1
|
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
|
2
|
Ha K, Loeb GB, Park M, Gupta M, Akiyama Y, Argiris J, Pinedo A, Park CH, Brandes N, Ritu F, Ye CJ, Reiter JF, Delling M. ADPKD-Causing Missense Variants in Polycystin-1 Disrupt Cell Surface Localization or Polycystin Channel Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.04.570035. [PMID: 38106161 PMCID: PMC10723288 DOI: 10.1101/2023.12.04.570035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the leading monogenic cause of kidney failure and affects millions of people worldwide. Despite the prevalence of this monogenic disorder, our limited mechanistic understanding of ADPKD has hindered therapeutic development. Here, we successfully developed bioassays that functionally classify missense variants in polycystin-1 (PC1). Strikingly, ADPKD pathogenic missense variants cluster into two major categories: 1) those that disrupt polycystin cell surface localization or 2) those that attenuate polycystin ion channel activity. We found that polycystin channels with defective surface localization could be rescued with a small molecule. We propose that small-molecule-based strategies to improve polycystin cell surface localization and channel function will be effective therapies for ADPKD patients.
Collapse
|
3
|
Xue C, Lv J, Yang B, Mei S, Xu J, Li X, Zhang L, Mao Z. Gene therapy in polycystic kidney disease: A promising future. J Transl Int Med 2024; 12:543-552. [PMID: 39802450 PMCID: PMC11720931 DOI: 10.1515/jtim-2024-0021] [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] [Indexed: 01/16/2025] Open
Abstract
Polycystic kidney disease (PKD) is a genetic disorder marked by numerous cysts in the kidneys, progressively impairing renal function. It is classified into autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD), with ADPKD being more common. Current treatments mainly focus on symptom relief and slowing disease progression, without offering a cure. Recent advancements in gene editing technologies, such as CRISPR-Cas9, have introduced new therapeutic possibilities for PKD. These approaches include miR-17 antisense oligonucleotides, adenovirus-mediated gene knockdown, Pkd1 gene or polycystin -1 C-terminal tail enhancement therapy, and 3-UTR miR-17 binding element by CRISPR-Cas9, which have shown potential in animal models and early clinical trials. Specifically for ARPKD, strategies like antisense oligonucleotide therapy targeting c-myc and CRISPR/ Cas9 knockdown of the P2rx7 gene have shown promise. Despite facing challenges such as technological limitations, ethical and legal issues, and high costs, gene therapy presents unprecedented hope for PKD treatment. Future interdisciplinary collaboration and international cooperation are essential for developing more effective treatment strategies for PKD patients.
Collapse
Affiliation(s)
- Cheng Xue
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Jiayi Lv
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Bo Yang
- Internal Medicine III (Nephrology), Naval Medical Center of PLA, Naval Medical University, Shanghai200433, China
| | - Shuqin Mei
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Jing Xu
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Xinming Li
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Liming Zhang
- Department of Nephrology, Zhabei Central Hospital of Jing’an District, Shanghai200120, China
| | - Zhiguo Mao
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| |
Collapse
|
4
|
Gan Z, van der Stelt I, Li W, Hu L, Song J, Grefte S, van de Westerlo E, Zhang D, van Schothorst EM, Claahsen-van der Grinten HL, Teerds KJ, Adjobo-Hermans MJW, Keijer J, Koopman WJH. Mitochondrial Nicotinamide Nucleotide Transhydrogenase: Role in Energy Metabolism, Redox Homeostasis, and Cancer. Antioxid Redox Signal 2024; 41:927-956. [PMID: 39585234 DOI: 10.1089/ars.2024.0694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Significance: Dimeric nicotinamide nucleotide transhydrogenase (NNT) is embedded in the mitochondrial inner membrane and couples the conversion of NADP+/NADH into NADPH/NAD+ to mitochondrial matrix proton influx. NNT was implied in various cancers, but its physiological role and regulation still remain incompletely understood. Recent Advances: NNT function was analyzed by studying: (1) NNT gene mutations in human (adrenal) glucocorticoid deficiency 4 (GCCD4), (2) Nnt gene mutation in C57BL/6J mice, and (3) the effect of NNT knockdown/overexpression in (cancer) cells. In these three models, altered NNT function induced both common and differential aberrations. Critical Issues: Information on NNT protein expression in GCCD4 patients is still scarce. Moreover, NNT expression levels are tissue-specific in humans and mice and the functional consequences of NNT deficiency strongly depend on experimental conditions. In addition, data from intact cells and isolated mitochondria are often unsuited for direct comparison. This prevents a proper understanding of NNT-linked (patho)physiology in GCCD4 patients, C57BL/6J mice, and cancer (cell) models, which complicates translational comparison. Future Directions: Development of mice with conditional NNT deletion, cell-reprogramming-based adrenal (organoid) models harboring specific NNT mutations, and/or NNT-specific chemical inhibitors/activators would be useful. Moreover, live-cell analysis of NNT substrate levels and mitochondrial/cellular functioning with fluorescent reporter molecules might provide novel insights into the conditions under which NNT is active and how this activity links to other metabolic and signaling pathways. This would also allow a better dissection of local signaling and/or compartment-specific (i.e., mitochondrial matrix, cytosol, nucleus) effects of NNT (dys)function in a cellular context. Antioxid. Redox Signal. 41, 927-956.
Collapse
Affiliation(s)
- Zhuohui Gan
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Inge van der Stelt
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Weiwei Li
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Liangyu Hu
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Jingyi Song
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Sander Grefte
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Els van de Westerlo
- Department of Medical BioSciences, Radboudumc, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Deli Zhang
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | | | | | - Katja J Teerds
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Merel J W Adjobo-Hermans
- Department of Medical BioSciences, Radboudumc, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Werner J H Koopman
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboudumc, Nijmegen, The Netherlands
- Department of Pediatrics, Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
5
|
Bjornstad P, Richard G, Choi YJ, Nowak KL, Steele C, Chonchol MB, Nadeau KJ, Vigers T, Pyle L, Tommerdahl K, van Raalte DH, Hilkin A, Driscoll L, Birznieks C, Hopp K, Wang W, Edelstein C, Nelson RG, Gregory AV, Kline TL, Blondin D, Gitomer B. Kidney Energetics and Cyst Burden in Autosomal Dominant Polycystic Kidney Disease: A Pilot Study. Am J Kidney Dis 2024; 84:286-297.e1. [PMID: 38621633 PMCID: PMC11344681 DOI: 10.1053/j.ajkd.2024.02.016] [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: 09/20/2023] [Revised: 02/20/2024] [Accepted: 02/28/2024] [Indexed: 04/17/2024]
Abstract
RATIONALE & OBJECTIVE In this pilot study, we hypothesized that autosomal dominant polycystic kidney disease (ADPKD) is characterized by impaired kidney oxidative metabolism that associates with kidney size and cyst burden. STUDY DESIGN Cross-sectional study. SETTING & PARTICIPANTS Twenty adults with ADPKD (age, 31±6 years; 65% women; body mass index [BMI], 26.8 [22.7-30.4] kg/m2; estimated glomerular filtration rate [eGFR, 2021 CKD-EPI creatinine], 103±18mL/min/1.73m2; height-adjusted total kidney volume [HTKV], 731±370mL/m; Mayo classifications 1B [5%], 1C [42%], 1D [21%], and 1E [32%]) and 11 controls in normal weight category (NWC) (age, 25±3 years; 45% women; BMI, 22.5 [21.7-24.2] kg/m2; eGFR, 113±15mL/min/1.73m2; HTKV, 159±31mL/m) at the University of Colorado Anschutz Medical Campus. PREDICTORS ADPKD status (yes/no) and severity (Mayo classifications). OUTCOME HTKV and cyst burden by magnetic resonance imaging, kidney oxidative metabolism, and perfusion by 11C-acetate positron emission tomography/computed tomography, insulin sensitivity by hyperinsulinemic-euglycemic clamps (presented as ratio of M-value of steady state insulin concentration [M/I]). ANALYTICAL APPROACH For categorical variables, χ2/Fisher's exact tests, and for continuous variables t tests/Mann-Whitney U tests. Pearson correlation was used to estimate the relationships between variables. RESULTS Compared with NWC individuals, the participants with ADPKD exhibited lower mean±SD M/I ratio (0.586±0.205 vs 0.424±0.171 [mg/kg lean/min]/(μIU/mL), P=0.04), lower median cortical perfusion (1.93 [IQR, 1.80-2.09] vs 0.68 [IQR, 0.47-1.04] mL/min/g, P<0.001) and lower median total kidney oxidative metabolism (0.17 [IQR, 0.16-0.19] vs. 0.14 [IQR, 0.12-0.15] min-1, P=0.001) in voxel-wise models excluding cysts. HTKV correlated inversely with cortical perfusion (r: -0.83, P < 0.001), total kidney oxidative metabolism (r: -0.61, P<0.001) and M/I (r: -0.41, P = 0.03). LIMITATIONS Small sample size and cross-sectional design. CONCLUSIONS Adults with ADPKD and preserved kidney function exhibited impaired renal perfusion and kidney oxidative metabolism across a wide range of cysts and kidney enlargements. FUNDING Grants from government (National Institutes of Health, Centers for Disease Control and Prevention) and not-for-profit (JDRF) entities. TRIAL REGISTRATION Registered at ClinicalTrials.gov with study numbers NCT04407481 and NCT04074668. PLAIN-LANGUAGE SUMMARY In our study, we explored how a common genetic kidney condition, autosomal dominant polycystic kidney disease (ADPKD), relates to kidney metabolism. ADPKD leads to the growth of numerous cysts in the kidneys, which can impact their ability to work properly. We wanted to understand the kidneys' ability to process oxygen and blood flow in ADPKD. Our approach involved using advanced imaging techniques to observe kidney metabolism and blood flow in people with ADPKD compared with healthy individuals. We discovered that those with ADPKD had significant changes in kidney oxygen metabolism even when their kidney function was still normal. These findings are crucial as they provide deeper insights into ADPKD, potentially guiding future treatments to target these changes.
Collapse
Affiliation(s)
- Petter Bjornstad
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado; Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado.
| | - Gabriel Richard
- Department of Medicine, Division of Neurology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Ye Ji Choi
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado; Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, Colorado
| | - Kristen L Nowak
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado; Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - Cortney Steele
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - Michel B Chonchol
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - Kristen J Nadeau
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado
| | - Timothy Vigers
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado; Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, Colorado
| | - Laura Pyle
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado; Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, Colorado
| | - Kalie Tommerdahl
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado; Barbara Davis Center for Diabetes, Section of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel H van Raalte
- Department of Endocrinology and Metabolism and Diabetes Center, Amsterdam University Medical Centers, VUMC, Amsterdam, the Netherlands
| | - Allison Hilkin
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado
| | - Lynette Driscoll
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado
| | - Carissa Birznieks
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado
| | - Katharina Hopp
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - Wei Wang
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - Charles Edelstein
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona
| | - Adriana V Gregory
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Timothy L Kline
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Denis Blondin
- Department of Medicine, Division of Neurology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Berenice Gitomer
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
6
|
Lichner Z, Ding M, Khare T, Dan Q, Benitez R, Praszner M, Song X, Saleeb R, Hinz B, Pei Y, Szászi K, Kapus A. Myocardin-Related Transcription Factor Mediates Epithelial Fibrogenesis in Polycystic Kidney Disease. Cells 2024; 13:984. [PMID: 38891116 PMCID: PMC11172104 DOI: 10.3390/cells13110984] [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: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Polycystic kidney disease (PKD) is characterized by extensive cyst formation and progressive fibrosis. However, the molecular mechanisms whereby the loss/loss-of-function of Polycystin 1 or 2 (PC1/2) provokes fibrosis are largely unknown. The small GTPase RhoA has been recently implicated in cystogenesis, and we identified the RhoA/cytoskeleton/myocardin-related transcription factor (MRTF) pathway as an emerging mediator of epithelium-induced fibrogenesis. Therefore, we hypothesized that MRTF is activated by PC1/2 loss and plays a critical role in the fibrogenic reprogramming of the epithelium. The loss of PC1 or PC2, induced by siRNA in vitro, activated RhoA and caused cytoskeletal remodeling and robust nuclear MRTF translocation and overexpression. These phenomena were also manifested in PKD1 (RC/RC) and PKD2 (WS25/-) mice, with MRTF translocation and overexpression occurring predominantly in dilated tubules and the cyst-lining epithelium, respectively. In epithelial cells, a large cohort of PC1/PC2 downregulation-induced genes was MRTF-dependent, including cytoskeletal, integrin-related, and matricellular/fibrogenic proteins. Epithelial MRTF was necessary for the paracrine priming of the fibroblast-myofibroblast transition. Thus, MRTF acts as a prime inducer of epithelial fibrogenesis in PKD. We propose that RhoA is a common upstream inducer of both histological hallmarks of PKD: cystogenesis and fibrosis.
Collapse
Affiliation(s)
- Zsuzsanna Lichner
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Mei Ding
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Tarang Khare
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Enrich Bioscience, Toronto, ON M5B 1T8, Canada
| | - Qinghong Dan
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Raquel Benitez
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Mercédesz Praszner
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Xuewen Song
- Division of Nephrology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Rola Saleeb
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Department of Laboratory Medicine and Pathobiology, Temerty School of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Boris Hinz
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - York Pei
- Division of Nephrology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Department of Laboratory Medicine and Pathobiology, Temerty School of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
7
|
Clerici S, Podrini C, Stefanoni D, Distefano G, Cassina L, Steidl ME, Tronci L, Canu T, Chiaravalli M, Spies D, Bell TA, Costa AS, Esposito A, D'Alessandro A, Frezza C, Bachi A, Boletta A. Inhibition of asparagine synthetase effectively retards polycystic kidney disease progression. EMBO Mol Med 2024; 16:1379-1403. [PMID: 38684863 PMCID: PMC11178866 DOI: 10.1038/s44321-024-00071-9] [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: 10/06/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
Polycystic kidney disease (PKD) is a genetic disorder characterized by bilateral cyst formation. We showed that PKD cells and kidneys display metabolic alterations, including the Warburg effect and glutaminolysis, sustained in vitro by the enzyme asparagine synthetase (ASNS). Here, we used antisense oligonucleotides (ASO) against Asns in orthologous and slowly progressive PKD murine models and show that treatment leads to a drastic reduction of total kidney volume (measured by MRI) and a prominent rescue of renal function in the mouse. Mechanistically, the upregulation of an ATF4-ASNS axis in PKD is driven by the amino acid response (AAR) branch of the integrated stress response (ISR). Metabolic profiling of PKD or control kidneys treated with Asns-ASO or Scr-ASO revealed major changes in the mutants, several of which are rescued by Asns silencing in vivo. Indeed, ASNS drives glutamine-dependent de novo pyrimidine synthesis and proliferation in cystic epithelia. Notably, while several metabolic pathways were completely corrected by Asns-ASO, glycolysis was only partially restored. Accordingly, combining the glycolytic inhibitor 2DG with Asns-ASO further improved efficacy. Our studies identify a new therapeutic target and novel metabolic vulnerabilities in PKD.
Collapse
Affiliation(s)
- Sara Clerici
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Christine Podrini
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy
- The BioArte Ltd, Laboratories at Malta Life Science Park (LS2.1.10, LS2.1.12-LS2.1.15), Triq San Giljan, San Gwann, SGN, 3000, Malta
| | - Davide Stefanoni
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Gianfranco Distefano
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Laura Cassina
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Maria Elena Steidl
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Laura Tronci
- Cogentech SRL Benefit Corporation, 20139, Milan, Italy
- IFOM ETS The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Tamara Canu
- Center for Experimental Imaging (CIS), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Marco Chiaravalli
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Daniel Spies
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy
- Center for Omics Sciences (COSR), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | | | - Ana Sh Costa
- MRC, Cancer Unit Cambridge, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
- Matterworks, Inc, 444 Somerville Avenue, Somerville, MA, 02143, USA
| | - Antonio Esposito
- Center for Experimental Imaging (CIS), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Christian Frezza
- Faculty of Medicine and University Hospital Cologne, Faculty of Mathematics and Natural Sciences, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Str. 26-50931, Cologne, Germany
| | - Angela Bachi
- IFOM ETS The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Alessandra Boletta
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
8
|
Chebib FT, Nowak KL, Chonchol MB, Bing K, Ghanem A, Rahbari-Oskoui FF, Dahl NK, Mrug M. Polycystic Kidney Disease Diet: What is Known and What is Safe. Clin J Am Soc Nephrol 2024; 19:664-682. [PMID: 37729939 PMCID: PMC11108253 DOI: 10.2215/cjn.0000000000000326] [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: 05/18/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disorder characterized by kidney cyst formation and progressive kidney function loss. Dietary interventions such as caloric restriction, intermittent fasting, and ketogenic diet have recently emerged as potential strategies to induce metabolic reprogramming and slow ADPKD progression. We review the available evidence supporting the efficacy and safety of these interventions in ADPKD. Dietary interventions show promise in managing ADPKD by improving metabolic health and reducing oxidative stress. However, while preclinical studies have shown favorable outcomes, limited clinical evidence supports their effectiveness. In addition, the long-term consequences of these dietary interventions, including their effect on adverse events in patients with ADPKD, remain uncertain. To optimize ADPKD management, patients are advised to follow a dietary regimen that aims to achieve or maintain an ideal body weight and includes high fluid intake, low sodium, and limited concentrated sweets. Caloric restriction seems particularly beneficial for patients with overweight or obesity because it promotes weight loss and improves metabolic parameters. Supplementation with curcumin, ginkgolide B, saponins, vitamin E, niacinamide, or triptolide has demonstrated uncertain clinical benefit in patients with ADPKD. Notably, β -hydroxybutyrate supplements have shown promise in animal models; however, their safety and efficacy in ADPKD require further evaluation through well-designed clinical trials. Therefore, the use of these supplements is not currently recommended for patients with ADPKD. In summary, dietary interventions such as caloric restriction, intermittent fasting, and ketogenic diet hold promise in ADPKD management by enhancing metabolic health. However, extensive clinical research is necessary to establish their effectiveness and long-term effects. Adhering to personalized dietary guidelines, including weight management and specific nutritional restrictions, can contribute to optimal ADPKD management. Future research should prioritize well-designed clinical trials to determine the benefits and safety of dietary interventions and supplementation in ADPKD.
Collapse
Affiliation(s)
- Fouad T. Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| | - Kristen L. Nowak
- Division of Renal Diseases and Hypertension, Polycystic Kidney Disease Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michel B. Chonchol
- Division of Renal Diseases and Hypertension, Polycystic Kidney Disease Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kristen Bing
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ahmad Ghanem
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| | | | - Neera K. Dahl
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Michal Mrug
- Division of Nephrology, Department of Medicine, Department of Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
9
|
Capelli I, Lerario S, Ciurli F, Berti GM, Aiello V, Provenzano M, La Manna G. Investigational agents for autosomal dominant polycystic kidney disease: preclinical and early phase study insights. Expert Opin Investig Drugs 2024; 33:469-484. [PMID: 38618918 DOI: 10.1080/13543784.2024.2342327] [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: 12/01/2023] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common inherited kidney condition caused by a single-gene mutation. It leads patients to kidney failure in more than 50% of cases by the age of 60, and, given the dominant inheritance, this disease is present in the family history in more than 90% of cases. AREAS COVERED This review aims to analyze the set of preclinical and early-phase studies to provide a general view of the current progress on ADPKD therapeutic options. Articles from PubMed and the current status of the trials listed in clinicaltrials.gov were examined for the review. EXPERT OPINION Many potential therapeutic targets are currently under study for the treatment of ADPKD. A few drugs have reached the clinical phase, while many are currently still in the preclinical phase. Organoids could be a novel approach to the study of drugs in this phase. Other than pharmacological options, very important developing approaches are represented by gene therapy and the use of MiRNA inhibitors.
Collapse
Affiliation(s)
- Irene Capelli
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Sarah Lerario
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Francesca Ciurli
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Gian Marco Berti
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Valeria Aiello
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Michele Provenzano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Gaetano La Manna
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
10
|
Tavakolidakhrabadi N, Aulicino F, May CJ, Saleem MA, Berger I, Welsh GI. Genome editing and kidney health. Clin Kidney J 2024; 17:sfae119. [PMID: 38766272 PMCID: PMC11099665 DOI: 10.1093/ckj/sfae119] [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: 02/14/2024] [Indexed: 05/22/2024] Open
Abstract
Genome editing technologies, clustered regularly interspaced short palindromic repeats (CRISPR)-Cas in particular, have revolutionized the field of genetic engineering, providing promising avenues for treating various genetic diseases. Chronic kidney disease (CKD), a significant health concern affecting millions of individuals worldwide, can arise from either monogenic or polygenic mutations. With recent advancements in genomic sequencing, valuable insights into disease-causing mutations can be obtained, allowing for the development of new treatments for these genetic disorders. CRISPR-based treatments have emerged as potential therapies, especially for monogenic diseases, offering the ability to correct mutations and eliminate disease phenotypes. Innovations in genome editing have led to enhanced efficiency, specificity and ease of use, surpassing earlier editing tools such as zinc-finger nucleases and transcription activator-like effector nucleases (TALENs). Two prominent advancements in CRISPR-based gene editing are prime editing and base editing. Prime editing allows precise and efficient genome modifications without inducing double-stranded DNA breaks (DSBs), while base editing enables targeted changes to individual nucleotides in both RNA and DNA, promising disease correction in the absence of DSBs. These technologies have the potential to treat genetic kidney diseases through specific correction of disease-causing mutations, such as somatic mutations in PKD1 and PKD2 for polycystic kidney disease; NPHS1, NPHS2 and TRPC6 for focal segmental glomerulosclerosis; COL4A3, COL4A4 and COL4A5 for Alport syndrome; SLC3A1 and SLC7A9 for cystinuria and even VHL for renal cell carcinoma. Apart from editing the DNA sequence, CRISPR-mediated epigenome editing offers a cost-effective method for targeted treatment providing new avenues for therapeutic development, given that epigenetic modifications are associated with the development of various kidney disorders. However, there are challenges to overcome, including developing efficient delivery methods, improving safety and reducing off-target effects. Efforts to improve CRISPR-Cas technologies involve optimizing delivery vectors, employing viral and non-viral approaches and minimizing immunogenicity. With research in animal models providing promising results in rescuing the expression of wild-type podocin in mouse models of nephrotic syndrome and successful clinical trials in the early stages of various disorders, including cancer immunotherapy, there is hope for successful translation of genome editing to kidney diseases.
Collapse
Affiliation(s)
| | - Francesco Aulicino
- BrisSynBio Bristol Synthetic Biology Centre, Biomedical Sciences, School of Biochemistry, Bristol Royal Hospital for Children
| | - Carl J May
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, UK
| | - Moin A Saleem
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, UK
- Department of Paediatric Nephrology, Bristol Royal Hospital for Children, Bristol, UK
| | - Imre Berger
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Gavin I Welsh
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, UK
| |
Collapse
|
11
|
Mahboobipour AA, Ala M, Safdari Lord J, Yaghoobi A. Clinical manifestation, epidemiology, genetic basis, potential molecular targets, and current treatment of polycystic liver disease. Orphanet J Rare Dis 2024; 19:175. [PMID: 38671465 PMCID: PMC11055360 DOI: 10.1186/s13023-024-03187-w] [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: 07/04/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Polycystic liver disease (PLD) is a rare condition observed in three genetic diseases, including autosomal dominant polycystic liver disease (ADPLD), autosomal dominant polycystic kidney disease (ADPKD), and autosomal recessive polycystic kidney disease (ARPKD). PLD usually does not impair liver function, and advanced PLD becomes symptomatic when the enlarged liver compresses adjacent organs or increases intra-abdominal pressure. Currently, the diagnosis of PLD is mainly based on imaging, and genetic testing is not required except for complex cases. Besides, genetic testing may help predict patients' prognosis, classify patients for genetic intervention, and conduct early treatment. Although the underlying genetic causes and mechanisms are not fully understood, previous studies refer to primary ciliopathy or impaired ciliogenesis as the main culprit. Primarily, PLD occurs due to defective ciliogenesis and ineffective endoplasmic reticulum quality control. Specifically, loss of function mutations of genes that are directly involved in ciliogenesis, such as Pkd1, Pkd2, Pkhd1, and Dzip1l, can lead to both hepatic and renal cystogenesis in ADPKD and ARPKD. In addition, loss of function mutations of genes that are involved in endoplasmic reticulum quality control and protein folding, trafficking, and maturation, such as PRKCSH, Sec63, ALG8, ALG9, GANAB, and SEC61B, can impair the production and function of polycystin1 (PC1) and polycystin 2 (PC2) or facilitate their degradation and indirectly promote isolated hepatic cystogenesis or concurrent hepatic and renal cystogenesis. Recently, it was shown that mutations of LRP5, which impairs canonical Wnt signaling, can lead to hepatic cystogenesis. PLD is currently treated by somatostatin analogs, percutaneous intervention, surgical fenestration, resection, and liver transplantation. In addition, based on the underlying molecular mechanisms and signaling pathways, several investigational treatments have been used in preclinical studies, some of which have shown promising results. This review discusses the clinical manifestation, complications, prevalence, genetic basis, and treatment of PLD and explains the investigational methods of treatment and future research direction, which can be beneficial for researchers and clinicians interested in PLD.
Collapse
Affiliation(s)
- Amir Ali Mahboobipour
- Tracheal Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moein Ala
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Javad Safdari Lord
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Yaghoobi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| |
Collapse
|
12
|
Fedeles BI, Bhardwaj R, Ishikawa Y, Khumsubdee S, Krappitz M, Gubina N, Volpe I, Andrade DC, Westergerling P, Staudner T, Campolo J, Liu SS, Dong K, Cai Y, Rehman M, Gallagher AR, Ruchirawat S, Croy RG, Essigmann JM, Fedeles SV, Somlo S. A synthetic agent ameliorates polycystic kidney disease by promoting apoptosis of cystic cells through increased oxidative stress. Proc Natl Acad Sci U S A 2024; 121:e2317344121. [PMID: 38241440 PMCID: PMC10823221 DOI: 10.1073/pnas.2317344121] [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: 10/09/2023] [Accepted: 11/15/2023] [Indexed: 01/21/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic cause of chronic kidney disease and the fourth leading cause of end-stage kidney disease, accounting for over 50% of prevalent cases requiring renal replacement therapy. There is a pressing need for improved therapy for ADPKD. Recent insights into the pathophysiology of ADPKD revealed that cyst cells undergo metabolic changes that up-regulate aerobic glycolysis in lieu of mitochondrial respiration for energy production, a process that ostensibly fuels their increased proliferation. The present work leverages this metabolic disruption as a way to selectively target cyst cells for apoptosis. This small-molecule therapeutic strategy utilizes 11beta-dichloro, a repurposed DNA-damaging anti-tumor agent that induces apoptosis by exacerbating mitochondrial oxidative stress. Here, we demonstrate that 11beta-dichloro is effective in delaying cyst growth and its associated inflammatory and fibrotic events, thus preserving kidney function in perinatal and adult mouse models of ADPKD. In both models, the cyst cells with homozygous inactivation of Pkd1 show enhanced oxidative stress following treatment with 11beta-dichloro and undergo apoptosis. Co-administration of the antioxidant vitamin E negated the therapeutic benefit of 11beta-dichloro in vivo, supporting the conclusion that oxidative stress is a key component of the mechanism of action. As a preclinical development primer, we also synthesized and tested an 11beta-dichloro derivative that cannot directly alkylate DNA, while retaining pro-oxidant features. This derivative nonetheless maintains excellent anti-cystic properties in vivo and emerges as the lead candidate for development.
Collapse
Affiliation(s)
- Bogdan I. Fedeles
- Departments of Biological Engineering, Chemistry and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Rishi Bhardwaj
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Yasunobu Ishikawa
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Sakunchai Khumsubdee
- Departments of Biological Engineering, Chemistry and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, Bangkok10210, Thailand
| | - Matteus Krappitz
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Nina Gubina
- Departments of Biological Engineering, Chemistry and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino142290, Russia
| | - Isabel Volpe
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Denise C. Andrade
- Departments of Biological Engineering, Chemistry and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Parisa Westergerling
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Tobias Staudner
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Jake Campolo
- Departments of Biological Engineering, Chemistry and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Sally S. Liu
- Departments of Biological Engineering, Chemistry and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Ke Dong
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Yiqiang Cai
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Michael Rehman
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Anna-Rachel Gallagher
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Somsak Ruchirawat
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, Bangkok10210, Thailand
| | - Robert G. Croy
- Departments of Biological Engineering, Chemistry and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
| | - John M. Essigmann
- Departments of Biological Engineering, Chemistry and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Sorin V. Fedeles
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| | - Stefan Somlo
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT06510
| |
Collapse
|
13
|
Walker RV, Yao Q, Xu H, Maranto A, Swaney KF, Ramachandran S, Li R, Cassina L, Polster BM, Outeda P, Boletta A, Watnick T, Qian F. Fibrocystin/Polyductin releases a C-terminal fragment that translocates into mitochondria and suppresses cystogenesis. Nat Commun 2023; 14:6513. [PMID: 37845212 PMCID: PMC10579373 DOI: 10.1038/s41467-023-42196-4] [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: 08/31/2022] [Accepted: 10/03/2023] [Indexed: 10/18/2023] Open
Abstract
Fibrocystin/Polyductin (FPC), encoded by PKHD1, is associated with autosomal recessive polycystic kidney disease (ARPKD), yet its precise role in cystogenesis remains unclear. Here we show that FPC undergoes complex proteolytic processing in developing kidneys, generating three soluble C-terminal fragments (ICDs). Notably, ICD15, contains a novel mitochondrial targeting sequence at its N-terminus, facilitating its translocation into mitochondria. This enhances mitochondrial respiration in renal epithelial cells, partially restoring impaired mitochondrial function caused by FPC loss. FPC inactivation leads to abnormal ultrastructural morphology of mitochondria in kidney tubules without cyst formation. Moreover, FPC inactivation significantly exacerbates renal cystogenesis and triggers severe pancreatic cystogenesis in a Pkd1 mouse mutant Pkd1V/V in which cleavage of Pkd1-encoded Polycystin-1 at the GPCR Proteolysis Site is blocked. Deleting ICD15 enhances renal cystogenesis without inducing pancreatic cysts in Pkd1V/V mice. These findings reveal a direct link between FPC and a mitochondrial pathway through ICD15 cleavage, crucial for cystogenesis mechanisms.
Collapse
Affiliation(s)
- Rebecca V Walker
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Qin Yao
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Hangxue Xu
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anthony Maranto
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kristen F Swaney
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sreekumar Ramachandran
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, Singapore, 117411, Singapore
| | - Laura Cassina
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Patricia Outeda
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Terry Watnick
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Feng Qian
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Lin CC, Menezes LF, Qiu J, Pearson E, Zhou F, Ishimoto Y, Anderson DE, Germino GG. In vivo Polycystin-1 interactome using a novel Pkd1 knock-in mouse model. PLoS One 2023; 18:e0289778. [PMID: 37540694 PMCID: PMC10403143 DOI: 10.1371/journal.pone.0289778] [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: 03/15/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023] Open
Abstract
PKD1 is the most commonly mutated gene causing autosomal dominant polycystic kidney disease (ADPKD). It encodes Polycystin-1 (PC1), a putative membrane protein that undergoes a set of incompletely characterized post-transcriptional cleavage steps and has been reported to localize in multiple subcellular locations, including the primary cilium and mitochondria. However, direct visualization of PC1 and detailed characterization of its binding partners remain challenging. We now report a new mouse model with HA epitopes and eGFP knocked-in frame into the endogenous mouse Pkd1 gene by CRISPR/Cas9. Using this model, we sought to visualize endogenous PC1-eGFP and performed affinity-purification mass spectrometry (AP-MS) and network analyses. We show that the modified Pkd1 allele is fully functional but the eGFP-tagged protein cannot be detected without signal amplification by secondary antibodies. Using nanobody-coupled beads and large quantities of tissue, AP-MS identified an in vivo PC1 interactome, which is enriched for mitochondrial proteins and components of metabolic pathways. These studies suggest this mouse model and interactome data will be useful to understand PC1 function, but that new methods and brighter tags will be required to track endogenous PC1.
Collapse
Affiliation(s)
- Cheng-Chao Lin
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Luis F. Menezes
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jiahe Qiu
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Elisabeth Pearson
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fang Zhou
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yu Ishimoto
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - D. Eric Anderson
- Advanced Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gregory G. Germino
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
15
|
Leitner BP, Lee WD, Zhu W, Zhang X, Gaspar RC, Li Z, Rabinowitz JD, Perry RJ. Tissue-specific reprogramming of glutamine metabolism maintains tolerance to sepsis. PLoS One 2023; 18:e0286525. [PMID: 37410734 PMCID: PMC10325078 DOI: 10.1371/journal.pone.0286525] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/17/2023] [Indexed: 07/08/2023] Open
Abstract
Reprogramming metabolism is of great therapeutic interest for reducing morbidity and mortality during sepsis-induced critical illness. Disappointing results from randomized controlled trials targeting glutamine and antioxidant metabolism in patients with sepsis have begged a deeper understanding of the tissue-specific metabolic response to sepsis. The current study sought to fill this gap. We analyzed skeletal muscle transcriptomics of critically ill patients, versus elective surgical controls, which revealed reduced expression of genes involved in mitochondrial metabolism and electron transport, with increases in glutathione cycling, glutamine, branched chain, and aromatic amino acid transport. We then performed untargeted metabolomics and 13C isotope tracing to analyze systemic and tissue specific metabolic phenotyping in a murine polymicrobial sepsis model. We found an increased number of correlations between the metabolomes of liver, kidney, and spleen, with loss of correlations between the heart and quadriceps and all other organs, pointing to a shared metabolic signature within vital abdominal organs, and unique metabolic signatures for muscles during sepsis. A lowered GSH:GSSG and elevated AMP:ATP ratio in the liver underlie the significant upregulation of isotopically labeled glutamine's contribution to TCA cycle anaplerosis and glutamine-derived glutathione biosynthesis; meanwhile, the skeletal muscle and spleen were the only organs where glutamine's contribution to the TCA cycle was significantly suppressed. These results highlight tissue-specific mitochondrial reprogramming to support liver energetic demands and antioxidant synthesis, rather than global mitochondrial dysfunction, as a metabolic consequence of sepsis.
Collapse
Affiliation(s)
- Brooks P. Leitner
- Department of Cellular & Molecular Physiology, Yale University, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Won D. Lee
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Chemistry, Princeton University, Princeton, New Jersey, United States of America
| | - Wanling Zhu
- Department of Cellular & Molecular Physiology, Yale University, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Xinyi Zhang
- Department of Cellular & Molecular Physiology, Yale University, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Rafael C. Gaspar
- Department of Cellular & Molecular Physiology, Yale University, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Zongyu Li
- Department of Cellular & Molecular Physiology, Yale University, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Joshua D. Rabinowitz
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Chemistry, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, New Jersey, United States of America
| | - Rachel J. Perry
- Department of Cellular & Molecular Physiology, Yale University, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|