK. She is currently a renal clinical research fellow at Imperial College and Hammersmith Hospital undertaking a PhD, funded by Kidney Research UK, looking at autoimmunity post-kidney transplantation.
As a kidney doctor, I realised that every day we were encountering patients with problems and issues that we did not know enough about or know the best way to treat. I realised the only way to improve this was through research. One of the areas I found we knew very little about was autoimmunity post-transplant; how people with autoimmunity do post-transplant, why certain diseases fire back and how we treat them, and if people post-transplant have a predisposition to autoimmunity, especially if they are treated with certain medications. Thanks to Kidney Research UK for looking into this in detail. Hopefully my findings will allow others to look at the same and similar subjects which will hopefully accelerate research.

Celine Grenier
Authors and affiliations (if published):
Celine Grenier1, Filipa M. Lopes1, Anna M Cueto-González2,3, Eulàlia Rovira-Moreno2,3, Romy Gander4, Alison M. Gurney5, Karen D. McCloskey6, Glenda M. Beaman7,8, William G. Newman7,8, Adrian S. Woolf1,9, Neil A. Roberts1.
1Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
2Department of Clinical and Molecular Genetics, Vall d'Hebron Campus Hospitalari, Barcelona, Catalonia, Spain
3Medicine Genetics Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.
4Department of Pediatric Surgery, Pediatric Urology and Renal Transplant Unit, University Hospital Vall D´Hebron Barcelona, Hospital Vall D´Hebron, Barcelona, Spain.
5Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
6Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
7Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK.
8Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, M13 9PL, UK.
9Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UK.
Background:
Urinary tract malformations account for half of all children with kidney failure. One such is urofacial, or Ochoa, syndrome (UFS), an autosomal recessive disease. Affected people have dyssynergic bladders in which the detrusor smooth muscle contracts against a bladder outflow tract that fails to dilate. Affected individuals experience incomplete voiding, with urosepsis and severe kidney failure. We discovered that around half of UFS individuals carry biallelic variants in HPSE2. Some others instead carry biallelic variants of LRIG2 encoding leucine rich repeats and immunoglobulin like domains 2. LRIG2 has been detected in human foetal bladder nerves.
Methods:
Barely a handful of families have been reported with UFS associated with LRIG2 variants, and we describe one new such family. Hypothesising that neurogenic defects underlie USF bladder dysfunction, we also undertook ex vivo physiology studies of Lrig2 homozygous mutant mice to learn more about pathobiology.
Results:
The index case was a six-year-old boy born to first cousin parents. Bilateral hydronephrosis was detected on foetal ultrasonogaphy. Neonatally, urinary tract infections occurred, and ultrasound revealed hydronephrosis with a thickened bladder wall. Cystography revealed vesicoureteral reflux, but the urethra appeared normal, excluding a diagnosis of posterior urethral valves. Aged three years, the index case was assessed by the genetics department and was noted that he had a grimace upon smiling, such that his eyes closed, and the corners of his mouth failed to elevate. A clinical diagnosis of UFS was made and HPSE2 and LRIG2 were sequenced. No variants were found in HPSE2 but a homozygous variant c.1939C>T (p.Arg647*), classified as pathogenic according to American College of Medical Genetics guidelines, was identified in LRIG2. Juvenile homozygous mutant Lrig2 mice displayed abnormal urination and enlarged bladders. Physiology experiments showed a neurogenic defect in relaxation of the bladder outflow tract. Lrig2 mutant mice also displayed abnormal contractile responses of the detrusor, the bladder muscle that normally causes the organ to void urine.
Conclusion:
Putting the family in the context of LRIG2 variants reported to date, it is clear that full-blown UFS occurs in the presence of stop or frameshift mutations but missense changes lead to bladder-limited disease without the grimace. This family also emphasises that the disease begins before birth. Our observations in mutant mice support the idea that UFS is a genetic autonomic neuropathy of the bladder. Our results may also inform future treatments for UFS, including viral vector mediated gene therapy.
Lay summary:
Around half of all children are born with kidneys, and sometimes also bladders, that have failed to mature properly before birth and in the early years after birth. In our study we make two important points. First, some such children carry mutations of genes that, in health, ensure that the bladder can store urine and then empty it during urination. This can lead to them getting urinary infections and kidney failure. Second, to better understand the disease we are studying mice that mimic the human disease. We found that these mice, which carry the same abnormal gene as goes wrong in the human disease, can also not empty the bladders. We also found defects in the nerves controlling bladder function in these mice. This leads to two, compounding, problems namely, the outflow to the bladder does not relax, nor can the body of the bladder contract properly. These results could lead, in the future, to a better management of the symptoms and potentially could help to find a cure. The best treatment may be to replace the abnormal gene with a normal one, with ‘gene therapy’, so that the bladder functions normally.
Biography:
In 2012, I started my work in research in France in Angers as a PhD student. There, I studied the adaptation of small arteries to the chronic variation of blood flow. After completing my PhD, I moved to the UK in Cambridge and worked on the involvement of molecular changes in aortas in Marfan syndrome. I later worked in Edinburgh on the effect of high salt consumption on the physiology of renal arteries. At this occasion, I also discovered how the kidney worked, and its central role in the maintenance of normal blood pressure. I now work in Manchester in a project funded by Kidney Research UK. I study a rare genetic disease, urofacial syndrome, where a bladder dysfunction could lead to kidney failure. At the moment, no treatments are available and only the symptoms can be treated. With our mice model of urofacial syndrome, I can understand the progression of this disease. We also study gene therapy to develop a treatment to potentially cure the disease in the future. In addition to my research work, Kidney Research UK funding also allows me to present these discoveries in international conferences.

Jess Ivy
Authors and affiliations (if published):
Georgios Krilis, Frances Turner, Hannah M Costello, Matthew A Bailey, Jessica R Ivy.
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 recognised as an 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 signalling, allowing the synchronisation 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. Therefore, we hypothesise that glucocorticoid treatment disrupts the rhythm of 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 and clamp plasma glucocorticoids at intermediate physiological levels. Mice were culled every two hours for 48 hours and renal arteries were collected for RNA sequencing. To detect circadian rhythmicity of genes, cosinor regression was performed using the algorithm, LimoRhyde. Gene set enrichment analysis (GSEA) was performed using a false discovery rate of <0.05.
Results:
In the control group 3% (465/14 425) of protein-coding genes were rhythmic. 210 of these genes lost their rhythm following treatment, including components of the molecular clock. GSEA analysis on a gene list ranked by glucocorticoid-induced loss of rhythmicity, revealed enrichment of genes related to “Circadian Rhythm”, “Extracellular Matrix Organisation” and “Oxidative Stress”. A surprising finding was that treatment doubled the proportion of rhythmic genes to 6% (919/14806), i.e. 774 genes gained rhythm in the absence of circadian glucocorticoid variation. These newly rhythmic genes had a reduced oscillation amplitude compared to the rhythmic genes in the control group. Moreover, the peak times of de novo rhythmic genes clustered either four hours after the lights went on or four hours after the lights went off. This suggests that the absence of a glucocorticoid rhythm unmasks genes that are strongly influenced by light.
Conclusion:
In the mouse renal artery, arrhythmic glucocorticoid signalling causes dampening of genes involved in normal circadian rhythms and induces de novo rhythms. Further studies will investigate how these gene changes contribute to rhythmic vascular function and BP dipping.
Lay summary:
Body clocks present throughout our body allow us to adapt to our rhythmic environment of light and dark. They control many physiological parameters, 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. Stress hormones also cause non-dipping and we can use these in experiments to model non-dipping. Blood vessels in the kidney are important for the regulation of normal blood pressure but nothing is known about the rhythmic genes that might exist in kidney blood vessels. In this experiment we showed that in mice, the kidney blood vessels have robust clocks, and these clocks are blunted with steroid treatment. This results in a set of normally rhythmic genes becoming arrhythmic (including genes involved in the body clock itself and genes that contribute to the structure of the kidney blood vessels) and surprisingly also caused a usually arrhythmic set of genes to become rhythmic (this included genes that are involved in energy production). Further experiments will investigate how these gene changes affect the rhythmic function of the blood vessels.
Biography:
I am a Kidney Research UK intermediate fellow at the University of Edinburgh. I completed my MSc and PhD at the University of Edinburgh 2016 and subsequently undertook a postdoctoral position before starting my Kidney Research UK funded fellowship in December 2019. I am attracted to renal research because of the kidney’s extraordinarily important role in the regulation of blood pressure. This is critical for the maintenance of cardiovascular health. But it is not just the level of blood pressure that is important. Loss of the circadian rhythm of blood pressure, called non-dipping, is estimated to be present in up to 60% of chronic kidney disease patients and independently predicts cardiovascular disease.
Little is known about how blood pressure rhythm is generated, and which mechanisms fail when this important rhythm is lost. My work accelerates research by aiming to fill this gap and focuses on renal and reno-vascular mechanisms of circadian blood pressure control using a combination of in vivo and molecular approaches. My work would not have been possible without support from Kidney Research UK. Working with Kidney Research UK has been a turning point in my research career and enabled me to start my own lab and pursue this exciting avenue of research.

Jessica Keeple
Authors and affiliations (if published):
Jessica Kepple1,2, Simon Davis2,3, Roman Fischer2,3, Jon Milton4,5, Rutger Ploeg4,5,6, Richard Cornall1,2, Katherine Bull1,2,6
1Wellcome Centre for Human Genetics, University of Oxford
2Nuffield Dept of Medicine, University of Oxford
3Target Discovery Institute, University of Oxford
4Oxford Transplant Biobank
5Nuffield Department of Surgery, University of Oxford
6Oxford Kidney Unit and Oxford Transplant Centre, Oxford University Hospitals Trust
Background:
Kidney disease is frequently associated with glomerular damage resulting in impaired filtration. Understanding disease related molecular changes which arise in the glomerulus is an essential prerequisite for the development of more targeted therapeutics. Transcriptomic and proteomic profiling of human renal tissue offers a unique opportunity to investigate drivers of kidney disease at a cellular level. To date, most studies have utilised a small number of nephrectomy sections, with challenges associated with limited biopsy material, technical dissociation bias against glomerular cells, and non-standardised methodology restricting the application of human biopsies in research. To develop tools that can be applied to larger sample sets and routine clinical diagnostics, our aim is to identify optimal experimental protocols for processing renal biopsy core samples, focusing on the assessment of less abundant glomeruli cell types.
Methods:
We obtained healthy kidney donor human renal biopsy cores via the Oxford Transplant Biobank and processed these for RNA sequencing and proteomics. Single cells or nuclei were isolated from fresh tissue to investigate cell type specific efficiency of different tissue dissociation protocols. RNA sequencing libraries were analysed using the R package Seurat, with glomeruli cell clusters evaluated for overall cell number and gene enrichment. In addition, glomerular regions from formalin-fixed paraffin-embedded (FFPE) 10um renal biopsy sections were isolated via targeted laser capture microdissection coupled with mass spectrometry (MS). Given the limited tissue, we conducted a titration experiment with varying numbers of isolated glomeruli to establish the minimum amount of material required for high quality proteomics. MS results were analysed using DI-ANN and Perseus software to assess relative protein abundance with different starting material.
Results:
Comparison of single-cell and single-nuclei RNA sequencing (scRNA-Seq, snRNA-Seq) results revealed the greatest cellular diversity in the snRNA-Seq dataset, containing more glomeruli cell types including mesangial cells and podocytes. We successfully isolated glomerular proteins from FFPE embedded renal tissue, quantifying around 4.5k unique proteins from a single human glomerulus. Titrating of starting material showed that isolation of 6-10 human glomeruli is sufficient to quantify over 5k proteins.
Conclusion:
Our results provide a framework for RNA-seq or MS experiments in glomerular disease using tissue from a single standard biopsy core, with possible applications to stored FFPE blocks of human biopsies for targeted proteomics screens. Future studies to isolate glomeruli using snRNA-Seq and LCM from biopsies in patients with kidney disease will provide novel cellular insight into renal cell specific pathways of importance in kidney disease.
Lay summary:
New methods now allow us to study which genes are active in individual cells. Applying these methods to human kidney samples could help transform diagnostics while potentially allowing for the discovery of new drug targets for kidney disease. For practical application, such methods need to work using the small samples typically taken during a kidney biopsy. However, until now, such samples have been considered inadequate for single cell methods, particularly for certain rare but important cell types, like those of the glomerulus, a key structure in the kidney that filters blood. The project goal is to identify methods which produce the highest quality results from a limited amount of human tissue. We measured changes in gene expression and proteins from kidney biopsy samples using different experimental methods. We can obtain gene expression data from more rare cell types, including those found in the glomerulus. Using a laser, we can also collect glomerular proteins by cutting out these sections. We only need to collect thin slices containing 6 human glomeruli to detect over 5000 different proteins. Our results provide a basis for future experiments testing gene expression and proteins changes in biopsy and stored tissue from people with kidney disease.
Biography:
I am currently an Oxford-Novo Nordisk postdoctoral research fellow in the lab of Dr Katherine Bull and Professor Richard Cornell (Oxford-NDM) and Dr Ramneek Gupta (NNRCO). The focus of my research is to identify early modulators of kidney disease in human renal transplant tissue. I am developing and utilising integrated transcriptomic and proteomic approaches to assess cellular alterations in human renal biopsies, with a particular focus on glomerular structures. My research interest in metabolic disease aligns well with renal research, as chronic kidney disease in patients places them at an increased risk for other metabolic diseases, like cardiovascular diseases, diabetes, and hypertension.
I hope to develop future integrated imaging ‘omics’ platforms that can be applied to limited renal samples for the investigation of causative disease pathways and possible drug targets. Being a part of the Kidney Research UK team affords me the opportunity to engage and collaborate with a diverse range of research topics and presenters. This collaborative partnership is critical for driving scientific advantages and patient focused research.
Authors and affiliations (if published):
Katie J. Mylonas*, Eoin D. O’Sullivan, Duncan Humphries, David P. Baird,Marie-Helena Docherty, Sarah A. Neely, Paul J. Krimpenfort, Anette Melk, Roland Schmitt, Sofia Ferreira-Gonzalez, Stuart J. Forbes, Jeremy Hughes, David A. Ferenbach
*University of Edinburgh
Background:
The ability of the kidney to regenerate successfully after injury is lost with advancing age, chronic kidney disease (CKD), and after irradiation. The factors responsible for this reduced regenerative capacity remain incompletely understood, with increasing interest in a potential role for cellular senescence in determining outcomes after injury. Here, we demonstrated correlations between senescent cell load and functional loss in human aging and chronic kidney diseases including radiation nephropathy.
Methods:
Published datasets of human CKD were analysed, examining levels of markers of cellular senescence in cohorts of aging and injured human kidneys. Human proximal tubular epithelial cells were grown and senescence induced via gamma irradiation in vitro. Senescence was also studied in vivo in naturally aged, injured and irradiated mice, in the presence and absence of the Bcl2/w/xL inhibitor ABT-263.
Results:
We dissected the causative role of senescence in the augmented fibrosis occurring after injury in aged and irradiated murine kidneys. In vitro studies on human proximal tubular epithelial cells and in vivo mouse studies demonstrated that senescent renal epithelial cells produced multiple components of the senescence-associated secretory phenotype including transforming growth factor beta1, induced fibrosis, and inhibited tubular proliferative capacity after injury. Treatment of aged and irradiated mice with the B cell lymphoma 2/w/xL inhibitor ABT-263 reduced senescent cell numbers and restored a regenerative phenotype in the kidneys with increased tubular proliferation, improved function, and reduced fibrosis after subsequent ischemia-reperfusion injury.
Conclusion:
Senescent cells are key determinants of renal regenerative capacity in mice and represent emerging treatment targets to protect aging and vulnerable kidneys in man.
Lay summary:
There is an urgent need for novel therapies to prevent progressive kidney scarring (fibrosis). In order to generate new drugs to give to humans, it is necessary to understand the basic pathways which drive kidney scarring.
In this study we explored the role of a particular kind of cell (termed a ‘senescent cell’) in driving kidney fibrosis. Any cell can become senescent in response to ageing and injury cues, and senescent cells are known to increase in aged and in injured human kidneys – meaning they are ‘in the right place at the right time’ to be involved in driving fibrosis.
The most important finding from this study was that when we gave a drug (ABT-263) to old and previously injured kidneys which killed the senescent cells whilst leaving the non-senescent cells intact. The ability of this kidney to recover from a subsequent injury was restored. This provides important evidence that an approach based on killing senescent cells could be efficacious in preventing kidney fibrosis in humans. Based on the results of this study, we have applied for and been granted further funding by Kidney Research UK to test the safety and efficacy of ABT263 on human kidneys which cannot be used for human transplantation, as the next step towards clinical trials.
Biography:
I graduated with a first-class honours degree from Queen’s University of Belfast. Post-graduation, I worked for four years as a research associate at DeCode Genetics in Iceland. I was then selected for the Wellcome Trust four-year PhD programme at the University of Edinburgh, where I carried out my PhD in Professor Judith Allen’s lab.
I then joined Professor Hughes’ lab investigating the role of macrophages in kidney injury, where I learned that kidney disease is a major cause of mortality and morbidity in the UK. Later, I studied the role of senescent cells in kidney disease with Dr David Ferenbach. My original observation that elimination of senescent cells prior to injury in the kidney is protective, and drives repair and regeneration, led to a first author paper published recently in Science Translational Medicine. This work also led to my recent fellowship supported by Kidney Research UK and the Chief Scientist Office, Scotland - “Driving kidney repair through modulation of senescent cells and macrophages”. Supported by Kidney Research UK, the work we do will improve our understanding of kidney disease, and ageing in general, and lead to new clinical treatments to save lives and make important contributions to future human health and quality of life.
Authors and affiliations (if published):
Ross A Campbell, Jeremy Hughes, David A Ferenbach and Katie J Mylonas.
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 that both natural ageing and total body irradiation (TBI) induces renal senescence, with administration of the senolytic ABT- 263 prior to renal ischaemia reperfusion injury (IRI) being protective; reducing tissue loss, fibrosis and inflammation, whilst promoting structural integrity/regeneration.
It was hypothesised that SCs compromise renal repair/regeneration after IRI in mice by driving excessive monocyte/macrophage recruitment and polarisation towards pro-inflammatory, rather than reparative, phenotypes.
Methods:
In vitro cultures: Human renal proximal tubular epithelial cells (PTECs) were exposed to 10 Gy radiation to induce senescence. Human macrophages were before exposed to conditioned medium from SC (SCCM) or control PTEC (Control CM) overnight. qPCR was then carried out for various parameters.
In vivo: Young mice were exposed to TBI. ABT-263 was administered by gavage (50 mg/kg/day). IRI surgery left renal pedicle was clipped using an atraumatic clamp for 15 min. Clodronate liposomes or vehicle were administered i.v. 24 hours post-IRI. At 7 days and 5 weeks, kidneys were recovered, various assays carried out e.g. flow cytometry, qPCR.
Results:
In vitro: Senescent PTECs produced significantly increased levels of monocyte chemoattractant protein (MCP)-1 (5-fold change vs control, p<0.01), indicating that epithelial senescence drives monocyte recruitment. Pro-inflammatory markers including CD80 (2-fold increase, p<0.05) and IL-1B (3-fold increase, p<0.05) were significantly increased in human macrophages after exposure to SCCM vs control CM.
In vivo: Flow cytometry at d7 and d35 post-IRI indicated that the presence of SCs drove persistent monocyte/macrophage recruitment (3-fold increase, p<0.05). Treatment with ABT-263 or clodronate liposomes significantly decreased F480+CD80+ monocyte-derived inflammatory macrophages recruitment to the kidney by d35 (both approx. 50% reduction, p<0.05). This correlated with an increase in kidney mass in ABT-263 (approx. x1.5 heavier; p<0.01) and/or clodronate-treated mice (approx.. x1.75 heavier; p<0.005). Collagen deposition measured by picrosirius red (PSR) IHC indicated less renal fibrosis with ABT-263 or clodronate treatment (3-10x lower; both p<0.05). qPCR of the renal tissue indicated less of various markers of senescence and inflammation in ABT-263 and/or clodronate treated vs control.
Conclusion:
Collectively, these results support the hypothesis that senescence drives pathogenic monocyte/macrophage recruitment and a proinflammatory, profibrotic phenotype in the kidney. Importantly, selective targeting of either SCs or macrophages reduced renal fibrosis and thus represent candidates for future clinical translation.
Lay summary:
Senescent cells have lost the ability to divide but accumulate with age and injury. Killing senescent cells increases healthy lifespan in mice. We found that ageing and irradiation of mice induced increased kidney cell senescence which was reversed with the novel drug ABT-263 that kills senescent cells. By reducing senescent cells, ABT-263 treatment protected kidneys from scarring after subsequent kidney injury and also increased repair and led to better kidney function.
White blood cells called macrophages appear in the kidney after injury and can become a type of cell that aids the healing process. However, excessive macrophages can also be damaging and cause a delay in, or even prevent, adequate healing. It is proposed that the ability of these macrophages to promote kidney repair is negatively affected by the presence of senescent cells. How senescent cells affect macrophages was tested, and whether kidney repair could be enhanced by removing senescent cells and excess macrophages.
it was found that senescent cells did negatively affect macrophages and by removing them and excess macrophages we could drive better kidney repair.
This work could lead to novel treatments to protect vulnerable older or chronically damaged kidneys from further damage, scarring and loss of function.
Biography:
I graduated with a first-class honours degree from Queen’s University of Belfast. Post-graduation, I worked for four years as a research associate at DeCode Genetics in Iceland. I was then selected for the Wellcome Trust four-Year PhD programme at the University of Edinburgh, where I carried out my PhD in Professor Judith Allen’s lab.
I then joined Professor Hughes’ lab investigating the role of macrophages in kidney injury, where I learned that kidney disease is a major cause of mortality and morbidity in the UK. Later, I studied the role of senescent cells in kidney disease with Dr David Ferenbach. My original observation that elimination of senescent cells prior to injury in the kidney is protective, and drives repair and regeneration, led to a first author paper published recently in Science Translational Medicine. This work also led to my recent fellowship supported by Kidney Research UK and the Chief Scientist Office, Scotland - “Driving kidney repair through modulation of senescent cells and macrophages”. Supported by Kidney Research UK, the work we do will improve our understanding of kidney disease, and ageing in general, and lead to new clinical treatments to save lives and make important contributions to future human health and quality of life.

Cathy Pogson
Authors and affiliations (if published):
Pogson, CM
Background:
People receiving haemodialysis, have the highest medicine burden of all chronically ill populations. Medicine burden exposes people to medication related problems, impacting on quality of life and healthcare experience. Frailty describes a persons’ ability to bounce back after an illness or injury. Polypharmacy is defined at taking five or more regular medicines daily. Medicines optimisation is the process of reviewing medicines to manage polypharmacy and improve outcomes. For people with frailty without kidney disease, medicines optimisation, is associated with decreased risk of death, decreased referral to nursing home, lower drug costs and improvements in patient’s perception of health. Patient centred care, incorporating a patient's values and preferences, and minimizing harm, is central to effective medicines optimisation. Reductions in polypharmacy may improve the quality of life for haemodialysis patients who suffer from the side effects of the multiple medicines prescribed.
Methods:
Evaluation of a systematic literature search (Cochrane, Google scholar, Delphi, CINAHL, Medline and via OVID, Embase, Amed and Ovid Emcare) to understand the impact of medicines optimisation (individualized medicine, medication management, polypharmacy, medication adherence, inappropriate medication) upon the healthcare experience (health-related quality of life, quality of life) of adult patients (frail older adults, elderly) receiving haemodialysis (dialysis; dialysis, renal; haemodialysis; renal dialysis; chronic renal failure).
Results:
Medicines optimisation for patients receiving haemodialysis identifies high numbers of potentially inappropriate medications and high numbers of omitted indicated medicines.
Use of the criteria from Screening Tool for Older Persons’ Prescriptions (STOPP) and Screening Tool to Alert Doctors to the Right Treatment (START), identifies significant numbers of medicines for review but does not improve medication adherence or the healthcare experience for people receiving haemodialysis.
Two other studies identified, each using different refined deprescribing tools, both significantly reducing polypharmacy. One study reports a significant improvement in patient reported Living with Medicines Score following deprescribing together with a (non-significant) improvement in adherence. The second study had no negative impact on self-reported patient satisfaction but did not capture impact upon quality of life.
Conclusion:
Medicines optimisation can improve the healthcare experience for people receiving haemodialysis. Medicines optimisation for haemodialysis patients needs investigating. This process must ensure that the health-related consequences of polypharmacy and the negative impact on quality of life are minimalised.
Lay summary:
Many people who receive haemodialysis take multiple medicines daily. Haemodialysis is when a machine filters your blood when your kidneys are not able to do this. This is one way to treat chronic kidney disease, when your kidneys are not working as well as they once did. Frailty is when you don’t bounce back quickly after an illness, accident, or stressful event. People with both can find it difficult to remember drug names, when to take them, understand what they are for and may find them difficult to manage.
Medicines are beneficial but people don’t always get the best from them. Taking too many medicines can cause problems. People with frailty are also more likely to get problems with medicines. These can be serious and cause people to become unwell, sometimes even causing admissions to hospital.
Medicines optimisation is reviewing medicines and offering safe alternatives. Optimising medicines for people receiving haemodialysis reduces the number of medicines they are asked to take. One study describes a better ‘living with medicines score’. We need to look further into the benefits of optimising medicines for these people. The harmful effect of medicines on quality of life must be made as small as possible.
Biography:
The provision of effective services and quality patient care depend upon evidence. Having worked as a renal clinical pharmacist for over seven years I have seen many opportunities to improve patient experiences and outcomes. In December 2021, I was awarded an NIHR Research Initiation grant to investigate whether we could improve polypharmacy for people living with advanced chronic kidney disease (CKD) and frailty, adopting medicines optimisation strategies used for people living with frailty but without CKD. My literature review revealed papers describing different strategies to cut polypharmacy, all single centre studies. The conclusion was there needs to be development of a specific tool to encompass prescribing practice within renal units.
Kidney Research UK provides the opportunity to join a community of experienced renal researchers and patients, committed to addressing the specific needs of kidney patients. Through peer support, sharing experiences and learning in a supported environment I will develop into an independent researcher. As a clinical academic, I will act as a role model, encouraging, supporting, and leading research and provide patients with the reassurance we are a research active department, delivering change for people with kidney disease.

Raina Ramnath
Authors and affiliations (if published):
Raina D. Ramnath, Eve Miller, Sevil Erarslan Catak, Gavin I. Welsh, Rebecca R. Foster and Simon C. Satchell.
Background:
The endothelial glycocalyx is a key determinant of vascular function, and studies have pointed to the glycosaminoglycan heparan sulphate (HS) as having a particularly important role in diabetic kidney disease. HS loss has been associated with kidney damage in human pilot studies. HS chains are assembled in the endoplasmic reticulum by the actions of a series of enzymes and exostosin (EXT) 1 is an essential and rate-limiting enzyme for HS chain polymerisation. We aimed to examine in detail the role of glomerular endothelial HS in normal kidney physiology.
Methods:
Eps Homology Domain (Ehd3) is highly and selectively expressed in glomerular endothelial cells in the kidney in comparison to other organs, tissues and cells, making Ehd3 a key tool in specifically targeting the glomerular endothelium. An inducible Ehd3 knock-in mouse model, expressing Cre-recombinase (Ehd3-eGFPCreERT2) under the Ehd3 promoter, was generated to excise floxed genes specifically in glomerular endothelial cells. The experimental and littermate control mice were injected intraperitoneally with 75 mg tamoxifen/kg body weight for five consecutive days. Glomerular albumin permeability, FACS, lectin and immunofluorescence staining were used to fully characterise the mouse model.
Results:
GFP staining, which denotes Cre recombinase expression, colocalises with CD31 staining, an endothelial marker, but not with nephrin, a podocyte marker. The colocalisation was confirmed by Pearson correlation coefficient analysis, establishing Cre recombinase in glomerular endothelial cells in the kidney. EXT1 gene expression was significantly reduced in FACS-glomerular endothelial cells. No significant change in EXT1 expression was observed in non-endothelial renal cells, pointing to the selective knockdown of EXT1 in glomerular endothelial cells. Lectin LEL staining, known to bind to N-acetylglucosamine ([GlcNAc]1-3) found on HS chains, shows reduced glomerular endothelial glycocalyx coverage but no change in other microvasculature, e.g. cardiac endothelial glycocalyx coverage, assessed by our novel confocal peak-to-peak analysis, confirming specificity to glomerular endothelial glycocalyx. Importantly, EXT1 knockdown leads to an increase in glomerular capillary albumin permeability, assessed by our sensitive ex vivo glomerular albumin permeability assay.
Conclusion:
We have successfully established an in vivo animal model that allows glomerular endothelial cells to be specifically targeted in the kidney. Moreover, our study shows for the first time that genetic deletion of EXT1 selectively in glomerular endothelial cells reduced endothelial glycocalyx HS coverage and increased leakage of albumin in the glomerular capillaries. Strategies targeted at restoring glomerular endothelial HS could be of potential therapeutic value in diabetic kidney disease.
Lay summary:
Blood vessels are vital to every organ in the body, including the kidney. In diabetes, blood vessel damage is very common, causing serious diseases including heart attacks, eye disease and kidney damage leading to kidney failure. Hence there is an urgent need to find better ways to protect blood vessels in diabetes.
All blood vessels are lined by the surface coat called the endothelial glycocalyx. This is a gel-like layer that protects and keeps blood vessels healthy. This layer is damaged in diabetes contributing to kidney disease. Studies suggest that endothelial glycocalyx component heparan sulphate (HS) might be playing a vital role in diabetic kidney damage. Hence, we aim to study HS damage specifically in kidney blood vessels. To that end, we successfully generated a transgenic animal model that has reduced HS in the kidney blood vessels. Our results show that these kidney blood vessels are leakier than the blood vessels in the control (healthy) animal with intact HS, confirming that HS reduction specifically in the kidney blood vessels contributes to kidney damage.
Our studies support that maintaining the levels of HS in the glycocalyx could be a treatment to prevent blood vessel damage in diabetes and protect from kidney injury.
Biography:
Renal research is a fascinating and complex field of research. It is intriguing how the different glomerular cell types/layers work together, through cross-talks, to filter blood to produce urine. And how damage to one layer affects the function of the whole glomerular filtration barrier. I am interested in thoroughly understanding how a delicate layer called the glycocalyx, located at the interphase between the blood and the endothelium, contributes to the glomerular filtration barrier. The glycocalyx is damaged in disease states, including diabetic kidney disease. This has led me to investigate the importance of one component of glycocalyx, the carbohydrate molecule heparan sulphate (HS), and to show that maintaining levels of HS in the glycocalyx could be a treatment to prevent glomerular filtration barrier damage and kidney injury in diabetes.
A better understanding of how glycocalyx loss leads to glomerular filtration damage and kidney injury will allow us to design strategies to protect or restore the glycocalyx and attenuate kidney damage. We are using transgenic animal models and immortalised human kidney cell line to understand the critical role HS plays in the kidney. Hence, drugs with HS protecting properties could be therapeutically attractive. HS is already being used in people in other situations, for example in pancreas islet transplants and kidney transplants. Technology is available to design and manufacture HS which can be taken in tablet form. So, once we have confirmed the importance and potential of HS, progress towards it being used to maintain healthy glycocalyx and protect people with diabetes will be relatively rapid, with benefits to patients starting to be realised.
Kidney Research UK has made a major contribution to my work and career progression. Its funding enabled me to investigate the enzymes (matrix metalloproteases (MMPs)) involved in glycocalyx damage in diabetic kidney disease, which led to a publication in Kidney International. Follow-on work, also funded by the charity, is evaluating the translational potential of clinically available drugs with MMP inhibition properties on the glycocalyx and determining whether they protect from kidney damage. Moreover, the charity has funded my salary and enabled me to progress my career to become an independent research leader.

Viktoriia Vasylchenko
Authors and affiliations (if published):
Viktoriia Vasylchenko1,2, Rebecca R. Foster1, Olena Kuchmenko2
1University of Bristol
2National University of Kyiv-Mohyla Academy
Background:
Chronic kidney disease changes cellular metabolism and oxidative status. A balance between pro- and antioxidant systems is disturbed and shifted towards pro-oxidant reactions. Early detection is crucial for the treatment of kidney diseases, so detection of proteins and enzymes that determine oxidative status, including a new uremic toxin, indoxyl sulfate, and citrulline, a byproduct of nitric oxide can be useful for quality monitoring and potentially for the prevention of disease progression for patients with chronic kidney disease. Therefore, our study aimed to quantify citrulline and indoxyl sulfate and correlate indicators of oxidative status with lipids indexes at different stages of chronic kidney disease and other classical clinical markers.
Methods:
Biochemical (spectrophotometrically and colourimetric) statistical (Tukey`s range test and Spearman). Number of samples was 417.
Results:
Citrulline content was 1.3 times lower in donors, in comparison with those who had chronic kidney disease in the first, second and terminal stages. In comparison with patients who were in the third and fourth stages of pathogenesis, this difference was even greater, the content of citrulline in their serum was exactly two times higher than that of donors. The correlation between the content of citrulline in the serum and indoxyl sulfate (0.694), as well as their combination with other indicators, deserves special attention. Thus, the content of citrulline in blood serum is also positively correlated with creatinine (0.794), the content of total cholesterol (0.665) and low-density lipoprotein cholesterol (0.65). As for indoxyl sulfate, it also has close relationships with the content of citrulline in urine (0.588), creatinine (0.752), haemoglobin (-0.701) and the content of total cholesterol (0.837), low-density lipoprotein (0.757) and high-density lipoprotein (-0.783). There is a strong negative correlation between indoxyl sulfate and haemoglobin, as well as indoxyl sulfate and high-density lipoprotein cholesterol. Because, indeed, with a decrease in the content of the latter, indoxyl sulfate increases rapidly.
Conclusion:
Oxidative status is primarily reflected in changes in both quantitative and qualitative protein and lipid components, as well as their interrelationship. The difference in quantity of indoxyl sulfate and citrulline, and their interrelationship, can complement the picture of pathogenesis and help early detection of chronic kidney disease. Both compounds can be determined non-invasively, which is an advantage.
Lay summary:
Chronic kidney disease is a condition in which patients undergo “oxidative stress”. Cholesterol balance, toxins in the blood and proteins leaking into the urine are the main symptoms. We aim to understand the relationship between these better so that we may improve patient monitoring and predict possible complications, and potentially reduce these complications using therapeutic approaches. According to our results, these oxidative stress changes increase with disease progression and become permanent, as the body’s defence mechanisms are depleted. Kidney damage is reflected by a build-up of citrulline and indoxyl sulphate in the blood and appear to be related to each other and “bad cholesterol”. We believe this reflects loss of normal metabolism within cells.
Biography:
My name is Viktoriia Vasylchenko. I am a molecular biologist from National University Kyiv-Mohyla Academy, Ukraine and I am a fourth year PhD student. I became interested in the process of free radical formation and redox reactions in my bachelor course. Since then, I have been working in this area with my supervisor, Professor Olena Kuchmenko, studying issues around the development of complications in cardiovascular diseases. One aspect was extremely interesting to me; the course of disease in patients who also had kidney disease. That is why I chose to study oxidative or redox status in chronic kidney disease for my doctoral thesis.
I am in the last year of my PhD, and I’m delighted to complete my PhD with Dr Becky Foster, at Bristol Renal, and begin a complimentary research project in her group. We are focusing on inflammation and vascular glycocalyx damage as a key mechanism in the development of proteinuria as a first step of kidney disease, which perfectly facilitates the acceleration of my research. Through the generosity of Kidney Research UK, I have an incredible opportunity to share and discuss my results with colleagues and form new potential collaborations for future work in this area.