1
|
Monteillet L, Perrot G, Evrard F, Miliano A, Silva M, Leblond A, Nguyen C, Terzi F, Mithieux G, Rajas F. Impaired Glucose Metabolism, Primary Cilium Defects, and Kidney Cystogenesis in Glycogen Storage Disease Type Ia. J Am Soc Nephrol 2024; 35:1639-1654. [PMID: 39141438 PMCID: PMC11617483 DOI: 10.1681/asn.0000000000000452] [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: 01/07/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024] Open
Abstract
Key Points Metabolism adaptations due to glucose-6 phosphate accumulation in glycogen storage disease type Ia kidneys, toward a Warburg-like metabolism, promoted cell proliferation. Metabolic perturbations directly affected primary cilium structure and cystogenesis in glycogen storage disease type Ia kidneys. Background Glycogen storage disease type Ia (GSDIa) is a rare metabolic disorder caused by mutations in the catalytic subunit of glucose-6 phosphatase (G6PC1). This leads to severe hypoglycemia, and most young patients with GSDIa develop CKD. The kidney pathology is characterized by the development of cysts, which typically occur at an advanced stage of CKD. Methods To elucidate the molecular mechanisms responsible for cyst formation, we characterized renal metabolism, molecular pathways involved in cell proliferation, and primary cilium integrity using mice in which G6pc1 was specifically deleted in the kidney from an in utero stage. Results GSDIa mice exhibited kidney fibrosis, high inflammation, and cyst formation, leading to kidney dysfunction. In addition, the loss of G6PC1 led to the ectopic accumulation of glycogen and lipids in the kidneys and a metabolic shift toward a Warburg-like metabolism. This metabolic adaptation was due to an excess of glucose-6 phosphate, which supports cell proliferation, driven by the mitogen-activated protein kinase/extracellular signal–regulated kinases and protein kinase B/mammalian target of rapamycin pathways. Treatment of GSDIa mice with rapamycin, a target of the mammalian target of rapamycin pathway, reduced cell proliferation and kidney damage. Our results also identified lipocalin 2 as a contributor to renal inflammation and an early biomarker of CKD progression in GSDIa mice. Its inactivation partially prevented kidney lesions in GSDIa. Importantly, primary cilium defects were observed in the kidneys of GSDIa mice. Conclusions Metabolic adaptations because of glucose-6 phosphate accumulation in GSDIa renal tubules, toward a Warburg-like metabolism, promoted cell proliferation and cyst formation in a similar manner to that observed in various cystic kidney diseases. This was associated with downregulation of primary cilium gene expression and, consequently, altered cilium morphology.
Collapse
Affiliation(s)
- Laure Monteillet
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Gwendoline Perrot
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Félicie Evrard
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Alexane Miliano
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Marine Silva
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Alicia Leblond
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Clément Nguyen
- Université de Paris Cité, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades, Département “Croissance et Signalisation,” Paris, France
| | - Fabiola Terzi
- Université de Paris Cité, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades, Département “Croissance et Signalisation,” Paris, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| |
Collapse
|
2
|
Grahammer F, Dumoulin B, Gulieva RE, Wu H, Xu Y, Sulaimanov N, Arnold F, Sandner L, Cordts T, Todkar A, Moulin P, Reichardt W, Puelles VG, Kramann R, Freedman BS, Busch H, Boerries M, Walz G, Huber TB. Cyclin-dependent kinase 4 drives cystic kidney disease in the absence of mTORC1 signaling activity. Kidney Int 2024; 106:856-869. [PMID: 39218392 DOI: 10.1016/j.kint.2024.08.021] [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: 12/19/2022] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Progression of cystic kidney disease has been linked to activation of the mTORC1 signaling pathway. Yet the utility of mTORC1 inhibitors to treat patients with polycystic kidney disease remains controversial despite promising preclinical data. To define the cell intrinsic role of mTORC1 for cyst development, the mTORC1 subunit gene Raptor was selectively inactivated in kidney tubular cells lacking cilia due to simultaneous deletion of the kinesin family member gene Kif3A. In contrast to a rapid onset of cyst formation and kidney failure in mice with defective ciliogenesis, both kidney function, cyst formation discerned by magnetic resonance imaging and overall survival were strikingly improved in mice additionally lacking Raptor. However, these mice eventually succumbed to cystic kidney disease despite mTORC1 inactivation. In-depth transcriptome analysis revealed the rapid activation of other growth-promoting signaling pathways, overriding the effects of mTORC1 deletion and identified cyclin-dependent kinase (CDK) 4 as an alternate driver of cyst growth. Additional inhibition of CDK4-dependent signaling by the CDK4/6 inhibitor Palbociclib markedly slowed disease progression in mice and human organoid models of polycystic kidney disease and potentiated the effects of mTORC1 deletion/inhibition. Our findings indicate that cystic kidneys rapidly adopt bypass mechanisms typically observed in drug resistant cancers. Thus, future clinical trials need to consider combinatorial or sequential therapies to improve therapeutic efficacy in patients with cystic kidney disease.
Collapse
Affiliation(s)
- Florian Grahammer
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Bernhard Dumoulin
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ramila E Gulieva
- Division of Nephrology, University of Washington School of Medicine, Seattle, Washington, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, Washington, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Hui Wu
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yaoxian Xu
- Institute of Experimental Medicine and Systems Biology, Medical Faculty, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Nurgazy Sulaimanov
- Department of Electrical Engineering and Information Technology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Frederic Arnold
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Lukas Sandner
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Tomke Cordts
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Abhijeet Todkar
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Pierre Moulin
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | - Wilfried Reichardt
- Department of Diagnostic and Interventional Radiology, Division of Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, Medical Faculty, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Benjamin S Freedman
- Division of Nephrology, University of Washington School of Medicine, Seattle, Washington, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, Washington, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Plurexa LLC, Seattle, Washington, USA
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between Deutsches Krebs Forschungs Zentrum (DKFZ) and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Gerd Walz
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany; Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Raya AI, Vidal A, López I, Rodríguez M, Aguilera-Tejero E, Pineda C. Phosphorus Restriction Prevents Rapamycin-Induced Kidney Damage in Rats. Am J Nephrol 2024; 56:48-57. [PMID: 39383849 DOI: 10.1159/000541411] [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: 08/01/2024] [Accepted: 09/08/2024] [Indexed: 10/11/2024]
Abstract
INTRODUCTION There are conflicting reports about the effect or rapamycin on the kidneys. Rapamycin is known to promote phosphaturia that may be associated to renal injury. METHODS Detailed histopathological studies were performed on the kidneys of rats with normal (control) and reduced (Nx) renal mass that were treated with rapamycin (1.3 mg/kg for 22 days) or placebo. The effect of rapamycin was also evaluated in control and Nx rats fed different amounts of phosphorus: 0.6% P (NP), 1.2% P (HP), and 0.2% P (LP). Quantitative scores of kidney lesions were obtained for interstitial nephritis (IN), tubular damage (TD), and nephrocalcinosis (NC). RESULTS When compared with placebo, rapamycin administration to Nx rats resulted in significant increases in IN (4.17 ± 0.74 vs. 1.51 ± 0.53%) and TD (14.45 ± 1.51 vs. 8.61 ± 1.83%). Rapamycin also increased NC both in control (0.86 ± 0.23 vs. 0.14 ± 0.06%) and Nx (0.86 ± 0.32 vs. 0.15 ± 0.14%) rats. In control rats receiving rapamycin, feeding HP aggravated IN (3.25 ± 0.48%), TD (22.47 ± 4.56%), and NC (3.66 ± 0.75%), while feeding LP prevented development of any renal lesions. In Nx rats treated with rapamycin, HP intake also increased IN (8.95 ± 1.94%), TD (26.86 ± 3.95%), and NC (2.77 ± 0.60%), whereas feeding LP reduced all lesions to lower levels than in rats fed NP. Rapamycin treatment increased fractional excretion of P (FEP), and an excellent correlation between scores for renal lesions and FEP was found. CONCLUSION Rapamycin has deleterious effects on kidney pathology causing lesions that are located mainly at tubular and tubulointerstitial level. Rapamycin-induced kidney damage is more evident in rats that already have decreased renal function and seems to be related to the phosphaturic effect of the drug. Dietary P restriction prevents kidney damage in rats treated with rapamycin.
Collapse
Affiliation(s)
- Ana I Raya
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Angela Vidal
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Ignacio López
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Mariano Rodríguez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Escolástico Aguilera-Tejero
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Carmen Pineda
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| |
Collapse
|
4
|
Su L, Chen T, Hu H, Xu Z, Luan X, Fu K, Ren Y, Sun D, Sun Y, Guo D. Notch3 as a novel therapeutic target for the treatment of ADPKD by regulating cell proliferation and renal cyst development. Biochem Pharmacol 2024; 224:116200. [PMID: 38604258 DOI: 10.1016/j.bcp.2024.116200] [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: 11/13/2023] [Revised: 01/22/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common monogenic kidney disease. Emerging research indicates that the Notch signaling pathway plays an indispensable role in the pathogenesis of numerous kidney diseases, including ADPKD. Herein, we identified that Notch3 but not other Notch receptors was overexpressed in renal tissues from mice with ADPKD and ADPKD patients. Inhibiting Notch3 with γ-secretase inhibitors, which block a proteolytic cleavage required for Notch3 activation, or shRNA knockdown of Notch3 significantly delayed renal cyst growth in vitro and in vivo. Subsequent mechanistic study elucidated that the cleaved intracellular domain of Notch3 (N3ICD) and Hes1 could bind to the PTEN promoter, leading to transcriptional inhibition of PTEN. This further activated the downstream PI3K-AKT-mTOR pathway and promoted renal epithelial cell proliferation. Overall, Notch3 was identified as a novel contributor to renal epithelial cell proliferation and cystogenesis in ADPKD. We envision that Notch3 represents a promising target for ADPKD treatment.
Collapse
Affiliation(s)
- Limin Su
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ting Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Hongtao Hu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Zifan Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xiande Luan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Kequan Fu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ying Ren
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Dong Sun
- Department of Urology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China.
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
5
|
Lan Q, Li J, Zhang H, Zhou Z, Fang Y, Yang B. Mechanistic complement of autosomal dominant polycystic kidney disease: the role of aquaporins. J Mol Med (Berl) 2024; 102:773-785. [PMID: 38668786 DOI: 10.1007/s00109-024-02446-4] [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: 01/09/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/21/2024]
Abstract
Autosomal dominant polycystic kidney disease is a genetic kidney disease caused by mutations in the genes PKD1 or PKD2. Its course is characterized by the formation of progressively enlarged cysts in the renal tubules bilaterally. The basic genetic explanation for autosomal dominant polycystic kidney disease is the double-hit theory, and many of its mechanistic issues can be explained by the cilia doctrine. However, the precise molecular mechanisms underpinning this condition's occurrence are still not completely understood. Experimental evidence suggests that aquaporins, a class of transmembrane channel proteins, including aquaporin-1, aquaporin-2, aquaporin-3, and aquaporin-11, are involved in the mechanism of autosomal dominant polycystic kidney disease. Aquaporins are either a potential new target for the treatment of autosomal dominant polycystic kidney disease, and further study into the physiopathological role of aquaporins in autosomal dominant polycystic kidney disease will assist to clarify the disease's pathophysiology and increase the pool of potential treatment options. We primarily cover pertinent findings on aquaporins in autosomal dominant polycystic kidney disease in this review.
Collapse
Affiliation(s)
- Qiumei Lan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Jie Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Hanqing Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Zijun Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Yaxuan Fang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Bo Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
- Department of Nephrology, The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, No.88, Changling Road, Xiqing District, Tianjin, 300193, China.
| |
Collapse
|
6
|
Srimai N, Tonum K, Soodvilai S. Activation of farnesoid X receptor retards expansion of renal collecting duct cell-derived cysts via inhibition of CFTR-mediated Cl - secretion. Am J Physiol Renal Physiol 2024; 326:F600-F610. [PMID: 38299213 DOI: 10.1152/ajprenal.00363.2023] [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: 11/07/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024] Open
Abstract
The transcription factor farnesoid X receptor (FXR) regulates energy metabolism. Specifically, FXR functions to regulate cystic fibrosis transmembrane conductance regulator (CFTR)-mediated Cl- secretion in intestinal epithelial cells. Therefore, this study aimed to investigate the role of FXR in CFTR-mediated Cl- secretion in renal tubular cells and to further elucidate its effects on renal cyst formation and growth. CFTR-mediated Cl- transport was evaluated via short-circuit current (ISC) measurements in Madin-Darby canine kidney (MDCK) cell monolayers and primary rat inner medullary collecting duct cells. The role of FXR in renal cyst formation and growth was determined by the MDCK cell-derived cyst model. Incubation with synthesized (GW4064) and endogenous (CDCA) FXR ligands reduced CFTR-mediated Cl- secretion in a concentration- and time-dependent manner. The inhibitory effect of FXR ligands was not due to the result of reduced cell viability and was attenuated by cotreatment with an FXR antagonist. FXR activation significantly decreased CFTR protein but not its mRNA. In addition, FXR activation inhibited CFTR-mediated Cl- secretion in primary renal collecting duct cells. FXR activation decreased ouabain-sensitive ISC without altering Na+-K+-ATPase mRNA and protein levels. Furthermore, FXR activation significantly reduced the number of cysts and renal cyst expansion. These inhibitory effects were correlated with a decrease in the expression of protein synthesis regulators mammalian target of rapamycin/S6 kinase. This study shows that FXR activation inhibits Cl- secretion in renal cells via inhibition of CFTR expression and retards renal cyst formation and growth. The discoveries point to a physiological role of FXR in the regulation of CFTR and a potential therapeutic application in polycystic kidney disease treatment.NEW & NOTEWORTHY The present study reveals that farnesoid X receptor (FXR) activation reduces microcyst formation and enlargement. This inhibitory effect of FXR activation is involved with decreased cell proliferation and cystic fibrosis transmembrane conductance regulator-mediated Cl- secretion in renal collecting duct cells. FXR might represent a novel target for the treatment of autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Nipitpon Srimai
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Kanlayanee Tonum
- Department of Physiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Sunhapas Soodvilai
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Excellent Center for Drug Discovery, Mahidol University, Bangkok, Thailand
| |
Collapse
|
7
|
Prosseda PP, Dannewitz Prosseda S, Tran M, Liton PB, Sun Y. Crosstalk between the mTOR pathway and primary cilia in human diseases. Curr Top Dev Biol 2023; 155:1-37. [PMID: 38043949 PMCID: PMC11227733 DOI: 10.1016/bs.ctdb.2023.09.004] [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] [Indexed: 12/05/2023]
Abstract
Autophagy is a fundamental catabolic process whereby excessive or damaged cytoplasmic components are degraded through lysosomes to maintain cellular homeostasis. Studies of mTOR signaling have revealed that mTOR controls biomass generation and metabolism by modulating key cellular processes, including protein synthesis and autophagy. Primary cilia, the assembly of which depends on kinesin molecular motors, serve as sensory organelles and signaling platforms. Given these pathways' central role in maintaining cellular and physiological homeostasis, a connection between mTOR and primary cilia signaling is starting to emerge in a variety of diseases. In this review, we highlight recent advances in our understanding of the complex crosstalk between the mTOR pathway and cilia and discuss its function in the context of related diseases.
Collapse
Affiliation(s)
- Philipp P Prosseda
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | | | - Matthew Tran
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Paloma B Liton
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States; Palo Alto Veterans Administration Medical Center, Palo Alto, CA, United States.
| |
Collapse
|
8
|
Zhou JX, Torres VE. Autosomal Dominant Polycystic Kidney Disease Therapies on the Horizon. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:245-260. [PMID: 37088527 DOI: 10.1053/j.akdh.2023.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 04/25/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the formation of numerous kidney cysts which leads to kidney failure. ADPKD is responsible for approximately 10% of patients with kidney failure. Overwhelming evidence supports that vasopressin and its downstream cyclic adenosine monophosphate signaling promote cystogenesis, and targeting vasopressin 2 receptor with tolvaptan and other antagonists ameliorates cyst growth in preclinical studies. Tolvaptan is the only drug approved by Food and Drug Administration to treat ADPKD patients at the risk of rapid disease progression. A major limitation of the widespread use of tolvaptan is aquaretic events. This review discusses the potential strategies to improve the tolerability of tolvaptan, the progress on the use of an alternative vasopressin 2 receptor antagonist lixivaptan, and somatostatin analogs. Recent advances in understanding the pathophysiology of PKD have led to new approaches of treatment via targeting different signaling pathways. We review the new pharmacotherapies and dietary interventions of ADPKD that are promising in the preclinical studies and investigated in clinical trials.
Collapse
|
9
|
Zhou JX, Torres VE. Drug repurposing in autosomal dominant polycystic kidney disease. Kidney Int 2023; 103:859-871. [PMID: 36870435 DOI: 10.1016/j.kint.2023.02.010] [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: 11/18/2022] [Revised: 01/23/2023] [Accepted: 02/07/2023] [Indexed: 03/06/2023]
Abstract
Autosomal dominant polycystic kidney disease is characterized by progressive kidney cyst formation that leads to kidney failure. Tolvaptan, a vasopressin 2 receptor antagonist, is the only drug approved to treat patients with autosomal dominant polycystic kidney disease who have rapid disease progression. The use of tolvaptan is limited by reduced tolerability from aquaretic effects and potential hepatotoxicity. Thus, the search for more effective drugs to slow down the progression of autosomal dominant polycystic kidney disease is urgent and challenging. Drug repurposing is a strategy for identifying new clinical indications for approved or investigational medications. Drug repurposing is increasingly becoming an attractive proposition because of its cost-efficiency and time-efficiency and known pharmacokinetic and safety profiles. In this review, we focus on the repurposing approaches to identify suitable drug candidates to treat autosomal dominant polycystic kidney disease and prioritization and implementation of candidates with high probability of success. Identification of drug candidates through understanding of disease pathogenesis and signaling pathways is highlighted.
Collapse
Affiliation(s)
- Julie Xia Zhou
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA; Mayo Clinic Robert M. and Billie Kelley Pirnie Translational Polycystic Kidney Disease Center, Rochester, Minnesota, USA.
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA; Mayo Clinic Robert M. and Billie Kelley Pirnie Translational Polycystic Kidney Disease Center, Rochester, Minnesota, USA.
| |
Collapse
|
10
|
Li LX, Zhang X, Zhang H, Agborbesong E, Zhou JX, Calvet JP, Li X. Single-Cell and CellChat Resolution Identifies Collecting Duct Cell Subsets and Their Communications with Adjacent Cells in PKD Kidneys. Cells 2022; 12:45. [PMID: 36611841 PMCID: PMC9818381 DOI: 10.3390/cells12010045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/02/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
ADPKD is a genetic disorder with a molecular complexity that remains poorly understood. In this study, we sampled renal cells to construct a comprehensive and spatiotemporally resolved gene expression atlas in whole Pkd1 mutant polycystic mouse kidneys at single-cell resolution. We characterized cell diversity and identified novel collecting duct (CD) cell subtypes in cystic kidneys. We further found that CD cells appear to take different cell fate trajectories, and the first and the most important step might take place around day 14 in Pkd1 homozygous kidneys. After that day, increased numbers of CD cells showed highly proliferative and fibrotic characteristics, as detected in later-stage Pkd1 homozygous kidneys, both of which should contribute to cyst growth and renal fibrosis. With a newly developed modeling algorithm, called CellChat Explorer, we identify cell-to-cell communication networks mediated by the ligand receptor, such as MIF-CD44/CD74, in cystic kidneys, and confirm them via the expression patterns of ligands and receptors in four major cell types, which addresses the key question as to whether and how Pkd1 mutant renal epithelial cells affect their neighboring cells. The allele-specific gene expression profiles show that the secretion of cytokines by Pkd1 mutant epithelial cells may affect the gene expression profiles in recipient cells via epigenetic mechanisms, and vice versa. This study can be used to drive precision therapeutic targeting of ADPKD.
Collapse
Affiliation(s)
- Linda Xiaoyan Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physical Medicine & Rehabilitation, Mayo Clinic, Rochester, MN 55905, USA
| | - Hongbing Zhang
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Ewud Agborbesong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Julie Xia Zhou
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - James P. Calvet
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
11
|
Zheng Q, Reid G, Eccles MR, Stayner C. Non-coding RNAs as potential biomarkers and therapeutic targets in polycystic kidney disease. Front Physiol 2022; 13:1006427. [PMID: 36203940 PMCID: PMC9531119 DOI: 10.3389/fphys.2022.1006427] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Polycystic kidney disease (PKD) is a significant cause of end-stage kidney failure and there are few effective drugs for treating this inherited condition. Numerous aberrantly expressed non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs), may contribute to PKD pathogenesis by participating in multiple intracellular and intercellular functions through post-transcriptional regulation of protein-encoding genes. Insights into the mechanisms of miRNAs and other ncRNAs in the development of PKD may provide novel therapeutic strategies. In this review, we discuss the current knowledge about the roles of dysregulated miRNAs and other ncRNAs in PKD. These roles involve multiple aspects of cellular function including mitochondrial metabolism, proliferation, cell death, fibrosis and cell-to-cell communication. We also summarize the potential application of miRNAs as biomarkers or therapeutic targets in PKD, and briefly describe strategies to overcome the challenges of delivering RNA to the kidney, providing a better understanding of the fundamental advances in utilizing miRNAs and other non-coding RNAs to treat PKD.
Collapse
Affiliation(s)
| | | | | | - Cherie Stayner
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
12
|
Shi H, Jin L, Li J, Liang K, Li X, Ye Z, Zhu X, Oliveira JM, Reis RL, Mao Z, Wu M. Mesoporous polydopamine nanoparticles for sustained release of rapamycin and reactive oxygen species scavenging to synergistically accelerate neurogenesis after spinal cord injury. J Mater Chem B 2022; 10:6351-6359. [PMID: 35942619 DOI: 10.1039/d2tb00841f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Spinal cord injury (SCI) is an intractable condition with complex pathological processes and poor prognosis. Reactive oxygen species (ROS) generation induced by the mammalian target of the rapamycin (mTOR) protein is one of the causes of secondary inflammation of SCI. Rapamycin (Rapa) is a pharmacological inhibitor of mTOR, which can inhibit ROS overproduction mediated by abnormal activation of the mTOR protein. Polydopamine, as a nanocarrier with excellent biological safety, has been reported to possess satisfactory ROS scavenging ability. Therefore, we designed a mesoporous polydopamine nanoparticle loaded with Rapa (mPDA@Rapa) for combination therapy, which simultaneously inhibited abnormally activated mTOR-mediated ROS production and eliminated already generated ROS. The synthesized mPDA nanoparticles could realize the effective encapsulation and sustained release of Rapa due to their mesoporous cavities and a hydrophobic benzene ring structure. In vitro experiments proved that mPDA@Rapa nanoparticles had a good ROS scavenging ability towards hydrogen peroxide and hydroxyl radicals. Furthermore, mPDA@Rapa also showed a good therapeutic effect in SCI model rats, which was evidenced by a smaller injury cavity, more coordinated hind limb movements, and a higher degree of neurogenesis and tissue regeneration. Our work provides a combined strategy to inhibit ROS overproduction and eliminate excess ROS, with potential applications not only in SCI, but also in other ROS-induced inflammations.
Collapse
Affiliation(s)
- Haifei Shi
- Department of Orthopedics, 1st Affiliated Hospital of Zhejiang University School of Medicine, Qingchun Road 79, Hangzhou, 310003, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Jinyi Li
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Kejiong Liang
- Department of Orthopedics, 1st Affiliated Hospital of Zhejiang University School of Medicine, Qingchun Road 79, Hangzhou, 310003, China
| | - Xigong Li
- Department of Orthopedics, 1st Affiliated Hospital of Zhejiang University School of Medicine, Qingchun Road 79, Hangzhou, 310003, China
| | - Ziqiang Ye
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Xinyue Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017, Barco GMR, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017, Barco GMR, Portugal
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Mengjie Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
13
|
Gupta S, Ozimek-Kulik JE, Phillips JK. Nephronophthisis-Pathobiology and Molecular Pathogenesis of a Rare Kidney Genetic Disease. Genes (Basel) 2021; 12:genes12111762. [PMID: 34828368 PMCID: PMC8623546 DOI: 10.3390/genes12111762] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/17/2022] Open
Abstract
The exponential rise in our understanding of the aetiology and pathophysiology of genetic cystic kidney diseases can be attributed to the identification of cystogenic genes over the last three decades. The foundation of this was laid by positional cloning strategies which gradually shifted towards next-generation sequencing (NGS) based screenings. This shift has enabled the discovery of novel cystogenic genes at an accelerated pace unlike ever before and, most notably, the past decade has seen the largest increase in identification of the genes which cause nephronophthisis (NPHP). NPHP is a monogenic autosomal recessive cystic kidney disease caused by mutations in a diverse clade of over 26 identified genes and is the most common genetic cause of renal failure in children. NPHP gene types present with some common pathophysiological features alongside a diverse range of extra-renal phenotypes associated with specific syndromic presentations. This review provides a timely update on our knowledge of this disease, including epidemiology, pathophysiology, anatomical and molecular features. We delve into the diversity of the NPHP causing genes and discuss known molecular mechanisms and biochemical pathways that may have possible points of intersection with polycystic kidney disease (the most studied renal cystic pathology). We delineate the pathologies arising from extra-renal complications and co-morbidities and their impact on quality of life. Finally, we discuss the current diagnostic and therapeutic modalities available for disease management, outlining possible avenues of research to improve the prognosis for NPHP patients.
Collapse
Affiliation(s)
- Shabarni Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
- Correspondence:
| | - Justyna E. Ozimek-Kulik
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
- School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW 2031, Australia
- Department of Paediatric Nephrology, Sydney Children’s Hospital Network, Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Jacqueline Kathleen Phillips
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
| |
Collapse
|
14
|
Madhivanan K, Ramadesikan S, Hsieh WC, Aguilar MC, Hanna CB, Bacallao RL, Aguilar RC. Lowe syndrome patient cells display mTOR- and RhoGTPase-dependent phenotypes alleviated by rapamycin and statins. Hum Mol Genet 2021; 29:1700-1715. [PMID: 32391547 DOI: 10.1093/hmg/ddaa086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/20/2020] [Accepted: 05/04/2020] [Indexed: 12/25/2022] Open
Abstract
Lowe syndrome (LS) is an X-linked developmental disease characterized by cognitive deficiencies, bilateral congenital cataracts and renal dysfunction. Unfortunately, this disease leads to the early death of affected children often due to kidney failure. Although this condition was first described in the early 1950s and the affected gene (OCRL1) was identified in the early 1990s, its pathophysiological mechanism is not fully understood and there is no LS-specific cure available to patients. Here we report two important signaling pathways affected in LS patient cells. While RhoGTPase signaling abnormalities led to adhesion and spreading defects as compared to normal controls, PI3K/mTOR hyperactivation interfered with primary cilia assembly (scenario also observed in other ciliopathies with compromised kidney function). Importantly, we identified two FDA-approved drugs able to ameliorate these phenotypes. Specifically, statins mitigated adhesion and spreading abnormalities while rapamycin facilitated ciliogenesis in LS patient cells. However, no single drug was able to alleviate both phenotypes. Based on these and other observations, we speculate that Ocrl1 has dual, independent functions supporting proper RhoGTPase and PI3K/mTOR signaling. Therefore, this study suggest that Ocrl1-deficiency leads to signaling defects likely to require combinatorial drug treatment to suppress patient phenotypes and symptoms.
Collapse
Affiliation(s)
- Kayalvizhi Madhivanan
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Swetha Ramadesikan
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Wen-Chieh Hsieh
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Mariana C Aguilar
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Claudia B Hanna
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Robert L Bacallao
- Division of Nephrology, Indiana University School of Medicine, 340 W 10th St #6200, Indianapolis, IN 46202, USA
| | - R Claudio Aguilar
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| |
Collapse
|
15
|
Nowak KL, Steele C, Gitomer B, Wang W, Ouyang J, Chonchol MB. Overweight and Obesity and Progression of ADPKD. Clin J Am Soc Nephrol 2021; 16:908-915. [PMID: 34117082 PMCID: PMC8216617 DOI: 10.2215/cjn.16871020] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/11/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES On the basis of earlier observations, we evaluated the association between overweight and obesity and rapid progression of autosomal dominant polycystic kidney disease in participants in the Tolvaptan Efficacy and Safety in Management of Autosomal Dominant Polycystic Kidney Disease and Its Outcomes (TEMPO) 3:4 trial. More importantly, we also determined whether efficacy of tolvaptan was attenuated in individuals with baseline overweight or obesity. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS A total of 1312 study participants with relatively early-stage autosomal dominant polycystic kidney disease (mean eGFR 78±22 ml/min per 1.73 m2) who were at high risk of rapid progression were categorized by body mass index (BMI; calculated using nonkidney weight) as normal weight (18.5-24.9 kg/m2; n=670), overweight (25.0-29.9 kg/m2; n=429), or obese (≥30 kg/m2; n=213). Linear and multinomial logistic regression models were used to determine the association of baseline overweight and obesity with change in total kidney volume (TKV) over the 3-year study period. RESULTS In fully adjusted models, higher BMI was associated with greater annual percent change in TKV (difference of 1.20 [95% confidence interval (95% CI), 0.85 to 1.55] per five-unit higher BMI). Overweight and obesity were associated with higher odds of annual percent change in TKV of ≥7% versus <5% (overweight: odds ratio, 2.04 [95% CI, 1.45 to 2.87]; obese: odds ratio, 4.31 [95% CI, 2.83 to 6.57] versus normal weight). eGFR decline did not differ according to BMI (fully adjusted difference in decline of -0.95 [95% CI, -2.32 to 0.40] ml/min per 1.73 m2 per year per five-unit higher BMI). The three-way interaction (treatment×time×BMI group) was not statistically significant in linear mixed models with an outcome of TKV (log-transformed estimated coefficient comparing the treatment effect for overweight versus normal weight: 0.56% [95% CI, -0.70% to 1.84%] per year; P=0.38; obese versus normal weight: 0.07% [95% CI, -1.47% to 1.63%] per year; P=0.93) or eGFR (estimated coefficient comparing overweight versus normal weight: -0.07 [95% CI, -0.95 to 0.82] ml/min per 1.73 m2 per year; P=0.88; obese versus normal weight: 0.22 [95% CI, -0.93 to 1.36] ml/min per 1.73 m2 per year; P=0.71). CONCLUSIONS Overweight and particularly obesity are strongly and independently associated with kidney growth, but not eGFR slope, in the TEMPO 3:4 trial, and tolvaptan efficacy is irrespective of BMI categorization. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER Tolvaptan Efficacy and Safety in Management of Autosomal Dominant Polycystic Kidney Disease and Its Outcomes (TEMPO) 3:4, NCT00428948.
Collapse
Affiliation(s)
- Kristen L. Nowak
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Cortney Steele
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Berenice Gitomer
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Wenchyi Wang
- Otsuka Pharmaceutical Development and Commercialization, Princeton, New Jersey
| | - John Ouyang
- Otsuka Pharmaceutical Development and Commercialization, Rockville, Maryland
| | | |
Collapse
|
16
|
Radadiya PS, Thornton MM, Puri RV, Yerrathota S, Dinh-Phan J, Magenheimer B, Subramaniam D, Tran PV, Zhu H, Bolisetty S, Calvet JP, Wallace DP, Sharma M. Ciclopirox olamine induces ferritinophagy and reduces cyst burden in polycystic kidney disease. JCI Insight 2021; 6:141299. [PMID: 33784251 PMCID: PMC8119220 DOI: 10.1172/jci.insight.141299] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 03/24/2021] [Indexed: 01/21/2023] Open
Abstract
Despite the recent launch of tolvaptan, the search for safer polycystic kidney disease (PKD) drugs continues. Ciclopirox (CPX) or its olamine salt (CPX-O) is contained in a number of commercially available antifungal agents. CPX is also reported to possess anticancer activity. Several mechanisms of action have been proposed, including chelation of iron and inhibition of iron-dependent enzymes. Here, we show that CPX-O inhibited in vitro cystogenesis of primary human PKD cyst-lining epithelial cells cultured in a 3D collagen matrix. To assess the in vivo role of CPX-O, we treated PKD mice with CPX-O. CPX-O reduced the kidney-to-body weight ratios of PKD mice. The CPX-O treatment was also associated with decreased cell proliferation, decreased cystic area, and improved renal function. Ferritin levels were markedly elevated in cystic kidneys of PKD mice, and CPX-O treatment reduced renal ferritin levels. The reduction in ferritin was associated with increased ferritinophagy marker nuclear receptor coactivator 4, which reversed upon CPX-O treatment in PKD mice. Interestingly, these effects on ferritin appeared independent of iron. These data suggest that CPX-O can induce ferritin degradation via ferritinophagy, which is associated with decreased cyst growth progression in PKD mice. Most importantly these data indicate that CPX-O has the potential to treat autosomal dominant PKD.
Collapse
Affiliation(s)
| | | | - Rajni V. Puri
- Department of Internal Medicine
- Jared Grantham Kidney Institute
| | | | | | - Brenda Magenheimer
- Jared Grantham Kidney Institute
- Department of Biochemistry and Molecular Biology
| | | | - Pamela V. Tran
- Jared Grantham Kidney Institute
- Department of Anatomy and Cell Biology, and
| | - Hao Zhu
- Jared Grantham Kidney Institute
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Subhashini Bolisetty
- Department of Internal Medicine, School of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - James P. Calvet
- Jared Grantham Kidney Institute
- Department of Biochemistry and Molecular Biology
| | | | | |
Collapse
|
17
|
Novel Potential Application of Chitosan Oligosaccharide for Attenuation of Renal Cyst Growth in the Treatment of Polycystic Kidney Disease. Molecules 2020; 25:molecules25235589. [PMID: 33261193 PMCID: PMC7730275 DOI: 10.3390/molecules25235589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
Chitosan oligosaccharide (COS), a natural polymer derived from chitosan, exerts several biological activities including anti-inflammation, anti-tumor, anti-metabolic syndrome, and drug delivery enhancer. Since COS is vastly distributed to kidney and eliminated in urine, it may have a potential advantage as the therapeutics of kidney diseases. Polycystic kidney disease (PKD) is a common genetic disorder characterized by multiple fluid-filled cysts, replacing normal renal parenchyma and leading to impaired renal function and end-stage renal disease (ESRD). The effective treatment for PKD still needs to be further elucidated. Interestingly, AMP-activated protein kinase (AMPK) has been proposed as a drug target for PKD. This study aimed to investigate the effect of COS on renal cyst enlargement and its underlying mechanisms. We found that COS at the concentrations of 50 and 100 µg/mL decreased renal cyst growth without cytotoxicity, as measured by MTT assay. Immunoblotting analysis showed that COS at 100 µg/mL activated AMPK, and this effect was abolished by STO-609, a calcium/calmodulin-dependent protein kinase kinase beta (CaMKKβ) inhibitor. Moreover, COS elevated the level of intracellular calcium. These results suggest that COS inhibits cyst progression by activation of AMPK via CaMKKβ. Therefore, COS may hold the potential for pharmaceutical application in PKD.
Collapse
|
18
|
Cyst Reduction by Melatonin in a Novel Drosophila Model of Polycystic Kidney Disease. Molecules 2020; 25:molecules25225477. [PMID: 33238462 PMCID: PMC7700119 DOI: 10.3390/molecules25225477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/14/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) causes progressive cystic degeneration of the renal tubules, the nephrons, eventually severely compromising kidney function. ADPKD is incurable, with half of the patients eventually needing renal replacement. Treatments for ADPKD patients are limited and new effective therapeutics are needed. Melatonin, a central metabolic regulator conserved across all life kingdoms, exhibits oncostatic and oncoprotective activity and no detected toxicity. Here, we used the Bicaudal C (BicC) Drosophila model of polycystic kidney disease to test the cyst-reducing potential of melatonin. Significant cyst reduction was found in the renal (Malpighian) tubules upon melatonin administration and suggest mechanistic sophistication. Similar to vertebrate PKD, the BicC fly PKD model responds to the antiproliferative drugs rapamycin and mimics of the second mitochondria-derived activator of caspases (Smac). Melatonin appears to be a new cyst-reducing molecule with attractive properties as a potential candidate for PKD treatment.
Collapse
|
19
|
Morita M, Kanasaki K. Sodium-glucose cotransporter-2 inhibitors for diabetic kidney disease: Targeting Warburg effects in proximal tubular cells. DIABETES & METABOLISM 2020; 46:353-361. [PMID: 32891754 DOI: 10.1016/j.diabet.2020.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/27/2020] [Accepted: 06/08/2020] [Indexed: 12/29/2022]
Abstract
Inhibitors of sodium-glucose cotransporter 2 (SGLT2) have undoubtedly shifted the paradigm for diabetes medicine and research and, especially, diabetic kidney disease (DKD). The pharmacological action of SGLT2 inhibitors is simply the release of glucose into urine; however, precisely how SGLT2 inhibitors contribute to the health of those with diabetes has still not been completely elucidated. Towards this end, the present review provides a novel insight into the action of SGLT2 inhibitors by highlighting a neglected fuel-burning system found in proximal tubular cells-'glycolysis'. In addition, exploring the details of the molecular mechanisms and clinical biomarkers of the organ protection conferred by SGLT2 inhibitors is now required to prepare for the next stage of clinical diabetes medicine-the 'post-SGLT2 inhibitor era'.
Collapse
Affiliation(s)
- Miwa Morita
- Department of Internal Medicine 1, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan
| | - Keizo Kanasaki
- Department of Internal Medicine 1, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan.
| |
Collapse
|
20
|
Testa F, Marchiò M, D’Amico R, Giovanella S, Ligabue G, Fontana F, Alfano G, Cappelli G, Biagini G, Magistroni R. GREASE II. A phase II randomized, 12-month, parallel-group, superiority study to evaluate the efficacy of a Modified Atkins Diet in Autosomal Dominant Polycystic Kidney Disease patients. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Reciprocal Regulation between Primary Cilia and mTORC1. Genes (Basel) 2020; 11:genes11060711. [PMID: 32604881 PMCID: PMC7349257 DOI: 10.3390/genes11060711] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022] Open
Abstract
In quiescent cells, primary cilia function as a mechanosensor that converts mechanic signals into chemical activities. This unique organelle plays a critical role in restricting mechanistic target of rapamycin complex 1 (mTORC1) signaling, which is essential for quiescent cells to maintain their quiescence. Multiple mechanisms have been identified that mediate the inhibitory effect of primary cilia on mTORC1 signaling. These mechanisms depend on several tumor suppressor proteins localized within the ciliary compartment, including liver kinase B1 (LKB1), AMP-activated protein kinase (AMPK), polycystin-1, and polycystin-2. Conversely, changes in mTORC1 activity are able to affect ciliogenesis and stability indirectly through autophagy. In this review, we summarize recent advances in our understanding of the reciprocal regulation of mTORC1 and primary cilia.
Collapse
|
22
|
Millet-Boureima C, Selber-Hnatiw S, Gamberi C. Drug discovery and chemical probing in Drosophila. Genome 2020; 64:147-159. [PMID: 32551911 DOI: 10.1139/gen-2020-0037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Flies are increasingly utilized in drug discovery and chemical probing in vivo, which are novel technologies complementary to genetic probing in fundamental biological studies. Excellent genetic conservation, small size, short generation time, and over one hundred years of genetics make Drosophila an attractive model for rapid assay readout and use of analytical amounts of compound, enabling the experimental iterations needed in early drug development at a fraction of time and costs. Here, we describe an effective drug-testing pipeline using adult flies that can be easily implemented to study several disease models and different genotypes to discover novel molecular insight, probes, quality lead compounds, and develop novel prototype drugs.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Susannah Selber-Hnatiw
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| |
Collapse
|
23
|
Zhang JQJ, Burgess J, Stepanova D, Saravanabavan S, Wong ATY, Kaldis P, Rangan GK. Role of cyclin-dependent kinase 2 in the progression of mouse juvenile cystic kidney disease. J Transl Med 2020; 100:696-711. [PMID: 31915367 DOI: 10.1038/s41374-019-0360-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022] Open
Abstract
A hallmark of polycystic kidney diseases (PKDs) is aberrant proliferation, which leads to the formation and growth of renal cysts. Proliferation is mediated by cyclin-dependent kinases (Cdks), and the administration of roscovitine (a pan-Cdk inhibitor) attenuates renal cystic disease in juvenile cystic kidney (jck) mice. Cdk2 is a key regulator of cell proliferation, but its specific role in PKD remains unknown. The aim of this study was to test the hypothesis that Cdk2 deficiency reduces renal cyst growth in PKD. Three studies were undertaken: (i) a time course (days 28, 56, and 84) of cyclin and Cdk activity was examined in jck mice and compared with wild-type mice; (ii) the progression was compared in jck mice with or without Cdk2 ablation from birth; and (iii) the effect of sirolimus (an antiproliferative agent) on Cdk2 activity in jck mice was investigated. Renal disease in jck mice was characterized by diffuse tubular cyst growth, interstitial inflammation and fibrosis, and renal impairment, peaking on day 84. Renal cell proliferation peaked during earlier stages of disease (days 28-56), whereas the expression of Cdk2-cyclin partners (A and E) and Cdk1 and 2 activity, was maximal in the later stages of disease (days 56-84). Cdk2 ablation did not attenuate renal disease progression and was associated with persistent Cdk1 activity. In contrast, the postnatal treatment of jck mice with sirolimus reduced both Cdk2 and Cdk1 activity and reduced renal cyst growth. In conclusion, (i) the kinetics of Cdk2 and Cdk2-cyclin partners did not correlate with proliferation in jck mice; and (ii) the absence of Cdk2 did not alter renal cyst growth, most likely due to compensation by Cdk1. Taken together, these data suggest that Cdk2 is dispensable for the proliferation of cystic epithelial cells and progression of PKD.
Collapse
Affiliation(s)
- Jennifer Qin Jing Zhang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia. .,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia.
| | - Jane Burgess
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Daria Stepanova
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Sayanthooran Saravanabavan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Annette T Y Wong
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science Technology and Research), Singapore, 138673, Republic of Singapore.,Department of Biochemistry, National University of Singapore (NUS), Singapore, 117597, Republic of Singapore
| | - Gopala K Rangan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| |
Collapse
|
24
|
Zimmerman KA, Hopp K, Mrug M. Role of chemokines, innate and adaptive immunity. Cell Signal 2020; 73:109647. [PMID: 32325183 DOI: 10.1016/j.cellsig.2020.109647] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
Polycystic Kidney Disease (PKD) triggers a robust immune system response including changes in both innate and adaptive immunity. These changes involve immune cells (e.g., macrophages and T cells) as well as cytokines and chemokines (e.g., MCP-1) that regulate the production, differentiation, homing, and various functions of these cells. This review is focused on the role of the immune system and its associated factors in the pathogenesis of PKDs as evidenced by data from cell-based systems, animal models, and PKD patients. It also highlights relevant pre-clinical and clinical studies that point to specific immune system components as promising candidates for the development of prognostic biomarkers and therapeutic strategies to improve PKD outcomes.
Collapse
Affiliation(s)
- Kurt A Zimmerman
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Katharina Hopp
- Department of Medicine, Division of Renal Diseases and Hypertension, Polycystic Kidney Disease Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michal Mrug
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Veterans Affairs Medical Center, Birmingham, AL 35233, USA.
| |
Collapse
|
25
|
Tao F, Jiang T, Tao H, Cao H, Xiang W. Primary cilia: Versatile regulator in cartilage development. Cell Prolif 2020; 53:e12765. [PMID: 32034931 PMCID: PMC7106963 DOI: 10.1111/cpr.12765] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/21/2019] [Accepted: 12/29/2019] [Indexed: 02/07/2023] Open
Abstract
Cartilage is a connective tissue in the skeletal system and has limited regeneration ability and unique biomechanical reactivity. The growth and development of cartilage can be affected by different physical, chemical and biological factors, such as mechanical stress, inflammation, osmotic pressure, hypoxia and signalling transduction. Primary cilia are multifunctional sensory organelles that regulate diverse signalling transduction and cell activities. They are crucial for the regulation of cartilage development and act in a variety of ways, such as react to mechanical stress, mediate signalling transduction, regulate cartilage‐related diseases progression and affect cartilage tumorigenesis. Therefore, research on primary cilia‐mediated cartilage growth and development is currently extremely popular. This review outlines the role of primary cilia in cartilage development in recent years and elaborates on the potential regulatory mechanisms from different aspects.
Collapse
Affiliation(s)
- Fenghua Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ting Jiang
- Department of Neurological Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hai Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hui Cao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Wei Xiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Natoli TA, Modur V, Ibraghimov-Beskrovnaya O. Glycosphingolipid metabolism and polycystic kidney disease. Cell Signal 2020; 69:109526. [PMID: 31911181 DOI: 10.1016/j.cellsig.2020.109526] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/21/2022]
Abstract
Sphingolipids and glycosphingolipids are classes of structurally and functionally important lipids that regulate multiple cellular processes, including membrane organization, proliferation, cell cycle regulation, apoptosis, transport, migration, and inflammatory signalling pathways. Imbalances in sphingolipid levels or subcellular localization result in dysregulated cellular processes and lead to the development and progression of multiple disorders, including polycystic kidney disease. This review will describe metabolic pathways of glycosphingolipids with a focus on the evidence linking glycosphingolipid mediated regulation of cell signalling, lipid microdomains, cilia, and polycystic kidney disease. We will discuss molecular mechanisms of glycosphingolipid dysregulation and their impact on cystogenesis. We will further highlight how modulation of sphingolipid metabolism can be translated into new approaches for the treatment of polycystic kidney disease and describe current clinical studies with glucosylceramide synthase inhibitors in Autosomal Dominant Polycystic Kidney Disease.
Collapse
Affiliation(s)
- Thomas A Natoli
- Rare and Neurological Disease Research, Sanofi-Genzyme, 49 New York Ave., Framingham, MA 01701, USA
| | - Vijay Modur
- Rare Disease Development, Sanofi-Genzyme, 50 Binney St., Cambridge, MA 02142, USA
| | | |
Collapse
|
27
|
Testa F, Magistroni R. ADPKD current management and ongoing trials. J Nephrol 2019; 33:223-237. [PMID: 31853789 DOI: 10.1007/s40620-019-00679-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/24/2019] [Indexed: 12/14/2022]
Abstract
Among the diseases that require renal replacement therapy (RRT), ADPKD is the fourth for incidence and prevalence. In Italy, there are at least 32,000 patients affected by ADPKD, of which about 2900 in dialysis. The pure costs of dialysis treatment for the Italian National Health Service can be conservatively estimated at 87 million euros per year. Even a modest slowdown in the evolution of the disease would obtain an important result in terms of reduction of health expenditure. In recent years, many new or repurposed drugs have been evaluated in clinical trials for ADPKD. In this review we will mainly focus on advanced stage clinical trials (phase 2 and 3). We have grouped these studies according to the molecular pathway addressed by the experimental drug or the therapeutic strategy. More than 10 years after the start of the first Phase III clinical trials in ADPKD, the first drug active in slowing disease progression is finally available. It cannot be considered a goal but only the beginning of a journey because of the significant side effects and the high cost of Tolvaptan. An exuberant basic research activity in the field, together with the large number of ongoing protocols, keep the nephrologists and their patients positive with regard to the discovery of new and better therapies in a not-too-distant future.
Collapse
Affiliation(s)
- Francesca Testa
- UOC Divisione di Nefrologia Dialisi e Trapianto, AOU Policlinico di Modena, Modena, Italy
| | - Riccardo Magistroni
- UOC Divisione di Nefrologia Dialisi e Trapianto, AOU Policlinico di Modena, Modena, Italy. .,Dipartimento Chirurgico Medico Odontoiatrico e di Scienze Morfologiche con Interesse Trapiantologico, Oncologico e di Medicina Rigenerativa, Università di Modena e Reggio Emilia, Modena, Italy.
| |
Collapse
|
28
|
Ghazi S, Polesel M, Hall AM. Targeting glycolysis in proliferative kidney diseases. Am J Physiol Renal Physiol 2019; 317:F1531-F1535. [DOI: 10.1152/ajprenal.00460.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Glycolytic activity is increased in proliferating cells, leading to the concept that glycolysis could be a therapeutic target in cystic diseases and kidney cancer. Preclinical studies using the glucose analog 2-deoxy-d-glucose have shown promise; however, inhibiting glycolysis in humans is unlikely to be without risks. While proximal tubules are predominantly aerobic, later segments are more glycolytic. Understanding exactly where and why glycolysis is important in the physiology of the distal nephron is thus crucial in predicting potential adverse effects of glycolysis inhibitors. Live imaging techniques could play an important role in the process of characterizing cellular metabolism in the functioning kidney. The goal of this review is to briefly summarize recent findings on targeting glycolysis in proliferative kidney diseases and to highlight the necessity for future research focusing on glycolysis in the healthy kidney.
Collapse
Affiliation(s)
- Susan Ghazi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | - Andrew M. Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Margaria JP, Campa CC, De Santis MC, Hirsch E, Franco I. The PI3K/Akt/mTOR pathway in polycystic kidney disease: A complex interaction with polycystins and primary cilium. Cell Signal 2019; 66:109468. [PMID: 31715259 DOI: 10.1016/j.cellsig.2019.109468] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 12/19/2022]
Abstract
Over-activation of the PI3K/Akt/mTOR network is a well-known pathogenic event that leads to hyper-proliferation. Pharmacological targeting of this pathway has been developed for the treatment of multiple diseases, including cancer. In polycystic kidney disease (PKD), the mTOR cascade promotes cyst growth by boosting proliferation, size and metabolism of kidney tubule epithelial cells. Therefore, mTOR inhibition has been tested in pre-clinical and clinical studies, but only the former showed positive results. This review reports recent discoveries describing the activity and molecular mechanisms of mTOR activation in tubule epithelial cells and cyst formation and discusses the evidence of an upstream regulation of mTOR by the PI3K/Akt axis. In particular, the complex interconnections of the PI3K/Akt/mTOR network with the principal signaling routes involved in the suppression of cyst formation are dissected. These interactions include the antagonism and the reciprocal negative regulation between mTOR complex 1 and the proteins whose deletion causes Autosomal Dominant PKD, the polycystins. In addition, the emerging role of phopshoinositides, membrane components modulated by PI3K, will be presented in the context of primary cilium signaling, cell polarization and protection from cyst formation. Overall, studies demonstrate that the activity of various members of the PI3K/Akt/mTOR network goes beyond the classical transduction of mitogenic signals and can impact several aspects of kidney tubule homeostasis and morphogenesis. These properties might be useful to guide the establishment of more effective treatment protocols to be tested in clinical trials.
Collapse
Affiliation(s)
- Jean Piero Margaria
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Carlo Cosimo Campa
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Maria Chiara De Santis
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Emilio Hirsch
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Irene Franco
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, 14157 Huddinge, Sweden.
| |
Collapse
|
30
|
Kou P, Wei S, Xiong F. Recent Advances of mTOR Inhibitors Use in Autosomal Dominant Polycystic Kidney Disease: Is the Road Still Open? Curr Med Chem 2019; 26:2962-2973. [PMID: 29600752 DOI: 10.2174/0929867325666180330094434] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/15/2017] [Accepted: 03/21/2018] [Indexed: 12/25/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD), the most common monogenic kidney disease, is caused by mutations in the PKD1, PKD2 or, in a very limited number of families, GANAB genes. Although cellular and molecular mechanisms of this disease have been understood in the past 20 years, specific therapy approaches remain very little. Both experimental and clinical studies show that the mammalian or mechanistic target of rapamycin (mTOR) pathway plays an important role during cyst formation and enlargement in ADPKD. Studies in rodent models of ADPKD showed that mTOR inhibitors had a significant and long-lasting decrease in kidney volume and amelioration in kidney function. In the past over ten years, researchers have been devoting continuously to test mTOR inhibitors efficacy and safety in both preclinical studies and clinical trials in patients with ADPKD. In this review, we will discuss the mTOR pathway thoroughly, mainly focusing on current advances in understanding its role in ADPKD, especially the recent progress of mTOR inhibitors use in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Pei Kou
- Department of Nephrology, Wuhan No.1 Hospital, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- Department of Nephrology, Wuhan No.1 Hospital, Wuhan, China
| |
Collapse
|
31
|
Weydert C, Decuypere JP, De Smedt H, Janssens P, Vennekens R, Mekahli D. Fundamental insights into autosomal dominant polycystic kidney disease from human-based cell models. Pediatr Nephrol 2019; 34:1697-1715. [PMID: 30215095 DOI: 10.1007/s00467-018-4057-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/23/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022]
Abstract
Several animal- and human-derived models are used in autosomal dominant polycystic kidney disease (ADPKD) research to gain insight in the disease mechanism. However, a consistent correlation between animal and human ADPKD models is lacking. Therefore, established human-derived models are relevant to affirm research results and translate findings into a clinical set-up. In this review, we give an extensive overview of the existing human-based cell models. We discuss their source (urine, nephrectomy and stem cell), immortalisation procedures, genetic engineering, kidney segmental origin and characterisation with nephron segment markers. We summarise the most studied pathways and lessons learned from these different ADPKD models. Finally, we issue recommendations for the derivation of human-derived cell lines and for experimental set-ups with these cell lines.
Collapse
Affiliation(s)
- Caroline Weydert
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
| | - Jean-Paul Decuypere
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
| | - Humbert De Smedt
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Peter Janssens
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
- Department of Nephrology, University Hospitals Brussels, Brussels, Belgium
| | - Rudi Vennekens
- VIB Center for Brain and Disease Research, Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Djalila Mekahli
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium.
- Department of Pediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
32
|
Testa F, Marchiò M, Belli M, Giovanella S, Ligabue G, Cappelli G, Biagini G, Magistroni R. A pilot study to evaluate tolerability and safety of a modified Atkins diet in ADPKD patients. PHARMANUTRITION 2019. [DOI: 10.1016/j.phanu.2019.100154] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
33
|
Metabolism and mitochondria in polycystic kidney disease research and therapy. Nat Rev Nephrol 2019; 14:678-687. [PMID: 30120380 DOI: 10.1038/s41581-018-0051-1] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common, potentially lethal, monogenic diseases and is caused predominantly by mutations in polycystic kidney disease 1 (PKD1) and PKD2, which encode polycystin 1 (PC1) and PC2, respectively. Over the decades-long course of the disease, patients develop large fluid-filled renal cysts that impair kidney function, leading to end-stage renal disease in ~50% of patients. Despite the identification of numerous dysregulated pathways in ADPKD, the molecular mechanisms underlying the renal dysfunction from mutations in PKD genes and the physiological functions of the polycystin proteins are still unclear. Alterations in cell metabolism have emerged in the past decade as a hallmark of ADPKD. ADPKD cells shift their mode of energy production from oxidative phosphorylation to alternative pathways, such as glycolysis. In addition, the polycystins seem to play regulatory roles in modulating mechanisms and machinery related to energy production and utilization, including AMPK, PPARα, PGC1α, calcium signalling at mitochondria-associated membranes, mTORC1, cAMP and CFTR-mediated ion transport as well as the expression of crucial components of the mitochondrial energy production apparatus. In this Review, we explore these metabolic changes and discuss in detail the relationship between energy metabolism and ADPKD pathogenesis and identify potential therapeutic targets.
Collapse
|
34
|
Therapeutic Use of mTOR Inhibitors in Renal Diseases: Advances, Drawbacks, and Challenges. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3693625. [PMID: 30510618 PMCID: PMC6231362 DOI: 10.1155/2018/3693625] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 09/07/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
The mammalian (or mechanistic) target of rapamycin (mTOR) pathway has a key role in the regulation of a variety of biological processes pivotal for cellular life, aging, and death. Impaired activity of mTOR complexes (mTORC1/mTORC2), particularly mTORC1 overactivation, has been implicated in a plethora of age-related disorders, including human renal diseases. Since the discovery of rapamycin (or sirolimus), more than four decades ago, advances in our understanding of how mTOR participates in renal physiological and pathological mechanisms have grown exponentially, due to both preclinical studies in animal models with genetic modification of some mTOR components as well as due to evidence coming from the clinical experience. The main clinical indication of rapamycin is as immunosuppressive therapy for the prevention of allograft rejection, namely, in renal transplantation. However, considering the central participation of mTOR in the pathogenesis of other renal disorders, the use of rapamycin and its analogs meanwhile developed (rapalogues) everolimus and temsirolimus has been viewed as a promising pharmacological strategy. This article critically reviews the use of mTOR inhibitors in renal diseases. Firstly, we briefly overview the mTOR components and signaling as well as the pharmacological armamentarium targeting the mTOR pathway currently available or in the research and development stages. Thereafter, we revisit the mTOR pathway in renal physiology to conclude with the advances, drawbacks, and challenges regarding the use of mTOR inhibitors, in a translational perspective, in four classes of renal diseases: kidney transplantation, polycystic kidney diseases, renal carcinomas, and diabetic nephropathy.
Collapse
|
35
|
Nantavishit J, Chatsudthipong V, Soodvilai S. Lansoprazole reduces renal cyst in polycystic kidney disease via inhibition of cell proliferation and fluid secretion. Biochem Pharmacol 2018; 154:175-182. [DOI: 10.1016/j.bcp.2018.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/04/2018] [Indexed: 12/19/2022]
|
36
|
Jo HS, Eum WS, Park EY, Ko JY, Kim DY, Kim DW, Shin MJ, Son O, Cho SB, Park JH, Lee CH, Yeo EJ, Yeo HJ, Choi YJ, Youn JK, Cho SW, Park J, Park JH, Choi SY. Effects of PEP-1-FK506BP on cyst formation in polycystic kidney disease. BMB Rep 2018; 50:460-465. [PMID: 28760196 PMCID: PMC5625693 DOI: 10.5483/bmbrep.2017.50.9.090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Indexed: 01/07/2023] Open
Abstract
Polycystic kidney disease (PKD) is one of the most common inherited disorders, involving progressive cyst formation in the kidney that leads to renal failure. FK506 binding protein 12 (FK506BP) is an immunophilin protein that performs multiple functions, including regulation of cell signaling pathways and survival. In this study, we determined the roles of PEP-1-FK506BP on cell proliferation and cyst formation in PKD cells. Purified PEP-1-FK506BP transduced into PKD cells markedly inhibited cell proliferation. Also, PEP-1-FK506BP drastically inhibited the expression levels of p-Akt, p-p70S6K, p-mTOR, and p-ERK in PKD cells. In a 3D-culture system, PEP-1-FK506BP significantly reduced cyst formation. Furthermore, the combined effects of rapamycin and PEP-1-FK506BP on cyst formation were markedly higher than the effects of individual treatments. These results suggest that PEP-1-FK506BP delayed cyst formation and could be a new therapeutic strategy for renal cyst formation in PKD. [BMB Reports 2017; 50(9): 460-465].
Collapse
Affiliation(s)
- Hyo Sang Jo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Eun Young Park
- Department of Biological Science, Sookmyung Women's University, Seoul 04310, Korea
| | - Je Young Ko
- Department of Biological Science, Sookmyung Women's University, Seoul 04310, Korea
| | - Do Yeon Kim
- Department of Biological Science, Sookmyung Women's University, Seoul 04310, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Ora Son
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Su Bin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Chi Hern Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jong Kyu Youn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jong Hoon Park
- Department of Biological Science, Sookmyung Women's University, Seoul 04310, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
37
|
Billot K, Coquil C, Villiers B, Josselin-Foll B, Desban N, Delehouzé C, Oumata N, Le Meur Y, Boletta A, Weimbs T, Grosch M, Witzgall R, Saunier S, Fischer E, Pontoglio M, Fautrel A, Mrug M, Wallace D, Tran PV, Trudel M, Bukanov N, Ibraghimov-Beskrovnaya O, Meijer L. Casein kinase 1ε and 1α as novel players in polycystic kidney disease and mechanistic targets for (R)-roscovitine and (S)-CR8. Am J Physiol Renal Physiol 2018. [PMID: 29537311 DOI: 10.1152/ajprenal.00489.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Following the discovery of (R)-roscovitine's beneficial effects in three polycystic kidney disease (PKD) mouse models, cyclin-dependent kinases (CDKs) inhibitors have been investigated as potential treatments. We have used various affinity chromatography approaches to identify the molecular targets of roscovitine and its more potent analog (S)-CR8 in human and murine polycystic kidneys. These methods revealed casein kinases 1 (CK1) as additional targets of the two drugs. CK1ε expression at the mRNA and protein levels is enhanced in polycystic kidneys of 11 different PKD mouse models as well as in human polycystic kidneys. A shift in the pattern of CK1α isoforms is observed in all PKD mouse models. Furthermore, the catalytic activities of both CK1ε and CK1α are increased in mouse polycystic kidneys. Inhibition of CK1ε and CK1α may thus contribute to the long-lasting attenuating effects of roscovitine and (S)-CR8 on cyst development. CDKs and CK1s may constitute a dual therapeutic target to develop kinase inhibitory PKD drug candidates.
Collapse
Affiliation(s)
- Katy Billot
- ManRos Therapeutics, Centre de Perharidy , Roscoff , France
| | | | | | - Béatrice Josselin-Foll
- CNRS "Protein Phosphorylation and Human Disease Group, Station Biologique, Roscoff Cedex, Bretagne , France
| | - Nathalie Desban
- CNRS "Protein Phosphorylation and Human Disease Group, Station Biologique, Roscoff Cedex, Bretagne , France
| | - Claire Delehouzé
- CNRS "Protein Phosphorylation and Human Disease Group, Station Biologique, Roscoff Cedex, Bretagne , France
| | - Nassima Oumata
- ManRos Therapeutics, Centre de Perharidy , Roscoff , France
| | - Yannick Le Meur
- Service de Néphrologie, Centre Hospitalier Universitaire La Cavale Blanche, Rue Tanguy Prigent, Brest Cedex, France
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, DIBIT San Raffaele Scientific Institute , Milan , Italy
| | - Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology, Neuroscience Research Institute, University of California Santa Barbara , Santa Barbara, California
| | - Melanie Grosch
- University of Regensburg, Institute for Molecular and Cellular Anatomy, Universitätsstr 31, Regensburg , Germany
| | - Ralph Witzgall
- University of Regensburg, Institute for Molecular and Cellular Anatomy, Universitätsstr 31, Regensburg , Germany
| | | | - Evelyne Fischer
- "Expression Génique, Développement et Maladies", Equipe 26/INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Institut Cochin, Département Génétique & Développement, Paris , France
| | - Marco Pontoglio
- "Expression Génique, Développement et Maladies", Equipe 26/INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Institut Cochin, Département Génétique & Développement, Paris , France
| | - Alain Fautrel
- Université de Rennes 1, H2P2 Histopathology Core Facility, Rennes Cedex, France
| | - Michal Mrug
- Division of Nephrology, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Veterans Affairs Medical Center , Birmingham, Alabama
| | - Darren Wallace
- University of Kansas Medical Center, The Jared Grantham Kidney Institute , Kansas City, Kansas
| | - Pamela V Tran
- University of Kansas Medical Center, The Jared Grantham Kidney Institute , Kansas City, Kansas.,University of Kansas Medical Center, Department of Anatomy and Cell Biology , Kansas City, Kansas
| | - Marie Trudel
- Institut de Recherches Cliniques de Montréal, Molecular Genetics and Development, Montreal, Quebec , Canada
| | - Nikolay Bukanov
- Sanofi Genzyme, Rare Renal and Bone Diseases, Framingham, Massachusetts
| | | | - Laurent Meijer
- ManRos Therapeutics, Centre de Perharidy , Roscoff , France
| |
Collapse
|
38
|
Skalická K, Hrčková G, Vaská A, Baranyaiová Á, Kovács L. Genetic defects in ciliary genes in autosomal dominant polycystic kidney disease. World J Nephrol 2018; 7:65-70. [PMID: 29527510 PMCID: PMC5838416 DOI: 10.5527/wjn.v7.i2.65] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 12/31/2017] [Accepted: 02/05/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the genetic defects of ciliary genes causing the loss of primary cilium in autosomal dominant polycystic kidney disease (ADPKD).
METHODS We analyzed 191 structural and functional genes of the primary cilium using next-generation sequencing analysis. We analyzed the kidney samples, which were obtained from 7 patients with ADPKD who underwent nephrectomy. Each sample contained polycystic kidney tissue and matched normal kidney tissue.
RESULTS In our study, we identified genetic defects in the 5 to 15 genes in each ADPKD sample. The most frequently identified defects were found in genes encoding centrosomal proteins (PCM1, ODF2, HTT and CEP89) and kinesin family member 19 (KIF19), which are important for ciliogenesis. In addition, pathogenic mutations in the PCM1 and KIF19 genes were found in all ADPKD samples. Interestingly, mutations in the genes encoding the intraflagellar transport proteins, which are the basis of animal models of ADPKD, were only rarely detected.
CONCLUSION The results of our study revealed the actual state of structural ciliary genes in human ADPKD tissues and provided valuable indications for further research.
Collapse
Affiliation(s)
- Katarína Skalická
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| | - Gabriela Hrčková
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| | - Anita Vaská
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| | - Ágnes Baranyaiová
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| | - László Kovács
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| |
Collapse
|
39
|
Li J, Lu D, Liu H, Williams BO, Overbeek PA, Lee B, Zheng L, Yang T. Sclt1 deficiency causes cystic kidney by activating ERK and STAT3 signaling. Hum Mol Genet 2018; 26:2949-2960. [PMID: 28486600 DOI: 10.1093/hmg/ddx183] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 05/03/2017] [Indexed: 12/31/2022] Open
Abstract
Ciliopathies form a group of inherited disorders sharing several clinical manifestations because of abnormal cilia formation or function, and few treatments have been successful against these disorders. Here, we report a mouse model with mutated Sclt1 gene, which encodes a centriole distal appendage protein important for ciliogenesis. Sodium channel and clathrin linker 1 (SCLT1) mutations were associated with the oral-facial-digital syndrome (OFD), an autosomal recessive ciliopathy. The Sclt1-/- mice exhibit typical ciliopathy phenotypes, including cystic kidney, cleft palate and polydactyly. Sclt1-loss decreases the number of cilia in kidney; increases proliferation and apoptosis of renal tubule epithelial cells; elevates protein kinase A, extracellular signal-regulated kinases, SMAD and signal transducer and activator of transcription 3 (STAT3) pathways; and enhances pro-inflammation and pro-fibrosis pathways with disease progression. Embryonic kidney cyst formation of Sclt1-/- mice was effectively reduced by an anti-STAT3 treatment using pyrimethamine. Overall, we reported a new mouse model for the OFD; and our data suggest that STAT3 inhibition may be a promising treatment for SCLT1-associated cystic kidney.
Collapse
Affiliation(s)
- Jianshuang Li
- Hubei Key Laboratory of Cell Homeostasis, Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China.,Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Di Lu
- Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Huadie Liu
- Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Bart O Williams
- Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | | | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Tao Yang
- Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
40
|
Takahashi K, Nagai T, Chiba S, Nakayama K, Mizuno K. Glucose deprivation induces primary cilium formation through mTORC1 inactivation. J Cell Sci 2018; 131:jcs.208769. [PMID: 29180513 DOI: 10.1242/jcs.208769] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/20/2017] [Indexed: 01/14/2023] Open
Abstract
Primary cilia are antenna-like sensory organelles extending from the surface of many cell types that play critical roles in tissue development and homeostasis. Here, we examined the effect of nutrient status on primary cilium formation. Glucose deprivation significantly increased the number of ciliated cells under both serum-fed and -starved conditions. Glucose deprivation-induced ciliogenesis was suppressed by overexpression of Rheb, an activator of the mammalian target of rapamycin complex-1 (mTORC1). Inactivating mTORC1 by rapamycin treatment or Raptor knockdown significantly promoted ciliogenesis. These results indicate that glucose deprivation promotes primary cilium formation through mTORC1 inactivation. Rapamycin treatment did not promote autophagy or degradation of OFD1, a negative regulator of ciliogenesis. In contrast, rapamycin treatment increased the level of the p27KIP1 (also known as CDKN1B) cyclin-dependent kinase inhibitor, and rapamycin-induced ciliogenesis was abrogated in p27KIP1-depleted cells. These results indicate that mTORC1 inactivation induces ciliogenesis through p27KIP1 upregulation, but not through autophagy. By contrast, glucose deprivation or rapamycin treatment shortened the cilium length. Thus, glucose deprivation and subsequent inactivation of mTORC1 play dual roles in ciliogenesis: triggering primary cilium formation and shortening cilium length.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Kengo Takahashi
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Tomoaki Nagai
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Shuhei Chiba
- Department of Genetic Disease Research, Osaka City University, Graduate School of Medicine, Osaka 545-8585, Japan
| | - Keiko Nakayama
- Department of Cell Proliferation, United Center for Advanced Research and Translational Medicine, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Kensaku Mizuno
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
41
|
Nowak KL, You Z, Gitomer B, Brosnahan G, Torres VE, Chapman AB, Perrone RD, Steinman TI, Abebe KZ, Rahbari-Oskoui FF, Yu ASL, Harris PC, Bae KT, Hogan M, Miskulin D, Chonchol M. Overweight and Obesity Are Predictors of Progression in Early Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2017; 29:571-578. [PMID: 29118087 DOI: 10.1681/asn.2017070819] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/08/2017] [Indexed: 01/12/2023] Open
Abstract
The association of overweight/obesity with disease progression in patients with autosomal dominant polycystic kidney disease (ADPKD) remains untested. We hypothesized that overweight/obesity associates with faster progression in early-stage ADPKD. Overall, 441 nondiabetic participants with ADPKD and an eGFR>60 ml/min per 1.73 m2 who participated in the Halt Progression of Polycystic Kidney Disease Study A were categorized on the basis of body mass index (BMI; calculated using nonkidney and nonliver weight) as normal weight (18.5-24.9 kg/m2; reference; n=192), overweight (25.0-29.9 kg/m2; n=168), or obese (≥30 kg/m2; n=81). We evaluated the longitudinal (5-year) association of overweight/obesity with change in total kidney volume (TKV) by magnetic resonance imaging using linear regression and multinomial logistic regression models. Among participants, mean±SD age was 37±8 years, annual percent change in TKV was 7.4%±5.1%, and BMI was 26.3±4.9 kg/m2 The annual percent change in TKV increased with increasing BMI category (normal weight: 6.1%±4.7%, overweight: 7.9%±4.8%, obese: 9.4%±6.2%; P<0.001). In the fully adjusted model, higher BMI associated with greater annual percent change in TKV (β=0.79; 95% confidence interval [95% CI], 0.18 to 1.39, per 5-unit increase in BMI). Overweight and obesity associated with increased odds of annual percent change in TKV ≥7% compared with <5% (overweight: odds ratio, 2.02; 95% CI, 1.15 to 3.56; obese: odds ratio, 3.76; 95% CI, 1.81 to 7.80). Obesity also independently associated with greater eGFR decline (slope) versus normal weight (fully adjusted β =-0.08; 95% CI, -0.15 to -0.02). In conclusion, overweight and, particularly, obesity are strongly and independently associated with rate of progression in early-stage ADPKD.
Collapse
Affiliation(s)
- Kristen L Nowak
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado;
| | - Zhiying You
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Berenice Gitomer
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Godela Brosnahan
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | | - Ronald D Perrone
- Division of Nephrology, Tufts University Medical Center, Boston, Massachusetts
| | - Theodore I Steinman
- Department of Medicine and Renal Division, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Kaleab Z Abebe
- Center for Clinical Trials & Data Coordination, Division of General Internal Medicine, and
| | | | - Alan S L Yu
- Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Kyongtae T Bae
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marie Hogan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Dana Miskulin
- Division of Nephrology, Tufts University Medical Center, Boston, Massachusetts
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
42
|
Primary Cilium-Dependent Signaling Mechanisms. Int J Mol Sci 2017; 18:ijms18112272. [PMID: 29143784 PMCID: PMC5713242 DOI: 10.3390/ijms18112272] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/13/2017] [Accepted: 10/25/2017] [Indexed: 01/02/2023] Open
Abstract
Primary cilia are hair-like organelles and play crucial roles in vertebrate development, organogenesis, health, and many genetic disorders. A primary cilium is a mechano-sensory organelle that responds to mechanical stimuli in the micro-environment. A cilium is also a chemosensor that senses chemical signals surrounding a cell. The overall function of a cilium is therefore to act as a communication hub to transfer extracellular signals into intracellular responses. Although intracellular calcium has been one of the most studied signaling messengers that transmit extracellular signals into the cells, calcium signaling by various ion channels remains a topic of interest in the field. This may be due to a broad spectrum of cilia functions that are dependent on or independent of utilizing calcium as a second messenger. We therefore revisit and discuss the calcium-dependent and calcium-independent ciliary signaling pathways of Hedgehog, Wnt, PDGFR, Notch, TGF-β, mTOR, OFD1 autophagy, and other GPCR-associated signaling. All of these signaling pathways play crucial roles in various cellular processes, such as in organ and embryonic development, cardiac functioning, planar cell polarity, transactivation, differentiation, the cell cycle, apoptosis, tissue homeostasis, and the immune response.
Collapse
|
43
|
Fitzgibbon WR, Dang Y, Bunni MA, Baicu CF, Zile MR, Mullick AE, Saigusa T. Attenuation of accelerated renal cystogenesis in Pkd1 mice by renin-angiotensin system blockade. Am J Physiol Renal Physiol 2017; 314:F210-F218. [PMID: 29021226 DOI: 10.1152/ajprenal.00389.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The intrarenal renin angiotensin system (RAS) is activated in polycystic kidney disease. We have recently shown in the Pkd1 mouse that Gen 2 antisense oligonucleotide (ASO), which suppresses angiotensinogen (Agt) synthesis, is efficacious in slowing kidney cyst formation compared with lisinopril. The aim of this current study was to determine 1) if unilateral nephrectomy accelerates cystogenesis in Pkd1 mice (as previously shown in cilia knockout mice) and 2) whether Agt ASO can slow the progression in this accelerated cystic mouse model. Adult Pkd1 conditional floxed allele mice expressing cre were administered tamoxifen, resulting in global knockout of Pkd1. Three weeks after tamoxifen injection, mice underwent left unilateral nephrectomy. Mice were then treated with Agt ASO (75 mg/kg per week) or aliskiren (20 mg/kg per day)+Agt ASO or control for 8 wk. Unilateral nephrectomy accelerated kidney cyst formation compared with nonnephrectomized mice. Both Agt ASO and Aliskiren+Agt ASO treatments significantly reduced plasma and urinary Agt levels. Blood pressure was lowest in Aliskiren+Agt ASO mice among all treatment groups, and the control group had the highest blood pressure. All mice developed significant kidney cysts at 8 wk after nephrectomy, but Agt ASO and Aliskiren+Agt ASO groups had fewer kidney cysts than controls. Renal pAkt, pS6 levels, and apoptosis were significantly suppressed in those receiving Agt ASO compared with controls. These results indicate that suppressing Agt using an ASO slowed the progression of accelerated cystic kidney disease induced by unilateral nephrectomy in Pkd1 mice by suppressing intrarenal RAS, mammalian target of rapamycin pathway, and cell proliferation.
Collapse
Affiliation(s)
- Wayne R Fitzgibbon
- Divison of Nephrology, Medical University of South Carolina , Charleston, South Carolina
| | - Yujing Dang
- Divison of Nephrology, Medical University of South Carolina , Charleston, South Carolina
| | - Marlene A Bunni
- Divison of Nephrology, Medical University of South Carolina , Charleston, South Carolina
| | - Catalin F Baicu
- Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center , Charleston, South Carolina
| | - Michael R Zile
- Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center , Charleston, South Carolina
| | | | - Takamitsu Saigusa
- Divison of Nephrology, Medical University of South Carolina , Charleston, South Carolina.,Division of Nephrology, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
44
|
Walz G. Role of primary cilia in non-dividing and post-mitotic cells. Cell Tissue Res 2017; 369:11-25. [PMID: 28361305 PMCID: PMC5487853 DOI: 10.1007/s00441-017-2599-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/20/2017] [Accepted: 02/27/2017] [Indexed: 12/12/2022]
Abstract
The essential role of primary (non-motile) cilia during the development of multi-cellular tissues and organs is well established and is underlined by severe disease manifestations caused by mutations in cilia-associated molecules that are collectively termed ciliopathies. However, the role of primary cilia in non-dividing and terminally differentiated, post-mitotic cells is less well understood. Although the prevention of cells from re-entering the cell cycle may represent a major chore, primary cilia have recently been linked to DNA damage responses, autophagy and mitochondria. Given this connectivity, primary cilia in non-dividing cells are well positioned to form a signaling hub outside of the nucleus. Such a center could integrate information to initiate responses and to maintain cellular homeostasis if cell survival is jeopardized. These more discrete functions may remain undetected until differentiated cells are confronted with emergencies.
Collapse
Affiliation(s)
- Gerd Walz
- Renal Division, Department of Medicine, University Freiburg Medical Center, Hugstetter Strasse 55, 79106, Freiburg, Germany.
| |
Collapse
|
45
|
Abstract
The primary cilium is a microtubule-based organelle that is considered to be a cellular antennae, because proteins related to multiple signaling pathways such as Wnt, PDGFRα, Hh, and mechanosignaling are localized to the membrane of the primary cilium. In the kidney, primary cilia extend from the cell membrane to the lumen of renal tubules to respond to fluidic stress. Recent studies have indicated that the disruption of ciliary proteins including polycystin-1 (PC1), polycystin-2 (PC2), and members of the intraflagellar transport (IFT) family induce the development of polycystic kidney disease (PKD), suggesting that the malformation or absence of primary cilia is a driving force of the onset of PKD. Therefore, in this chapter, the renal cystogenesis mechanism induced by cilia defects and pathogenic ciliary proteins associated with PKD development will be described.
Collapse
Affiliation(s)
- Je Yeong Ko
- Molecular Medicine Laboratory, Department of Life systems, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-gu, Seoul, 04310, South Korea.
| |
Collapse
|
46
|
Michgehl U, Pavenstädt H, Vollenbröker B. Cross talk between the Crumbs complex and Hippo signaling in renal epithelial cells. Pflugers Arch 2017; 469:917-926. [PMID: 28612137 DOI: 10.1007/s00424-017-2004-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
Cell polarity has a crucial role in organizing cells into tissues and in mediating transport processes and cell-cell communication. Especially the cells of the nephron require apicobasal polarity to establish and maintain their barrier function. The Crumbs complex including the integral membrane protein Crumbs, as well as Pals1 and Patj, is essential for the establishment of apicobasal polarity. The interactions of the core proteins and the interplay with other processes have been characterized in various epithelial cell lines in detail. Notably, Crb2 and Crb3 are expressed within the kidney and play an important role in the proper function of podocytes and tubules, respectively. The interaction of polarity proteins and components of the Hippo pathway-an evolutionarily highly conserved kinase cascade regulating cell proliferation, organ size, and tissue regeneration-has been discovered recently. Here, we discuss potential molecular and physiological links between the Crumbs complex and the Hippo pathway in renal cells.
Collapse
Affiliation(s)
- U Michgehl
- Internal Medicine D, University Hospital of Muenster, Albert-Schweitzer-Campus 1, A14, D-48149, Muenster, Germany.
| | - H Pavenstädt
- Internal Medicine D, University Hospital of Muenster, Albert-Schweitzer-Campus 1, A14, D-48149, Muenster, Germany
| | - B Vollenbröker
- Internal Medicine D, University Hospital of Muenster, Albert-Schweitzer-Campus 1, A14, D-48149, Muenster, Germany
| |
Collapse
|
47
|
Gamberi C, Hipfner DR, Trudel M, Lubell WD. Bicaudal C mutation causes myc and TOR pathway up-regulation and polycystic kidney disease-like phenotypes in Drosophila. PLoS Genet 2017; 13:e1006694. [PMID: 28406902 PMCID: PMC5390980 DOI: 10.1371/journal.pgen.1006694] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 03/15/2017] [Indexed: 12/26/2022] Open
Abstract
Progressive cystic kidney degeneration underlies diverse renal diseases, including the most common cause of kidney failure, autosomal dominant Polycystic Kidney Disease (PKD). Genetic analyses of patients and animal models have identified several key drivers of this disease. The precise molecular and cellular changes underlying cystogenesis remain, however, elusive. Drosophila mutants lacking the translational regulator Bicaudal C (BicC, the fly ortholog of vertebrate BICC1 implicated in renal cystogenesis) exhibited progressive cystic degeneration of the renal tubules (so called “Malpighian” tubules) and reduced renal function. The BicC protein was shown to bind to Drosophila (d-) myc mRNA in tubules. Elevation of d-Myc protein levels was a cause of tubular degeneration in BicC mutants. Activation of the Target of Rapamycin (TOR) kinase pathway, another common feature of PKD, was found in BicC mutant flies. Rapamycin administration substantially reduced the cystic phenotype in flies. We present new mechanistic insight on BicC function and propose that Drosophila may serve as a genetically tractable model for dissecting the evolutionarily-conserved molecular mechanisms of renal cystogenesis. Polycystic kidney disease (PKD) is a degenerative, potentially lethal, genetic malady that affects 12.5 million people world-wide for which there is no cure. In the kidney, PKD causes the formation of prominent, fluid-filled cysts the growth of which damages progressively kidney function. Crucial to PKD development, mutations in the PKD1 and PKD2 genes cause renal cystic degeneration via factors and mechanisms that are only partially known. This manuscript reports novel insights into the molecular mechanisms of the evolutionarily conserved RNA binding protein BicC, which has been implicated in vertebrate cystic kidney diseases. The BicC mutants of the fruit fly Drosophila melanogaster recapitulate crucial characteristics of PKD. A clear link between BicC and PKD has begun to emerge, in part because both PKD1 patients and Pkd1 mice exhibit reduced BicC function. This first in kind Drosophila model of renal cystogenesis offers strong potential to decipher the complex mechanisms of the molecular and cellular changes causing renal cyst formation.
Collapse
Affiliation(s)
- Chiara Gamberi
- Department of Biology, Concordia University, Montréal, QC, Canada
- * E-mail:
| | - David R. Hipfner
- Institut de recherches cliniques de Montréal, 110 Pine Avenue West, Montréal, QC, Canada
- Département de médecine, Université de Montréal, Montréal, QC, Canada
| | - Marie Trudel
- Institut de recherches cliniques de Montréal, 110 Pine Avenue West, Montréal, QC, Canada
- Département de médecine, Université de Montréal, Montréal, QC, Canada
| | | |
Collapse
|
48
|
Cordido A, Besada-Cerecedo L, García-González MA. The Genetic and Cellular Basis of Autosomal Dominant Polycystic Kidney Disease-A Primer for Clinicians. Front Pediatr 2017; 5:279. [PMID: 29326913 PMCID: PMC5741702 DOI: 10.3389/fped.2017.00279] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/07/2017] [Indexed: 12/14/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common genetic disorders worldwide. In recent decades, the field has undergone a revolution, starting with the identification of causal ADPKD genes, including PKD1, PKD2, and the recently identified GANAB. In addition, advances defining the genetic mechanisms, protein localization and function, and the identification of numerous pathways involved in the disease process, have contributed to a better understanding of this illness. Together, this has led to a better prognosis, diagnosis, and treatment in clinical practice. In this mini review, we summarize and discuss new insights about the molecular mechanisms underlying ADPKD, including its genetics, protein function, and cellular pathways.
Collapse
Affiliation(s)
- Adrián Cordido
- Grupo de Genética y Biología del Desarrollo de las Enfermedades Renales, Laboratorio de Nefrología (n.° 11), Instituto de Investigación Sanitaria (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Lara Besada-Cerecedo
- Grupo de Genética y Biología del Desarrollo de las Enfermedades Renales, Laboratorio de Nefrología (n.° 11), Instituto de Investigación Sanitaria (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Miguel A García-González
- Grupo de Genética y Biología del Desarrollo de las Enfermedades Renales, Laboratorio de Nefrología (n.° 11), Instituto de Investigación Sanitaria (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| |
Collapse
|
49
|
Abstract
Primary cilia are small, antenna-like structures that detect mechanical and chemical cues and transduce extracellular signals. While mammalian primary cilia were first reported in the late 1800s, scientific interest in these sensory organelles has burgeoned since the beginning of the twenty-first century with recognition that primary cilia are essential to human health. Among the most common clinical manifestations of ciliary dysfunction are renal cysts. The molecular mechanisms underlying renal cystogenesis are complex, involving multiple aberrant cellular processes and signaling pathways, while initiating molecular events remain undefined. Autosomal Dominant Polycystic Kidney Disease is the most common renal cystic disease, caused by disruption of polycystin-1 and polycystin-2 transmembrane proteins, which evidence suggests must localize to primary cilia for proper function. To understand how the absence of these proteins in primary cilia may be remediated, we review intracellular trafficking of polycystins to the primary cilium. We also examine the controversial mechanisms by which primary cilia transduce flow-mediated mechanical stress into intracellular calcium. Further, to better understand ciliary function in the kidney, we highlight the LKB1/AMPK, Wnt, and Hedgehog developmental signaling pathways mediated by primary cilia and misregulated in renal cystic disease.
Collapse
|
50
|
Ta MHT, Schwensen KG, Foster S, Korgaonkar M, Ozimek-Kulik JE, Phillips JK, Peduto A, Rangan GK. Effects of TORC1 Inhibition during the Early and Established Phases of Polycystic Kidney Disease. PLoS One 2016; 11:e0164193. [PMID: 27723777 PMCID: PMC5056751 DOI: 10.1371/journal.pone.0164193] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/21/2016] [Indexed: 01/01/2023] Open
Abstract
The disease-modifying effects of target of rapamycin complex 1 (TORC1) inhibitors during different stages of polycystic kidney disease (PKD) are not well defined. In this study, male Lewis Polycystic Kidney Disease (LPK) rats (a genetic ortholog of human NPHP9, phenotypically characterised by diffuse distal nephron cystic growth) and Lewis controls received either vehicle (V) or sirolimus (S, 0.2 mg/kg by intraperitoneal injection 5 days per week) during the early (postnatal weeks 3 to 10) or late stages of disease (weeks 10 to 20). In early-stage disease, sirolimus reduced kidney enlargement (by 63%), slowed the rate of increase in total kidney volume (TKV) in serial MRI by 78.2% (LPK+V: 132.3±59.7 vs. LPK+S: 28.8±12.0% per week) but only partly reduced the percentage renal cyst area (by 19%) and did not affect the decline in endogenous creatinine clearance (CrCl) in LPK rats. In late-stage disease, sirolimus reduced kidney enlargement (by 22%) and the rate of increase in TKV by 71.8% (LPK+V: 13.1±6.6 vs. LPK+S: 3.7±3.7% per week) but the percentage renal cyst area was unaltered, and the CrCl only marginally better. Sirolimus reduced renal TORC1 activation but not TORC2, NF-κB DNA binding activity, CCL2 or TNFα expression, and abnormalities in cilia ultrastructure, hypertension and cardiac disease were also not improved. Thus, the relative treatment efficacy of TORC1 inhibition on kidney enlargement was consistent at all disease stages, but the absolute effect was determined by the timing of drug initiation. Furthermore, cystic microarchitecture, renal function and cardiac disease remain abnormal with TORC1 inhibition, indicating that additional approaches to normalise cellular dedifferentiation, inflammation and hypertension are required to completely arrest the progression of PKDs.
Collapse
Affiliation(s)
- Michelle H. T. Ta
- Michael Stern Translational Laboratory for Polycystic Kidney Disease, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Kristina G. Schwensen
- Michael Stern Translational Laboratory for Polycystic Kidney Disease, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Sheryl Foster
- Department of Radiology, University of Sydney at Westmead Hospital, Sydney, Australia
- Faculty of Health Sciences, University of Sydney, Sydney, Australia
| | - Mayuresh Korgaonkar
- Brain Dynamics Centre, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Justyna E. Ozimek-Kulik
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Jacqueline K. Phillips
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Anthony Peduto
- Department of Radiology, University of Sydney at Westmead Hospital, Sydney, Australia
| | - Gopala K. Rangan
- Michael Stern Translational Laboratory for Polycystic Kidney Disease, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
- Department of Renal Medicine, Westmead Hospital, Western Sydney Local Heath District, Westmead, Sydney, Australia
- * E-mail:
| |
Collapse
|