1
|
Kędzierska-Kapuza K, Łopuszyńska I, Niewiński G, Franek E, Szczuko M. The Influence of Non-Pharmacological and Pharmacological Interventions on the Course of Autosomal Dominant Polycystic Kidney Disease. Nutrients 2024; 16:3216. [PMID: 39339816 PMCID: PMC11434835 DOI: 10.3390/nu16183216] [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: 08/18/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Polycystic kidney disease (PKD) includes autosomal dominant (ADPKD) and autosomal recessive (ARPKD) forms, both of which are primary genetic causes of kidney disease in adults and children. ADPKD is the most common hereditary kidney disease, with a prevalence of 329 cases per million in Europe. This condition accounts for 5-15% of end-stage chronic kidney disease (ESKD) cases, and in developed countries such as Poland, 8-10% of all dialysis patients have ESKD due to ADPKD. The disease is caused by mutations in the PKD1 and PKD2 genes, with PKD1 mutations responsible for 85% of cases, leading to a more aggressive disease course. Recent research suggests that ADPKD involves a metabolic defect contributing to cystic epithelial proliferation and cyst growth. Aim: This review explores the interplay between metabolism, obesity, and ADPKD, discussing dietary and pharmacological strategies that target these metabolic abnormalities to slow disease progression. Conclusion: Metabolic reprogramming therapies, including GLP-1 analogs and dual agonists of GIP/GLP-1 or glucagon/GLP-1 receptors, show promise, though further research is needed to understand their potential in ADPKD treatment fully.
Collapse
Affiliation(s)
- Karolina Kędzierska-Kapuza
- Department of Internal Diseases, Endocrinology and Diabetology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Inga Łopuszyńska
- Department of Gastroenterological Surgery and Transplantology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Grzegorz Niewiński
- Department of Gastroenterological Surgery and Transplantology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Edward Franek
- Department of Internal Diseases, Endocrinology and Diabetology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Małgorzata Szczuko
- Department of Human Nutrition and Metabolomic, Pomeranian Medical University, 24 W. Broniewskiego St., 71-460 Szczecin, Poland
| |
Collapse
|
2
|
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
|
3
|
Devlin L, Dhondurao Sudhindar P, Sayer JA. Renal ciliopathies: promising drug targets and prospects for clinical trials. Expert Opin Ther Targets 2023; 27:325-346. [PMID: 37243567 DOI: 10.1080/14728222.2023.2218616] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/12/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Renal ciliopathies represent a collection of genetic disorders characterized by deficiencies in the biogenesis, maintenance, or functioning of the ciliary complex. These disorders, which encompass autosomal dominant polycystic kidney disease (ADPKD), autosomal recessive polycystic kidney disease (ARPKD), and nephronophthisis (NPHP), typically result in cystic kidney disease, renal fibrosis, and a gradual deterioration of kidney function, culminating in kidney failure. AREAS COVERED Here we review the advances in basic science and clinical research into renal ciliopathies which have yielded promising small compounds and drug targets, within both preclinical studies and clinical trials. EXPERT OPINION Tolvaptan is currently the sole approved treatment option available for ADPKD patients, while no approved treatment alternatives exist for ARPKD or NPHP patients. Clinical trials are presently underway to evaluate additional medications in ADPKD and ARPKD patients. Based on preclinical models, other potential therapeutic targets for ADPKD, ARPKD, and NPHP look promising. These include molecules targeting fluid transport, cellular metabolism, ciliary signaling and cell-cycle regulation. There is a real and urgent clinical need for translational research to bring novel treatments to clinical use for all forms of renal ciliopathies to reduce kidney disease progression and prevent kidney failure.
Collapse
Affiliation(s)
- Laura Devlin
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Praveen Dhondurao Sudhindar
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Renal Services, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle Upon Tyne, UK
| |
Collapse
|
4
|
Reiterová J, Tesař V. Autosomal Dominant Polycystic Kidney Disease: From Pathophysiology of Cystogenesis to Advances in the Treatment. Int J Mol Sci 2022; 23:ijms23063317. [PMID: 35328738 PMCID: PMC8949594 DOI: 10.3390/ijms23063317] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/28/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic renal disease, with an estimated prevalence between 1:1000 and 1:2500. It is mostly caused by mutations of the PKD1 and PKD2 genes encoding polycystin 1 (PC1) and polycystin 2 (PC2) that regulate cellular processes such as fluid transport, differentiation, proliferation, apoptosis and cell adhesion. Reduction of calcium ions and induction of cyclic adenosine monophosphate (sAMP) promote cyst enlargement by transepithelial fluid secretion and cell proliferation. Abnormal activation of MAPK/ERK pathway, dysregulated signaling of heterotrimeric G proteins, mTOR, phosphoinositide 3-kinase, AMPK, JAK/STAT activator of transcription and nuclear factor kB (NF-kB) are involved in cystogenesis. Another feature of cystic tissue is increased extracellular production and recruitment of inflammatory cells and abnormal connections among cells. Moreover, metabolic alterations in cystic cells including defective glucose metabolism, impaired beta-oxidation and abnormal mitochondrial activity were shown to be associated with cyst expansion. Although tolvaptan has been recently approved as a drug that slows ADPKD progression, some patients do not tolerate tolvaptan because of frequent aquaretic. The advances in the knowledge of multiple molecular pathways involved in cystogenesis led to the development of animal and cellular studies, followed by the development of several ongoing randomized controlled trials with promising drugs. Our review is aimed at pathophysiological mechanisms in cystogenesis in connection with the most promising drugs in animal and clinical studies.
Collapse
Affiliation(s)
- Jana Reiterová
- Department of Nephrology, First Faculty of Medicine, Charles University, General University Hospital in Prague, 128 08 Prague, Czech Republic;
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, General University Hospital in Prague, 128 08 Prague, Czech Republic
| | - Vladimír Tesař
- Department of Nephrology, First Faculty of Medicine, Charles University, General University Hospital in Prague, 128 08 Prague, Czech Republic;
- Correspondence:
| |
Collapse
|
5
|
Capuano I, Buonanno P, Riccio E, Amicone M, Pisani A. Therapeutic advances in ADPKD: the future awaits. J Nephrol 2021; 35:397-415. [PMID: 34009558 DOI: 10.1007/s40620-021-01062-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a heterogeneous genetic disorder included in ciliopathies, representing the fourth cause of end stage renal disease (ESRD), with an estimated prevalence between 1:1000 and 1:2500. It is mainly caused by mutations in the PKD1 and PKD2 genes encoding for polycystin 1 (PC1) and polycystin 2 (PC2), which regulate differentiation, proliferation, survival, apoptosis, and autophagy. The advances in the knowledge of multiple molecular pathways involved in the pathophysiology of ADPKD led to the development of several treatments which are currently under investigation. Recently, the widespread approval of tolvaptan and, in Italy, of long-acting release octreotide (octreotide-LAR), represents but the beginning of the new therapeutic management of ADPKD patients. Encouraging results are expected from ongoing randomized controlled trials (RCTs), which are investigating not only drugs acting on the calcium/cyclic adenosin monoposphate (cAMP) pathway, the most studied target so far, but also molecules targeting specific pathophysiological pathways (e.g. epidermal growth factor (EGF) receptor, AMP-activated protein kinase (AMPK) and KEAP1-Nrf2) and sphingolipids. Moreover, studies on animal models and cultured cells have also provided further promising therapeutic strategies based on the role of intracellular calcium, cell cycle regulation, MAPK pathway, epigenetic DNA, interstitial inflammation, and cell therapy. Thus, in a near future, tailored therapy could be the key to changing the natural history of ADPKD thanks to the vigorous efforts that are being made to implement clinical and preclinical studies in this field. Our review aimed to summarize the spectrum of drugs that are available in the clinical practice and the most promising molecules undergoing clinical, animal, and cultured cell studies.
Collapse
Affiliation(s)
- Ivana Capuano
- Chair of Nephrology "Federico II", Department of Public Health, University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy.
| | - Pasquale Buonanno
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples, Naples, Italy
| | - Eleonora Riccio
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Maria Amicone
- Chair of Nephrology "Federico II", Department of Public Health, University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Antonio Pisani
- Chair of Nephrology "Federico II", Department of Public Health, University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
| |
Collapse
|
6
|
Mao Z, Valluru MK, Ong ACM. Drug repurposing in autosomal dominant polycystic kidney disease: back to the future with pioglitazone. Clin Kidney J 2021; 14:1715-1718. [PMID: 34221378 PMCID: PMC8243263 DOI: 10.1093/ckj/sfab062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Indexed: 11/16/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of end-stage kidney failure. At present, only one drug, tolvaptan, has been approved for use to slow disease progression, but its use is limited by reduced tolerability and idiosyncratic liver toxicity. Thiazolidinediones were first developed as insulin-sensitizers but also regulate gene transcription in multiple tissues, leading to systemic effects on metabolism, inflammation and vascular reactivity. In this issue, Blazer-Yost et al. report the results of a single-centre Phase 1b double-blind placebo-controlled crossover study of the peroxisome proliferator-activated receptor γ (PPAR-γ) agonist pioglitazone in 18 ADPKD patients. Encouragingly, there were no major safety signals, although evidence of efficacy could not be demonstrated due to the small sample size. We review the preclinical evidence for the use of PPAR-γ agonists in ADPKD and speculate on the likely beneficial and adverse clinical effects of this interesting class of compounds in a future trial.
Collapse
Affiliation(s)
- Zhiguo Mao
- Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Manoj K Valluru
- Department of Infection, Immunity and Cardiovascular Disease, Kidney Genetics Group, Academic Nephrology Unit, University of Sheffield Medical School, Sheffield, UK
| | - Albert C M Ong
- Department of Infection, Immunity and Cardiovascular Disease, Kidney Genetics Group, Academic Nephrology Unit, University of Sheffield Medical School, Sheffield, UK
| |
Collapse
|
7
|
Blazer-Yost BL, Bacallao RL, Erickson BJ, LaPradd ML, Edwards ME, Sheth N, Swinney K, Ponsler-Sipes KM, Moorthi RN, Perkins SM, Torres VE, Moe SM. A randomized phase 1b cross-over study of the safety of low-dose pioglitazone for treatment of autosomal dominant polycystic kidney disease. Clin Kidney J 2021; 14:1738-1746. [PMID: 34221381 PMCID: PMC8243264 DOI: 10.1093/ckj/sfaa232] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Indexed: 01/10/2023] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common monogenetic disorders in humans and is characterized by numerous fluid-filled cysts that grow slowly, resulting in end-stage renal disease in the majority of patients. Preclinical studies have indicated that treatment with low-dose thiazolidinediones, such as pioglitazone, decrease cyst growth in rodent models of PKD. Methods This Phase 1b cross-over study compared the safety of treatment with a low dose (15 mg) of the peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist pioglitazone or placebo in PKD patients, with each treatment given for 1 year. The study monitored known side effects of PPAR-γ agonist treatment, including fluid retention and edema. Liver enzymes and risk of hypoglycemia were assessed throughout the study. As a secondary objective, the efficacy of low-dose pioglitazone was followed using a primary assessment of total kidney volume (TKV), blood pressure (BP) and kidney function. Results Eighteen patients were randomized and 15 completed both arms. Compared with placebo, allocation to pioglitazone resulted in a significant decrease in total body water as assessed by bioimpedance analysis {mean difference 0.16 Ω [95% confidence interval (CI) 0.24–2.96], P = 0.024} and no differences in episodes of heart failure, clinical edema or change in echocardiography. Allocation to pioglitazone led to no difference in the percent change in TKV of −3.5% (95% CI −8.4–1.4, P = 0.14), diastolic BP and microalbumin:creatinine ratio. Conclusions In this small pilot trial in people with ADPKD but without diabetes, pioglitazone 15 mg was found to be as safe as placebo. Larger and longer-term randomized trials powered to assess effects on TKV are needed.
Collapse
Affiliation(s)
- Bonnie L Blazer-Yost
- Department of Biology, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Robert L Bacallao
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush Veteran Administration Health Center, Indianapolis, IN, USA
| | | | - Michelle L LaPradd
- Department of Biostatistics, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Marie E Edwards
- Department of Medicine, Division of Nephrology, Mayo Clinic, Rochester, MN, USA
| | - Nehal Sheth
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kim Swinney
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kristen M Ponsler-Sipes
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ranjani N Moorthi
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Susan M Perkins
- Department of Biostatistics, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Vicente E Torres
- Department of Medicine, Division of Nephrology, Mayo Clinic, Rochester, MN, USA
| | - Sharon M Moe
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush Veteran Administration Health Center, Indianapolis, IN, USA
| |
Collapse
|
8
|
Nowak KL, Hopp K. Metabolic Reprogramming in Autosomal Dominant Polycystic Kidney Disease: Evidence and Therapeutic Potential. Clin J Am Soc Nephrol 2020; 15:577-584. [PMID: 32086281 PMCID: PMC7133124 DOI: 10.2215/cjn.13291019] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Autosomal dominant polycystic kidney disease is characterized by progressive development and enlargement of kidney cysts, leading to ESKD. Because the kidneys are under high metabolic demand, it is not surprising that mounting evidence suggests that a metabolic defect exists in in vitro and animal models of autosomal dominant polycystic kidney disease, which likely contributes to cystic epithelial proliferation and subsequent cyst growth. Alterations include defective glucose metabolism (reprogramming to favor aerobic glycolysis), dysregulated lipid and amino acid metabolism, impaired autophagy, and mitochondrial dysfunction. Limited evidence supports that cellular kidney metabolism is also dysregulated in humans with autosomal dominant polycystic kidney disease. There are notable overlapping features and pathways among metabolism, obesity, and/or autosomal dominant polycystic kidney disease. Both dietary and pharmacologic-based strategies targeting metabolic abnormalities are being considered as therapies to slow autosomal dominant polycystic kidney disease progression and are attractive, particularly given the slowly progressive nature of the disease. Dietary strategies include daily caloric restriction, intermittent fasting, time-restricted feeding, a ketogenic diet, and 2-deoxy-glucose as well as alterations to nutrient availability. Pharmacologic-based strategies include AMP-activated kinase activators, sodium glucose cotransporter-2 inhibitors, niacinamide, and thiazolidenediones. The results from initial clinical trials targeting metabolism are upcoming and anxiously awaited within the scientific and polycystic kidney disease communities. There continues to be a need for additional mechanistic studies to better understand the role of dysregulated metabolism in autosomal dominant polycystic kidney disease and for subsequent translation to clinical trials. Beyond single-intervention trials focused on metabolic reprograming in autosomal dominant polycystic kidney disease, great potential also exists by combining metabolic-focused therapeutic approaches with compounds targeting other signaling cascades altered in autosomal dominant polycystic kidney disease, such as tolvaptan.
Collapse
Affiliation(s)
- Kristen L Nowak
- Division of Renal Diseases and Hypertension, Polycystic Kidney Disease Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Katharina Hopp
- Division of Renal Diseases and Hypertension, Polycystic Kidney Disease Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
9
|
Kanhai AA, Bange H, Verburg L, Dijkstra KL, Price LS, Peters DJM, Leonhard WN. Renal cyst growth is attenuated by a combination treatment of tolvaptan and pioglitazone, while pioglitazone treatment alone is not effective. Sci Rep 2020; 10:1672. [PMID: 32015419 PMCID: PMC6997373 DOI: 10.1038/s41598-020-58382-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/09/2020] [Indexed: 12/30/2022] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is one of the most common monogenic disorders, characterized by the progressive formation of fluid-filled cysts. Tolvaptan is an approved drug for ADPKD patients, but is also associated with multiple side effects. The peroxisome proliferator-activator receptor gamma (PPARγ) agonist pioglitazone slows disease progression in the PCK rat model for PKD. Here, we tested whether a combination treatment of relevant doses of tolvaptan and pioglitazone leads to improved efficacy in an adult-onset PKD mouse model. Tolvaptan indeed slowed PKD progression, but the combination treatment was not more effective than tolvaptan alone. In addition, although pioglitazone raised plasma levels of its surrogate drug marker adiponectin, the drug unexpectedly failed to slow PKD progression. The pioglitazone target PPARγ was expressed at surprisingly low levels in mouse, rat and human kidneys. Other pioglitazone targets were more abundantly expressed, but this pattern was comparable across various species. The data suggest that several potential pharmacokinetic and pharmacodynamic (PK/PD) differences between different species may underlie whether or not pioglitazone is able to slow PKD progression. The ongoing phase II clinical trial with low-dose pioglitazone treatment (NCT02697617) will show whether pioglitazone is a suitable drug candidate for ADPKD treatment.
Collapse
Affiliation(s)
- Anish A Kanhai
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Lotte Verburg
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Kyra L Dijkstra
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.
| | - Wouter N Leonhard
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
10
|
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: 121] [Impact Index Per Article: 24.2] [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
|
11
|
Molinari E, Sayer JA. Emerging treatments and personalised medicine for ciliopathies associated with cystic kidney disease. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1372282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Elisa Molinari
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - John A. Sayer
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
12
|
Boldt C, Röschel T, Himmerkus N, Plain A, Bleich M, Labes R, Blum M, Krause H, Magheli A, Giesecke T, Mutig K, Rothe M, Weldon SM, Dragun D, Schunck WH, Bachmann S, Paliege A. Vasopressin lowers renal epoxyeicosatrienoic acid levels by activating soluble epoxide hydrolase. Am J Physiol Renal Physiol 2016; 311:F1198-F1210. [PMID: 27681558 DOI: 10.1152/ajprenal.00062.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 09/19/2016] [Indexed: 11/22/2022] Open
Abstract
Activation of the thick ascending limb (TAL) Na+-K+-2Cl- cotransporter (NKCC2) by the antidiuretic hormone arginine vasopressin (AVP) is an essential mechanism of renal urine concentration and contributes to extracellular fluid and electrolyte homeostasis. AVP effects in the kidney are modulated by locally and/or by systemically produced epoxyeicosatrienoic acid derivates (EET). The relation between AVP and EET metabolism has not been determined. Here, we show that chronic treatment of AVP-deficient Brattleboro rats with the AVP V2 receptor analog desmopressin (dDAVP; 5 ng/h, 3 days) significantly lowered renal EET levels (-56 ± 3% for 5,6-EET, -50 ± 3.4% for 11,12-EET, and -60 ± 3.7% for 14,15-EET). The abundance of the principal EET-degrading enzyme soluble epoxide hydrolase (sEH) was increased at the mRNA (+160 ± 37%) and protein levels (+120 ± 26%). Immunohistochemistry revealed dDAVP-mediated induction of sEH in connecting tubules and cortical and medullary collecting ducts, suggesting a role of these segments in the regulation of local interstitial EET signals. Incubation of murine kidney cell suspensions with 1 μM 14,15-EET for 30 min reduced phosphorylation of NKCC2 at the AVP-sensitive threonine residues T96 and T101 (-66 ± 5%; P < 0.05), while 14,15-DHET had no effect. Concomitantly, isolated perfused cortical thick ascending limb pretreated with 14,15-EET showed a 30% lower transport current under high and a 70% lower transport current under low symmetric chloride concentrations. In summary, we have shown that activation of AVP signaling stimulates renal sEH biosynthesis and enzyme activity. The resulting reduction of EET tissue levels may be instrumental for increased NKCC2 transport activity during AVP-induced antidiuresis.
Collapse
Affiliation(s)
- Christin Boldt
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tom Röschel
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nina Himmerkus
- Department of Physiology, Christian-Albrechts-University, Kiel, Germany
| | - Allein Plain
- Department of Physiology, Christian-Albrechts-University, Kiel, Germany
| | - Markus Bleich
- Department of Physiology, Christian-Albrechts-University, Kiel, Germany
| | - Robert Labes
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maximilian Blum
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Hans Krause
- Department of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ahmed Magheli
- Department of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Torsten Giesecke
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kerim Mutig
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Steven M Weldon
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut
| | - Duska Dragun
- Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany; and.,Berlin Institute of Health, Berlin, Germany
| | | | - Sebastian Bachmann
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Paliege
- Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany; and .,Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
13
|
Flaig SM, Gattone VH, Blazer-Yost BL. Inhibition of cyst growth in PCK and Wpk rat models of polycystic kidney disease with low doses of peroxisome proliferator-activated receptor γ agonists. J Transl Int Med 2016; 4:118-126. [PMID: 28191533 DOI: 10.1515/jtim-2016-0028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The studies were designed to test the efficacy of two peroxisome proliferator-activated receptor γ (PPARγ) agonists in two rodent models of polycystic kidney disease (PKD). MATERIALS AND METHODS The PCK rat is a slowly progressing cystic model while the Wpk-/- rat is a rapidly progressing model. PCK rats were fed with a pharmacological (0.4 mg/kg body weight [BW]) and a sub-pharmacological (0.04 mg/kg BW) dose of rosiglitazone (week 4-28). Wpk-/- rats were fed with pharmacological (2.0 mg/kg BW) and sub-pharmacologic (0.2 mg/kg BW) doses of pioglitazone from day 5 to 18. At termination, kidney weights of treated versus untreated cystic animals were used to determine efficacy. The current studies were also compared with previous studies containing higher doses of PPARγ agonists. The concentrations used in the animals were calculated with reference to equivalent human doses for both drugs. RESULTS The current studies demonstrate: 1) that low, pharmacologically relevant, doses of the PPARγ agonists effectively inhibit cyst growth; 2) there is a class action of the drugs with both commercially available PPARγ agonists, rosiglitazone, and pioglitazone, inhibiting cyst growth; 3) the drugs showed efficacy in two different preclinical cystic models. In the PCK rat, animals fed with a sub-pharmacological dose of rosiglitazone for 24 weeks had significantly lower kidney weights than untreated animals (3.68 ± 0.13 g vs. 4.17 ± 0. 11 g, respectively, P < 0.01) while treatment with a pharmacologic dose had no significant effect on kidney weight. The rapidly progressing Wpk-/- rats were fed with pharmacological and sub-pharmacologic doses of pioglitazone from day 5 to 18 and the kidneys were compared with non-treated, cystic animals. Kidney weights on the pharmacologic dose were not statistically lower than the untreated animals while rats fed a sub-pharmacologic dose showed a significant decrease compared with untreated animals (3.35 ± 0.15 g vs. 4.55 ± 0.46 g, respectively, P = 0.045). CONCLUSION Concentrations of PPARγ agonists below the human equivalent diabetic doses are effective in slowing cyst growth in two rodent models of PKD.
Collapse
Affiliation(s)
- Stephanie M Flaig
- Biology Department, Indiana University Purdue University Indianapolis, IN 46202, United States of America
| | - Vincent H Gattone
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America
| | - Bonnie L Blazer-Yost
- Biology Department, Indiana University Purdue University Indianapolis, IN 46202, United States of America; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America
| |
Collapse
|
14
|
Goltsman I, Khoury EE, Winaver J, Abassi Z. Does Thiazolidinedione therapy exacerbate fluid retention in congestive heart failure? Pharmacol Ther 2016; 168:75-97. [PMID: 27598860 DOI: 10.1016/j.pharmthera.2016.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ever-growing global burden of congestive heart failure (CHF) and type 2 diabetes mellitus (T2DM) as well as their co-existence necessitate that anti-diabetic pharmacotherapy will modulate the cardiovascular risk inherent to T2DM while complying with the accompanying restrictions imposed by CHF. The thiazolidinedione (TZD) family of peroxisome proliferator-activated receptor γ (PPARγ) agonists initially provided a promising therapeutic option in T2DM owing to anti-diabetic efficacy combined with pleiotropic beneficial cardiovascular effects. However, the utility of TZDs in T2DM has declined in the past decade, largely due to concomitant adverse effects of fluid retention and edema formation attributed to salt-retaining effects of PPARγ activation on the nephron. Presumably, the latter effects are potentially deleterious in the context of pre-existing fluid retention in CHF. However, despite a considerable body of evidence on mechanisms responsible for TZD-induced fluid retention suggesting that this class of drugs is rightfully prohibited from use in CHF patients, there is a paucity of experimental and clinical studies that investigate the effects of TZDs on salt and water homeostasis in the CHF setting. In an attempt to elucidate whether TZDs actually exacerbate the pre-existing fluid retention in CHF, our review summarizes the pathophysiology of fluid retention in CHF. Moreover, we thoroughly review the available data on TZD-induced fluid retention and proposed mechanisms in animals and patients. Finally, we will present recent studies challenging the common notion that TZDs worsen renal salt and water retention in CHF.
Collapse
Affiliation(s)
- Ilia Goltsman
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Emad E Khoury
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Joseph Winaver
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Laboratory Medicine, Rambam Human Health Care Campus, Haifa, Israel.
| |
Collapse
|
15
|
Zhou L, Panasiuk A, Downton M, Zhao D, Yang B, Jia Z, Yang T. Systemic PPARγ deletion causes severe disturbance in fluid homeostasis in mice. Physiol Genomics 2015; 47:541-7. [PMID: 26330489 DOI: 10.1152/physiolgenomics.00066.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/27/2015] [Indexed: 01/29/2023] Open
Abstract
The pharmacological action of peroxisome proliferator-activated receptor (PPAR)γ in promoting sodium and water retention is well documented as highlighted by the major side-effect of body weight gain and edema associated with thiazolidinedione use. However, a possible physiological role of PPARγ in regulation of fluid metabolism has not been reported by previous studies. Here we analyzed fluid metabolism in inducible whole-body PPARγ knockout mice. The null mice developed severe polydipsia and polyuria, reduced urine osmolality, and modest hyperphagia. The phenomenon persisted during 3 days of pair feeding and pair drinking, accompanied by progressive weight loss. After 24 h water deprivation, the null mice had a lower urine osmolality, a higher urine volume, a greater weight loss, and a greater rise in hematocrit than the floxed control. Urinary vasopressin (AVP) excretion was not different between the genotypes under basal condition or after WD. The response of urine osmolality to acute and chronic 1-desamino-8-D-arginine vasopressin treatment was attenuated in the null mice, but the total abundance or phosphorylation of aquaporin 2 (AQP2) in the kidney or AVP-induced cAMP production in inner medullary collecting duct suspensions was unaffected. Overall, PPARγ participates in physiological control of fluid homeostasis through an unknown mechanism involving cAMP/AQP2-independent enhancement of AVP response.
Collapse
Affiliation(s)
- Li Zhou
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Alexandra Panasiuk
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Maicy Downton
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Daqiang Zhao
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhanjun Jia
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| |
Collapse
|
16
|
Raksaseri P, Chatsudthipong V, Muanprasat C, Soodvilai S. Activation of liver X receptors reduces CFTR-mediated Cl(-) transport in kidney collecting duct cells. Am J Physiol Renal Physiol 2013; 305:F583-91. [PMID: 23720350 DOI: 10.1152/ajprenal.00579.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Liver X receptors (LXRs) are transcription factors belonging to the nuclear receptor super family, which act as regulators of lipid and glucose metabolism. However, LXRs have been shown to regulate the function of transporters in the kidney, including the Na-Pi cotransporter, organic anion transporter, and epithelial Na(+) channel. In this report, we demonstrated the ability of LXR ligands, both endogenous [22 (R)-hydroxycholesterol] and synthetic (T0901317 and GW3965), to reduce CFTR-mediated Cl(-) secretion in a type I Madin-Darby canine kidney (MDCK) cell line and in murine primary inner medullary collecting duct (IMCD) cells, based on measurements of [Arg(8)]-vasopressin-induced Cl(-) current. However, treatment of MDCK cell monolayers with 5 μM T0901317 for 24 h did not alter ouabain-senstive current or Na(+)-K(+)-ATPase-α protein content. Furthermore, basolateral membranes permeabilization of MDCK cell monolayers still resulted in a decrease in apical Cl(-) current stimulated by both [Arg(8)]-vasopressin and 8-cholorophenyl-thio-cAMP, indicating that the factor(s) encoded by the target gene(s) of agonist-activated LXRs might be located at the apical membrane. Western blot analysis revealed that inhibition of Cl(-) secretion occurred via a decrease in CFTR protein, which was not due to downregulation of its mRNA expression. In addition, both synthetic LXR agonists significantly retarded the growth of forskolin-induced cysts formed in MDCK cells cultured in collagen gel. This is the first evidence showing that ligand-activated LXRs are capable of downregulating CFTR-mediated Cl(-) secretion of kidney cells and of retarding cyst growth in a MDCK cell model.
Collapse
Affiliation(s)
- Promporn Raksaseri
- Dept. of Physiology, Faculty of Science, Mahidol Univ., Rama VI Road, Bangkok 10400, Thailand.
| | | | | | | |
Collapse
|
17
|
Bełtowski J, Rachańczyk J, Włodarczyk M. Thiazolidinedione-induced fluid retention: recent insights into the molecular mechanisms. PPAR Res 2013; 2013:628628. [PMID: 23577024 PMCID: PMC3614122 DOI: 10.1155/2013/628628] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/19/2013] [Indexed: 01/11/2023] Open
Abstract
Peroxisome proliferator-activated receptor- γ (PPAR γ ) agonists such as rosiglitazone and pioglitazone are used to improve insulin sensitivity in patients with diabetes mellitus. However, thiazolidinediones induce fluid retention, edema, and sometimes precipitate or exacerbate heart failure in a subset of patients. The mechanism through which thiazolidinediones induce fluid retention is controversial. Most studies suggest that this effect results from the increase in tubular sodium and water reabsorption in the kidney, but the role of specific nephron segments and sodium carriers involved is less clear. Some studies suggested that PPAR γ agonist stimulates Na(+) reabsorption in the collecting duct by activating epithelial Na(+) channel (ENaC), either directly or through serum and glucocorticoid-regulated kinase-1 (SGK-1). However, other studies did not confirm this mechanism and even report the suppression of ENaC. Alternative mechanisms in the collecting duct include stimulation of non-ENaC sodium channel or inhibition of chloride secretion to the tubular lumen. In addition, thiazolidinediones may augment sodium reabsorption in the proximal tubule by stimulating the expression and activity of apical Na(+)/H(+) exchanger-3 and basolateral Na(+)-HCO3 (-) cotransporter as well as of Na(+),K(+)-ATPase. These effects are mediated by PPAR γ -induced nongenomic transactivation of the epidermal growth factor receptor and downstream extracellular signal-regulated kinases (ERK).
Collapse
Affiliation(s)
- Jerzy Bełtowski
- Department of Pathophysiology, Medical University of Lublin, 8 Jaczewskiego, 20090 Lublin, Poland
| | - Jolanta Rachańczyk
- Department of Pathophysiology, Medical University of Lublin, 8 Jaczewskiego, 20090 Lublin, Poland
| | - Mirosław Włodarczyk
- Department of Pathophysiology, Medical University of Lublin, 8 Jaczewskiego, 20090 Lublin, Poland
| |
Collapse
|
18
|
Yuajit C, Homvisasevongsa S, Chatsudthipong L, Soodvilai S, Muanprasat C, Chatsudthipong V. Steviol reduces MDCK Cyst formation and growth by inhibiting CFTR channel activity and promoting proteasome-mediated CFTR degradation. PLoS One 2013; 8:e58871. [PMID: 23536832 PMCID: PMC3594167 DOI: 10.1371/journal.pone.0058871] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 02/07/2013] [Indexed: 01/26/2023] Open
Abstract
Cyst enlargement in polycystic kidney disease (PKD) involves cAMP-activated proliferation of cyst-lining epithelial cells and transepithelial fluid secretion into the cyst lumen via cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel. This study aimed to investigate an inhibitory effect and detailed mechanisms of steviol and its derivatives on cyst growth using a cyst model in Madin-Darby canine kidney (MDCK) cells. Among 4 steviol-related compounds tested, steviol was found to be the most potent at inhibiting MDCK cyst growth. Steviol inhibition of cyst growth was dose-dependent; steviol (100 microM) reversibly inhibited cyst formation and cyst growth by 72.53.6% and 38.2±8.5%, respectively. Steviol at doses up to 200 microM had no effect on MDCK cell viability, proliferation and apoptosis. However, steviol acutely inhibited forskolin-stimulated apical chloride current in MDCK epithelia, measured with the Ussing chamber technique, in a dose-dependent manner. Prolonged treatment (24 h) with steviol (100 microM) also strongly inhibited forskolin-stimulated apical chloride current, in part by reducing CFTR protein expression in MDCK cells. Interestingly, proteasome inhibitor, MG-132, abolished the effect of steviol on CFTR protein expression. Immunofluorescence studies demonstrated that prolonged treatment (24 h) with steviol (100 microM) markedly reduced CFTR expression at the plasma membrane. Taken together, the data suggest that steviol retards MDCK cyst progression in two ways: first by directly inhibiting CFTR chloride channel activity and second by reducing CFTR expression, in part, by promoting proteasomal degradation of CFTR. Steviol and related compounds therefore represent drug candidates for treatment of polycystic kidney disease.
Collapse
Affiliation(s)
- Chaowalit Yuajit
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Sureeporn Homvisasevongsa
- Division of Physical Science, Faculty of Science and Technology, Huachiew Chalermprakiet University, Samutprakarn, Thailand
| | - Lisa Chatsudthipong
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Sunhapas Soodvilai
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Research Center of Transport Protein for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Chatchai Muanprasat
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Research Center of Transport Protein for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Varanuj Chatsudthipong
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Research Center of Transport Protein for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
19
|
Irazabal MV, Torres VE. Experimental therapies and ongoing clinical trials to slow down progression of ADPKD. Curr Hypertens Rev 2013; 9:44-59. [PMID: 23971644 PMCID: PMC4067974 DOI: 10.2174/1573402111309010008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/19/2012] [Accepted: 11/30/2012] [Indexed: 12/19/2022]
Abstract
The improvement of imaging techniques over the years has contributed to the understanding of the natural history of autosomal dominant polycystic kidney disease, and facilitated the observation of its structural progression. Advances in molecular biology and genetics have made possible a greater understanding of the genetics, molecular, and cellular pathophysiologic mechanisms responsible for its development and have laid the foundation for the development of potential new therapies. Therapies targeting genetic mechanisms in ADPKD have inherent limitations. As a result, most experimental therapies at the present time are aimed at delaying the growth of the cysts and associated interstitial inflammation and fibrosis by targeting tubular epithelial cell proliferation and fluid secretion by the cystic epithelium. Several interventions affecting many of the signaling pathways disrupted in ADPKD have been effective in animal models and some are currently being tested in clinical trials.
Collapse
Affiliation(s)
- Maria V. Irazabal
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester MN, USA
| | - Vicente E. Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester MN, USA
| |
Collapse
|
20
|
Tan CD, Smolenski RT, Harhun MI, Patel HK, Ahmed SG, Wanisch K, Yáñez-Muñoz RJ, Baines DL. AMP-activated protein kinase (AMPK)-dependent and -independent pathways regulate hypoxic inhibition of transepithelial Na+ transport across human airway epithelial cells. Br J Pharmacol 2013; 167:368-82. [PMID: 22509822 DOI: 10.1111/j.1476-5381.2012.01993.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary transepithelial Na(+) transport is reduced by hypoxia, but in the airway the regulatory mechanisms remain unclear. We investigated the role of AMPK and ROS in the hypoxic regulation of apical amiloride-sensitive Na(+) channels and basolateral Na(+) K(+) ATPase activity. EXPERIMENTAL APPROACH H441 human airway epithelial cells were used to examine the effects of hypoxia on Na(+) transport, AMP : ATP ratio and AMPK activity. Lentiviral constructs were used to modify cellular AMPK abundance and activity; pharmacological agents were used to modify cellular ROS. KEY RESULTS AMPK was activated by exposure to 3% or 0.2% O(2) for 60 min in cells grown in submerged culture or when fluid (0.1 mL·cm(-2) ) was added to the apical surface of cells grown at the air-liquid interface. Only 0.2% O(2) activated AMPK in cells grown at the air-liquid interface. AMPK activation was associated with elevation of cellular AMP:ATP ratio and activity of the upstream kinase LKB1. Hypoxia inhibited basolateral ouabain-sensitive I(sc) (I(ouabain) ) and apical amiloride-sensitive Na(+) conductance (G(Na+) ). Modification of AMPK activity prevented the effect of hypoxia on I(ouabain) (Na(+) K(+) ATPase) but not apical G(Na+) . Scavenging of superoxide and inhibition of NADPH oxidase prevented the effect of hypoxia on apical G(Na+) (epithelial Na(+) channels). CONCLUSIONS AND IMPLICATIONS Hypoxia activates AMPK-dependent and -independent pathways in airway epithelial cells. Importantly, these pathways differentially regulate apical Na(+) channels and basolateral Na(+) K(+) ATPase activity to decrease transepithelial Na(+) transport. Luminal fluid potentiated the effect of hypoxia and activated AMPK, which could have important consequences in lung disease conditions.
Collapse
Affiliation(s)
- C D Tan
- Pharmacology and Cell Physiology Research Group, Division of Biomedical Sciences, St George's University of London, Cranmer Terrace, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Gondzik V, Weber WM, Awayda MS. Coupling of epithelial Na+ and Cl- channels by direct and indirect activation by serine proteases. Am J Physiol Cell Physiol 2012; 303:C936-46. [PMID: 22914644 DOI: 10.1152/ajpcell.00395.2011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The mammalian collecting duct (CD) is continuously exposed to urinary proteases. The CD expresses an epithelial Na(+) channel (ENaC) that is activated after cleavage by serine proteases. ENaC also exists at the plasma membrane in the uncleaved form, rendering activation by extracellular proteases an important mechanism for regulating Na(+) transport. Many exogenous and a small number of endogenous extracellular serine proteases have been shown to activate the channel. Recently, kallikrein 1 (KLK1) was shown to increase γENaC cleavage in the native CD indicating a possible direct role of this endogenous protease in Na(+) homeostasis. To explore this process, we examined the coordinated effect of this protease on Na(+) and Cl(-) transport in a polarized renal epithelial cell line (Madin-Darby canine kidney). We also examined the role of native urinary proteases in this process. Short-circuit current (I(sc)) was used to measure transport of these ions. The I(sc) exhibited an ENaC-dependent Na(+) component that was amiloride blockable and a cystic fibrosis transmembrane conductance regulator (CFTR)-dependent Cl(-) component that was blocked by inhibitor 172. Apical application of trypsin, an exogenous S1 serine protease, activated I(ENaC) but was without effects on I(CFTR). Subtilisin an exogenous S8 protease that mimics endogenous furin-type proteases activated both currents. A similar activation was also observed with KLK1 and native rat urinary proteases. Activation with urinary proteases occurred within minutes and at protease concentrations similar to those in the CD indicating physiological significance of this process. ENaC activation was irreversible and mediated by enhanced cleavage of γENaC. The activation of CFTR was indirect and likely dependent on activation of an endogenous apical membrane protease receptor. Collectively, these data demonstrate coordinated stimulation of separate Na(+) and Cl(-) transport pathways in renal epithelia by extracellular luminal proteases. They also indicate that baseline urinary proteolytic activity is sufficient to modify Na(+) and Cl(-) transport in these epithelia.
Collapse
Affiliation(s)
- Veronika Gondzik
- Dept. of Physiology and Biophysics, SUNY at Buffalo, 124 Sherman Hall, 3435 Main St., Buffalo NY 14214, USA
| | | | | |
Collapse
|
22
|
Li H, Yang W, Mendes F, Amaral MD, Sheppard DN. Impact of the cystic fibrosis mutation F508del-CFTR on renal cyst formation and growth. Am J Physiol Renal Physiol 2012; 303:F1176-86. [PMID: 22874761 DOI: 10.1152/ajprenal.00130.2012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In autosomal dominant polycystic kidney disease (ADPKD), cystic fibrosis transmembrane conductance regulator (CFTR), the protein product of the gene defective in cystic fibrosis (CF), plays a crucial role in fluid accumulation, which promotes cyst swelling. Several studies have identified individuals afflicted by both ADPKD and CF. Two studies suggested that CF mutations might attenuate the severity of ADPKD, whereas a third found no evidence of a protective effect. In this study, we investigated the impact of the commonest CF mutation F508del-CFTR on the formation and growth of renal cysts. As a model system, we used Madin-Darby canine kidney (MDCK) epithelial cells engineered to express wild-type and F508del human CFTR. We grew MDCK cysts in collagen gels in the presence of the cAMP agonist forskolin and measured transepithelial resistance and Cl(-) secretion with the Ussing chamber technique and assayed cell proliferation using nonpolarized MDCK cells. When compared with untransfected MDCK cells, cells expressing wild-type CFTR generated substantial numbers of large cysts, which grew markedly over time. By contrast, MDCK cells expressing F508del-CFTR formed very few tiny cysts that failed to enlarge. Interestingly, treatment of F508del-CFTR cysts with the CFTR corrector VRT-325 and the CFTR corrector-potentiator VRT-532 increased the number, but not size, of F508del-CFTR cysts, possibly because VRT-325 inhibited strongly cell proliferation. Based on its effects on transepithelial resistance, Cl(-) secretion, and cell proliferation, we conclude that the F508del-CFTR mutation disrupts cyst formation and growth by perturbing strongly fluid accumulation within the cyst lumen without compromising epithelial integrity.
Collapse
Affiliation(s)
- Hongyu Li
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, UK
| | | | | | | | | |
Collapse
|
23
|
Dekkers JF, van der Ent CK, Kalkhoven E, Beekman JM. PPARγ as a therapeutic target in cystic fibrosis. Trends Mol Med 2012; 18:283-91. [PMID: 22494945 DOI: 10.1016/j.molmed.2012.03.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/06/2012] [Accepted: 03/12/2012] [Indexed: 12/31/2022]
Abstract
Cystic fibrosis (CF) is characterized by a proinflammatory pulmonary condition that may result from increased infections and altered intracellular metabolism in CFTR-deficient cells. The lipid-activated transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) has well-established roles in immune cell function and inflammatory modulation and has been demonstrated to play an important role in the heightened inflammatory response in CF cells. Here, we summarize current literature describing PPARγ-dependent alterations of CF cells and discuss the potential of PPARγ ligands for treating CF.
Collapse
Affiliation(s)
- Johanna F Dekkers
- Department of Pediatric Pulmonology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
24
|
Chang MY, Ong ACM. Mechanism-based therapeutics for autosomal dominant polycystic kidney disease: recent progress and future prospects. Nephron Clin Pract 2011; 120:c25-34; discussion c35. [PMID: 22205396 DOI: 10.1159/000334166] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease, accounting for up to 10% of patients on renal replacement therapy. There are presently no proven treatments for ADPKD and an effective disease-modifying drug would have significant implications for patients and their families. Since the identification of PKD1 and PKD2, there has been an explosion in knowledge identifying new disease mechanisms and testing new drugs. Currently, the three major treatment strategies are to: (1) reduce cAMP levels; (2) inhibit cell proliferation, and (3) reduce fluid secretion. Several compounds shown to be effective in preclinical models have already undergone clinical trials and more are planned. In addition, a whole raft of other compounds have been developed from preclinical studies. The purpose of this paper is to evaluate the results of recent published trials, review current trials and highlight the most promising compounds in the pipeline. There appears to be no shortage of potential candidates, but several key issues need to be addressed to facilitate clinical translation.
Collapse
Affiliation(s)
- Ming-Yang Chang
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | | |
Collapse
|
25
|
Campioli E, Batarseh A, Li J, Papadopoulos V. The endocrine disruptor mono-(2-ethylhexyl) phthalate affects the differentiation of human liposarcoma cells (SW 872). PLoS One 2011; 6:e28750. [PMID: 22205965 PMCID: PMC3244402 DOI: 10.1371/journal.pone.0028750] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 11/14/2011] [Indexed: 01/04/2023] Open
Abstract
Esters of phthalic acid (phthalates) are largely used in industrial plastics, medical devices, and pharmaceutical formulations. They are easily released from plastics into the environment and can be found in measurable levels in human fluids. Phthalates are agonists for peroxisome proliferator-activated receptors (PPARs), through which they regulate translocator protein (TSPO; 18 kDa) transcription in a tissue-specific manner. TSPO is a drug- and cholesterol-binding protein involved in mitochondrial respiration, steroid formation, and cell proliferation. TSPO has been shown to increase during differentiation and decrease during maturation in mouse adipocytes. The purpose of this study was to establish the effect of mono-(2-ethylhexyl) phthalate (MEHP) on the differentiation of human SW 872 preadipocyte cells, and examine the role of TSPO in the process. After 4 days of treatment with 10 µM MEHP, we observed changes in the transcription of acetyl-CoA carboxylase alpha, adenosine triphosphate citrate lyase, glucose transporters 1 and 4, and the S100 calcium binding protein B, all of which are markers of preadipocyte differentiation. These observed gene expression changes coincided with a decrease in cellular proliferation without affecting cellular triglyceride content. Taken together, these data suggest that MEHP exerts a differentiating effect on human preadipocytes. Interestingly, MEHP was able to temporarily increase TSPO mRNA levels through the PPAR-α and β/δ pathways. These results suggest that TSPO can be considered an important player in the differentiation process itself, or alternatively a factor whose presence is essential for adipocyte development.
Collapse
Affiliation(s)
- Enrico Campioli
- Research Institute of the McGill University Health Center and the Departments of Medicine, Biochemistry, and Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Amani Batarseh
- Research Institute of the McGill University Health Center and the Departments of Medicine, Biochemistry, and Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Jiehan Li
- Research Institute of the McGill University Health Center and the Departments of Medicine, Biochemistry, and Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Vassilios Papadopoulos
- Research Institute of the McGill University Health Center and the Departments of Medicine, Biochemistry, and Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
26
|
Blazer-Yost BL, Blacklock BJ, Flaig S, Bacallao RL, Gattone VH. Lysophosphatidic acid is a modulator of cyst growth in autosomal dominant polycystic kidney disease. Cell Physiol Biochem 2011; 28:1255-64. [PMID: 22179013 DOI: 10.1159/000335857] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2011] [Indexed: 12/31/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the slow growth of multiple fluid-filled cysts predominately in the kidney tubules and liver bile ducts. Elucidation of mechanisms that control cyst growth will provide the basis for rational therapeutic intervention. We used electrophysiological methods to identify lysophosphatidic acid (LPA) as a component of cyst fluid and serum that stimulates secretory Cl- transport in the epithelial cell type that lines renal cysts. LPA effects are manifested through receptors located on the basolateral membrane of the epithelial cells resulting in stimulation of channel activity in the apical membrane. Concentrations of LPA measured in human ADPKD cyst fluid and in normal serum are sufficient to maximally stimulate ion transport. Thus, cyst fluid seepage and/or leakage of vascular LPA into the interstitial space are capable of stimulating epithelial cell secretion resulting in cyst enlargement. These observations are particularly relevant to the rapid decline in renal function in late-stage disease and to the "third hit" hypothesis that renal injury exacerbates cyst growth.
Collapse
Affiliation(s)
- Bonnie L Blazer-Yost
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | | | | | | | | |
Collapse
|
27
|
Goltsman I, Wang X, LaVallie ER, DiBlasio-Smith EA, Ovcharenko E, Hoffman A, Abassi Z, Feuerstein GZ, Winaver J. Effects of Chronic Rosiglitazone Treatment on Renal Handling of Salt and Water in Rats With Volume-Overload Congestive Heart Failure. Circ Heart Fail 2011; 4:345-54. [DOI: 10.1161/circheartfailure.110.960179] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
The side effects of fluid retention and edema of the thiazolidinedione (TZD) class of peroxisome proliferator-activated receptor-γ agonists limit their use in patients with congestive heart failure (CHF). The present study aims to explore whether chronic treatment with the TZD compound rosiglitazone (RGZ) is associated with worsening of salt and water retention in male Sprague-Dawley rats with aorto-caval fistula, an experimental model of volume-overload CHF.
Methods and Results—
The effects of oral RGZ (30 mg/kg per day for 4 weeks) in CHF rats on plasma volume, cumulative sodium excretion, renal expression of Na
+
channels and transporters, and selected biomarkers of CHF were compared with those in CHF rats and sham-operated control rats treated with vehicle only (n=7 to 10). Additionally, the response to acute saline loading (3.5% of body weight) was evaluated after 2 weeks of treatment by renal clearance methodology. Chronic RGZ treatment caused no further increase in plasma volume compared with vehicle-treated CHF rats. Moreover, no increase in renal expression of Na
+
transport-linked channels/transporters was observed in response to RGZ. Cumulative sodium excretion was enhanced in CHF rats after RGZ and by another TZD compound, pioglitazone. In response to saline loading, RGZ-treated animals displayed a higher natriuretic/diuretic response than did vehicle-treated rats. Chronic RGZ treatment was not associated with any deterioration in selected biomarkers of CHF, whereas indices of cardiac hypertrophy and blood pressure were improved.
Conclusions—
Chronic RGZ treatment was not associated with worsening of fluid retention or cardiac status in rats with experimental volume-overload CHF. Rather, RGZ appeared to improve renal handling of salt and water in rats with CHF.
Collapse
Affiliation(s)
- Ilia Goltsman
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| | - Xinkang Wang
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| | - Edward R. LaVallie
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| | - Elizabeth A. DiBlasio-Smith
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| | - Elena Ovcharenko
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| | - Aaron Hoffman
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| | - Zaid Abassi
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| | - Giora Z. Feuerstein
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| | - Joseph Winaver
- From the Department of Physiology and Biophysics, Faculty of Medicine, Technion-IIT, Haifa, Israel (I.G., E.O., A.H., Z.A., J.W.); Translational Medicine, Pfizer, Collegeville, PA (X.W., G.Z.F.); and Biological Technologies, Pfizer, Cambridge, MA (E.R.L., E.A.D.-S.)
| |
Collapse
|
28
|
Pioglitazone Attenuates Cystic Burden in the PCK Rodent Model of Polycystic Kidney Disease. PPAR Res 2010; 2010:274376. [PMID: 21052534 PMCID: PMC2968120 DOI: 10.1155/2010/274376] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 10/11/2010] [Indexed: 02/08/2023] Open
Abstract
Polycystic kidney disease (PKD) is a genetic disorder characterized by growth of fluid-filled cysts predominately in kidney tubules and liver bile ducts. Currently, the clinical management of PKD is limited to cyst aspiration, surgical resection or organ transplantation. Based on an observation that PPARγ agonists such as pioglitazone and rosiglitazone decrease mRNA levels of a Cl− transport protein, CFTR (cystic fibrosis transmembrane conductance regulator), and the Cl− secretory response to vasopressin in cultured renal cells, it is hypothesized that PPARγ agonists will inhibit cyst growth. The current studies show that a 7- or 14-week pioglitazone feeding regimen inhibits renal and hepatic bile duct cyst growth in the PCK rat, a rodent model orthologous to human PKD. These studies provide proof of concept for the mechanism of action of the PPARγ agonists and suggest that this class of drugs may be effective in controlling both renal and hepatic cyst growth and fibrosis in PKD.
Collapse
|
29
|
Regulation of ENaC-Mediated Sodium Reabsorption by Peroxisome Proliferator-Activated Receptors. PPAR Res 2010; 2010:703735. [PMID: 20613963 PMCID: PMC2896859 DOI: 10.1155/2010/703735] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 03/16/2010] [Accepted: 04/14/2010] [Indexed: 12/14/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of a steroid hormone receptor superfamily that responds to changes in lipid and glucose homeostasis. Peroxisomal proliferator-activated receptor subtype γ (PPARγ) has received much attention as the target for antidiabetic drugs, as well as its role in responding to endogenous compounds such as prostaglandin J2. However, thiazolidinediones (TZDs), the synthetic agonists of the PPARγ are tightly associated with fluid retention and edema, as potentially serious side effects. The epithelial sodium channel (ENaC) represents the rate limiting step for sodium absorption in the renal collecting duct. Consequently, ENaC is a central effector impacting systemic blood volume and pressure. The role of PPARγ agonists on ENaC activity remains controversial. While PPARγ agonists were shown to stimulate ENaC-mediated renal salt absorption, probably via Serum- and Glucocorticoid-Regulated Kinase 1 (SGK1), other studies reported that PPARγ agonist-induced fluid retention is independent of ENaC activity. The current paper provides new insights into the control and function of ENaC and ENaC-mediated sodium transport as well as several other epithelial channels/transporters by PPARs and particularly PPARγ. The potential contribution of arachidonic acid (AA) metabolites in PPAR-dependent mechanisms is also discussed.
Collapse
|
30
|
Blazer-Yost BL. PPARgamma Agonists: Blood Pressure and Edema. PPAR Res 2009; 2010:785369. [PMID: 20069049 PMCID: PMC2801011 DOI: 10.1155/2010/785369] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 11/23/2009] [Indexed: 12/23/2022] Open
Abstract
Peroxisome proliferator activated receptor gamma (PPARgamma) agonists are widely used in the treatment of type 2 diabetes. Side effects of drug treatment include both fluid retention and a lowering of blood pressure. Data from animal and human studies suggest that these effects arise, at least in part, from drug-induced changes in the kidney. In order to capitalize on the positive aspect (lowering of blood pressure) and exclude the negative one (fluid retention), it is necessary to understand the mechanisms of action underlying each of the effects. When interpreted with known physiological principles, current hypotheses regarding potential mechanisms produce enigmas that are difficult to resolve. This paper is a summary of the current understanding of PPARgamma agonist effects on both blood pressure and fluid retention from a renal perspective and concludes with the newest studies that suggest alternative pathways within the kidney that could contribute to the observed drug-induced effects.
Collapse
Affiliation(s)
- Bonnie L. Blazer-Yost
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, SL 358 Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Nofziger C, Blazer-Yost BL. PPARgamma agonists, modulation of ion transporters, and fluid retention. J Am Soc Nephrol 2009; 20:2481-3. [PMID: 19820124 DOI: 10.1681/asn.2009060673] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
32
|
Pavlov TS, Levchenko V, Karpushev AV, Vandewalle A, Staruschenko A. Peroxisome Proliferator-Activated Receptor γ Antagonists Decrease Na+ Transport via the Epithelial Na+ Channel. Mol Pharmacol 2009; 76:1333-40. [DOI: 10.1124/mol.109.056911] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|