1
|
Borghol AH, Bou Antoun MT, Hanna C, Salih M, Rahbari-Oskoui FF, Chebib FT. Autosomal dominant polycystic kidney disease: an overview of recent genetic and clinical advances. Ren Fail 2025; 47:2492374. [PMID: 40268755 DOI: 10.1080/0886022x.2025.2492374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common inherited kidney disease, characterized by the progressive development of multiple kidney cysts, leading to a gradual decline in kidney function. ADPKD is also the fourth leading cause of kidney failure (KF) in adults. In addition to kidney manifestations, ADPKD is associated with various extrarenal features, including liver cysts, cardiovascular abnormalities, intracranial aneurysms, and chronic pain with significant impact on patients' quality of life. While several disease-modifying agents have been tested in ADPKD, tolvaptan remains the only approved drug by the US Food and Drug Administration. The Mayo Imaging Classification is currently the most practical tool for predicting rate of kidney disease progression in ADPKD. This review provides a comprehensive overview of ADPKD, focusing on its genetics, pathophysiology, clinical presentation, management, and prognostic tools. Advances in diagnostic imaging and genetic testing have improved the early detection of ADPKD, allowing better classification of patients and prediction of KF. The review also discusses current therapeutic approaches to ADPKD, including tolvaptan, a vasopressin V2-receptor antagonist. Additionally, we address specific issues in children and pregnant individuals with ADPKD. Despite substantial progress in understanding ADPKD, there is a large need for additional effective treatments and prognostic markers to provide a more personalized care for these patients.
Collapse
Affiliation(s)
- Abdul Hamid Borghol
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Florida PKD Center of Excellence, Jacksonville, FL, USA
| | - Marie Therese Bou Antoun
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Florida PKD Center of Excellence, Jacksonville, FL, USA
| | - Christian Hanna
- Division of Pediatric Nephrology and Hypertension, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mahdi Salih
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Fouad T Chebib
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Florida PKD Center of Excellence, Jacksonville, FL, USA
| |
Collapse
|
2
|
Cordido A, Nuñez-González L, Lamas-González O, Vizoso-González M, Bravo S, Díaz C, Banales JM, García-González MA. Therapeutic opportunities in polycystic kidney and liver disease through extracellular matrix dynamics. Biochem Pharmacol 2025; 236:116858. [PMID: 40081770 DOI: 10.1016/j.bcp.2025.116858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Autosomal Dominant and Autosomal Recessive Polycystic Kidney Disease (ADPKD and ARPKD) are, respectively, common and rare forms of polycystic disorders, characterized by the formation and progressive growth of cysts from tubules in the kidneys and bile ducts in the liver. Alterations in the extracellular matrix (ECM) and in the activity of matrix metalloproteases (MMPs), both associated with fibrosis, have been shown to be important factors in cystic growth and progression of these diseases. We used tandem mass spectrometry (LC-MS/MS) to identify the most enriched proteins and pathways in an orthologous rapidly progressive mouse model of ADPKD: Pkd1flox/floxTamCre. This information was used to discover and validate novel therapeutic targets in orthologous models of ADPKD (Pkd1flox/floxTamCre) and ARPKD (Pkdh1del3-4/del3-4). ECM related pathways and expression levels of MMPs were among the most dysregulated cellular processes in polycystic kidney and liver. Selective inhibition of MMPs by marimastat (MTT) altered the ECM response and resulted in inhibition of collecting duct-derived cyst growth, delay of global kidney cyst progression and rescue of liver phenotype by normalized MMPs expression and significant reduction in fibrosis. This phenotypic improvement was further enhanced by treatment of MTT and tolvaptan, indicating an additive benefit to targeting the fibrotic and growth pathways in cysts. As conclusion, targeting of MMPs are important in ECM dysregulation and offers a new potential therapeutic strategy for both kidney and bile duct derived fibrocystic disease in ADPKD and ARPKD. Such approaches can have additive benefits with other treatment approaches, such as tolvaptan.
Collapse
Affiliation(s)
- Adrian Cordido
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, N°11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| | - Laura Nuñez-González
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, N°11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| | - Olaya Lamas-González
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, N°11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
| | - Marta Vizoso-González
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, N°11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| | - Susana Bravo
- Proteomics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
| | - Candido Díaz
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, N°11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; Nephrology Service, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| | - Miguel A García-González
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, N°11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain; RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain.
| |
Collapse
|
3
|
Clerici S, Boletta A. Metabolic reprogramming in polycystic kidney disease and other renal ciliopathies. EMBO Mol Med 2025:10.1038/s44321-025-00239-x. [PMID: 40263625 DOI: 10.1038/s44321-025-00239-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
Primary cilia are solitary organelles formed by a microtubule-based skeleton protruding in a single copy on the surface of most cells. Alterations in their function cause a plethora of human conditions collectively called the ciliopathies. The kidney is frequently and severely affected in the ciliopathies, presenting with a spectrum of phenotypes. Cyst formation is a common trait of all renal ciliopathies. Besides this common manifestation, however, the renal ciliopathies present with profoundly different phenotypes, resulting in either polycystic kidney disease (PKD) or nephronophthisis (NPH) phenotypes. The past decade has seen a surge of studies highlighting metabolic reprogramming as a major feature of PKD, with a distinct involvement of mitochondrial dysfunction. This discovery has brought forward the development of novel therapeutic approaches. More recent evidence suggests that primary cilia modulate the mitochondrial production of energy in response to environmental cues. Here, we summarize the evidence available to date and propose a more general involvement of metabolic and mitochondrial alterations in the renal ciliopathies that might in principle help defining the profoundly different, and potentially opposite, manifestations observed.
Collapse
Affiliation(s)
- Sara Clerici
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
4
|
Nikonorova IA, desRanleau E, Jacobs KC, Saul J, Walsh JD, Wang J, Barr MM. Polycystins recruit cargo to distinct ciliary extracellular vesicle subtypes in C. elegans. Nat Commun 2025; 16:2899. [PMID: 40180912 PMCID: PMC11968823 DOI: 10.1038/s41467-025-57512-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
Therapeutic use of tiny extracellular vesicles (EVs) requires understanding cargo loading mechanisms. Here, we use a modular proximity labeling approach to identify the cargo of ciliary EVs associated with the transient receptor potential channel polycystin-2 PKD-2 of C. elegans. Polycystins are conserved ciliary proteins and cargo of EVs; dysfunction causes polycystic kidney disease in humans and mating deficits in C. elegans. We discover that polycystins localize with specific cargo on ciliary EVs: polycystin-associated channel-like protein PACL-1, dorsal and ventral polycystin-associated membrane C-type lectins PAMLs, and conserved tumor necrosis factor receptor-associated factor (TRAF) TRF-1 and TRF-2. Loading of these components to EVs relies on polycystin-1 LOV-1. Our modular EV-TurboID approach can be applied in both cell- and tissue-specific manners to define the composition of distinct EV subtypes, addressing a major challenge of the EV field.
Collapse
Affiliation(s)
- Inna A Nikonorova
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - Elizabeth desRanleau
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Katherine C Jacobs
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Josh Saul
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Jonathon D Walsh
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Juan Wang
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Maureen M Barr
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
5
|
Mustafa RA, Kawtharany H, Kalot MA, Lumpkins CY, Kimminau KS, Creed C, Fowler K, Perrone RD, Jaure A, Cho Y, Baron D, Yu ASL. Establishing Meaningful Patient-Centered Outcomes with Relevance for Patients with Polycystic Kidney Disease: Patient, Caregiver, and Researcher Priorities for Research in Polycystic Kidney Disease. KIDNEY360 2025; 6:573-582. [PMID: 39808496 DOI: 10.34067/kid.0000000695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Key Points
Participants are less concerned about mortality than they are about kidney health.Psychological effect emerged as the most important theme regarding the effects of polycystic kidney disease on patients.
Background
Patient involvement in research can help to ensure that the evidence generated aligns with their needs and priorities. In the Establishing Meaningful Patient-Centered Outcomes with Relevance for Patients with Polycystic Kidney Disease (PKD) project, we aimed to identify patient-important outcomes and discuss the effect of PKD on patients.
Methods
Nine focus groups were held with adult patients with PKD, caregivers, and clinical or research experts in PKD. We used a nominal, multivote technique to rank patient-important outcomes to be prioritized by future PKD research. We conducted a thematic analysis of verbatim transcriptions to identify themes regarding the effect of PKD on their daily lives. Other focus group topics included insurability and patient engagement.
Results
Ninety patients and/or caregivers and eight clinicians and/or researchers participated in the focus groups. Nine focus groups yielded 35 outcomes important to patients that were grouped into six categories, ranked in order of importance: kidney health, comorbidities, lifestyle, psychological effect, family and awareness, and mortality. Regarding the effect of PKD on the patient's daily lives, we identified five themes, listed in order of importance: psychological effect, effect on daily living, issues affecting decision making, health care-related issues, and PKD-specific testing dilemmas.
Conclusions
This study of stakeholder engagement in patients with PKD revealed important priorities and values that should be considered for future research and when caring for patients with PKD. Future research should focus on kidney health and managing comorbidities in patients with PKD. This will help to bridge the knowledge gap and develop meaningful comparative effectiveness research in PKD.
Collapse
Affiliation(s)
- Reem A Mustafa
- Division of Nephrology and Hypertension,Department of Internal Medicine, University of Kansas Medical Centre, Kansas City, Kansas
| | - Hassan Kawtharany
- Evidence-Based Practice and Impact Center, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Mohamad A Kalot
- Evidence-Based Practice and Impact Center, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | | | - Kim S Kimminau
- Department of Family and Community Medicine and Office of the EVCHA/Dean, School of Medicine, University of Missouri, Columbia, Missouri
| | - Cathy Creed
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Kevin Fowler
- The Voice of the Patient, Inc., Saint Louis, Missouri
| | - Ronald D Perrone
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Allison Jaure
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - Yeoungjee Cho
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - David Baron
- Polycystic Kidney Disease Foundation, Kansas City, Missouri
| | - Alan S L Yu
- Division of Nephrology and Hypertension,Department of Internal Medicine, University of Kansas Medical Centre, Kansas City, Kansas
| |
Collapse
|
6
|
Fernandez JM, Rodriguez-Pérez JC, Lorenzo-Medina MM, Rodriguez-Esparragon F, Quevedo-Reina JC, Hernandez-Socorro CR. Longitudinal assessment of measured and estimated glomerular filtration-rate in autosomal dominant polycystic kidney disease: Real practice experience. World J Nephrol 2025; 14:99044. [PMID: 40134648 PMCID: PMC11755237 DOI: 10.5527/wjn.v14.i1.99044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/07/2024] [Accepted: 12/27/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Equations for estimation glomerular filtration rate (eGFR) have been associated with poor clinical performance and their clinical accuracy and reliability have been called into question. AIM To assess the longitudinal changes in measured glomerular filtration rate (mGFR) in patients with autosomal dominant polycystic kidney disease (ADPKD). METHODS Analysis of an ambispective data base conducted on consecutive patients diagnosed with ADPKD. The mGFR was assessed by iohexol clearance; while eGFR was calculated by three different formulas: (1) The chronic kidney disease epidemiology collaboration (CKD-EPI); (2) Modification of diet in renal disease (MDRD); and (3) The 24-hour urine creatinine clearance (CrCl). The primary end-points were the mean change in mGFR between the baseline and final visit, as well as the comparison of the mean change in mGFR with the change estimated by the different formulas. RESULTS Thirty-seven patients were included in the study. As compared to baseline, month-6 mGFR was significantly decrease by -4.4 mL/minute ± 10.3 mL/minute (P = 0.0132). However, the CKD-EPI, MDRD, and CrCl formulas underestimated this change by 48.3%, 89.0%, and 45.8% respectively, though none of these differences reached statistical significance (P = 0.3647; P = 0.0505; and P = 0.736, respectively). The discrepancies between measured and estimated glomerular filtration rate values, as evaluated by CKD-EPI (r = 0.29, P = 0.086); MDRD (r = 0.19, P = 0.272); and CrCl (r = 0.09, P = 0.683), were not correlated with baseline mGFR values. CONCLUSION This study indicated that eGFR inaccurately reflects the decline in mGFR and cannot reliably track changes over time. This poses significant challenges for clinical decision-making, particularly in treatment strategies.
Collapse
Affiliation(s)
- Juan M Fernandez
- Medical Manager Southern Europe, Baxter Healthcare Ltd., Madrid 28830, Spain
- Escuela de Doctorado, Universidad De Las Palmas De Gran Canaria, Las Palmas 35001, Canary Islands, Spain
| | - José C Rodriguez-Pérez
- Department of Research, Universidad Fernando Pessoa Canarias, Las Palmas 35450, Canary Islands, Spain
| | - M Mercedes Lorenzo-Medina
- Department of Clinical Chemistry, Hospital Universitario De Gran Canaria Doctor Negrín, Las Palmas 35010, Canary Islands, Spain
| | - Fancisco Rodriguez-Esparragon
- Department of Research, Hospital Universitario De Gran Canaria Doctor Negrín, Las Palmas 35010, Canary Islands, Spain
| | - Juan C Quevedo-Reina
- Department of Nephrology, Hospital Universitario De Gran Canaria Doctor Negrín, Las Palmas 35010, Canary Islands, Spain
| | - Carmen R Hernandez-Socorro
- Department of Radiology, Hospital Universitario De Gran Canaria Doctor Negrín, Las Palmas 35010, Canary Islands, Spain
| |
Collapse
|
7
|
Ji Y, Liu S, Zhang Y, Min Y, Wei L, Guan C, Yu H, Zhang Z. Lysine crotonylation in disease: mechanisms, biological functions and therapeutic targets. Epigenetics Chromatin 2025; 18:13. [PMID: 40119392 PMCID: PMC11929287 DOI: 10.1186/s13072-025-00577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/24/2025] [Indexed: 03/24/2025] Open
Abstract
Lysine crotonylation (Kcr), a previously unknown post-translational modification (PTM), plays crucial roles in regulating cellular processes, including gene expression, chromatin remodeling, and cellular metabolism. Kcr is associated with various diseases, including neurodegenerative disorders, cancer, cardiovascular conditions, and metabolic syndromes. Despite advances in identifying crotonylation sites and their regulatory enzymes, the molecular mechanisms by which Kcr influences disease progression remain poorly understood. Understanding the interplay between Kcr and other acylation modifications may reveal opportunities for developing targeted therapies. This review synthesizes current research on Kcr, focusing on its regulatory mechanisms and disease associations, and highlights insights into future exploration in epigenetics and therapeutic interventions.
Collapse
Affiliation(s)
- Yu Ji
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Shanshan Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Yiqiao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Yiyang Min
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Luyang Wei
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Chengjian Guan
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Huajing Yu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
8
|
Alshriem LA, Buqaileh R, Alorjani Q, AbouAlaiwi W. Ciliary Ion Channels in Polycystic Kidney Disease. Cells 2025; 14:459. [PMID: 40136708 PMCID: PMC11941060 DOI: 10.3390/cells14060459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Polycystic kidney disease (PKD) is the most common hereditary disorder that disrupts renal function and frequently progresses to end-stage renal disease. Recent advances have elucidated the critical role of primary cilia and ciliary ion channels, including transient receptor potential (TRP) channels, cystic fibrosis transmembrane conductance regulator (CFTR), and polycystin channels, in the pathogenesis of PKD. While some channels primarily function as chloride conductance channels (e.g., CFTR), others primarily regulate calcium (Ca+2) homeostasis. These ion channels are essential for cellular signaling and maintaining the normal kidney architecture. Dysregulation of these pathways due to genetic mutations in PKD1 and PKD2 leads to disrupted Ca+2 and cAMP signaling, aberrant fluid secretion, and uncontrolled cellular proliferation, resulting in tubular cystogenesis. Understanding the molecular mechanisms underlying these dysfunctions has opened the door for innovative therapeutic strategies, including TRPV4 activators, CFTR inhibitors, and calcimimetics, to mitigate cyst growth and preserve renal function. This review summarizes the current knowledge on the roles of ciliary ion channels in PKD pathophysiology, highlights therapeutic interventions targeting these channels, and identifies future research directions for improving patient outcomes.
Collapse
Affiliation(s)
- Lubna A. Alshriem
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (L.A.A.); (R.B.); (Q.A.)
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Raghad Buqaileh
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (L.A.A.); (R.B.); (Q.A.)
| | - Qasim Alorjani
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (L.A.A.); (R.B.); (Q.A.)
| | - Wissam AbouAlaiwi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (L.A.A.); (R.B.); (Q.A.)
| |
Collapse
|
9
|
Cadnapaphornchai MA, Dell KM, Gimpel C, Guay-Woodford LM, Gulati A, Hartung EA, Liebau MC, Mallett AJ, Marlais M, Mekahli D, Piccirilli A, Seeman T, Tindal K, Winyard PJD. Polycystic Kidney Disease in Children: The Current Status and the Next Horizon. Am J Kidney Dis 2025:S0272-6386(25)00772-3. [PMID: 40113156 DOI: 10.1053/j.ajkd.2025.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 03/22/2025]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD) are inherited disorders that share many features such as kidney cysts, hypertension, urinary concentrating defects, and progressive chronic kidney disease. Underlying pathogenic mechanisms for both include cilia dysfunction and dysregulated intracellular signaling. ADPKD has been traditionally regarded as an adult-onset disease, whereas ARPKD has been classically described as an infantile or childhood condition. However, clinicians must recognize that both disorders can present across all age groups ranging from fetal life and infancy to childhood and adolescence, as well as adulthood. Here we highlight the points of overlap and distinct features for these disorders with respect to pathogenesis, diagnostic modalities (radiological and genetic), clinical assessment, and early therapeutic management. In particular, we consider key issues at two critical points for transition of care, i.e., fetal life to infancy and adolescence to adulthood. These timepoints are poorly covered in the extant literature. Therefore, we recommend guiding principles for transitions of clinical care at these critical junctures in the lifespan. While there is no cure for polycystic kidney disease (PKD), recent insights into pathogenic mechanisms have identified promising therapeutic targets that are currently being evaluated in a growing portfolio of clinical trials. We summarize the key findings from these largely adult-based trials and discuss the implications for designing child-focused studies. Finally, we look forward to the next horizon for childhood PKD, highlighting gaps in our current knowledge, and discussing future directions and strategies to attenuate the full burden of disease for children affected with PKD.
Collapse
Affiliation(s)
| | - Katherine M Dell
- Cleveland Clinic Children's Institute and Case Western Reserve University, Cleveland, USA
| | | | - Lisa M Guay-Woodford
- Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, USA.
| | - Ashima Gulati
- Children's National Hospital and the George Washington University, Washington, DC, USA
| | - Erum A Hartung
- Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, USA
| | - Max C Liebau
- University Hospital Cologne and the University of Cologne, Cologne, Germany
| | - Andrew J Mallett
- Townsville University Hospital and James Cook University , Queensland, Australia
| | - Matko Marlais
- Great Ormond Street Hospital for Children and UCL Great Ormond Street Institute of Child Health, London, UK
| | - Djalila Mekahli
- KU Leuven University and UZ Leuven Hospital, Leuven, Belgium
| | | | - Tomas Seeman
- Charles University and Ostrava University, Prague and Ostrava, Czech Republic
| | | | | |
Collapse
|
10
|
Devlin LA, Dewhurst RM, Sudhindar PD, Sayer JA. Renal ciliopathies. Curr Top Dev Biol 2025; 163:229-305. [PMID: 40254346 DOI: 10.1016/bs.ctdb.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Primary cilia are essential cellular organelles with pivotal roles in many signalling pathways. Here we provide an overview of the role of primary cilia within the kidney, starting with primary ciliary structure and key protein complexes. We then highlight the specialised functions of primary cilia, emphasising their role in a group of diseases known as renal ciliopathies. These conditions include forms of polycystic kidney disease, nephronophthisis, and other syndromic ciliopathies, such as Joubert syndrome and Bardet-Biedl syndrome. We explore models of renal ciliopathies, both in vitro and in vivo, shedding light on the molecular mechanisms underlying these diseases including Wnt and Hedgehog signalling pathways, inflammation, and cellular metabolism. Finally, we discuss therapeutic approaches, from current treatments to cutting-edge preclinical research and clinical trials.
Collapse
Affiliation(s)
- Laura A Devlin
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rebecca M Dewhurst
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Praveen D Sudhindar
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John A Sayer
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Renal Services, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; National Institute for Health Research, Newcastle Biomedical Research Centre, Newcastle Upon Tyne, United Kingdom.
| |
Collapse
|
11
|
Yang Y, Liu H, Yuan H, Lyu K, Zhong H, Li Y, Cao D, Zhao W, Zhang H, Xiong B, Chen D, Guo D. Design of Selective BRD4 Inhibitors for the Treatment of Autosomal Dominant Polycystic Kidney Disease. J Med Chem 2025; 68:5257-5274. [PMID: 39945752 DOI: 10.1021/acs.jmedchem.4c02128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Epigenetic modulation plays a pivotal role in restraining tumor progression by governing gene expression and protein function. Autosomal dominant polycystic kidney disease (ADPKD), characterized by neoplastic-like progression, can be managed by inhibiting cyst expansion. Of note, the epigenetic regulator BRD4 has been implicated in ADPKD's development. Our prior research unveiled a class of (pyrazol-3-yl) pyrimidin-4-amine compounds as potent BRD4 inhibitors with additional kinase inhibition, which might induce unwanted biological activities. To address this, this study focused on creating selective BRD4 inhibitors through structure-guided design, minimizing off-target kinase interactions. Specifically, compound 23 emerged as an efficacious and selective BRD4 inhibitor in cellular and embryonic kidney models of ADPKD, along with encouraging outcomes in murine models. Collectively, these results highlight the therapeutic potential of targeted BRD4 inhibition as a safe and efficacious strategy for managing ADPKD.
Collapse
Affiliation(s)
- Yueyue Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongli Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| | - Haoxing Yuan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| | - Kaikai Lyu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haiyang Zhong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| | - Yanlian Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Danyan Cao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wenchao Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| | - Haoran Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| | - Bing Xiong
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Danqi Chen
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
12
|
Tran U, Streets AJ, Smith D, Decker E, Kirschfink A, Izem L, Hassey JM, Rutland B, Valluru MK, Bräsen JH, Ott E, Epting D, Eisenberger T, Ong AC, Bergmann C, Wessely O. BICC1 Interacts with PKD1 and PKD2 to Drive Cystogenesis in ADPKD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.27.608867. [PMID: 39253489 PMCID: PMC11383298 DOI: 10.1101/2024.08.27.608867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is primarily of adult-onset and caused by pathogenic variants in PKD1 or PKD2 . Yet, disease expression is highly variable and includes very early-onset PKD presentations in utero or infancy. In animal models, the RNA-binding molecule Bicc1 has been shown to play a crucial role in the pathogenesis of PKD. To study the interaction between BICC1, PKD1 and PKD2 we combined biochemical approaches, knockout studies in mice and Xenopus, genetic engineered human kidney cells as well as genetic association studies in a large ADPKD cohort. We first demonstrated that BICC1 physically binds to the proteins Polycystin-1 and -2 encoded by PKD1 and PKD2 via distinct protein domains. Furthermore, PKD was aggravated in loss-of-function studies in Xenopus and mouse models resulting in more severe disease when Bicc1 was depleted in conjunction with Pkd1 or Pkd2 . Finally, in a large human patient cohort, we identified a sibling pair with a homozygous BICC1 variant and patients with very early onset PKD (VEO-PKD) that exhibited compound heterozygosity of BICC1 in conjunction with PKD1 and PKD2 variants. Genome editing demonstrated that these BICC1 variants were hypomorphic in nature and impacted disease-relevant signaling pathways. These findings support the hypothesis that BICC1 cooperates functionally with PKD1 and PKD2 , and that BICC1 variants may aggravate PKD severity highlighting RNA metabolism as an important new concept for disease modification in ADPKD.
Collapse
|
13
|
Yang G, Schock ME, Wall PW, Lu Y, Cui D. Bilateral Polycystic Kidney Disease and Inferior Vena Cava (IVC) Thrombosis: A Cadaveric Study. Cureus 2025; 17:e80765. [PMID: 40248555 PMCID: PMC12004418 DOI: 10.7759/cureus.80765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic kidney disease and one of the most common causes of end-stage kidney disease. The occurrence of bilateral polycystic kidney disease with liver cysts is uncommon. A case of bilateral polycystic kidney disease, found to have liver cysts and inferior vena cava thrombosis in a cadaveric study, is presented in this study. Dissection was performed on a 90-year-old Caucasian male cadaver. Both kidneys were enlarged and contained multiple cysts with normal renal tissue interposed. When comparing the two kidneys, the cysts on the left were notably larger. There was a partial absence of renal capsules. Additionally, a polycystic liver and an inferior vena cava filter were found during the dissection. The patient's history of venous thrombosis is indicated by the previously inserted inferior vena cava filter.
Collapse
Affiliation(s)
- Gongchao Yang
- Department of Advanced Biomedical Education, University of Mississippi Medical Center, Jackson, USA
| | - Morgan E Schock
- School of Medicine, University of Mississippi Medical Center, Jackson, USA
| | - Peyton W Wall
- School of Medicine, University of Mississippi Medical Center, Jackson, USA
| | - Yuefeng Lu
- Department of Advanced Biomedical Education, University of Mississippi Medical Center, Jackson, USA
| | - Dongmei Cui
- Department of Advanced Biomedical Education, University of Mississippi Medical Center, Jackson, USA
| |
Collapse
|
14
|
Scholes G, Prawer Y, Ryan J, Verma K, Jayasinghe K. Exploring new gene-disease associations in polycystic kidney disease: a case report highlighting the importance of a precise genomic diagnosis. J Nephrol 2025; 38:765-769. [PMID: 39382785 DOI: 10.1007/s40620-024-02113-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Affiliation(s)
- Gemma Scholes
- Department of Nephrology, Monash Health, Clayton, VIC, Australia.
| | - Yael Prawer
- Department of Genetics, Monash Health, Clayton, VIC, Australia
| | - Jessica Ryan
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
| | - Kunal Verma
- Department of Genetics, Monash Health, Clayton, VIC, Australia
| | - Kushani Jayasinghe
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Monash University, Clayton, VIC, Australia
| |
Collapse
|
15
|
Noishiki H, Yamauchi H, Komaki K, Kusaba T, Tamagaki K. A Rare Coexisting Presentation of Autosomal Dominant Polycystic Kidney Disease With Rapid Deterioration of Renal Function and Neurofibromatosis Type 1. Cureus 2025; 17:e79931. [PMID: 40051696 PMCID: PMC11883721 DOI: 10.7759/cureus.79931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2025] [Indexed: 03/09/2025] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common hereditary kidney disorders, characterized by progressive cyst development. Neurofibromatosis type 1 (NF1) is another autosomal dominant disorder, characterized by café-au-lait spots, neurofibromas, and multisystem involvement. We report the case of an 18-year-old male with ADPKD and NF1, referred due to progressively worsening renal dysfunction. His initial estimated glomerular filtration rate (eGFR) was 71.9 mL/min/1.73m², with MRI showing bilateral cystic renal enlargement (total kidney volume: 758 mL). One year later, his eGFR declined to 56.7 mL/min/1.73m², and kidney volume increased by 10.4% over one year. Tolvaptan was initiated, and he remains under follow-up. Mutations in the PKD1/PKD2, which are responsible for ADPKD, affect intracellular signaling, including the mammalian target of the rapamycin (mTOR) pathway, leading to cyst formation and progression, while NF1 mutations overactivate the Ras proteins. His disease progression was more severe than that of his father with ADPKD alone, suggesting NF1 may have accelerated cyst enlargement. The co-occurrence of ADPKD and NF1 is extremely rare, with only a few cases reported in the past.
Collapse
Affiliation(s)
- Haruna Noishiki
- Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, JPN
| | - Hiroko Yamauchi
- Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, JPN
| | - Kazumi Komaki
- Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, JPN
| | - Tetsuro Kusaba
- Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, JPN
| | - Keiichi Tamagaki
- Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, JPN
| |
Collapse
|
16
|
Matarneh A, Akkari A, Sardar S, Miller R, Verma N, Ghahramani N, Farooq U. A Challenging Case of Inferior Vena Cava Compression in an Adult Polycystic Kidney Disease Patient. A Case Report. Clin Case Rep 2025; 13:e70283. [PMID: 40034720 PMCID: PMC11873366 DOI: 10.1002/ccr3.70283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/01/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
Adult polycystic kidney disease (ADPKD) is a multi-system genetic disorder characterized by the development and progressive enlargement of fluid-filled cysts in both kidneys, along with other organs. As one of the main causes of kidney failure, ADPKD can progress to end-stage renal disease (ESRD), with over 50% of affected individuals progressing to ESRD by age 50. The symptoms in ADPKD are variable, with some patients experiencing nonspecific signs, while others present with symptoms related to the mass effect of enlarged kidneys on surrounding structures. This case report highlights an unusual presentation of ADPKD in a patient who developed symptoms of inferior vena cava (IVC) compression. Remarkably, these symptoms improved after bilateral nephrectomies, suggesting that bilateral nephrectomy would provide help in these situations.
Collapse
Affiliation(s)
- Ahmad Matarneh
- Department of NephrologyPenn State Milton S Hershey Medical CenterHersheyPennsylvaniaUSA
| | - Abdelraouf Akkari
- Department of NephrologyPenn State Milton S Hershey Medical CenterHersheyPennsylvaniaUSA
| | - Sundus Sardar
- Department of NephrologyPenn State Milton S Hershey Medical CenterHersheyPennsylvaniaUSA
| | - Ronald Miller
- Department of NephrologyPenn State Milton S Hershey Medical CenterHersheyPennsylvaniaUSA
| | - Navin Verma
- Department of NephrologyPenn State Milton S Hershey Medical CenterHersheyPennsylvaniaUSA
| | - Nasrollah Ghahramani
- Department of NephrologyPenn State Milton S Hershey Medical CenterHersheyPennsylvaniaUSA
| | - Umar Farooq
- Department of NephrologyPenn State Milton S Hershey Medical CenterHersheyPennsylvaniaUSA
| |
Collapse
|
17
|
Yamaguchi H, Meyer MD, Barrell WB, Faisal M, Berdeaux R, Liu KJ, Komatsu Y. The primary cilia: Orchestrating cranial neural crest cell development. Differentiation 2025; 142:100818. [PMID: 39500655 PMCID: PMC11911094 DOI: 10.1016/j.diff.2024.100818] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 03/17/2025]
Abstract
Primary cilia (hereafter "cilia") are microtubule-based antenna-like organelles projecting from the surface of vertebrate cells. Cilia can serve as cellular antennae controlling cell growth and differentiation. Absent or dysfunctional cilia frequently lead to craniofacial anomalies known as craniofacial ciliopathies. However, the detailed pathological mechanisms of craniofacial ciliopathies remain unclear. This perspective discusses our current understanding of the role of cilia in cranial neural crest cells. We also describe potential mechanisms of ciliogenesis in cranial neural crest cells, which may contribute to unraveling the complex pathogenesis of craniofacial ciliopathies.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX, USA
| | - William B Barrell
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, UK
| | - Maryam Faisal
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Bioengineering, Rice University George R. Brown School of Engineering, 77005, Houston, TX, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; CellChorus INC, Houston, TX, USA
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, UK
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 77030, Houston, TX, USA.
| |
Collapse
|
18
|
Vitulano C, Forcina G, Colosimo S, Frattolillo V, Villani AV, Marzuillo P, Miraglia Del Giudice E, Di Sessa A. A miRNA-Based Approach in Autosomal Dominant Polycystic Kidney Disease: Challenges and Insights from Adult to Pediatric Evidence. Mol Diagn Ther 2025; 29:183-193. [PMID: 39820940 DOI: 10.1007/s40291-024-00761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2024] [Indexed: 01/19/2025]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) represents the most common inherited kidney disorder leading to kidney failure in a significant percentage of patients over time. Although previously considered as an adult disease, robust evidence demonstrated that clinical manifestations might occur during childhood and adolescence. Therefore, early identification and treatment of the disease are of cardinal importance for pediatricians to ensure the best long-term outcomes. To date, licensed treatment options are limited but promising potential therapeutic targets are emerging. Among these, an intriguing pathophysiological role for microRNAs as small molecules with a critical role in regulating gene expression has been considered possible in ADPKD. Indeed, numerous circulating microRNAs have been found to be dysregulated in ADPKD, suggesting their potential role as biomarkers and therapeutic targets. Based on this background, further detailed insights into the mechanisms of miRNAs contributing to ADPKD development might pave the way for their effective application as a targeted treatment in young patients with ADPKD. We aimed to summarize the most recent evidence in this fascinating research area, providing a comprehensive overview of the current landscape of specific microRNAs in ADPKD as a potential innovative therapeutic strategy for these young patients.
Collapse
Affiliation(s)
- Caterina Vitulano
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 4, 80138, Naples, Italy
| | - Gianmario Forcina
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 4, 80138, Naples, Italy
| | - Simone Colosimo
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 4, 80138, Naples, Italy
| | - Vittoria Frattolillo
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 4, 80138, Naples, Italy
| | - Annalisa Valentina Villani
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 4, 80138, Naples, Italy
| | - Pierluigi Marzuillo
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 4, 80138, Naples, Italy
| | - Emanuele Miraglia Del Giudice
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 4, 80138, Naples, Italy
| | - Anna Di Sessa
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 4, 80138, Naples, Italy.
| |
Collapse
|
19
|
Bracciamà V, Vaisitti T, Mioli F, Faini AC, Del Prever GMB, Martins VH, Camilla R, Mattozzi F, Pieretti S, Luca M, Romeo CM, Saglia C, Migliorero M, Arruga F, Carli D, Amoroso A, Lonardi P, Deaglio S, Peruzzi L. Matching clinical and genetic data in pediatric patients at risk of developing cystic kidney disease. Pediatr Nephrol 2025; 40:743-753. [PMID: 39384646 PMCID: PMC11747002 DOI: 10.1007/s00467-024-06548-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/11/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Cystic kidney disease is a heterogeneous group of hereditary and non-hereditary pathologic conditions, associated with the development of renal cysts. These conditions may be present both in children and adults. Cysts can even be observed already during the prenatal age, and pediatric patients with cysts need to be clinically monitored. An early clinical and genetic diagnosis is therefore mandatory for optimal patient management. The aim of this study was to perform genetic analyses in patients with echographic evidence of kidney cysts to provide an early molecular diagnosis. METHODS A cohort of 70 pediatric patients was enrolled and clinically studied at the time of first recruitment and at follow-up. Genetic testing by clinical exome sequencing was performed and a panel of genes responsible for "cystic kidneys" was analyzed to identify causative variants. Sanger validation and segregation studies were exploited for the final classification of the variants and accurate genetic counseling. RESULTS Data showed that 53/70 of pediatric patients referred with a clinical suspicion of cystic kidney disease presented a causative genetic variant. In a significant proportion of the cohort (24/70), evidence of hyper-echogenic/cystic kidneys was already present in the prenatal period, even in the absence of a positive family history. CONCLUSIONS This study suggests that cystic kidney disease may develop since the very early stages of life and that screening programs based on ultrasound scans and genetic testing play a critical role in diagnosis, allowing for better clinical management and tailored genetic counseling to the family.
Collapse
Affiliation(s)
- Valeria Bracciamà
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Tiziana Vaisitti
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Fiorenza Mioli
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Angelo Corso Faini
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giulia Margherita Brach Del Prever
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vitor Hugo Martins
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Roberta Camilla
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Francesca Mattozzi
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Silvia Pieretti
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Maria Luca
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Carmelo Maria Romeo
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Claudia Saglia
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Martina Migliorero
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Francesca Arruga
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Diana Carli
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Antonio Amoroso
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Pietro Lonardi
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Silvia Deaglio
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Licia Peruzzi
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| |
Collapse
|
20
|
Macke EL, Miller AR, Colwell CM, Gonzalez MH, Hunter J, Venkata LPR, Walker L, Wheeler G, Wilson RK, Mardis ER, Miller KE, Mathew MT, Chaudhari BP, Akkari Y. Optical Genome Mapping (OGM) Identifies Multiple Structural Variants in a Case With Atypical Phelan-McDermid Syndrome. Am J Med Genet A 2025; 197:e63929. [PMID: 39535355 DOI: 10.1002/ajmg.a.63929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/11/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Here we describe a neonate exhibiting hypotonia, macrocephaly, renal cysts, and respiratory failure requiring tracheostomy and ventilator support. Genetic analysis via rapid genome sequencing (rGS) identified a loss on chromosome 4 encompassing polycystin-2 (PKD2) and a loss on chromosome 22 encompassing SH3 and Multiple Ankyrin Repeat Domains 3 (SHANK3), indicative of Phelan-McDermid syndrome. Further analysis via traditional karyotyping, Optical Genome Mapping (OGM), and PacBio long-read sequencing revealed a more complex landscape of chromosomal rearrangements in this individual, including a balanced 3;12 translocation, and an unbalanced 17;22 translocation. The proband's phenotypic presentation is thought to be the result of Phelan-McDermid syndrome and represents an expansion of the described phenotypes to include significant respiratory failure. This study underscores the challenges and importance of comprehensive genetic testing in elucidating complex presentations and highlights the need for complementary testing methods to overcome limitations in resolution.
Collapse
Affiliation(s)
- Erica L Macke
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Anthony R Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Caitlyn M Colwell
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Maria Hernandez Gonzalez
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jesse Hunter
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Lakshmi Prakruthi Rao Venkata
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Lauren Walker
- Division of Genetics and Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Gregory Wheeler
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Richard K Wilson
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Elaine R Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Katherine E Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Mariam T Mathew
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Bimal P Chaudhari
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Division of Neonatology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Yassmine Akkari
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
21
|
Fan CN, Chung CH, Chien WC, Tsao CH, Weng TH, Wu KL, Chiang WF, Yen CC, Chan JS, Hsiao PJ. Impact of Chronic Kidney Disease on Aortic Dissection in Patients with Polycystic Kidney Disease: A Fifteen-year Nationwide Population-based Cohort Study in Taiwan. Int J Med Sci 2025; 22:1493-1503. [PMID: 40093803 PMCID: PMC11905265 DOI: 10.7150/ijms.106518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/15/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Aortic dissection is a life-threatening condition associated with polycystic kidney disease (PKD). Additionally, PKD often progresses to chronic kidney disease (CKD), a known risk factor for cardiovascular disease. However, the impact of CKD on aortic dissection, particularly in patients with PKD, remains unclear. This study aims to investigate the effects of both CKD and PKD on aortic dissection. Materials and methods: This nationwide, population-based, retrospective cohort study used data from the National Health Insurance Research Database (NHIRD) in Taiwan. The primary outcome evaluated in this study was the cumulative incidence of aortic dissection, compared between PKD patients and a control group without PKD over a 15-year follow-up period. CKD subgroup analyses were performed to further assess the impact of CKD progression on the development of aortic dissection. Results: From 2000 to 2015, this study included 9,192 PKD patients and 36,768 matched controls without PKD from the NHIRD. Our findings demonstrated that PKD patients who developed aortic dissection had a higher incidence of comorbidities, including hypertension and coronary artery disease. Aortic dissection was more prevalent among male patients, individuals over 45 years of age, and those in the lowest insured premium group. PKD patients had a 2.53-fold higher adjusted hazard ratio (HR) for developing aortic dissection compared to the control group (95% CI: 1.74 to 3.66, p < 0.001). Notably, PKD patients with concurrent hypertension had a 7.77-fold increased risk of aortic dissection (95% CI: 4.97 to 12.13, p < 0.001). In CKD subgroup analyses, PKD patients without CKD and those with CKD had adjusted HRs of 1.74 and 3.38, respectively (p < 0.001). Among PKD patients with CKD, those who initiated hemodialysis (HD) and those who did not showed adjusted HRs of 3.95 and 2.74, respectively, for aortic dissection (p < 0.001). Conclusion: These findings indicate that the risk of aortic dissection in PKD patients significantly increases with CKD progression. Additionally, hypertension is an independent risk factor for aortic dissection in PKD patients. Careful management of blood pressure and strategies to prevent CKD progression may reduce the incidence of aortic dissection in this population.
Collapse
Affiliation(s)
- Chia-Ning Fan
- Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- Department of Surgery, National Defense Medical Center, Division of Cardiovascular Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | - Chi-Hsiang Chung
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Wu-Chien Chien
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Chang-Huei Tsao
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Microbiology & Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Tzu-Hsuan Weng
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lin Wu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Wen-Fang Chiang
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan
| | - Chih-Chien Yen
- Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- Department of Surgery, National Defense Medical Center, Division of Cardiovascular Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | - Jenq-Shyong Chan
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan
| | - Po-Jen Hsiao
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan
- Department of Life Sciences, National Central University, Taoyuan City, Taiwan
| |
Collapse
|
22
|
Hedger G, Yen HY. The Influence of Phosphoinositide Lipids in the Molecular Biology of Membrane Proteins: Recent Insights from Simulations. J Mol Biol 2025; 437:168937. [PMID: 39793883 PMCID: PMC7617384 DOI: 10.1016/j.jmb.2025.168937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/29/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
The phosphoinositide family of membrane lipids play diverse and critical roles in eukaryotic molecular biology. Much of this biological activity derives from interactions of phosphoinositide lipids with integral and peripheral membrane proteins, leading to modulation of protein structure, function, and cellular distribution. Since the discovery of phosphoinositides in the 1940s, combined molecular biology, biophysical, and structural approaches have made enormous progress in untangling this vast and diverse cellular network of interactions. More recently, in silico approaches such as molecular dynamics simulations have proven to be an asset in prospectively identifying, characterising, explaining the structural basis of these interactions, and in the best cases providing atomic level testable hypotheses on how such interactions control the function of a given membrane protein. This review details a number of recent seminal discoveries in phosphoinositide biology, enabled by advanced biomolecular simulation, and its integration with molecular biology, biophysical, and structural biology approaches. The results of the simulation studies agree well with experimental work, and in a number of notable cases have arrived at the key conclusion several years in advance of the experimental structures. SUMMARY: Hedger and Yen review developments in simulations of phosphoinositides and membrane proteins.
Collapse
Affiliation(s)
- George Hedger
- Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, SW7 2AZ, UK.
| | - Hsin-Yung Yen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Department of Chemistry, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK
| |
Collapse
|
23
|
Gittus M, Haley H, Harris T, Borrows S, Padmanabhan N, Gale D, Simms R, Williams T, Acquaye A, Wong A, Chan M, Lee E, Ong AC. Commentary: Tolvaptan for Autosomal Dominant Polycystic Kidney Disease (ADPKD) - an update. BMC Nephrol 2025; 26:79. [PMID: 39953521 PMCID: PMC11827152 DOI: 10.1186/s12882-025-03960-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/10/2025] [Indexed: 02/17/2025] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) affects up to 70 000 people in the UK and the most common inherited cause of end-stage kidney disease (ESKD). It is generally a late-onset multisystem disorder characterised by bilateral kidney cysts, liver cysts and an increased risk of intracranial aneurysms. Approximately 50% of people with ADPKD reach ESKD by age 60. Disease-associated pain, discomfort, fatigue, emotional distress and, impaired mobility can impact health-related quality of life. The approval of tolvaptan, a vasopressin V2 receptor antagonist, has greatly advanced the care for people with ADPKD, shifting the focus from general chronic kidney disease management to targeted therapeutic approaches. While guidance from NICE and SMC provides a foundational framework, this is not clear or comprehensive enough to offer practical guidance for healthcare professionals in real-world settings. This commentary expands on the previous United Kingdom Kidney Association (UKKA) commentary in 2016 with an updated evidence base, the incorporation of real-world data and expert opinion to provide practical guidance to healthcare professionals. Through co-development with people affected by ADPKD, it now incorporates valuable patient perspectives and offers practical recommendations for the UK kidney community seeking to harmonise the quality of care of all people with ADPKD.
Collapse
Affiliation(s)
- Matt Gittus
- University of Sheffield, Sheffield, United Kingdom.
- Sheffield Teaching Hospitals NHS Trust, Sheffield, United Kingdom.
| | - Helen Haley
- University Hospitals of North Midlands, Birmingham, United Kingdom
| | | | - Sarah Borrows
- Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | | | - Danny Gale
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Roslyn Simms
- Sheffield Teaching Hospitals NHS Trust, Sheffield, United Kingdom
| | | | - Aaron Acquaye
- Hull and East Yorkshire Hospitals NHS Trust, Hull, United Kingdom
| | - Alisa Wong
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Melanie Chan
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Eduardo Lee
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Albert Cm Ong
- University of Sheffield, Sheffield, United Kingdom.
- Sheffield Teaching Hospitals NHS Trust, Sheffield, United Kingdom.
| |
Collapse
|
24
|
Klemens CA, Fedoriuk M, Semenikhina M, Stefanenko M, Zietara A, Levchenko V, Dissanayake LV, Palygin O, Staruschenko A. Electrolyte and metabolite composition of cystic fluid from a rat model of ARPKD. Commun Biol 2025; 8:230. [PMID: 39948436 PMCID: PMC11825955 DOI: 10.1038/s42003-025-07631-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Fluid-filled cysts are the key feature of polycystic kidney disease, which eventually leads to renal failure. We analyzed the composition of cyst fluid from a rat model of autosomal recessive polycystic kidney disease, the PCK rat, and identified sexual differences. Our results demonstrate that the ion composition of cyst fluid differs from that of urine or plasma. Untargeted metabolomics combined with transcriptomic data identified tryptophan metabolism, enzyme metabolism, steroid hormone biosynthesis, and fatty acid metabolism as pathways differing between male and female PCK rats. We quantified 42 amino acids in the cyst fluid (PCK only), plasma, and urine of male and female PCK rats and Sprague Dawley rats. Taurine was the most concentrated amino acid present in the cyst fluid, and PCK rat urinary taurine excretion was over 3-fold greater than Sprague Dawley rats. Understanding the composition of cyst fluid provides valuable insights into disease pathophysiology and may help identify potential dietary or pharmacological interventions to mitigate disease progression and improve patient outcomes.
Collapse
Affiliation(s)
- Christine A Klemens
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, USA.
| | - Mykhailo Fedoriuk
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | - Mariia Stefanenko
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Adrian Zietara
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Oleg Palygin
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, USA.
- James A. Haley Veterans' Hospital, Tampa, FL, USA.
| |
Collapse
|
25
|
Wang W, Chen J, Zhan L, Zou H, Wang L, Guo M, Gao H, Xu J, Wu W. Iron and ferroptosis in kidney disease: molecular and metabolic mechanisms. Front Immunol 2025; 16:1531577. [PMID: 39975561 PMCID: PMC11835690 DOI: 10.3389/fimmu.2025.1531577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Maintaining iron homeostasis is necessary for kidney functioning. There is more and more research indicating that kidney disease is often caused by iron imbalance. Over the past decade, ferroptosis' role in mediating the development and progression of renal disorders, such as acute kidney injury (renal ischemia-reperfusion injury, drug-induced acute kidney injury, severe acute pancreatitis induced acute kidney injury and sepsis-associated acute kidney injury), chronic kidney disease (diabetic nephropathy, renal fibrosis, autosomal dominant polycystic kidney disease) and renal cell carcinoma, has come into focus. Thus, knowing kidney iron metabolism and ferroptosis regulation may enhance disease therapy. In this review, we discuss the metabolic and molecular mechanisms of iron signaling and ferroptosis in kidney disease. We also explore the possible targets of ferroptosis in the therapy of renal illness, as well as their existing limitations and future strategies.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jingdi Chen
- Department of orthopedics, The Airborne Military Hospital, Wuhan, Hubei, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Handong Zou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mengmeng Guo
- The First Clinical College of Wuhan University, Wuhan, Hubei, China
| | - Hang Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
26
|
Zhong J, Liu J, Mutchler AL, Yang H, Kirabo A, Shelton EL, Kon V. Moving toward a better understanding of renal lymphatics: challenges and opportunities. Pediatr Nephrol 2025:10.1007/s00467-025-06692-7. [PMID: 39899153 DOI: 10.1007/s00467-025-06692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025]
Abstract
The development of lymphatic-specific markers has enabled detailed visualization of the lymphatic vascular network that has greatly enhanced our ability to explore this often-overlooked system. Lymphatics remove fluid, solutes, macromolecules, and cells from the interstitium and return them to circulation. The kidneys have lymphatics. As in other organs, the kidney lymphatic vessels are highly sensitive to changes in the local microenvironment. The sensitivity to its milieu may be especially relevant in kidneys because they are central in regulating fluid homeostasis and clearance of metabolites delivered into and eliminated from the renal interstitial compartment. Numerous physiologic conditions and diseases modify the renal interstitial volume, pressure, and composition that can, in turn, influence the growth and function of the renal lymphatics. The impact of the renal microenvironment is further heightened by the fact that kidneys are encapsulated. This review considers the development, structure, and function of the renal lymphatic vessels and explores how factors within the kidney interstitial compartment modify their structure and functionality. Moreover, although currently there are no pharmaceutical agents that specifically target the lymphatic network, we highlight several medications currently used in children with kidney disease and hypertension that have significant but underappreciated effects on lymphatics.
Collapse
Affiliation(s)
- Jianyong Zhong
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jing Liu
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA
- Department of Nephrology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Ashley L Mutchler
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Haichun Yang
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elaine L Shelton
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA
| | - Valentina Kon
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA.
| |
Collapse
|
27
|
Guerriero CJ, Carattino MD, Sharp KG, Kantz LJ, Gresko NP, Caplan MJ, Brodsky JL. Identification of polycystin 2 missense mutants targeted for endoplasmic reticulum-associated degradation. Am J Physiol Cell Physiol 2025; 328:C483-C499. [PMID: 39714991 DOI: 10.1152/ajpcell.00776.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disorder leading to end-stage renal disease. ADPKD arises from mutations in the PKD1 and PKD2 genes, which encode polycystin 1 (PC1) and polycystin 2 (PC2), respectively. PC2 is a nonselective cation channel, and disease-linked mutations disrupt normal cellular processes, including signaling and fluid secretion. In this study, we investigate whether disease-causing missense mutations compromise PC2 folding, an event that can lead to endoplasmic reticulum-associated degradation (ERAD). To this end, we first developed a new yeast PC2 expression system. We show that the yeast system provides a tractable model to investigate PC2 biogenesis and that a disease-associated PC2 mutant, D511V, exhibits increased polyubiquitination and accelerated proteasome-dependent degradation compared with wild-type PC2. In contrast to wild-type PC2, the PC2 D511V variant also failed to improve the growth of yeast strains that lack endogenous potassium transporters, highlighting a loss of channel function at the cell surface and a new assay for loss-of-function PKD2 variants. In HEK293 cells, both D511V along with another disease-associated mutant, R322Q, were targeted for ERAD. Consistent with defects in protein folding, the surface localization of these PC2 variants was increased by incubation at low-temperature in HEK293 cells, underscoring the potential to pharmacologically rescue these and perhaps other misfolded PC2 alleles. Together, our study supports the hypothesis that select PC2 missense variants are degraded by ERAD, the potential for screening PKD2 alleles in a new genetic system, and the possibility that chemical chaperone-based therapeutic interventions might be used to treat ADPKD.NEW & NOTEWORTHY This study indicates that select missense mutations in PC2, a protein that when mutated leads to ADPKD, result in protein misfolding and degradation via the ERAD pathway. Our work leveraged a new yeast model and an HEK293 cell model to discover the mechanism underlying PC2 instability and demonstrates the potential for pharmacological rescue. We also suggest that targeting the protein misfolding phenotype with chemical chaperones may offer new therapeutic strategies to manage ADPKD-related protein dysfunction.
Collapse
Affiliation(s)
- Christopher J Guerriero
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Marcelo D Carattino
- Departments of Medicine and Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Katherine G Sharp
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Luke J Kantz
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Nikolay P Gresko
- Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
28
|
Xu B, Staruschenko A. Endoplasmic reticulum-associated degradation: a novel therapeutic avenue for ADPKD. Am J Physiol Cell Physiol 2025; 328:C574-C575. [PMID: 39751181 DOI: 10.1152/ajpcell.00982.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025]
Affiliation(s)
- Biyang Xu
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida, United States
- James A. Haley Veteran's Hospital, Tampa, Florida, United States
| |
Collapse
|
29
|
Torres VE, Ahn C, Barten TRM, Brosnahan G, Cadnapaphornchai MA, Chapman AB, Cornec-Le Gall E, Drenth JPH, Gansevoort RT, Harris PC, Harris T, Horie S, Liebau MC, Liew M, Mallett AJ, Mei C, Mekahli D, Odland D, Ong ACM, Onuchic LF, Pei YPC, Perrone RD, Rangan GK, Rayner B, Torra R, Balk EM, Gordon CE, Earley A, Mustafa RA, Devuyst O. KDIGO 2025 clinical practice guideline for the evaluation, management, and treatment of autosomal dominant polycystic kidney disease (ADPKD): executive summary. Kidney Int 2025; 107:234-254. [PMID: 39848746 DOI: 10.1016/j.kint.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 01/25/2025]
Abstract
The Kidney Disease: Improving Global Outcomes (KDIGO) 2025 Clinical Practice Guideline for the Evaluation, Management, and Treatment of Autosomal Dominant Polycystic Kidney Disease (ADPKD) represents the first KDIGO guideline on this subject. Its scope includes nomenclature, diagnosis, prognosis, and prevalence; kidney manifestations; chronic kidney disease (CKD) management and progression, kidney failure, and kidney replacement therapy; therapies to delay progression of kidney disease; polycystic liver disease; intracranial aneurysms and other extrarenal manifestations; lifestyle and psychosocial aspects; pregnancy and reproductive issues; pediatric issues; and approaches to the management of people with ADPKD. The guideline has been developed with patient partners, clinicians, and researchers around the world, with the goal to generate a useful resource for healthcare providers and patients by providing actionable recommendations. The development of this guideline followed an explicit process of evidence review and appraisal, based on a rigorous, formal systematic literature review. The strength of recommendations follows the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. The guideline also provides practice points serving to direct clinical care or activities relating to areas for which a systematic review was not conducted. Limitations of the evidence are discussed. Research recommendations to address gaps in knowledge, and implications for policy and payment, are provided. The guideline targets a broad audience of healthcare providers, people living with ADPKD, and stakeholders involved in the various aspects of ADPKD care.
Collapse
Affiliation(s)
- Vicente E Torres
- Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.
| | - Curie Ahn
- Laboratory of Transplantation Immunology, Seoul National University, Seoul, South Korea
| | - Thijs R M Barten
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Godela Brosnahan
- Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Melissa A Cadnapaphornchai
- Division of Nephrology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Arlene B Chapman
- Section of Nephrology, University of Chicago, Chicago, Illinois, USA
| | - Emilie Cornec-Le Gall
- Department of Nephrology, Centre de reference MAladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), University Hospital of Brest, Brest, France; Division of Nephrology, Inserm UMR1078, Brest, France
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ron T Gansevoort
- Department of Nephrology, University of Groningen, Groningen, The Netherlands
| | - Peter C Harris
- Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tess Harris
- Polycystic Kidney Disease Charity UK, London, UK
| | - Shigeo Horie
- Department of Urology, Juntendo University, Tokyo, Japan
| | - Max C Liebau
- Department of Pediatrics, Center for Rare Diseases, Medical Faculty, Cologne, Germany; Department of Pediatrics, Center for Family Health, Medical Faculty, Cologne, Germany; Department of Pediatrics, Center for Molecular Medicine, Medical Faculty, Cologne, Germany; University Hospital Cologne, Cologne, Germany
| | | | - Andrew J Mallett
- Department of Renal Medicine, Townsville University Hospital, Townsville, Queensland, Australia
| | - Changlin Mei
- Department of Nephrology, Changzheng Hospital, Shanghai, China
| | - Djalila Mekahli
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Pediatric Nephrology, University Hospital Leuven, Leuven, Belgium
| | | | - Albert C M Ong
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Luiz F Onuchic
- Division of Nephrology, Universidade de São Paulo, São Paulo, Brazil
| | - York P-C Pei
- Division of Nephrology, University of Toronto, Toronto, Ontario, Canada
| | - Ronald D Perrone
- Division of Nephrology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Gopala K Rangan
- Department of Renal Medicine, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Brian Rayner
- Division of Nephrology and Hypertension, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Roser Torra
- Department of Nephrology, Fundació Puigvert, IR Sant Pau, Barcelona, Spain
| | - Ethan M Balk
- Center for Evidence Synthesis in Health, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Craig E Gordon
- Division of Nephrology, Tufts University School of Medicine, Boston, Massachusetts, USA; Center for Evidence Synthesis in Health, Brown University School of Public Health, Providence, Rhode Island, USA
| | | | - Reem A Mustafa
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Kansas, Kansas City, Kansas, USA
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Division of Nephrology, Cliniques universitaires Saint-Luc, UCLouvain Medical School, Brussels, Belgium.
| |
Collapse
|
30
|
Devuyst O, Ahn C, Barten TR, Brosnahan G, Cadnapaphornchai MA, Chapman AB, Cornec-Le Gall E, Drenth JP, Gansevoort RT, Harris PC, Harris T, Horie S, Liebau MC, Liew M, Mallett AJ, Mei C, Mekahli D, Odland D, Ong AC, Onuchic LF, P-C Pei Y, Perrone RD, Rangan GK, Rayner B, Torra R, Mustafa R, Torres VE. KDIGO 2025 Clinical Practice Guideline for the Evaluation, Management, and Treatment of Autosomal Dominant Polycystic Kidney Disease (ADPKD). Kidney Int 2025; 107:S1-S239. [PMID: 39848759 DOI: 10.1016/j.kint.2024.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 01/25/2025]
|
31
|
Stanajic-Petrovic G, Keck M, Barbe P, Urman A, Correia E, Isnard P, Duong Van Huyen JP, Chmeis K, Diarra SS, Palea S, Theodoro F, Nguyen AL, Castelli F, Pruvost A, Zhao W, Mendre C, Mouillac B, Bienaimé F, Robin P, Kessler P, Llorens-Cortes C, Servent D, Nozach H, Maillère B, Guo D, Truillet C, Gilles N. A Snake Toxin Derivative for Treatment of Hyponatremia and Polycystic Kidney Diseases. J Am Soc Nephrol 2025; 36:181-192. [PMID: 39431458 PMCID: PMC11801765 DOI: 10.1681/asn.0000000505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Key Points MQ232, a disulfide-bond reticulated peptide derived from a natural snake toxin, was optimized as a new aquaretic drug candidate. MQ232 showed very low acute and chronic toxicity in rat and a biodistribution in mice strongly in favor of the kidney organs. MQ232 induced a sole aquaretic effect and demonstrated high in vivo activities on hyponatremia and polycystic kidney disease models. Background Vaptans were developed at the end of the previous century as vasopressin type 2 receptor antagonists. Tolvaptan is the most prescribed vaptan for hyponatremia and autosomal dominant polycystic kidney disease (ADPKD). However, its use is not as widespread as it should be due to price issues, a narrow therapeutic window, and some side effects. With the aim of discovering new efficient and safer vasopressin type 2 receptor antagonists, we screened animal venoms and identified several peptide toxins. Among them, mambaquaretin 1 (MQ1) displayed unique biological properties in that regard that it was the starting point for the development of a potential drug candidate. Methods Human T-cell assays and bioinformatics were used to mitigate MQ1 immunogenicity risk. MQ232 biodistribution in mice was performed by positron emission tomography. Pharmacodynamics, pharmacokinetics, and acute and chronic toxicity tests were performed on control rats. A rat experimental model of desmopressin-induced hyponatremia, ex vivo mice model of kidney cysts, and mice orthologous model of ADPKD were used to validate MQ232 efficacy in these pathologies. Results Three mutations were introduced in MQ1 to mitigate its immunogenicity risk. A fourth gain-of-function mutation was added to generate MQ232. MQ232's safety was demonstrated by a first toxic dose as high as 3000 nmol/kg and a strong kidney organ selectivity by positron emission tomography imaging, while showing almost no interaction with the liver. MQ232's efficacy was first demonstrated with an effective dose of 3 nmol/kg in a hyponatremic model and then in polycystic kidney models, on which MQ232 significantly reduced cyst growth. Conclusions We demonstrated, using diverse translational techniques and minimizing animal use, MQ232's safety and efficacy in several rodent models of hyponatremia and ADPKD.
Collapse
Affiliation(s)
- Goran Stanajic-Petrovic
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Mathilde Keck
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Peggy Barbe
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Apolline Urman
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Evelyne Correia
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Pierre Isnard
- Anatomie et Cytologie Pathologiques, CHU Necker-Enfants Malades, Paris, France
| | | | - Khawla Chmeis
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | | | - Stefano Palea
- Humana Biosciences, Prologue Biotech, Labège, France
| | - Frederic Theodoro
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Anvi-Laëtitia Nguyen
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Florence Castelli
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Alain Pruvost
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Wenchao Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | | | - Bernard Mouillac
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Frank Bienaimé
- Service d'Explorations Fonctionnelles, Département Croissance et Signalisation, Institut Necker Enfants Malades, INSERM U1151, CNRS UMR 8253, Université de Paris Cité, Hôpital Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Philippe Robin
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Pascal Kessler
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Catherine Llorens-Cortes
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Denis Servent
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Hervé Nozach
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Bernard Maillère
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Charles Truillet
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Nicolas Gilles
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| |
Collapse
|
32
|
Elhassan EAE, O'Donoghue D, Heneghan S, Teltsh O, Sarihan S, Osman SM, Clince M, Synnott D, Craig S, Hudson A, Doyle B, Lappin D, Sexton DJ, Casserly L, Holian J, Magee C, Denton M, Sweeney C, Awan A, McCann E, Cavalleri GL, Benson KA, Conlon PJ. Phenotypic outcomes of PKD1 compared with non-PKD1 genetically confirmed autosomal dominant polycystic kidney disease. J Nephrol 2025:10.1007/s40620-024-02184-3. [PMID: 39881088 DOI: 10.1007/s40620-024-02184-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/29/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Autosomal Dominant Polycystic Kidney Disease (ADPKD) represents the most common monogenic cause of kidney failure. While identifying genetic variants predicts disease progression, characterization of recently described ADPKD-like variants is limited. We explored disease progression and genetic spectrum of genetically-confirmed ADPKD families with PKD1 and non-PKD1 variants. METHODS In this observational study, we evaluated the clinical (ADPKD-related complications, estimated glomerular filtration rate (eGFR) decline, and progression to kidney failure), radiological (height-adjusted total kidney volume (ht-TKV)), and genetic characteristics of ADPKD families referred to the Irish Kidney Gene Project. Logistic regression and Kaplan-Meier analyses examined relationships between genetic variants and disease progression. RESULTS Genomic sequencing was performed on 261 ADPKD families, and 75.8% (198/261 families, comprising 391 individuals) were identified to harbor pathogenic/likely pathogenic variants; 74.2% (147/198) PKD1 families and 23.2% (46/198) non-PKD1 families, which include PKD2 (n = 29 families), IFT140 variants (n = 4), ALG5, DNAJB11 and NEK8 (n = 3 each), ALG8 and ALG9 variants (n = 2 each). The remaining 2.6% (5/198) accounted for non-ADPKD variants. Compared to PKD1, non-PKD1 families were characterized by a milder phenotype; milder eGFR decline (- 1.4 mL/min/1.73m2/year vs. - 3.2; p < 0.001), smaller ht-TKV (449.7 mL/m vs. 1769; p 0.002) and a delayed progression towards kidney failure (73 vs. 52 years; HR: 0.12, p < 0.001 [95% CI: 0.07-0.19]). ADPKD-NEK8 heterozygotes demonstrated earlier progression to kidney failure (average age 8 vs. 49 years for PKD1; Bonferroni-corrected p 0.017). CONCLUSION Non-PKD1 variants have heterogeneous phenotypic and genotypic attributes resulting in milder disease, although ADPKD-NEK8 is an important exception with early progression.
Collapse
Affiliation(s)
- Elhussein A E Elhassan
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland.
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Darragh O'Donoghue
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
| | - Sophia Heneghan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland
| | - Omri Teltsh
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland
| | - Sahin Sarihan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland
| | - Shohdan M Osman
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michelle Clince
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
| | - David Synnott
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
| | - Sophie Craig
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
| | - Amy Hudson
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
| | - Brendan Doyle
- Department of Pathology, Beaumont Hospital, Dublin, Ireland
- Department of Pathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David Lappin
- Department of Nephrology, University Hospital Galway, Galway, Ireland
| | - Donal J Sexton
- Department of Nephrology, St Jame's Hospital, Dublin, Ireland
- Trinity Health Kidney Center, Trinity College Dublin, Dublin, Ireland
| | - Liam Casserly
- Department of Renal Medicine, University Hospital Limerick, Limerick, Ireland
| | - John Holian
- Nephrology Department St Vincent's University Hospital, Dublin, Ireland
| | - Colm Magee
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
| | - Mark Denton
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
| | - Clodagh Sweeney
- Department for Paediatric Nephrology and Transplantation, Children's Health Ireland, Dublin, Ireland
| | - Atif Awan
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department for Paediatric Nephrology and Transplantation, Children's Health Ireland, Dublin, Ireland
| | - Emma McCann
- The Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Gianpiero L Cavalleri
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland
- FutureNeuro SFI Research Centre, Royal College of Surgeons, Dublin, Ireland
| | - Katherine A Benson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland
| | - Peter J Conlon
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
33
|
Watson D, Mentch F, Billings J, Ostberg K, March ME, Kalish JM, Li D, Cannon I, Guay-Woodford LM, Hartung E, Strong A. Elucidating the Molecular Landscape of Cystic Kidney Disease: Old Friends, New Friends and Some Surprises. Am J Med Genet A 2025:e64011. [PMID: 39888183 DOI: 10.1002/ajmg.a.64011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Cystic kidney diseases (CyKD) are a diverse group of disorders affecting more than 1 in 1000 individuals. Over 120 genes are implicated, primarily encoding components of the primary cilium, transcription factors, and morphogens. Prognosis varies greatly by molecular diagnosis. Causal variants are not identified in 10%-60% of individuals due to our limited understanding of CyKD. To elucidate the molecular landscape of CyKD, we queried the CAG Biobank using the ICD10 codes N28.1, Q61.1, Q61.11, Q61.19, Q61.2, Q61.3, and Q61.8 to identify individuals with CyKD. One hundred eight individuals met clinical criteria for CyKD and underwent proband-only exome sequencing. Causal variants were identified in 86/108 (80%) individuals. The most common molecular diagnoses were PKD1-related autosomal dominant polycystic kidney disease (32/108; 30%) and autosomal recessive polycystic kidney disease (21/108; 19%). Other common molecular diagnoses were ciliopathy syndromes (7/108; 6.5%) and Tuberous Sclerosis (6/108; 5.6%). Seven individuals had variants in genes not previously associated with CyKD (7/108; 6.5%). Candidate genes were identified in five individuals (5/108; 4.5%). Discordance between molecular and clinical diagnosis was present in two individuals. We demonstrate a high molecular diagnosis rate in individuals with CyKD that can result in diagnostic reclassification, supporting a role for genetic testing in CyKD.
Collapse
Affiliation(s)
- Deborah Watson
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Frank Mentch
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan Billings
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kayleigh Ostberg
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michael E March
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jennifer M Kalish
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - India Cannon
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lisa M Guay-Woodford
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Nephrology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Erum Hartung
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Nephrology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Alanna Strong
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
Guadarrama E, Vanoye CG, DeCaen PG. Defining the Polycystin Pharmacophore Through HTS & Computational Biophysics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632808. [PMID: 39868095 PMCID: PMC11761769 DOI: 10.1101/2025.01.13.632808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Background and Purpose Polycystins (PKD2, PKD2L1) are voltage-gated and Ca2+-modulated members of the transient receptor potential (TRP) family of ion channels. Loss of PKD2L1 expression results in seizure-susceptibility and autism-like features in mice, whereas variants in PKD2 cause autosomal dominant polycystic kidney disease. Despite decades of evidence clearly linking their dysfunction to human disease and demonstrating their physiological importance in the brain and kidneys, the polycystin pharmacophore remains undefined. Contributing to this knowledge gap is their resistance to drug screening campaigns, which are hindered by these channels' unique subcellular trafficking to organelles such as the primary cilium. PKD2L1 is the only member of the polycystin family to form constitutively active ion channels on the plasma membrane when overexpressed. Experimental Approach HEK293 cells stably expressing PKD2L1 F514A were pharmacologically screened via high-throughput electrophysiology to identify potent polycystin channel modulators. In-silico docking analysis and mutagenesis were used to define the receptor sites of screen hits. Inhibition by membrane-impermeable QX-314 was used to evaluate PKD2L1's binding site accessibility. Key Results Screen results identify potent PKD2L1 antagonists with divergent chemical core structures and highlight striking similarities between the molecular pharmacology of PKD2L1 and voltage-gated sodium channels. Docking analysis, channel mutagenesis, and physiological recordings identify an open-state accessible lateral fenestration receptor within the pore, and a mechanism of inhibition that stabilizes the PKD2L1 inactivated state. Conclusion and Implication Outcomes establish the suitability of our approach to expand our chemical knowledge of polycystins and delineates novel receptor moieties for the development of channel-specific antagonists in TRP channel research.
Collapse
Affiliation(s)
- Eduardo Guadarrama
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Carlos G. Vanoye
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Paul G. DeCaen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
35
|
Nikolsky KS, Kopylov AT, Nakhod VI, Potoldykova NV, Enikeev DV, Butkova TV, Kulikova LI, Malsagova KA, Rudnev VR, Petrovskiy DV, Izotov AA, Kaysheva AL. Plasma proteome fingerprint in kidney diseases. Front Mol Biosci 2025; 11:1494779. [PMID: 39896931 PMCID: PMC11782039 DOI: 10.3389/fmolb.2024.1494779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction Kidney diseases pose a serious healthcare problem because of their high prevalence, worsening of patients' quality of life, and high mortality. Patients with kidney diseases are often asymptomatic until disease progression starts. Expensive renal replacement therapy options, such as dialysis or kidney transplant, are required for end-stage kidney disease. Early diagnosis of kidney pathology is crucial for slowing down or curbing further damage. This study aimed to analyze the features of the protein composition of blood plasma in patients with the most common kidney pathologies: kidney calculus, kidney cyst, and kidney cancer. Methods The study involved 75 subjects. Proteins associated with kidney pathologies (CFB, SERPINA3, HPX, HRG, SERPING1, HBB, ORM2, and CP) were proposed. These proteins are important participants of complement and coagulation cascade activation and lipid metabolism. Results The revealed phosphorylated proteoforms (CFB, C4A/C4B, F2, APOB, TTR, and NRAP) were identified. For them, modification sites were mapped on 3D protein models, and the potential role in formation of complexes with native partner proteins was assessed. Discussion The study demonstrates that the selected kidney pathologies have a similar proteomic profile, and patients can be classified into kidney pathology groups with an accuracy of (70-80)%.
Collapse
Affiliation(s)
- Kirill S. Nikolsky
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Arthur T. Kopylov
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Valeriya I. Nakhod
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Natalia V. Potoldykova
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Dmitry V. Enikeev
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Tatiana V. Butkova
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Liudmila I. Kulikova
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Kristina A. Malsagova
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Vladimir R. Rudnev
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Denis V. Petrovskiy
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Alexander A. Izotov
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Anna L. Kaysheva
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
36
|
Jiang D, Li P, Lu Y, Tao J, Hao X, Wang X, Wu W, Xu J, Zhang H, Li X, Chen Y, Jin Y, Zhang L. A feedback loop between Paxillin and Yorkie sustains Drosophila intestinal homeostasis and regeneration. Nat Commun 2025; 16:570. [PMID: 39794306 PMCID: PMC11724037 DOI: 10.1038/s41467-024-55255-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/04/2024] [Indexed: 01/13/2025] Open
Abstract
Balanced self-renewal and differentiation of stem cells are crucial for maintaining tissue homeostasis, but the underlying mechanisms of this process remain poorly understood. Here, from an RNA interference (RNAi) screen in adult Drosophila intestinal stem cells (ISCs), we identify a factor, Pax, which is orthologous to mammalian PXN, coordinates the proliferation and differentiation of ISCs during both normal homeostasis and injury-induced midgut regeneration in Drosophila. Loss of Pax promotes ISC proliferation while suppressing its differentiation into absorptive enterocytes (ECs). Mechanistically, our findings demonstrate that Pax is a conserved target gene of the Hippo signaling pathway in both Drosophila and mammals. Subsequent investigations have revealed Pax interacts with Yki and enhances its cytoplasmic localization, thereby establishing a feedback regulatory mechanism that attenuates Yki activity and ultimately inhibits ISCs proliferation. Additionally, Pax induces the differentiation of ISCs into ECs by activating Notch expression, thus facilitating the differentiation process. Overall, our study highlights Pax as a pivotal component of the Hippo and Notch pathways in regulating midgut homeostasis, shedding light on this growth-related pathway in tissue maintenance and intestinal function.
Collapse
Affiliation(s)
- Dan Jiang
- The Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Pengyue Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi Lu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiaxin Tao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xue Hao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaodong Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Wei Wu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jinjin Xu
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Haoen Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaoyu Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yixing Chen
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yunyun Jin
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China.
| | - Lei Zhang
- The Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China.
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China.
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
37
|
Seufert F, Pérez-Hernández G, Pándy-Szekeres G, Guixà-González R, Langenhan T, Gloriam DE, Hildebrand PW. Generic residue numbering of the GAIN domain of adhesion GPCRs. Nat Commun 2025; 16:246. [PMID: 39747076 PMCID: PMC11697300 DOI: 10.1038/s41467-024-55466-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
The GPCR autoproteolysis inducing (GAIN) domain is an ancient protein fold ubiquitous in adhesion G protein-coupled receptors (aGPCR). It contains a tethered agonist necessary and sufficient for receptor activation. The GAIN domain is a hotspot for pathological mutations. However, the low primary sequence conservation of GAIN domains has thus far hindered the knowledge transfer across different GAIN domains in human receptors as well as species orthologs. Here, we present a scheme for generic residue numbering of GAIN domains, based on structural alignments of over 14,000 modeled GAIN domain structures. This scheme is implemented in the GPCR database (GPCRdb) and elucidates the domain topology across different aGPCRs and their homologs in a large panel of species. We identify conservation hotspots and statistically cancer-enriched positions in human aGPCRs and show the transferability of positional and structural information between GAIN domain homologs. The GAIN-GRN scheme provides a robust strategy to allocate structural homologies at the primary and secondary levels also to GAIN domains of polycystic kidney disease 1/PKD1-like proteins, which now renders positions in both GAIN domain types comparable to one another. In summary, our work enables researchers to generate hypothesis and rationalize experiments related to GAIN domain function and pathology.
Collapse
Affiliation(s)
- Florian Seufert
- Institute for Medical Physics and Biophysics, Leipzig University, Medical Faculty, Leipzig, Germany
| | - Guillermo Pérez-Hernández
- Institute for Medical Physics and Biophysics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gáspár Pándy-Szekeres
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark
- Medicinal Chemistry Research Group, HUN-REN Research Center for Natural Sciences, Magyar Tudósok körútja 2., Budapest, Hungary
| | - Ramon Guixà-González
- Institute for Medical Physics and Biophysics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Tobias Langenhan
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
- Comprehensive Cancer Center Central Germany (CCCG), Leipzig, Germany
- Institute of Biology, Faculty of Life Sciences, Leipzig University, Leipzig, Germany
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark.
| | - Peter W Hildebrand
- Institute for Medical Physics and Biophysics, Leipzig University, Medical Faculty, Leipzig, Germany.
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Leipzig, Germany.
| |
Collapse
|
38
|
Page J, Moore N, Broderick G. A Computational Protocol for the Knowledge-Based Assessment and Capture of Pathologies. Methods Mol Biol 2025; 2868:265-284. [PMID: 39546235 DOI: 10.1007/978-1-0716-4200-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
We propose that one of the main hurdles in delivering comprehensively informed care results from the challenges surrounding the extraction, representation, and retention of prior clinical experience and basic medical knowledge, as well as its translation into time- and context-informed actionable interventions. While emerging applications in artificial intelligence-based techniques, for example, large language models, offer impressive pattern association capabilities, they often fall short in producing human-readable explanations crucial to their integration into clinical care. Moreover, they require large well-defined and well-integrated data sets that typically conflict with the availability of such data in all but a few areas of medicine, for example, medical imaging and neuroimaging, noninvasive monitoring of bio-electrical activity, etc. In this chapter, we argue that approximate reasoning rooted in the knowledge that is explainable to the human clinician may offer attractive avenues for the introduction of such knowledge in a systematic way that supports formal retention, sharing, and reuse of new clinical and basic medical experience. We outline a conceptual protocol that targets the use of sparse and disparate data of different types and from different sources, seamlessly drawing on our collective experience and that of others. We illustrate the utility of such an integrative approach by applying the latter to the assessment and reconciliation of data from different experimental models, human and animal, in the example use case of a complex health condition.
Collapse
Affiliation(s)
- Jeffrey Page
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, USA
| | - Nadia Moore
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, USA
| | - Gordon Broderick
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, USA.
- Vaccine and Infectious Disease Organization (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
39
|
Qin K, Wang Q, Qing J, Li Y, Gong H, Zha Z, Zhou B, Li Y. Analysis of mutations in Chinese patients with polycystic kidney disease by targeted exome sequencing. Genes Dis 2025; 12:101246. [PMID: 39430052 PMCID: PMC11490921 DOI: 10.1016/j.gendis.2024.101246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/21/2024] [Indexed: 10/22/2024] Open
Affiliation(s)
- Kaili Qin
- Department of Nephrology, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030012, China
| | - Qian Wang
- Shanxi Provincial Key Laboratory of Kidney Disease, Shanxi Provincial People’s Hospital, Taiyuan 030012, China
| | - Jianbo Qing
- Department of Nephrology, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030012, China
| | - Yaheng Li
- Shanxi Provincial Key Laboratory of Kidney Disease, Shanxi Provincial People’s Hospital, Taiyuan 030012, China
| | - Hao Gong
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Zhijian Zha
- Third Clinical School, Shanxi University of Traditional Chinese Medicine, No. 89, Sec. 1, Jinzi Road, Wanbailin District, Taiyuan City, Shanxi Province, China
| | - Bingrui Zhou
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030012, China
- Department of Nephrology, Hejin Municipal People’s Hospital, Yuncheng 043300, China
- Core Laboratory, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030012, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan 030012, China
| |
Collapse
|
40
|
Hakeem A, Yang S. Regulation of INPP5E in Ciliogenesis, Development, and Disease. Int J Biol Sci 2025; 21:579-594. [PMID: 39781470 PMCID: PMC11705637 DOI: 10.7150/ijbs.99010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/03/2024] [Indexed: 01/30/2025] Open
Abstract
Inositol polyphosphate-5-phosphatase E (INPP5E) is a 5-phosphatase critically involved in diverse physiological processes, including embryonic development, neurological function, immune regulation, hemopoietic cell dynamics, and macrophage proliferation, differentiation, and phagocytosis. Mutations in INPP5E cause Joubert and Meckel-Gruber syndromes in humans; these are characterized by brain malformations, microphthalmia, situs inversus, skeletal abnormalities, and polydactyly. Recent studies have demonstrated the key role of INPP5E in governing intracellular processes like endocytosis, exocytosis, vesicular trafficking, and membrane dynamics. Moreover, it regulates cellular signaling pathways by dephosphorylating the 5-phosphate of phosphatidylinositol-3,4,5-trisphosphate, phosphatidylinositol 4,5-bisphosphate, and phosphatidylinositol 3,5-bisphosphate. Despite recent advances, knowledge gaps persist regarding the function and molecular mechanism of INPP5E in various cells and species. This review integrates recent findings on the role of INPP5E in regulating cellular function, development, and the pathogenesis of various human disorders, emphasizing the molecular mechanism by which INPP5E regulates primary cilia assembly and function and critical signaling pathways. Identifying the importance of INPP5E in healthy and diseased states can advance our understanding of cellular processes and disease pathogenesis and provide a foundation for developing targeted therapeutic interventions.
Collapse
Affiliation(s)
- Abdulaziz Hakeem
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, USA
- Department of Basic and Translation Science, School of Dentistry, Umm Al Qura University, Saudi Arabia
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, USA
- The Penn Center for Musculoskeletal Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Center for Innovation & Precision Dentistry, Penn Dental Medicine and School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
41
|
Chakraborty A, Yu ASL. Prospects for gene therapy in polycystic kidney disease. Curr Opin Nephrol Hypertens 2025; 34:121-127. [PMID: 39499052 PMCID: PMC11606769 DOI: 10.1097/mnh.0000000000001030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
PURPOSE OF REVIEW We aim to provide an updated perspective on the recent advancements in gene therapy for polycystic kidney disease (PKD), a genetic disorder with significant morbidity. Given the rapid evolution of gene therapy technologies and their potential for treating inherited diseases, this review explores the therapeutic prospects and challenges in applying these technologies to PKD. RECENT FINDINGS Significant progress has been made in understanding the genetic underpinnings of PKD, making it a prime candidate for gene therapy. Re-expression of the PKD genes, treatment with the C-terminal tail of polycystin 1 protein and antagomir therapy against miR-17 have shown promise in reducing cyst formation and preserving kidney function. The rapid development of gene-editing tools, antisense oligonucleotide-based strategies, programmable RNA, and advanced gene delivery systems has opened new possibilities for PKD treatment. However, challenges such as off-target effects, delivery efficiency, and long-term safety remain significant barriers to clinical application. SUMMARY Current research highlights the transformative potential of gene therapy for PKD. Ongoing studies are crucial to overcoming existing challenges and translating these findings into clinical practice. We highlight the need for multidisciplinary efforts to optimize gene-editing technologies and ensure their safety and efficacy in treating PKD.
Collapse
Affiliation(s)
- Anubhav Chakraborty
- The Jared Grantham Kidney Institute
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Alan S L Yu
- The Jared Grantham Kidney Institute
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
42
|
Trant J, Sanchez G, McDermott JP, Blanco G. The cystogenic effects of ouabain in autosomal dominant polycystic kidney disease require cell caveolae. Exp Cell Res 2025; 444:114356. [PMID: 39586486 DOI: 10.1016/j.yexcr.2024.114356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
We have previously shown that the hormone ouabain is a circulating factor which can accelerate the progression of autosomal dominant polycystic kidney disease (ADPKD). At physiologic concentrations, ouabain increases cyst area and fibrosis in kidneys from ADPKD but not wildtype mice. These effects are due to an increased affinity for ouabain by its receptor, Na,K-ATPase (NKA), in the kidneys of ADPKD mice which leads to over-activation of NKA signaling function. Previous studies suggested that ouabain's stimulation of NKA signal transduction is mediated by NKA located within cell caveolae. Here, we determined whether caveolae are involved in the ouabain-induced progression of ADPKD cysts. We generated an ADPKD mouse with a global knockout of the main structural component of caveolae, caveolin-1 (CAV1), which we confirmed lacks caveolae in the kidney. When given physiological amounts of ouabain for 5 months, Pkd1RC/RCCav1-/- mice did not exhibit any changes in cyst progression, contrasting with the Pkd1RC/RC mice which showed a significant increase in cystic area and kidney fibrosis. Also, measures of ouabain-induced cell proliferation, including the number of Ki67-positive nuclei and phosphorylation of the extracellular regulated kinase (ERK) and protein kinase B (Akt), did not increase in the Pkd1RC/RCCav1-/- mice compared with the Pkd1RC/RC mice. Moreover, the abnormally increased affinity for ouabain of NKA in Pkd1RC/RC mice was restored to wildtype levels in the Pkd1RC/RCCav1-/- mice. This work highlights the role of caveolae in ouabain-induced NKA signaling and ADPKD cyst progression.
Collapse
Affiliation(s)
- Jordan Trant
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gladis Sanchez
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffery P McDermott
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gustavo Blanco
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
43
|
Sun Y, Zou Q, Yu H, Yi X, Dou X, Yang Y, Liu Z, Yang H, Jia J, Chen Y, Sun SK, Zhang L. Melanin-like nanoparticles slow cyst growth in ADPKD by dual inhibition of oxidative stress and CREB. EMBO Mol Med 2025; 17:169-192. [PMID: 39567834 PMCID: PMC11730739 DOI: 10.1038/s44321-024-00167-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Melanin-like nanoparticles (MNPs) have recently emerged as valuable agents in antioxidant therapy due to their excellent biocompatibility and potent capacity to scavenge various reactive oxygen species (ROS). However, previous studies have mainly focused on acute ROS-related diseases, leaving a knowledge gap regarding their potential in chronic conditions. Furthermore, apart from their well-established antioxidant effects, it remains unclear whether MNPs target other intracellular molecular pathways. In this study, we synthesized ultra-small polyethylene glycol-incorporated Mn2+-chelated MNP (MMPP). We found that MMPP traversed the glomerular filtration barrier and specifically accumulated in renal tubules. Autosomal dominant polycystic kidney disease (ADPKD) is a chronic genetic disorder closely associated with increased oxidative stress and featured by the progressive enlargement of cysts originating from various segments of the renal tubules. Treatment with MMPP markedly attenuated oxidative stress levels, inhibited cyst growth, thereby improving renal function. Interestingly, we found that MMPP effectively inhibits a cyst-promoting gene program downstream of the cAMP-CREB pathway, a crucial signaling pathway implicated in ADPKD progression. Mechanistically, we observed that MMPP directly binds to the bZIP DNA-binding domain of CREB, leading to competitive inhibition of CREB's DNA binding ability and subsequent reduction in CREB target gene expression. In summary, our findings identify an intracellular target of MMPP and demonstrate its potential for treating ADPKD by simultaneously targeting oxidative stress and CREB transcriptional activity.
Collapse
Affiliation(s)
- Yongzhan Sun
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Quan Zou
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Huizheng Yu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaoping Yi
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xudan Dou
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yu Yang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiheng Liu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Hong Yang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junya Jia
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yupeng Chen
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin, China.
| | - Lirong Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
44
|
Márquez-Nogueras KM, Elliott B, Thuo P, DiNello E, Knutila RM, Fritzmann GE, Vuchkovska V, Flury S, Willis M, Chapman AB, Cao Q, Barefield DY, Kuo IY. Cardiac Localized Polycystin-2 in the Natriuretic Peptide Signaling Pathway and Hypertension. J Am Soc Nephrol 2025; 36:34-47. [PMID: 39302726 PMCID: PMC11706566 DOI: 10.1681/asn.0000000000000490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
Key Points Cardiac localized polycystin facilitates natriuretic peptide signaling pathways. Hypertension associated with autosomal dominant polycystic kidney disease may arise from impaired cardiac natriuretic peptide signaling. Background Hypertension is seen in 70% of patients with autosomal dominant polycystic kidney disease by age of 30 years before decline in kidney function. However, cardiac origins of hypertension, such as the natriuretic peptide signaling pathway, have not been fully investigated. We hypothesized that cardiomyocyte localized polycystin proteins contribute to production of natriuretic peptides, and loss of this pathway would contribute to hypertension. Methods Telemetry, echocardiography, and a molecular analysis of the natriuretic peptide pathway from left ventricular tissue of cardiomyocyte specific knockout models of polycystin-2 (cPC2-KO) mice and Cre control littermates were conducted. Complementary studies were conducted in ex vivo murine hearts, engineered heart tissue with human iPSCs driven into cardiomyocytes with CRISPR/Cas9 knockout of PKD2 and in in vitro cell lines. Results cPC2-KO mice demonstrated diurnal hypertension. Circulating atrial natriuretic peptide (ANP) and brain natriuretic peptide were unchanged between cPC2-KO and Cre mice. Analysis of the pathways involved in production, maturation, and activity of natriuretic peptides identified decreased transcription of chromogranin B, PCSK6, NPR1, and NFAT genes in cPC2-KOs. Human iPSC-derived cardiomyocytes with PC2-KO failed to produce ANP. Re-expression of polycystin-2 in a myoblast cell line, but not pathogenic forms of polycystin-2, restored ANP production. Conclusions Natriuretic peptide production required cardiac localized polycystin-2, and loss of this pathway may contribute to the development of hypertension in autosomal dominant polycystic kidney disease. Podcast This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2024_10_08_ASN0000000000000490.mp3
Collapse
Affiliation(s)
- Karla M. Márquez-Nogueras
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Brandon Elliott
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Paula Thuo
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Elisabeth DiNello
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Ryne M. Knutila
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Geena E. Fritzmann
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
- Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois
| | - Virdjinija Vuchkovska
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Sarah Flury
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Monte Willis
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Arlene B. Chapman
- Section of Nephrology, Biological Sciences Division, Department of Medicine and Institute for Translational Medicine, University of Chicago, Chicago, Illinois
| | - Quan Cao
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
- Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois
| | - David Y. Barefield
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
- Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois
| | - Ivana Y. Kuo
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
- Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois
| |
Collapse
|
45
|
Russell LG, Kolatsi‐Joannou M, Wilson L, Chandler JC, Tejedor NP, Stagg G, Price KL, Rowan CJ, Crompton T, Rosenblum ND, Winyard PJD, Long DA. Reduction of elevated Gli3 does not alter the progression of autosomal recessive polycystic kidney disease. Physiol Rep 2025; 13:e70191. [PMID: 39823139 PMCID: PMC11738646 DOI: 10.14814/phy2.70191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025] Open
Abstract
Polycystic kidney diseases (PKD) are genetic disorders which disrupt kidney architecture and function. Autosomal recessive PKD (ARPKD) is a rare form of PKD, caused by mutations in PKHD1, and clinically more severe than the more common autosomal dominant PKD (ADPKD). Prior studies have implicated Hedgehog (Hh) signaling in ADPKD, with increased levels of Hh components in experimental ADPKD and reduced cystogenesis following pharmacological Hh inhibition. In contrast, the role of the Hh pathway in ARPKD is poorly understood. We hypothesized that Hh pathway activity would be elevated during ARPKD pathogenesis, and its modulation may slow disease progression. We utilized Cpk mice which phenocopy ARPKD and generated a PKHD1-mutant spheroid model in human collecting ducts. Significantly elevated levels of the Hh transcriptional effector Gli3 were found in Cpk mice, a finding replicated in PKHD1-mutant spheroids. In Cpk mice, total GLI3 and GLI3 repressor protein levels were also increased. Reduction of increased Gli3 levels via heterozygous genetic deletion in Cpk mice did not affect cyst formation. Additionally, lowering GLI3 transcripts to wildtype levels did not influence PKHD1-mutant spheroid size. Collectively, these data suggest attenuation of elevated Gli3 does not modulate murine and human models of ARPKD.
Collapse
Affiliation(s)
- Lauren G. Russell
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| | - Maria Kolatsi‐Joannou
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| | - Laura Wilson
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| | - Jennifer C. Chandler
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| | - Nuria Perretta Tejedor
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| | - Georgie Stagg
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| | - Karen L. Price
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| | - Christopher J. Rowan
- Department of Paediatrics, Program in Developmental and Stem Cell Biology, Hospital for Sick ChildrenUniversity of TorontoTorontoOntarioCanada
| | - Tessa Crompton
- Infection, Immunity and Inflammation Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
| | - Norman D. Rosenblum
- Department of Paediatrics, Program in Developmental and Stem Cell Biology, Hospital for Sick ChildrenUniversity of TorontoTorontoOntarioCanada
| | - Paul J. D. Winyard
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| | - David A. Long
- Developmental Biology and Cancer Research and Teaching DepartmentUniversity College London, Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthUniversity College LondonLondonUK
| |
Collapse
|
46
|
Ning N, Liu Z, Li X, Liu Y, Song W. Progress of Induced Pluripotent Stem Cell-Derived Renal Organoids in Clinical Application. KIDNEY DISEASES (BASEL, SWITZERLAND) 2025; 11:1-10. [PMID: 40093027 PMCID: PMC11908814 DOI: 10.1159/000541919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/03/2024] [Indexed: 03/19/2025]
Abstract
Background Kidney disease has become a growing public health problem worldwide, and there is an urgent need to develop reliable models for investigating novel and effective treatment strategies. In recent years, kidney organoids, as novel models different from traditional two-dimensional cells and model animals, have attracted more and more attention. Current advances have allowed the generation of kidney organoids from the directed differentiation of induced pluripotent stem cells (iPSCs), which possess similar characteristics to embryonic stem cells, but bypass ethical constraints and have a wide range of sources. Summary Herein, the methods of generating renal organoids from iPSCs, the applications of iPSC-derived renal organoids in disease modeling, drug effectiveness detection, and regenerative medicine as well as the challenges were reviewed. Key Messages iPSC-derived renal organoids can be used to model kidney diseases and are great models for studying kidney injury and toxicity. Many efforts are needed to finally apply organoids into clinical application.
Collapse
Affiliation(s)
- Na Ning
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, China
- Fuwei Biotechnology (Shandong) Co., LTD, Jinan, China
| | - Zhiting Liu
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Fuwei Biotechnology (Shandong) Co., LTD, Jinan, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yi Liu
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, China
| | - Wei Song
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
47
|
Li D, Dawson J, Gunton JE. Therapeutic Potential of Ketogenic Interventions for Autosomal-Dominant Polycystic Kidney Disease: A Systematic Review. Nutrients 2024; 17:145. [PMID: 39796576 PMCID: PMC11723166 DOI: 10.3390/nu17010145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Recent findings have highlighted that abnormal energy metabolism is a key feature of autosomal-dominant polycystic kidney disease (ADPKD). Emerging evidence suggests that nutritional ketosis could offer therapeutic benefits, including potentially slowing or even reversing disease progression. This systematic review aims to synthesise the literature on ketogenic interventions to evaluate the impact in ADPKD. METHODS A systematic search was conducted in Medline, Embase, and Scopus using relevant Medical Subject Headings (MeSH) and keywords. Studies assessing ketogenic interventions in the management of ADPKD in both human and animal models were selected for data extraction and analysis. RESULTS Three animal reports and six human studies were identified. Ketogenic diets (KD) significantly slowed polycystic kidney disease (PKD) progression in rats with improved renal function and reduced cystic areas. There was reduced renal fibrosis and cell proliferation. The supplementation of beta-hydroxybutyrate (BHB) in rats also reduced PKD progression in a dose-dependent manner. Human studies (n = 129) on KD in ADPKD reported consistent body mass index (BMI) reduction across trials, with an average weight loss of ∼4 kg. Improvements in blood pressure were also noted. Ketosis was achieved in varying degrees. Effects on kidney function (eGFR) were beneficial. Results for kidney volume were mixed but most studies were underpowered for this outcome. Lipid profiles showed increases in total cholesterol (∼1 mmol/L) and LDL cholesterol (∼0.4 mmol/L) in most studies. Safety concerns such as "keto flu" symptoms, elevated uric acid levels, and occasional kidney stones were noted. Overall feasibility and adherence to the KD were rated positively by most participants. CONCLUSIONS Human studies are promising; however, they have been limited by small sample sizes and short durations. Larger, long-term trials are needed to assess the efficacy, adherence, and safety of ketogenic diets in people with ADPKD.
Collapse
Affiliation(s)
- Donglai Li
- Centre for Diabetes, Obesity and Endocrinology Research (CDOER), Westmead Institute for Medical Research, Westmead, Sydney, NSW 2145, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2066, Australia
| | - Jessica Dawson
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW 2050, Australia;
- Department Nutrition and Dietetics, St George Hospital, Sydney, NSW 2217, Australia
| | - Jenny E. Gunton
- Centre for Diabetes, Obesity and Endocrinology Research (CDOER), Westmead Institute for Medical Research, Westmead, Sydney, NSW 2145, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2066, Australia
- Department of Diabetes and Endocrinology, Room 2040, Clinical Sciences Corridor, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia
| |
Collapse
|
48
|
Tavakolidakhrabadi N, Ding WY, Saleem MA, Welsh GI, May C. Gene therapy and kidney diseases. Mol Ther Methods Clin Dev 2024; 32:101333. [PMID: 39434922 PMCID: PMC11492605 DOI: 10.1016/j.omtm.2024.101333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Chronic kidney disease (CKD) poses a significant global health challenge, projected to become one of the leading causes of death by 2040. Current treatments primarily manage complications and slow progression, highlighting the urgent need for personalized therapies targeting the disease-causing genes. Our increased understanding of the underlying genomic changes that lead to kidney diseases coupled with recent successful gene therapies targeting specific kidney cells have turned gene therapy and genome editing into a promising therapeutic approach for treating kidney disease. This review paper reflects on different delivery routes and systems that can be exploited to target specific kidney cells and the ways that gene therapy can be used to improve kidney health.
Collapse
Affiliation(s)
- Nadia Tavakolidakhrabadi
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Wen Y. Ding
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Moin A. Saleem
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
- Department of Paediatric Nephrology, Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust, Upper Maudlin Street, Bristol BS2 8BJ, UK
| | - Gavin I. Welsh
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Carl May
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| |
Collapse
|
49
|
Ventayol-Guirado M, Torres L, Asensio-Landa V, Pérez-Granero Á, Madrid MI, Hernandez-Rodriguez J, Llull-Alberti MV, Lumbreras J, Escribà S, Pons M, Roldan J, Martínez-López I, Heine-Suñer D, Santos-Simarro F. Atypical noncontiguous TSC2/PKD1 gene deletions presenting as tuberous sclerosis/polycystic kidney disease contiguous gene syndrome. Am J Med Genet A 2024; 194:e63830. [PMID: 39095963 DOI: 10.1002/ajmg.a.63830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
Tuberous sclerosis complex (TSC) and autosomal dominant polycystic kidney disease (ADPKD) are genetically distinct disorders typically associated with pathogenic variants in TSC1 and TSC2 for the former and PKD1 and PKD2 for the latter. TSC2 and PKD1 lie adjacent to each other, and large deletions comprising both genes lead to TSC2/PKD1 contiguous gene deletion syndrome (CGS). In this study, we describe a young female patient exhibiting symptoms of TSC2/PKD1 CGS in which genetic analysis disclosed two noncontiguous partial gene deletions in TSC2 and PKD1 that putatively are responsible for the manifestations of the syndrome. Further analysis revealed that both deletions appear to be de novo on the maternal chromosome, presumably with a germline origin. Despite extensive analysis, no maternal chromosomal rearrangement triggering these pathogenic variants was detected. This case elucidates a unique pathogenesis for TSC2/PKD1 CGS, diverging from the common contiguous deletions typically observed, marking the first reported instance of TSC2/PKD1 CGS caused by independent, functionally significant partial gene deletions.
Collapse
Affiliation(s)
- Marc Ventayol-Guirado
- Health Research Institute of the Balearic Islands (IdISBa), Genomics of Health research group, Palma, Spain
| | - Laura Torres
- Health Research Institute of the Balearic Islands (IdISBa), Genomics of Health research group, Palma, Spain
- Hospital Universitari Son Espases, Unit of Molecular Diagnostics and Clinical Genetics, Palma, Spain
| | - Victor Asensio-Landa
- Health Research Institute of the Balearic Islands (IdISBa), Genomics of Health research group, Palma, Spain
- Hospital Universitari Son Espases, Unit of Molecular Diagnostics and Clinical Genetics, Palma, Spain
| | - Ángeles Pérez-Granero
- Hospital Universitari Son Espases, Unit of Molecular Diagnostics and Clinical Genetics, Palma, Spain
| | - Maria Isabel Madrid
- Hospital Universitari Son Espases, Unit of Molecular Diagnostics and Clinical Genetics, Palma, Spain
| | - Jessica Hernandez-Rodriguez
- Health Research Institute of the Balearic Islands (IdISBa), Genomics of Health research group, Palma, Spain
- Hospital Universitari Son Espases, Unit of Molecular Diagnostics and Clinical Genetics, Palma, Spain
| | | | - Javier Lumbreras
- Pediatrics Department, Hospital Universitari Son Espases, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Multidisciplinary Pediatric Research Group, Palma, Spain
| | - Silvia Escribà
- Pediatrics Department, Hospital Universitari Son Espases, Palma, Spain
| | - Monserrat Pons
- Pediatrics Department, Hospital Universitari Son Espases, Palma, Spain
| | - Jordi Roldan
- Radiology Department, Hospital Universitari Son Espases, Palma, Spain
| | - Iciar Martínez-López
- Health Research Institute of the Balearic Islands (IdISBa), Genomics of Health research group, Palma, Spain
- Hospital Universitari Son Espases, Unit of Molecular Diagnostics and Clinical Genetics, Palma, Spain
| | - Damian Heine-Suñer
- Health Research Institute of the Balearic Islands (IdISBa), Genomics of Health research group, Palma, Spain
- Hospital Universitari Son Espases, Unit of Molecular Diagnostics and Clinical Genetics, Palma, Spain
| | - Fernando Santos-Simarro
- Health Research Institute of the Balearic Islands (IdISBa), Genomics of Health research group, Palma, Spain
- Hospital Universitari Son Espases, Unit of Molecular Diagnostics and Clinical Genetics, Palma, Spain
| |
Collapse
|
50
|
Waiser J, Klotsche J, Glander P, Schmidt D, Naik M, Liefeldt L, Budde K, Halbritter J, Halleck F, Zukunft B, Peters R, Friedersdorff F, Lachmann N, Eckardt KU, d'Anjou L, Bachmann F. Kidney transplantation in patients with polycystic kidney disease: increased risk of infection does not compromise graft and patient survival. Clin Kidney J 2024; 17:sfae330. [PMID: 39664987 PMCID: PMC11630747 DOI: 10.1093/ckj/sfae330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Indexed: 12/13/2024] Open
Abstract
Background Patients with autosomal dominant polycystic kidney disease (ADPKD) represent >10% of patients awaiting kidney transplantation. These patients are prone to potentially severe urinary tract (UTI) and liver cyst infections after transplantation. Whether such infections compromise outcome is unclear. Methods Between 2000 and 2017 we performed 193 kidney transplantations in patients with ADPKD. In 189 patients, we assessed the occurrence, frequency, and severity of infection episodes requiring inpatient treatment and their impact on graft and patient outcomes compared with 189 matched controls. Risk factors were analyzed by uni- and multivariable analyses. Results During a mean observation period of 77 months UTIs occurred more frequently in ADPKD patients (39.1% vs. 26.7%, P = .022; 0.8 ± 1.4 vs. 0.5 ± 1.1 episodes, P < .001). Eight ADPKD patients suffered from 19 episodes of liver cyst infection. Steroid medication (RR 3.04; P < .001) and recipient age (RR 1.05; P = .003) increased the risk for UTI/urosepsis, while nephrectomy reduced it (unilateral, RR 0.60; P = .088; bilateral, RR 0.45; P = .020). Patient survival was similar in both groups. The risk of graft failure was lower in ADPKD patients [hazard ratio (HR) 0.67; P = .047] due to a lower risk of death-censored graft loss (HR 0.47; P = .014). Donor age (HR 1.34; P = .002) and rejection (HR 8.47; P < .001) were risk factors for death-censored graft loss. Conclusions ADPKD patients are at increased risk of UTI and liver cyst infection after transplantation. Steroid medication and recipient age seem to increase the risk of UTI/urosepsis, while nephrectomy seems to reduce it. Nevertheless, patient survival was similar compared to non-ADPKD patients and death-censored graft survival even better.
Collapse
Affiliation(s)
- Johannes Waiser
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jens Klotsche
- German Rheumatism Research Center Berlin – a Leibniz Institute, Berlin, Germany
| | - Petra Glander
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Danilo Schmidt
- Business Unit IT, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel Naik
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lutz Liefeldt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jan Halbritter
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bianca Zukunft
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robert Peters
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frank Friedersdorff
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Lachmann
- Institute of Transfusion Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leonie d'Anjou
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|