51
|
Polycystins as components of large multiprotein complexes of polycystin interactors. Cell Signal 2020; 72:109640. [PMID: 32305669 DOI: 10.1016/j.cellsig.2020.109640] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/27/2022]
Abstract
Naturally occurring mutations in two separate genes, PKD1 and PKD2, are responsible for the vast majority of all cases of autosomal dominant polycystic kidney disease (ADPKD), one of the most common genetic diseases affecting 1 in 1000 Americans. The hallmark of ADPKD is the development of epithelial cysts in the kidney, liver, and pancreas. PKD1 encodes a large plasma membrane protein (PKD1, PC1, or Polycystin-1) with a long extracellular domain and has been speculated to function as an atypical G protein coupled receptor. PKD2 encodes an ion channel of the Transient Receptor Potential superfamily (TRPP2, PKD2, PC2, or Polycystin-2). Despite the identification of these genes more than 20 years ago, the molecular function of their encoded proteins and the mechanism(s) by which mutations in PKD1 and PKD2 cause ADPKD remain elusive. Genetic, biochemical, and functional evidence suggests they form a multiprotein complex present in multiple locations in the cell, including the plasma membrane, endoplasmic reticulum, and the primary cilium. Over the years, numerous interacting proteins have been identified using directed and unbiased approaches, and shown to modulate function, cellular localization, and protein stability and turnover of Polycystins. Delineation of the molecular composition of the Polycystin complex can have a significant impact on understanding their cellular function in health and disease states and on the identification of more specific and effective therapeutic targets.
Collapse
|
52
|
Ta CM, Vien TN, Ng LCT, DeCaen PG. Structure and function of polycystin channels in primary cilia. Cell Signal 2020; 72:109626. [PMID: 32251715 DOI: 10.1016/j.cellsig.2020.109626] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
Abstract
Variants in genes which encode for polycystin-1 and polycystin-2 cause most forms of autosomal dominant polycystic disease (ADPKD). Despite our strong understanding of the genetic determinants of ADPKD, we do not understand the structural features which govern the function of polycystins at the molecular level, nor do we understand the impact of most disease-causing variants on the conformational state of these proteins. These questions have remained elusive because polycystins localize to several organelle membranes, including the primary cilia. Primary cilia are microtubule based organelles which function as cellular antennae. Polycystin-2 and related polycystin-2 L1 are members of the transient receptor potential (TRP) ion channel family, and form distinct ion channels in the primary cilia of disparate cell types which can be directly measured. Polycystin-1 has both ion channel and adhesion G-protein coupled receptor (GPCR) features-but its role in forming a channel complex or as a channel subunit chaperone is undetermined. Nonetheless, recent polycystin structural determination by cryo-EM has provided a molecular template to understand their biophysical regulation and the impact of disease-causing variants. We will review these advances and discuss hypotheses regarding the regulation of polycystin channel opening by their structural domains within the context of the primary cilia.
Collapse
Affiliation(s)
- Chau My Ta
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 E Superior, Chicago, IL 60611, USA
| | - Thuy N Vien
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 E Superior, Chicago, IL 60611, USA
| | - Leo C T Ng
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 E Superior, Chicago, IL 60611, USA
| | - Paul G DeCaen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 E Superior, Chicago, IL 60611, USA.
| |
Collapse
|
53
|
Abstract
Mutations in the polycystins PC1 or PC2 cause autosomal dominant polycystic kidney disease (ADPKD), which is characterized by the formation of fluid-filled renal cysts that disrupt renal architecture and function, ultimately leading to kidney failure in the majority of patients. Although the genetic basis of ADPKD is now well established, the physiological function of polycystins remains obscure and a matter of intense debate. The structural determination of both the homomeric PC2 and heteromeric PC1-PC2 complexes, as well as the electrophysiological characterization of PC2 in the primary cilium of renal epithelial cells, provided new valuable insights into the mechanisms of ADPKD pathogenesis. Current findings indicate that PC2 can function independently of PC1 in the primary cilium of renal collecting duct epithelial cells to form a channel that is mainly permeant to monovalent cations and is activated by both membrane depolarization and an increase in intraciliary calcium. In addition, PC2 functions as a calcium-activated calcium release channel at the endoplasmic reticulum membrane. Structural studies indicate that the heteromeric PC1-PC2 complex comprises one PC1 and three PC2 channel subunits. Surprisingly, several positively charged residues from PC1 occlude the ionic pore of the PC1-PC2 complex, suggesting that pathogenic polycystin mutations might cause ADPKD independently of an effect on channel permeation. Emerging reports of novel structural and functional findings on polycystins will continue to elucidate the molecular basis of ADPKD.
Collapse
|
54
|
Lea WA, McGreal K, Sharma M, Parnell SC, Zelenchuk L, Charlesworth MC, Madden BJ, Johnson KL, McCormick DJ, Hogan MC, Ward CJ. Analysis of the polycystin complex (PCC) in human urinary exosome-like vesicles (ELVs). Sci Rep 2020; 10:1500. [PMID: 32001768 PMCID: PMC6992733 DOI: 10.1038/s41598-020-58087-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 01/05/2020] [Indexed: 12/21/2022] Open
Abstract
The polycystin-1 (PC1), polycystin-2 (PC2) and fibrocystin proteins, the respective products of the PKD1, PKD2 and PKHD1 genes, are abundant in urinary exosome-like vesicles (ELVs) where they form the polycystin complex (PCC). ELVs are 100 nm diameter membrane vesicles shed into the urine by the cells lining the nephron. Using MS/MS analysis of ELVs from individuals with PKD1 mutations and controls, we show that in addition to the well-described GPS/GAIN cleavage event in PC1 at 3048 aa and the proprotein convertase cleavage (PPC) event in fibrocystin at 3616 aa, there are multiple other cleavage events in these proteins. The C-terminal 11 transmembrane portion of PC1 undergoes three cleavage events in vivo. The absence of peptides from the C-terminal cytoplasmic tail of fibrocystin implies a cleavage event close to its single TM domain prior to loading onto the ELVs. There is also evidence that the C-terminal tail of PC2 is also cleaved in ELVs. Native gel analysis of the PCC shows that the entire complex is > 2 MDa in size and that N-terminal GPS/GAIN cleaved PC1 and PPC cleaved fibrocystin ectodomains can be released under non-reducing conditions and resolve at 300 kDa. This paper shows that the three major human cystogene proteins are detectable in human urinary ELVs and that all three undergo post-translational proteolytic processing. Human urinary ELVs may be a useful source of material in the search for proteins that interact with the PCC.
Collapse
Affiliation(s)
- Wendy A Lea
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kerri McGreal
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Madhulika Sharma
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Stephen C Parnell
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Lesya Zelenchuk
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - M Cristine Charlesworth
- Mayo Proteomic Core, Medical Sciences Building, Ms 3-121, Mayo Clinic, 200 First Street, SW Rochester, MN, 55905, USA
| | - Benjamin J Madden
- Mayo Proteomic Core, Medical Sciences Building, Ms 3-121, Mayo Clinic, 200 First Street, SW Rochester, MN, 55905, USA
| | - Kenneth L Johnson
- Mayo Proteomic Core, Medical Sciences Building, Ms 3-121, Mayo Clinic, 200 First Street, SW Rochester, MN, 55905, USA
| | - Daniel J McCormick
- Mayo Proteomic Core, Medical Sciences Building, Ms 3-121, Mayo Clinic, 200 First Street, SW Rochester, MN, 55905, USA
| | - Marie C Hogan
- Division of Nephrology, Department of Internal Medicine, Mayo Clinic, Rochester, USA
| | - Christopher J Ward
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
55
|
A case of cerebral infarction caused by painless acute aortic dissection in autosomal dominant polycystic kidney disease. CEN Case Rep 2020; 9:177-181. [PMID: 31989455 DOI: 10.1007/s13730-020-00450-4] [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: 11/10/2019] [Accepted: 01/14/2020] [Indexed: 10/25/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disorder, characterized by the progressive formation of renal cysts. Although ADPKD is strongly associated with cerebral and cardiovascular complications, cerebral ischemia caused by dissection of thoracic and carotid arteries has rarely been reported. We report the case of a 71-year-old Japanese woman who complained of hemiparesis. She required maintenance hemodialysis therapy with a background of ADPKD. Cerebral infarction was initially diagnosed by excluding intracranial hemorrhage and aneurysm rupture that are recognized as common complications of ADPKD and thereby anticoagulation therapy was initiated. However, the patient was suspected as having painless aortic dissection because a chest X-ray examination showed expanded upper mediastinum. Sequential vascular imagings revealed dissection of the aorta, originating from brachiocephalic trunk to the right common carotid artery with mediastinal hematoma. The patient died from progression of dissection. Herein, we described a case of the ADPKD patient that an acute aortic dissection without any pain induced the occlusion of supplying vessels to the brain, resulting in cerebral ischemic symptoms. A high level of clinical vigilance for an acute aortic dissection should be maintained in the ADPKD population with sudden onset of neurological symptoms even in the absence of pain. Furthermore, the initiation of anticoagulation treatment for cerebral ischemia which may aggravate the risk of further dissection requires careful consideration.
Collapse
|
56
|
Blaustein MP, Hamlyn JM. Ouabain, endogenous ouabain and ouabain-like factors: The Na + pump/ouabain receptor, its linkage to NCX, and its myriad functions. Cell Calcium 2020; 86:102159. [PMID: 31986323 DOI: 10.1016/j.ceca.2020.102159] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 12/12/2022]
Abstract
In this brief review we discuss some aspects of the Na+ pump and its roles in mediating the effects of ouabain and endogenous ouabain (EO): i) in regulating the cytosolic Ca2+ concentration ([Ca2+]CYT) via Na/Ca exchange (NCX), and ii) in activating a number of protein kinase (PK) signaling cascades that control a myriad of cell functions. Importantly, [Ca2+]CYT and the other signaling pathways intersect at numerous points because of the influence of Ca2+ and calmodulin in modulating some steps in those other pathways. While both mechanisms operate in virtually all cells and tissues, this article focuses primarily on their functions in the cardiovascular system, the central nervous system (CNS) and the kidneys.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
57
|
Intraflagellar transport 20: New target for the treatment of ciliopathies. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118641. [PMID: 31893523 DOI: 10.1016/j.bbamcr.2019.118641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/17/2019] [Accepted: 12/25/2019] [Indexed: 11/22/2022]
Abstract
Cilia are ubiquitous in mammalian cells. The formation and assembly of cilia depend on the normal functioning of the ciliary transport system. In recent years, various proteins involved in the intracellular transport of the cilium have attracted attention, as many diseases are caused by disorders in cilia formation. Intraflagellar transport 20 (IFT20) is a subunit of IFT complex B, which contains approximately 20 protein particles. Studies have shown that defects in IFT20 are associated with numerous system -related diseases, such as those of the urinary system, cardiovascular system, skeletal system, nervous system, immune system, reproductive system, and respiratory system. This review summarizes current research on IFT20.We describe studies related to the role of IFT20 in cilia formation and discuss new targets for treating diseases associated with ciliary dysplasia.
Collapse
|
58
|
Hasan R, Leo MD, Muralidharan P, Mata-Daboin A, Yin W, Bulley S, Fernandez-Peña C, MacKay CE, Jaggar JH. SUMO1 modification of PKD2 channels regulates arterial contractility. Proc Natl Acad Sci U S A 2019; 116:27095-27104. [PMID: 31822608 PMCID: PMC6936352 DOI: 10.1073/pnas.1917264116] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PKD2 (polycystin-2, TRPP1) channels are expressed in a wide variety of cell types and can regulate functions, including cell division and contraction. Whether posttranslational modification of PKD2 modifies channel properties is unclear. Similarly uncertain are signaling mechanisms that regulate PKD2 channels in arterial smooth muscle cells (myocytes). Here, by studying inducible, cell-specific Pkd2 knockout mice, we discovered that PKD2 channels are modified by SUMO1 (small ubiquitin-like modifier 1) protein in myocytes of resistance-size arteries. At physiological intravascular pressures, PKD2 exists in approximately equal proportions as either nonsumoylated (PKD2) or triple SUMO1-modifed (SUMO-PKD2) proteins. SUMO-PKD2 recycles, whereas unmodified PKD2 is surface-resident. Intravascular pressure activates voltage-dependent Ca2+ influx that stimulates the return of internalized SUMO-PKD2 channels to the plasma membrane. In contrast, a reduction in intravascular pressure, membrane hyperpolarization, or inhibition of Ca2+ influx leads to lysosomal degradation of internalized SUMO-PKD2 protein, which reduces surface channel abundance. Through this sumoylation-dependent mechanism, intravascular pressure regulates the surface density of SUMO-PKD2-mediated Na+ currents (INa) in myocytes to control arterial contractility. We also demonstrate that intravascular pressure activates SUMO-PKD2, not PKD2, channels, as desumoylation leads to loss of INa activation in myocytes and vasodilation. In summary, this study reveals that PKD2 channels undergo posttranslational modification by SUMO1, which enables physiological regulation of their surface abundance and pressure-mediated activation in myocytes and thus control of arterial contractility.
Collapse
Affiliation(s)
- Raquibul Hasan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - M. Dennis Leo
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | | | - Alejandro Mata-Daboin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Wen Yin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Simon Bulley
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Carlos Fernandez-Peña
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Charles E. MacKay
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| |
Collapse
|
59
|
Brill AL, Ehrlich BE. Polycystin 2: A calcium channel, channel partner, and regulator of calcium homeostasis in ADPKD. Cell Signal 2019; 66:109490. [PMID: 31805375 DOI: 10.1016/j.cellsig.2019.109490] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/30/2019] [Accepted: 12/01/2019] [Indexed: 01/26/2023]
Abstract
Polycystin 2 (PC2) is one of two main protein types responsible for the underlying etiology of autosomal dominant polycystic kidney disease (ADPKD), the most prevalent monogenic renal disease in the world. This debilitating and currently incurable condition is caused by loss-of-function mutations in PKD2 and PKD1, the genes encoding for PC2 and Polycystin 1 (PC1), respectively. Two-hit mutation events in these genes lead to renal cyst formation and eventual kidney failure, the main hallmarks of ADPKD. Though much is known concerning the physiological consequences and dysfunctional signaling mechanisms resulting from ADPKD development, to best understand the requirement of PC2 in maintaining organ homeostasis, it is important to recognize how PC2 acts under normal conditions. As such, an array of work has been performed characterizing the endogenous function of PC2, revealing it to be a member of the transient receptor potential (TRP) channel family of proteins. As a TRP protein, PC2 is a nonselective, cation-permeant, calcium-sensitive channel expressed in all tissue types, where it localizes primarily on the endoplasmic reticulum (ER), primary cilia, and plasma membrane. In addition to its channel function, PC2 interacts with and acts as a regulator of a number of other channels, ultimately further affecting intracellular signaling and leading to dysfunction in its absence. In this review, we describe the biophysical and physiological properties of PC2 as a cation channel and modulator of intracellular calcium channels, along with how these properties are altered in ADPKD.
Collapse
Affiliation(s)
- Allison L Brill
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Barbara E Ehrlich
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA; Department of Pharmacology, Yale University, New Haven, CT, USA.
| |
Collapse
|
60
|
Abstract
Primary cilia project in a single copy from the surface of most vertebrate cell types; they detect and transmit extracellular cues to regulate diverse cellular processes during development and to maintain tissue homeostasis. The sensory capacity of primary cilia relies on the coordinated trafficking and temporal localization of specific receptors and associated signal transduction modules in the cilium. The canonical Hedgehog (HH) pathway, for example, is a bona fide ciliary signalling system that regulates cell fate and self-renewal in development and tissue homeostasis. Specific receptors and associated signal transduction proteins can also localize to primary cilia in a cell type-dependent manner; available evidence suggests that the ciliary constellation of these proteins can temporally change to allow the cell to adapt to specific developmental and homeostatic cues. Consistent with important roles for primary cilia in signalling, mutations that lead to their dysfunction underlie a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which affect many different tissues and organs of the body. In this Review, we highlight central mechanisms by which primary cilia coordinate HH, G protein-coupled receptor, WNT, receptor tyrosine kinase and transforming growth factor-β (TGFβ)/bone morphogenetic protein (BMP) signalling and illustrate how defects in the balanced output of ciliary signalling events are coupled to developmental disorders and disease progression.
Collapse
|
61
|
Structural insights into group II TRP channels. Cell Calcium 2019; 86:102107. [PMID: 31841954 DOI: 10.1016/j.ceca.2019.102107] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 01/01/2023]
Abstract
The seven members of the TRP channel superfamily are divided into two main groups with five members comprising group I (TRPC/V/M/N/A) and TRPML (TRP MucoLipin) and TRPP (TRP Polycystin) making up group II. Group II channels share a high sequence homology on their transmembrane domains and are distinct from group I members as they contain a large luminal/extracellular domain between transmembrane helix 1 (S1) and S2. Since 2016, there are more than ten research papers reporting various structures of group II channels by either cryo-EM or X-ray crystallography. These studies along with recent functional analysis by the other groups have considerably strengthened our knowledge on TRPML and TRPP channels. In this review, we summarize and discuss these reports providing molecular insights into the group II TRP channel family.
Collapse
|
62
|
Stayner C, Brooke DG, Bates M, Eccles MR. Targeted Therapies for Autosomal Dominant Polycystic Kidney Disease. Curr Med Chem 2019; 26:3081-3102. [PMID: 29737248 DOI: 10.2174/0929867325666180508095654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is the most common life-threatening genetic disease in humans, affecting approximately 1 in 500 people. ADPKD is characterized by cyst growth in the kidney leading to progressive parenchymal damage and is the underlying pathology in approximately 10% of patients requiring hemodialysis or transplantation for end-stage kidney disease. The two proteins that are mutated in ADPKD, polycystin-1 and polycystin-2, form a complex located on the primary cilium and the plasma membrane to facilitate calcium ion release in the cell. There is currently no Food and Drug Administration (FDA)-approved therapy to cure or slow the progression of the disease. Rodent ADPKD models do not completely mimic the human disease, and therefore preclinical results have not always successfully translated to the clinic. Moreover, the toxicity of many of these potential therapies has led to patient withdrawals from clinical trials. RESULTS Here, we review compounds in clinical trial for treating ADPKD, and we examine the feasibility of using a kidney-targeted approach, with potential for broadening the therapeutic window, decreasing treatment-associated toxicity and increasing the efficacy of agents that have demonstrated activity in animal models. We make recommendations for integrating kidney- targeted therapies with current treatment regimes, to achieve a combined approach to treating ADPKD. CONCLUSION Many compounds are currently in clinical trial for ADPKD yet, to date, none are FDA-approved for treating this disease. Patients could benefit from efficacious pharmacotherapy, especially if it can be kidney-targeted, and intensive efforts continue to be focused on this goal.
Collapse
Affiliation(s)
- Cherie Stayner
- Department of Pathology, Dunedin School of Medicine, University of Otago, 270 Great King Street, Dunedin 9054, New Zealand
| | - Darby G Brooke
- Cawthron Institute, 98 Halifax Street East, Nelson 7010, New Zealand
| | - Michael Bates
- Department of Pathology, Dunedin School of Medicine, University of Otago, 270 Great King Street, Dunedin 9054, New Zealand
| | - Michael R Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, 270 Great King Street, Dunedin 9054, New Zealand
| |
Collapse
|
63
|
Ciliary exclusion of Polycystin-2 promotes kidney cystogenesis in an autosomal dominant polycystic kidney disease model. Nat Commun 2019; 10:4072. [PMID: 31492868 PMCID: PMC6731238 DOI: 10.1038/s41467-019-12067-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 08/08/2019] [Indexed: 01/08/2023] Open
Abstract
The human PKD2 locus encodes Polycystin-2 (PC2), a TRPP channel that localises to several distinct cellular compartments, including the cilium. PKD2 mutations cause Autosomal Dominant Polycystic Kidney Disease (ADPKD) and affect many cellular pathways. Data underlining the importance of ciliary PC2 localisation in preventing PKD are limited because PC2 function is ablated throughout the cell in existing model systems. Here, we dissect the ciliary role of PC2 by analysing mice carrying a non-ciliary localising, yet channel-functional, PC2 mutation. Mutants develop embryonic renal cysts that appear indistinguishable from mice completely lacking PC2. Despite not entering the cilium in mutant cells, mutant PC2 accumulates at the ciliary base, forming a ring pattern consistent with distal appendage localisation. This suggests a two-step model of ciliary entry; PC2 first traffics to the cilium base before TOP domain dependent entry. Our results suggest that PC2 localisation to the cilium is necessary to prevent PKD. The molecular role of ciliary Polycystin-2 (PC2) in cyst formation and polycystic kidney disease (ADKPD) is unclear. Here, the authors identify a PC2 mutant lacking ciliary localisation but with active Ca2+ channel function in mice, that is sufficient to generate an ADPKD phenotype.
Collapse
|
64
|
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
|
65
|
Ma M, Legué E, Tian X, Somlo S, Liem KF. Cell-Autonomous Hedgehog Signaling Is Not Required for Cyst Formation in Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2019; 30:2103-2111. [PMID: 31451534 DOI: 10.1681/asn.2018121274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/15/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND PKD1 or PKD2, the two main causal genes for autosomal dominant polycystic kidney disease (ADPKD), encode the multipass transmembrane proteins polycystin-1 (PC1) and polycystin-2 (PC2), respectively. Polycystins localize to the primary cilium, an organelle essential for cell signaling, including signal transduction of the Hedgehog pathway. Mutations in ciliary genes that build and maintain the cilium also cause renal cystic disease through unknown pathways. Although recent studies have found alterations in Hedgehog signaling in ADPKD-related models and tissues, the relationship between Hedgehog and polycystic kidney disease is not known. METHODS To examine the potential role of cell-autonomous Hedgehog signaling in regulating kidney cyst formation in vivo in both early- and adult-onset mouse models of ADPKD, we used conditional inactivation of Pkd1 combined with conditional modulation of Hedgehog signaling components in renal epithelial cells, where mutations in Pkd1 initiate cyst formation. After increasing or decreasing levels of Hedgehog signaling in cells that underwent inactivation of Pkd1, we evaluated the effects of these genetic manipulations on quantitative parameters of polycystic kidney disease severity. RESULTS We found that in Pkd1 conditional mutant mouse kidneys, neither downregulation nor activation of the Hedgehog pathway in epithelial cells along the nephron significantly influenced the severity of the polycystic kidney phenotype in mouse models of developmental or adult-onset of ADPKD. CONCLUSIONS These data suggest that loss of Pkd1 function results in kidney cysts through pathways that are not affected by the activity of the Hedgehog pathway.
Collapse
Affiliation(s)
- Ming Ma
- Departments of Internal Medicine
| | - Emilie Legué
- Pediatrics, and.,Vertebrate Developmental Biology Program, Yale University, New Haven, Connecticut
| | - Xin Tian
- Departments of Internal Medicine
| | | | - Karel F Liem
- Pediatrics, and .,Vertebrate Developmental Biology Program, Yale University, New Haven, Connecticut
| |
Collapse
|
66
|
Sang D, Bai S, Yin S, Jiang S, Ye L, Hou W, Yao Y, Wang H, Shen Y, Shen B, Du J. Role of TRPP2 in mouse airway smooth muscle tension and respiration. Am J Physiol Lung Cell Mol Physiol 2019; 317:L466-L474. [PMID: 31411061 DOI: 10.1152/ajplung.00513.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The transient receptor potential polycystin-2 (TRPP2) is encoded by the Pkd2 gene, and mutation of this gene can cause autosomal dominant polycystic kidney disease (ADPKD). Some patients with ADPKD experience extrarenal manifestations, including radiologic and clinical bronchiectasis. We hypothesized that TRPP2 may regulate airway smooth muscle (ASM) tension. Thus, we used smooth muscle-Pkd2 conditional knockout (Pkd2SM-CKO) mice to investigate whether TRPP2 regulated ASM tension and whether TRPP2 deficiency contributed to bronchiectasis associated with ADPKD. Compared with wild-type mice, Pkd2SM-CKO mice breathed more shallowly and faster, and their cross-sectional area ratio of bronchi to accompanying pulmonary arteries was higher, suggesting that TRPP2 may regulate ASM tension and contribute to the occurrence of bronchiectasis in ADPKD. In a bioassay examining isolated tracheal ring tension, no significant difference was found for high-potassium-induced depolarization of the ASM between the two groups, indicating that TRPP2 does not regulate depolarization-induced ASM contraction. By contrast, carbachol-induced contraction of the ASM derived from Pkd2SM-CKO mice was significantly reduced compared with that in wild-type mice. In addition, relaxation of the carbachol-precontracted ASM by isoprenaline, a β-adrenergic receptor agonist that acts through the cAMP/adenylyl cyclase pathway, was also significantly attenuated in Pkd2SM-CKO mice compared with that in wild-type mice. Thus, TRPP2 deficiency suppressed both contraction and relaxation of the ASM. These results provide a potential target for regulating ASM tension and for developing therapeutic alternatives for some ADPKD complications of the respiratory system or for independent respiratory disease, especially bronchiectasis.
Collapse
Affiliation(s)
- Dacheng Sang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Department of Orthopedic Surgery, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Suwen Bai
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Sheng Yin
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Department of Neurosurgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Sen Jiang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Department of Neurosurgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Li Ye
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wenxuan Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yanheng Yao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Haoran Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yonggang Shen
- Nursing Faculty, Anhui Health College, Chizhou, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Juan Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
67
|
Saigusa T, Yue Q, Bunni MA, Bell PD, Eaton DC. Loss of primary cilia increases polycystin-2 and TRPV4 and the appearance of a nonselective cation channel in the mouse cortical collecting duct. Am J Physiol Renal Physiol 2019; 317:F632-F637. [PMID: 31313950 DOI: 10.1152/ajprenal.00210.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Flow-related bending of cilia results in Ca2+ influx through a polycystin-1 (Pkd1) and polycystin-2 (Pkd2) complex, both of which are members of the transient receptor potential (TRP) family (TRPP1 and TRPP2, respectively). Deletion of this complex as well as cilia result in polycystic kidney disease. The Ca2+ influx pathway has been previously characterized in immortalized collecting duct cells without cilia and found to be a 23-pS channel that was a multimere of TRPP2 and TRPV4. The purpose of the present study was to determine if this TRPP2 and TRPV4 multimere exists in vivo. Apical channel activity was measured using the patch-clamp technique from isolated split-open cortical collecting ducts from adult conditional knockout mice with (Ift88flox/flox) or without (Ift88-/-) cilia. Single tubules were isolated for measurements of mRNA for Pkd1, Pkd2, Trpv4, and epithelial Na+ channel subunits. The predominant channel activity from Ift88flox/flox mice was from epithelial Na+ channel [5-pS Na+-selective channels with long mean open times (475.7 ± 83.26 ms) and open probability > 0.2]. With the loss of cilia, the predominant conductance was a 23-pS nonselective cation channel (reversal potential near 0) with a short mean open time (72 ± 17 ms), open probability < 0.08, and a characteristic flickery opening. Loss of cilia increased mRNA levels for Pkd2 and Trpv4 from single isolated cortical collecting ducts. In conclusion, 23-pS channels exist in vivo, and activity of this channel is elevated with loss of cilia, consistent with previous finding of an elevated-unregulated Ca2+-permeable pathway at the apical membrane of collecting duct cells that lack cilia.
Collapse
Affiliation(s)
- Takamitsu Saigusa
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Qiang Yue
- Division of Nephrology, Department of Medicine, Emory University, Atlanta, Georgia
| | - Marlene A Bunni
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Douglas C Eaton
- Division of Nephrology, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
68
|
Winokurow N, Schumacher S. A role for polycystin-1 and polycystin-2 in neural progenitor cell differentiation. Cell Mol Life Sci 2019; 76:2851-2869. [PMID: 30895336 PMCID: PMC11105687 DOI: 10.1007/s00018-019-03072-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/17/2019] [Accepted: 03/14/2019] [Indexed: 10/27/2022]
Abstract
Polycystin-1 (PC1) and polycystin-2 (PC2) are transmembrane proteins encoded by the Pkd1 and Pkd2 genes, respectively. Mutations in these genes are causative for the development of autosomal-dominant polycystic kidney disease. A prominent feature of this disease is an unbalanced cell proliferation. PC1 and PC2 physically interact to form a complex, which localizes to the primary cilia of renal epithelial cells. Recently, PC1 and PC2 have also been described to be present in primary cilia of radial glial cells (RGCs) and to contribute to the planar cell polarity of late RGCs and E1 ependymal cells. As neural progenitor cells (NPCs), early RGCs have to balance proliferation for expansion, or for self-renewal and differentiation to generate neurons. It is not known whether the polycystins play a role in this process. Here, we show that PC1 and PC2 are expressed in RGCs of the developing mouse cerebral cortex during neurogenesis. Loss-of-function analysis and cell-based assays reveal that a reduction of PC1 or PC2 expression leads to increased NPC proliferation, while the differentiation to neurons becomes impaired. The increased NPC proliferation is preceded by enhanced Notch signaling and accompanied by a rise in the number of symmetric cell divisions. The transcription factor STAT3 seems to be mechanistically important for polycystin signaling in NPCs as either STAT3 knockdown or inhibition of STAT3 function abrogates the increased proliferation driven by reduced polycystin expression. Our findings indicate that PC1 and PC2 are critical for maintaining a balance between proliferation and differentiation of NPCs.
Collapse
Affiliation(s)
- Natalie Winokurow
- Institute of Molecular and Cellular Anatomy, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Stefan Schumacher
- Institute of Molecular and Cellular Anatomy, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
69
|
Valentine MS, Yano J, Van Houten J. A Novel Role for Polycystin-2 (Pkd2) in P. tetraurelia as a Probable Mg 2+ Channel Necessary for Mg 2+-Induced Behavior. Genes (Basel) 2019; 10:genes10060455. [PMID: 31207979 PMCID: PMC6627415 DOI: 10.3390/genes10060455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/05/2019] [Accepted: 06/11/2019] [Indexed: 01/26/2023] Open
Abstract
A human ciliopathy gene codes for Polycystin-2 (Pkd2), a non-selective cation channel. Here, the Pkd2 channel was explored in the ciliate Paramecium tetraurelia using combinations of RNA interference, over-expression, and epitope-tagging, in a search for function and novel interacting partners. Upon depletion of Pkd2, cells exhibited a phenotype similar to eccentric (XntA1), a Paramecium mutant lacking the inward Ca2+-dependent Mg2+ conductance. Further investigation showed both Pkd2 and XntA localize to the cilia and cell membrane, but do not require one another for trafficking. The XntA-myc protein co-immunoprecipitates Pkd2-FLAG, but not vice versa, suggesting two populations of Pkd2-FLAG, one of which interacts with XntA. Electrophysiology data showed that depletion and over-expression of Pkd2 led to smaller and larger depolarizations in Mg2+ solutions, respectively. Over-expression of Pkd2-FLAG in the XntA1 mutant caused slower swimming, supporting an increase in Mg2+ permeability, in agreement with the electrophysiology data. We propose that Pkd2 in P. tetraurelia collaborates with XntA for Mg2+-induced behavior. Our data suggest Pkd2 is sufficient and necessary for Mg2+ conductance and membrane permeability to Mg2+, and that Pkd2 is potentially a Mg2+-permeable channel.
Collapse
Affiliation(s)
- Megan S Valentine
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, NY 12901, USA.
| | - Junji Yano
- University of Vermont, Department of Biology, 120 Marsh Life Science, 109 Carrigan Drive, Burlington, VT 05405, USA.
| | - Judith Van Houten
- University of Vermont, Department of Biology, 120 Marsh Life Science, 109 Carrigan Drive, Burlington, VT 05405, USA.
| |
Collapse
|
70
|
Tajhya R, Delling M. New insights into ion channel-dependent signalling during left-right patterning. J Physiol 2019; 598:1741-1752. [PMID: 31106399 DOI: 10.1113/jp277835] [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: 02/13/2019] [Accepted: 04/23/2019] [Indexed: 01/20/2023] Open
Abstract
The left-right organizer (LRO) in the mouse consists of pit cells within the depression, located at the end of the developing notochord, also known as the embryonic node and crown cells lining the outer periphery of the node. Cilia on pit cells are posteriorly tilted, rotate clockwise and generate leftward fluid flow. Primary cilia on crown cells are required to interpret the directionality of fluid movement and initiate flow-dependent gene transcription. Crown cells express PC1-L1 and PC2, which may form a heteromeric polycystin channel complex on primary cilia. It is still only poorly understood how fluid flow activates the ciliary polycystin complex. Besides polycystin channels voltage gated channels like HCN4 and KCNQ1 have been implicated in establishing asymmetry. How this electrical network of ion channels initiates left-sided signalling cascades and differential gene expression is currently only poorly defined.
Collapse
Affiliation(s)
- Rajeev Tajhya
- Department of Physiology, University of California, 1550 4th Street, San Francisco, CA, 94518, USA
| | - Markus Delling
- Department of Physiology, University of California, 1550 4th Street, San Francisco, CA, 94518, USA
| |
Collapse
|
71
|
Andries A, Daenen K, Jouret F, Bammens B, Mekahli D, Van Schepdael A. Oxidative stress in autosomal dominant polycystic kidney disease: player and/or early predictor for disease progression? Pediatr Nephrol 2019; 34:993-1008. [PMID: 30105413 DOI: 10.1007/s00467-018-4004-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/19/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), caused by mutations in PKD1 or PKD2 genes, is the most common hereditary renal disease. Renal manifestations of ADPKD are gradual cyst development and kidney enlargement ultimately leading to end-stage renal disease. ADPKD also causes extrarenal manifestations, including endothelial dysfunction and hypertension. Both of these complications are linked with reduced nitric oxide levels related to excessive oxidative stress (OS). OS, defined as disturbances in the prooxidant/antioxidant balance, is harmful to cells due to the excessive generation of highly reactive oxygen and nitrogen free radicals. Next to endothelial dysfunction and hypertension, there is cumulative evidence that OS occurs in the early stages of ADPKD. In the current review, we aim to summarize the cardiovascular complications and the relevance of OS in ADPKD and, more specifically, in the early stages of the disease. First, we will briefly introduce the link between ADPKD and the early cardiovascular complications including hypertension. Secondly, we will describe the potential role of OS in the early stages of ADPKD and its possible importance beyond the chronic kidney disease (CKD) effect. Finally, we will discuss some pharmacological agents capable of reducing reactive oxygen species and OS, which might represent potential treatment targets for ADPKD.
Collapse
Affiliation(s)
- Asmin Andries
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
| | - Kristien Daenen
- Department of Microbiology and Immunology, Laboratory of Nephrology, KU Leuven - University of Leuven, 3000, Leuven, Belgium.,Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, 3000, Leuven, Belgium
| | - François Jouret
- Department of Internal Medicine, Division of Nephrology, University of Liège Hospital (ULg CHU), Liège, Belgium.,Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Science, University of Liège, Liège, Belgium
| | - Bert Bammens
- Department of Microbiology and Immunology, Laboratory of Nephrology, KU Leuven - University of Leuven, 3000, Leuven, Belgium.,Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Djalila Mekahli
- Department of Development and Regeneration, Laboratory of Pediatrics, PKD Group, KU Leuven - University of Leuven, 3000, Leuven, Belgium.,Department of Pediatric Nephrology, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Ann Van Schepdael
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, KU Leuven - University of Leuven, 3000, Leuven, Belgium
| |
Collapse
|
72
|
Parnell SC, Magenheimer BS, Maser RL, Pavlov TS, Havens MA, Hastings ML, Jackson SF, Ward CJ, Peterson KR, Staruschenko A, Calvet JP. A mutation affecting polycystin-1 mediated heterotrimeric G-protein signaling causes PKD. Hum Mol Genet 2019; 27:3313-3324. [PMID: 29931260 PMCID: PMC6140781 DOI: 10.1093/hmg/ddy223] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/05/2018] [Indexed: 12/16/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the growth of renal cysts that ultimately destroy kidney function. Mutations in the PKD1 and PKD2 genes cause ADPKD. Their protein products, polycystin-1 (PC1) and polycystin-2 (PC2) have been proposed to form a calcium-permeable receptor-channel complex; however the mechanisms by which they function are almost completely unknown. Most mutations in PKD1 are truncating loss-of-function mutations or affect protein biogenesis, trafficking or stability and reveal very little about the intrinsic biochemical properties or cellular functions of PC1. An ADPKD patient mutation (L4132Δ or ΔL), resulting in a single amino acid deletion in a putative G-protein binding region of the PC1 C-terminal cytosolic tail, was found to significantly decrease PC1-stimulated, G-protein-dependent signaling in transient transfection assays. Pkd1ΔL/ΔL mice were embryo-lethal suggesting that ΔL is a functionally null mutation. Kidney-specific Pkd1ΔL/cond mice were born but developed severe, postnatal cystic disease. PC1ΔL protein expression levels and maturation were comparable to those of wild type PC1, and PC1ΔL protein showed cell surface localization. Expression of PC1ΔL and PC2 complexes in transfected CHO cells failed to support PC2 channel activity, suggesting that the role of PC1 is to activate G-protein signaling to regulate the PC1/PC2 calcium channel.
Collapse
Affiliation(s)
- Stephen C Parnell
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brenda S Magenheimer
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Robin L Maser
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tengis S Pavlov
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, MI, USA
| | | | - Michelle L Hastings
- Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Stephen F Jackson
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Christopher J Ward
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kenneth R Peterson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - James P Calvet
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
73
|
Kim K, Trott JF, Gao G, Chapman A, Weiss RH. Plasma metabolites and lipids associate with kidney function and kidney volume in hypertensive ADPKD patients early in the disease course. BMC Nephrol 2019; 20:66. [PMID: 30803434 PMCID: PMC6388487 DOI: 10.1186/s12882-019-1249-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/06/2019] [Indexed: 01/09/2023] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disease and is characterized by gradual cyst growth and expansion, increase in kidney volume with an ultimate decline in kidney function leading to end stage renal disease (ESRD). Given the decades long period of stable kidney function while cyst growth occurs, it is important to identify those patients who will progress to ESRD. Recent data from our and other laboratories have demonstrated that metabolic reprogramming may play a key role in cystic epithelial proliferation resulting in cyst growth in ADPKD. Height corrected total kidney volume (ht-TKV) accurately reflects cyst burden and predicts future loss of kidney function. We hypothesize that specific plasma metabolites will correlate with eGFR and ht-TKV early in ADPKD, both predictors of disease progression, potentially indicative of early physiologic derangements of renal disease severity. Methods To investigate the predictive role of plasma metabolites on eGFR and/or ht-TKV, we used a non-targeted GC-TOF/MS-based metabolomics approach on hypertensive ADPKD patients in the early course of their disease. Patient data was obtained from the HALT-A randomized clinical trial at baseline including estimated glomerular filtration rate (eGFR) and measured ht-TKV. To identify individual metabolites whose intensities are significantly correlated with eGFR and ht-TKV, association analyses were performed using linear regression with each metabolite signal level as the primary predictor variable and baseline eGFR and ht-TKV as the continuous outcomes of interest, while adjusting for covariates. Significance was determined by Storey’s false discovery rate (FDR) q-values to correct for multiple testing. Results Twelve metabolites significantly correlated with eGFR and two triglycerides significantly correlated with baseline ht-TKV at FDR q-value < 0.05. Specific significant metabolites, including pseudo-uridine, indole-3-lactate, uric acid, isothreonic acid, and creatinine, have been previously shown to accumulate in plasma and/or urine in both diabetic and cystic renal diseases with advanced renal insufficiency. Conclusions This study identifies metabolic derangements in early ADPKD which may be prognostic for ADPKD disease progression. Clinical trial HALT Progression of Polycystic Kidney Disease (HALT PKD) Study A; Clinical www.clinicaltrials.gov identifier: NCT00283686; first posted January 30, 2006, last update posted March 19, 2015. Electronic supplementary material The online version of this article (10.1186/s12882-019-1249-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kyoungmi Kim
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Josephine F Trott
- Division of Nephrology, Department of Internal Medicine, University of California, Genome and Biomedical Sciences Building, Room 6311, 451 Health Sciences Dr, Davis, CA, 95616, USA
| | - Guimin Gao
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Arlene Chapman
- Nephrology Section, University of Chicago, Chicago, IL, USA
| | - Robert H Weiss
- Division of Nephrology, Department of Internal Medicine, University of California, Genome and Biomedical Sciences Building, Room 6311, 451 Health Sciences Dr, Davis, CA, 95616, USA. .,Cancer Center, University of California, Davis, CA, USA. .,Medical Service, VA Northern California Health Care System, Sacramento, CA, USA.
| |
Collapse
|
74
|
Hwang SH, Somatilaka BN, Badgandi H, Palicharla VR, Walker R, Shelton JM, Qian F, Mukhopadhyay S. Tulp3 Regulates Renal Cystogenesis by Trafficking of Cystoproteins to Cilia. Curr Biol 2019; 29:790-802.e5. [PMID: 30799239 DOI: 10.1016/j.cub.2019.01.047] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/09/2018] [Accepted: 01/17/2019] [Indexed: 12/25/2022]
Abstract
Polycystic kidney disease proteins, polycystin-1 and polycystin-2, localize to primary cilia. Polycystin knockouts have severe cystogenesis compared to ciliary disruption, whereas simultaneous ciliary loss suppresses excessive cyst growth. These data suggest the presence of a cystogenic activator that is inhibited by polycystins and an independent but relatively minor cystogenic inhibitor, either of which are cilia dependent. However, current genetic models targeting cilia completely ablate the compartment, making it difficult to uncouple cystoprotein function from ciliary localization. Thus, the role of cilium-generated signaling in cystogenesis is unclear. We recently demonstrated that the tubby family protein Tulp3 determines ciliary trafficking of polycystins in kidney collecting duct cells without affecting protein levels or cilia. Here, we demonstrate that embryonic-stage, nephron-specific Tulp3 knockout mice developed cystic kidneys, while retaining intact cilia. Cystic kidneys showed increased mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), mTOR, and persistently high cyclic AMP (cAMP) signaling, suggesting contribution of multiple factors to cystogenesis. Based on kidney-to-body-weight ratio, cystic index, and epithelial proliferation in developing tubules or cysts, the severity of cystogenesis upon Tulp3 deletion was intermediate between that caused by loss of polycystin-1 or cilia. However, concomitant Tulp3 loss did not inhibit cystogenesis in polycystin-1 knockouts, unlike ciliary disruption. Interestingly, ciliary trafficking of the small guanosine triphosphatase (GTPase) Arl13b, loss of which causes cystogenic severity similar to ciliary loss, was reduced prior to cyst initiation. Thus, we propose that cystogenesis in Tulp3 mutants results from a reduction of ciliary levels of polycystins, Arl13b, and Arl13b-dependent lipidated cargoes. Arl13b might be the ciliary factor that represses cystogenesis distinct from polycystins.
Collapse
Affiliation(s)
- Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Bandarigoda N Somatilaka
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Hemant Badgandi
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rebecca Walker
- Division of Nephrology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - John M Shelton
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Feng Qian
- Division of Nephrology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
75
|
Woodward OM, Watnick T. Molecular Structure of the PKD Protein Complex Finally Solved. Am J Kidney Dis 2019; 73:620-623. [PMID: 30704879 DOI: 10.1053/j.ajkd.2018.12.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 12/20/2018] [Indexed: 01/16/2023]
Affiliation(s)
- Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD.
| | - Terry Watnick
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD.
| |
Collapse
|
76
|
Meng J, Xu Y, Shen X, Liang C. A novel frameshift PKD1 mutation in a Chinese patient with autosomal dominant polycystic kidney disease and azoospermia: A case report. Exp Ther Med 2019; 17:507-511. [PMID: 30651829 DOI: 10.3892/etm.2018.6946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 10/03/2018] [Indexed: 11/05/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is primarily caused by mutations in polycystin 1, transient receptor potential channel interacting (PKD1) and PKD2, and characterized by numerous cysts in various organs, primarily the kidneys and liver. The present case report is on a 33-year-old Chinese male patient who suffered from abdominal pain and hypertension, and presented with long-term infertility. Laboratory tests indicated that the patient had a normal renal function, while abdominal computed tomography demonstrated that the patient had enlarged kidneys with a volume of 1,127.21 cm3. In a semen analysis, no sperm was detected, while a subsequent testicular biopsy analysis demonstrated numerous mature sperms with progressive motility which suggests that the cysts of the epididymis and the dilated seminal vesicles may have obstructed the ejaculation of semen. Genetic testing identified that a novel missense mutation (c.9053delT) that was responsible for the disease. ADPKD has various disease severities, which depend on whether there is a PKD1 or PKD2 mutation and whether the mutation impairs the function of the polycystin protein. Therefore, genetic testing is important for the clinical diagnosis and prognosis of ADPKD patients, as well as prenatal diagnosis.
Collapse
Affiliation(s)
- Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yuchen Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xufeng Shen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
77
|
Merrick D, Mistry K, Wu J, Gresko N, Baggs JE, Hogenesch JB, Sun Z, Caplan MJ. Polycystin-1 regulates bone development through an interaction with the transcriptional coactivator TAZ. Hum Mol Genet 2019; 28:16-30. [PMID: 30215740 PMCID: PMC6298236 DOI: 10.1093/hmg/ddy322] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/03/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023] Open
Abstract
Polycystin-1 (PC1), encoded by the PKD1 gene that is mutated in the autosomal dominant polycystic kidney disease, regulates a number of processes including bone development. Activity of the transcription factor RunX2, which controls osteoblast differentiation, is reduced in Pkd1 mutant mice but the mechanism governing PC1 activation of RunX2 is unclear. PC1 undergoes regulated cleavage that releases its C-terminal tail (CTT), which translocates to the nucleus to modulate transcriptional pathways involved in proliferation and apoptosis. We find that the cleaved CTT of PC1 (PC1-CTT) stimulates the transcriptional coactivator TAZ (Wwtr1), an essential coactivator of RunX2. PC1-CTT physically interacts with TAZ, stimulating RunX2 transcriptional activity in pre-osteoblast cells in a TAZ-dependent manner. The PC1-CTT increases the interaction between TAZ and RunX2 and enhances the recruitment of the p300 transcriptional co-regulatory protein to the TAZ/RunX2/PC1-CTT complex. Zebrafish injected with morpholinos directed against pkd1 manifest severe bone calcification defects and a curly tail phenotype. Injection of messenger RNA (mRNA) encoding the PC1-CTT into pkd1-morphant fish restores bone mineralization and reduces the severity of the curly tail phenotype. These effects are abolished by co-injection of morpholinos directed against TAZ. Injection of mRNA encoding a dominant-active TAZ construct is sufficient to rescue both the curly tail phenotype and the skeletal defects observed in pkd1-morpholino treated fish. Thus, TAZ constitutes a key mechanistic link through which PC1 mediates its physiological functions.
Collapse
Affiliation(s)
- David Merrick
- Department of Cellular and Molecular Physiology, New Haven, CT USA
- Department of Cell Biology, Norcross, GA USA
| | - Kavita Mistry
- Department of Cellular and Molecular Physiology, New Haven, CT USA
| | - Jingshing Wu
- Department of Cellular and Molecular Physiology, New Haven, CT USA
| | - Nikolay Gresko
- Department of Cellular and Molecular Physiology, New Haven, CT USA
| | | | - John B Hogenesch
- Divisions of Perinatal Biology and Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH USA
| | - Zhaoxia Sun
- Department of Genetics, Yale University School of Medicine, New Haven, CT USA
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, New Haven, CT USA
- Department of Cell Biology, Norcross, GA USA
| |
Collapse
|
78
|
Prasad H, Dang DK, Kondapalli KC, Natarajan N, Cebotaru V, Rao R. NHA2 promotes cyst development in an in vitro model of polycystic kidney disease. J Physiol 2019; 597:499-519. [PMID: 30242840 PMCID: PMC6332743 DOI: 10.1113/jp276796] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/31/2018] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Significant and selective up-regulation of the Na+ /H+ exchanger NHA2 (SLC9B2) was observed in cysts of patients with autosomal dominant polycystic kidney disease. Using the MDCK cell model of cystogenesis, it was found that NHA2 increases cyst size. Silencing or pharmacological inhibition of NHA2 inhibits cyst formation in vitro. Polycystin-1 represses NHA2 expression via Ca2+ /NFAT signalling whereas the dominant negative membrane-anchored C-terminal fragment (PC1-MAT) increased NHA2 levels. Drugs (caffeine, theophylline) and hormones (vasopressin, aldosterone) known to exacerbate cysts elicit NHA2 expression. Taken together, the findings reveal NHA2 as a potential new player in salt and water homeostasis in the kidney and in the pathogenesis of polycystic kidney disease. ABSTRACT Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 and PKD2 encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively. The molecular pathways linking polycystins to cyst development in ADPKD are still unclear. Intracystic fluid secretion via ion transporters and channels plays a crucial role in cyst expansion in ADPKD. Unexpectedly, we observed significant and selective up-regulation of NHA2, a member of the SLC9B family of Na+ /H+ exchangers, that correlated with cyst size and disease severity in ADPKD patients. Using three-dimensional cultures of MDCK cells to model cystogenesis in vitro, we showed that ectopic expression of NHA2 is causal to increased cyst size. Induction of PC1 in MDCK cells inhibited NHA2 expression with concordant inhibition of Ca2+ influx through store-dependent and -independent pathways, whereas reciprocal activation of Ca2+ influx by the dominant negative membrane-anchored C-terminal tail fragment of PC1 elevated NHA2. We showed that NHA2 is a target of Ca2+ /NFAT signalling and is transcriptionally induced by methylxanthine drugs such as caffeine and theophylline, which are contraindicated in ADPKD patients. Finally, we observed robust induction of NHA2 by vasopressin, which is physiologically consistent with increased levels of circulating vasopressin and up-regulation of vasopressin V2 receptors in ADPKD. Our findings have mechanistic implications on the emerging use of vasopressin V2 receptor antagonists such as tolvaptan as safe and effective therapy for polycystic kidney disease and reveal a potential new regulator of transepithelial salt and water transport in the kidney.
Collapse
Affiliation(s)
- Hari Prasad
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Donna K. Dang
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Kalyan C. Kondapalli
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Niranjana Natarajan
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Valeriu Cebotaru
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Rajini Rao
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
79
|
Bulley S, Fernández-Peña C, Hasan R, Leo MD, Muralidharan P, Mackay CE, Evanson KW, Moreira-Junior L, Mata-Daboin A, Burris SK, Wang Q, Kuruvilla KP, Jaggar JH. Arterial smooth muscle cell PKD2 (TRPP1) channels regulate systemic blood pressure. eLife 2018; 7:42628. [PMID: 30511640 PMCID: PMC6281320 DOI: 10.7554/elife.42628] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/22/2018] [Indexed: 01/12/2023] Open
Abstract
Systemic blood pressure is determined, in part, by arterial smooth muscle cells (myocytes). Several Transient Receptor Potential (TRP) channels are proposed to be expressed in arterial myocytes, but it is unclear if these proteins control physiological blood pressure and contribute to hypertension in vivo. We generated the first inducible, smooth muscle-specific knockout mice for a TRP channel, namely for PKD2 (TRPP1), to investigate arterial myocyte and blood pressure regulation by this protein. Using this model, we show that intravascular pressure and α1-adrenoceptors activate PKD2 channels in arterial myocytes of different systemic organs. PKD2 channel activation in arterial myocytes leads to an inward Na+ current, membrane depolarization and vasoconstriction. Inducible, smooth muscle cell-specific PKD2 knockout lowers both physiological blood pressure and hypertension and prevents pathological arterial remodeling during hypertension. Thus, arterial myocyte PKD2 controls systemic blood pressure and targeting this TRP channel reduces high blood pressure.
Collapse
Affiliation(s)
- Simon Bulley
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Carlos Fernández-Peña
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Raquibul Hasan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - M Dennis Leo
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Padmapriya Muralidharan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Charles E Mackay
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Kirk W Evanson
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Luiz Moreira-Junior
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Alejandro Mata-Daboin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Sarah K Burris
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Qian Wang
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Korah P Kuruvilla
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Jonathan H Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| |
Collapse
|
80
|
Lea WA, Parnell SC, Wallace DP, Calvet JP, Zelenchuk LV, Alvarez NS, Ward CJ. Human-Specific Abnormal Alternative Splicing of Wild-Type PKD1 Induces Premature Termination of Polycystin-1. J Am Soc Nephrol 2018; 29:2482-2492. [PMID: 30185468 DOI: 10.1681/asn.2018040442] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/06/2018] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The major form of autosomal dominant polycystic kidney disease is caused by heterozygous mutations in PKD1, the gene that encodes polycystin-1 (PC1). Unlike PKD1 genes in the mouse and most other mammals, human PKD1 is unusual in that it contains two long polypyrimidine tracts in introns 21 and 22 (2.5 kbp and 602 bp, respectively; 97% cytosine and thymine). Although these polypyrimidine tracts have been shown to form thermodynamically stable segments of triplex DNA that can cause DNA polymerase stalling and enhance the local mutation rate, the efficiency of transcription and splicing across these cytosine- and thymine-rich introns has been unexplored. METHODS We used RT-PCR and Western blotting (using an mAb to the N terminus) to probe splicing events over exons 20-24 in the mouse and human PKD1 genes as well as Nanopore sequencing to confirm the presence of multiple splice forms. RESULTS Analysis of PC1 indicates that humans, but not mice, have a smaller than expected protein product, which we call Trunc_PC1. The findings show that Trunc_PC1 is the protein product of abnormal differential splicing across introns 21 and 22 and that 28.8%-61.5% of PKD1 transcripts terminate early. CONCLUSIONS The presence of polypyrimidine tracts decreases levels of full-length PKD1 mRNA from normal alleles. In heterozygous individuals, low levels of full-length PC1 may reduce polycystin signaling below a critical "cystogenic" threshold.
Collapse
Affiliation(s)
- Wendy A Lea
- The Jared Grantham Kidney Institute and Departments of.,Internal Medicine
| | - Stephen C Parnell
- The Jared Grantham Kidney Institute and Departments of.,Biochemistry and Molecular Biology
| | - Darren P Wallace
- The Jared Grantham Kidney Institute and Departments of.,Internal Medicine.,Molecular and Integrative Physiology, and
| | - James P Calvet
- The Jared Grantham Kidney Institute and Departments of.,Biochemistry and Molecular Biology
| | - Lesya V Zelenchuk
- The Jared Grantham Kidney Institute and Departments of.,Internal Medicine
| | - Nehemiah S Alvarez
- Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas; and.,De Novo Genomics, Kansas City, Kansas
| | - Christopher J Ward
- The Jared Grantham Kidney Institute and Departments of .,Internal Medicine.,Biochemistry and Molecular Biology
| |
Collapse
|
81
|
Di Mise A, Ranieri M, Centrone M, Venneri M, Tamma G, Valenti D, Valenti G. Activation of the Calcium-Sensing Receptor Corrects the Impaired Mitochondrial Energy Status Observed in Renal Polycystin-1 Knockdown Cells Modeling Autosomal Dominant Polycystic Kidney Disease. Front Mol Biosci 2018; 5:77. [PMID: 30197885 PMCID: PMC6117232 DOI: 10.3389/fmolb.2018.00077] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/03/2018] [Indexed: 12/19/2022] Open
Abstract
Autosomal Dominant Polycistic kidney Disease (ADPKD) is a renal channelopathy due to loss-of-function mutations in the PKD1 or PKD2 genes, encoding polycystin-1 (PC1) or polycystin-2 (PC2), respectively. PC1 is a large protein found predominantly on the plasma membrane where interacts with different proteins, including PC2. PC2 is a smaller integral membrane protein also expressed in intracellular organelles, acting as a non-selective cation channel permeable to calcium. Both PC1 and PC2 are also localized to the primary cilium of renal epithelial cells serving as mechanosensor that controls calcium influx through the plasma membrane and regulates intracellular calcium release from the endoplasmic reticulum. The mechanisms by which PC1/2 dysfunction leads to ADPKD needs still to be clarified. We have recently reported that selective Calcium-Sensing Receptor (CaSR) activation in human conditionally immortalized Proximal Tubular Epithelial cells deficient for PC1 (ciPTEC-PC1KD), deriving from urine sediments reduces intracellular cAMP and mTOR activity, and increases intracellular calcium reversing the principal ADPKD dysregulations. Reduced cellular free calcium found in ADPKD can, on the other hand, affect mitochondrial function and ATP production and, interestingly, a relationship between mitochondria and renal polycystic diseases have been suggested. By using ciPTEC-PC1KD as experimental tool modeling of ADPKD, we show here that, compared with wild type cells, ciPTEC-PC1KD have significantly lower mitochondrial calcium levels associated with a severe deficit in mitochondrial ATP production, secondary to a multilevel impairment of oxidative phosphorylation. Notably, selective CaSR activation with the calcimimetic NPS-R568 increases mitochondrial calcium content close to the levels found in resting wild type cells, and fully recovers the cell energy deficit associated to the PC1 channel disruption. Treatment of ciPTEC-PC1KD with 2-APB, an IP3R inhibitor, prevented the rescue of bioenergetics deficit induced by CaSR activation supporting a critical role of IP3Rs in driving ER-to-mitochondria Ca2+ shuttle. Together these data indicate that, besides reversing the principal dysregulations considered the most proximal events in ADPKD pathogenesis, selective CaSR activation in PKD1 deficient cells restores altered mitochondrial function that, in ADPKD, is known to facilitate cyst formation. These findings identify CaSR as a potential therapeutic target.
Collapse
Affiliation(s)
- Annarita Di Mise
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Marianna Ranieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Mariangela Centrone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Maria Venneri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy.,Istituto Nazionale di Biostrutture e Biosistemi, Rome, Italy
| | - Daniela Valenti
- Institute of Biomembranes Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy.,Istituto Nazionale di Biostrutture e Biosistemi, Rome, Italy.,Center of Excellence in Comparative Genomics, University of Bari, Bari, Italy
| |
Collapse
|
82
|
Su Q, Hu F, Ge X, Lei J, Yu S, Wang T, Zhou Q, Mei C, Shi Y. Structure of the human PKD1-PKD2 complex. Science 2018; 361:science.aat9819. [DOI: 10.1126/science.aat9819] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022]
Abstract
Mutations in two genes, PKD1 and PKD2, account for most cases of autosomal dominant polycystic kidney disease, one of the most common monogenetic disorders. Here we report the 3.6-angstrom cryo–electron microscopy structure of truncated human PKD1-PKD2 complex assembled in a 1:3 ratio. PKD1 contains a voltage-gated ion channel (VGIC) fold that interacts with PKD2 to form the domain-swapped, yet noncanonical, transient receptor potential (TRP) channel architecture. The S6 helix in PKD1 is broken in the middle, with the extracellular half, S6a, resembling pore helix 1 in a typical TRP channel. Three positively charged, cavity-facing residues on S6b may block cation permeation. In addition to the VGIC, a five–transmembrane helix domain and a cytosolic PLAT domain were resolved in PKD1. The PKD1-PKD2 complex structure establishes a framework for dissecting the function and disease mechanisms of the PKD proteins.
Collapse
|
83
|
Morthorst SK, Christensen ST, Pedersen LB. Regulation of ciliary membrane protein trafficking and signalling by kinesin motor proteins. FEBS J 2018; 285:4535-4564. [PMID: 29894023 DOI: 10.1111/febs.14583] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/09/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022]
Abstract
Primary cilia are antenna-like sensory organelles that regulate a substantial number of cellular signalling pathways in vertebrates, both during embryonic development as well as in adulthood, and mutations in genes coding for ciliary proteins are causative of an expanding group of pleiotropic diseases known as ciliopathies. Cilia consist of a microtubule-based axoneme core, which is subtended by a basal body and covered by a bilayer lipid membrane of unique protein and lipid composition. Cilia are dynamic organelles, and the ability of cells to regulate ciliary protein and lipid content in response to specific cellular and environmental cues is crucial for balancing ciliary signalling output. Here we discuss mechanisms involved in regulation of ciliary membrane protein trafficking and signalling, with main focus on kinesin-2 and kinesin-3 family members.
Collapse
|
84
|
Zheng W, Yang X, Hu R, Cai R, Hofmann L, Wang Z, Hu Q, Liu X, Bulkley D, Yu Y, Tang J, Flockerzi V, Cao Y, Cao E, Chen XZ. Hydrophobic pore gates regulate ion permeation in polycystic kidney disease 2 and 2L1 channels. Nat Commun 2018; 9:2302. [PMID: 29899465 PMCID: PMC5998024 DOI: 10.1038/s41467-018-04586-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/01/2018] [Indexed: 01/20/2023] Open
Abstract
PKD2 and PKD1 genes are mutated in human autosomal dominant polycystic kidney disease. PKD2 can form either a homomeric cation channel or a heteromeric complex with the PKD1 receptor, presumed to respond to ligand(s) and/or mechanical stimuli. Here, we identify a two-residue hydrophobic gate in PKD2L1, and a single-residue hydrophobic gate in PKD2. We find that a PKD2 gain-of-function gate mutant effectively rescues PKD2 knockdown-induced phenotypes in embryonic zebrafish. The structure of a PKD2 activating mutant F604P by cryo-electron microscopy reveals a π- to α-helix transition within the pore-lining helix S6 that leads to repositioning of the gate residue and channel activation. Overall the results identify hydrophobic gates and a gating mechanism of PKD2 and PKD2L1. Mutations in the cation channel PKD2 cause human autosomal dominant polycystic kidney disease but its channel function and gating mechanism are poorly understood. Here authors study PKD2 using electrophysiology and cryo-EM, which identifies hydrophobic gates and proposes a gating mechanism for PKD2.
Collapse
Affiliation(s)
- Wang Zheng
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.,Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Xiaoyong Yang
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Ruikun Hu
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Ruiqi Cai
- Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Laura Hofmann
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, 66421, Germany
| | - Zhifei Wang
- Department of Biological Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qiaolin Hu
- Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Xiong Liu
- Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - David Bulkley
- Keck Advanced Microscopy Laboratory and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Yong Yu
- Department of Biological Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, 66421, Germany
| | - Ying Cao
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA.
| | - Xing-Zhen Chen
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China. .,Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
85
|
Abstract
Mutations in PKD1 and PKD2 are the leading cause of autosomal dominant polycystic kidney disease (ADPKD). In this issue of Genes & Development, a report by Cai and colleagues (pp. 781-793) reveals new insight into the molecular basis by which PKD1 deficiency leads to cystic kidney pathogenesis. By using extensive mouse genetic analyses coupled with in vitro cystic assays, the investigators delineate a RhoA-YAP-c-Myc signaling axis as a key downstream from PKD1 deficiency in ADPKD pathogenesis. Their findings provide evidence that the Hippo pathway could be a potential target for treating ADPKD.
Collapse
Affiliation(s)
- Shenghong Ma
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| | - Kun-Liang Guan
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
86
|
Katsianou MA, Skondra FG, Gargalionis AN, Piperi C, Basdra EK. The role of transient receptor potential polycystin channels in bone diseases. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:246. [PMID: 30069448 DOI: 10.21037/atm.2018.04.10] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Transient receptor potential (TRP) channels are cation channels which act as molecular sensors that enable cells to detect and respond to a plethora of mechanical and environmental cues. TRPs are involved in various physiological processes, such as mechanosensation, non-inception and thermosensation, while mutations in genes encoding them can lead to pathological conditions, called "channelopathies". The subfamily of transient receptor potential polycystins (TRPPs), Polycystin 1 (PC1, TRPP1) and Polycystin 2 (PC2, TRPP2), act as mechanoreceptors, sensing external mechanical forces, including strain, stretch and fluid shear stress, triggering a cascade of signaling pathways involved in osteoblastogenesis and ultimately bone formation. Both in vitro studies and research on animal models have already identified their implications in bone homeostasis. However, uncertainty veiling the role of polycystins (PCs) in bone disease urges studies to elucidate further their role in this field. Mutations in TRPPs have been related to autosomal polycystic kidney disease (ADKPD) and research groups try to identify their role beyond their well-established contribution in kidney disease. Such an elucidation would be beneficial for identifying signaling pathways where polycystins are involved in bone diseases related to exertion of mechanical forces such as osteoporosis, osteopenia and craniosynostosis. A better understanding of the implications of TRPPs in bone diseases would possibly lay the cornerstone for effective therapeutic schemes.
Collapse
Affiliation(s)
- Maria A Katsianou
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Foteini G Skondra
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios N Gargalionis
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthimia K Basdra
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
87
|
Modeling Renal Disease "On the Fly". BIOMED RESEARCH INTERNATIONAL 2018; 2018:5697436. [PMID: 29955604 PMCID: PMC6000847 DOI: 10.1155/2018/5697436] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/17/2018] [Indexed: 12/22/2022]
Abstract
Detoxification is a fundamental function for all living organisms that need to excrete catabolites and toxins to maintain homeostasis. Kidneys are major organs of detoxification that maintain water and electrolyte balance to preserve physiological functions of vertebrates. In insects, the renal function is carried out by Malpighian tubules and nephrocytes. Due to differences in their circulation, the renal systems of mammalians and insects differ in their functional modalities, yet carry out similar biochemical and physiological functions and share extensive genetic and molecular similarities. Evolutionary conservation can be leveraged to model specific aspects of the complex mammalian kidney function in the genetic powerhouse Drosophila melanogaster to study how genes interact in diseased states. Here, we compare the human and Drosophila renal systems and present selected fly disease models.
Collapse
|
88
|
Aboudehen K, Farahani S, Kanchwala M, Chan SC, Avdulov S, Mickelson A, Lee D, Gearhart MD, Patel V, Xing C, Igarashi P. Long noncoding RNA Hoxb3os is dysregulated in autosomal dominant polycystic kidney disease and regulates mTOR signaling. J Biol Chem 2018; 293:9388-9398. [PMID: 29716997 DOI: 10.1074/jbc.ra118.001723] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a debilitating disease that is characterized by the accumulation of numerous fluid-filled cysts in the kidney. ADPKD is primarily caused by mutations in two genes, PKD1 and PKD2 Long noncoding RNAs (lncRNA), defined by a length >200 nucleotides and absence of a long ORF, have recently emerged as epigenetic regulators of development and disease; however, their involvement in PKD has not been explored previously. Here, we performed deep RNA-Seq to identify lncRNAs that are dysregulated in two orthologous mouse models of ADPKD (kidney-specific Pkd1 and Pkd2 mutant mice). We identified a kidney-specific, evolutionarily conserved lncRNA called Hoxb3os that was down-regulated in cystic kidneys from Pkd1 and Pkd2 mutant mice. The human ortholog HOXB3-AS1 was down-regulated in cystic kidneys from ADPKD patients. Hoxb3os was highly expressed in renal tubules in adult WT mice, whereas its expression was lost in the cyst epithelium of mutant mice. To investigate the function of Hoxb3os, we utilized CRISPR/Cas9 to knock out its expression in mIMCD3 cells. Deletion of Hoxb3os resulted in increased phosphorylation of mTOR and its downstream targets, including p70 S6 kinase, ribosomal protein S6, and the translation repressor 4E-BP1. Consistent with activation of mTORC1 signaling, Hoxb3os mutant cells displayed increased mitochondrial respiration. The Hoxb3os mutant phenotype was partially rescued upon re-expression of Hoxb3os in knockout cells. These findings identify Hoxb3os as a novel lncRNA that is down-regulated in ADPKD and regulates mTOR signaling and mitochondrial respiration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Micah D Gearhart
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, 55455 and
| | - Vishal Patel
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Chao Xing
- McDermott Center for Human Growth and Development and.,Bioinformatics, and.,Departments of Clinical Sciences
| | | |
Collapse
|
89
|
Liu X, Vien T, Duan J, Sheu SH, DeCaen PG, Clapham DE. Polycystin-2 is an essential ion channel subunit in the primary cilium of the renal collecting duct epithelium. eLife 2018; 7:33183. [PMID: 29443690 PMCID: PMC5812715 DOI: 10.7554/elife.33183] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 02/01/2018] [Indexed: 01/08/2023] Open
Abstract
Mutations in the polycystin genes, PKD1 or PKD2, results in Autosomal Dominant Polycystic Kidney Disease (ADPKD). Although a genetic basis of ADPKD is established, we lack a clear understanding of polycystin proteins’ functions as ion channels. This question remains unsolved largely because polycystins localize to the primary cilium – a tiny, antenna-like organelle. Using a new ADPKD mouse model, we observe primary cilia that are abnormally long in cells associated with cysts after conditional ablation of Pkd1 or Pkd2. Using primary cultures of collecting duct cells, we show that polycystin-2, but not polycystin-1, is a required subunit for the ion channel in the primary cilium. The polycystin-2 channel preferentially conducts K+ and Na+; intraciliary Ca2+, enhances its open probability. We introduce a novel method for measuring heterologous polycystin-2 channels in cilia, which will have utility in characterizing PKD2 variants that cause ADPKD.
Collapse
Affiliation(s)
- Xiaowen Liu
- Department of Cardiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Thuy Vien
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, United States
| | - Jingjing Duan
- Department of Cardiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Shu-Hsien Sheu
- Department of Cardiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States.,Department of Pathology, Boston Children's Hospital, Boston, United States
| | - Paul G DeCaen
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, United States
| | - David E Clapham
- Department of Cardiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
90
|
Chang A, Abderemane-Ali F, Hura GL, Rossen ND, Gate RE, Minor DL. A Calmodulin C-Lobe Ca 2+-Dependent Switch Governs Kv7 Channel Function. Neuron 2018; 97:836-852.e6. [PMID: 29429937 DOI: 10.1016/j.neuron.2018.01.035] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/07/2017] [Accepted: 01/12/2018] [Indexed: 12/22/2022]
Abstract
Kv7 (KCNQ) voltage-gated potassium channels control excitability in the brain, heart, and ear. Calmodulin (CaM) is crucial for Kv7 function, but how this calcium sensor affects activity has remained unclear. Here, we present X-ray crystallographic analysis of CaM:Kv7.4 and CaM:Kv7.5 AB domain complexes that reveal an Apo/CaM clamp conformation and calcium binding preferences. These structures, combined with small-angle X-ray scattering, biochemical, and functional studies, establish a regulatory mechanism for Kv7 CaM modulation based on a common architecture in which a CaM C-lobe calcium-dependent switch releases a shared Apo/CaM clamp conformation. This C-lobe switch inhibits voltage-dependent activation of Kv7.4 and Kv7.5 but facilitates Kv7.1, demonstrating that mechanism is shared by Kv7 isoforms despite the different directions of CaM modulation. Our findings provide a unified framework for understanding how CaM controls different Kv7 isoforms and highlight the role of membrane proximal domains for controlling voltage-gated channel function. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Aram Chang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Greg L Hura
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Nathan D Rossen
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Rachel E Gate
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; California Institute for Quantitative Biomedical Research, University of California San Francisco, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
91
|
Global versus local mechanisms of temperature sensing in ion channels. Pflugers Arch 2018; 470:733-744. [PMID: 29340775 DOI: 10.1007/s00424-017-2102-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/15/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023]
Abstract
Ion channels turn diverse types of inputs, ranging from neurotransmitters to physical forces, into electrical signals. Channel responses to ligands generally rely on binding to discrete sensor domains that are coupled to the portion of the channel responsible for ion permeation. By contrast, sensing physical cues such as voltage, pressure, and temperature arises from more varied mechanisms. Voltage is commonly sensed by a local, domain-based strategy, whereas the predominant paradigm for pressure sensing employs a global response in channel structure to membrane tension changes. Temperature sensing has been the most challenging response to understand and whether discrete sensor domains exist for pressure and temperature has been the subject of much investigation and debate. Recent exciting advances have uncovered discrete sensor modules for pressure and temperature in force-sensitive and thermal-sensitive ion channels, respectively. In particular, characterization of bacterial voltage-gated sodium channel (BacNaV) thermal responses has identified a coiled-coil thermosensor that controls channel function through a temperature-dependent unfolding event. This coiled-coil thermosensor blueprint recurs in other temperature sensitive ion channels and thermosensitive proteins. Together with the identification of ion channel pressure sensing domains, these examples demonstrate that "local" domain-based solutions for sensing force and temperature exist and highlight the diversity of both global and local strategies that channels use to sense physical inputs. The modular nature of these newly discovered physical signal sensors provides opportunities to engineer novel pressure-sensitive and thermosensitive proteins and raises new questions about how such modular sensors may have evolved and empowered ion channel pores with new sensibilities.
Collapse
|
92
|
Deletion of Pkd1 in renal stromal cells causes defects in the renal stromal compartment and progressive cystogenesis in the kidney. J Transl Med 2017; 97:1427-1438. [PMID: 28892094 DOI: 10.1038/labinvest.2017.97] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/29/2017] [Accepted: 08/02/2017] [Indexed: 12/15/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), caused by PKD1 and PKD2 gene mutations, is one of the most common genetic diseases, affecting up to 1 in 500 people. Mutations of PKD1 account for over 85% of ADPKD cases. However, mechanisms of disease progression and explanations for the wide range in disease phenotype remain to be elucidated. Moreover, functional roles of PKD1 in the renal stromal compartment are poorly understood. In this work, we tested if Pkd1 is essential for development and maintenance of the renal stromal compartment and if this role contributes to pathogenesis of polycystic kidney disease using a novel tissue-specific knockout mouse model. We demonstrate that deletion of Pkd1 from renal stromal cells using Foxd1-driven Cre causes a spectrum of defects in the stromal compartment, including excessive apoptosis/proliferation and extracellular matrix deficiency. Renal vasculature was also defective. Further, mutant mice showed epithelial changes and progressive cystogenesis in adulthood modeling human ADPKD. Altogether, we provide robust evidence to support indispensable roles for Pkd1 in development and maintenance of stromal cell derivatives by using a novel ADPKD model. Moreover, stromal compartment defects caused by Pkd1 deletion might serve as an important mechanism for pathogenesis of ADPKD.
Collapse
|
93
|
Xiao Z, Baudry J, Cao L, Huang J, Chen H, Yates CR, Li W, Dong B, Waters CM, Smith JC, Quarles LD. Polycystin-1 interacts with TAZ to stimulate osteoblastogenesis and inhibit adipogenesis. J Clin Invest 2017; 128:157-174. [PMID: 29202470 DOI: 10.1172/jci93725] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/17/2017] [Indexed: 01/15/2023] Open
Abstract
The molecular mechanisms that transduce the osteoblast response to physical forces in the bone microenvironment are poorly understood. Here, we used genetic and pharmacological experiments to determine whether the polycystins PC1 and PC2 (encoded by Pkd1 and Pkd2) and the transcriptional coactivator TAZ form a mechanosensing complex in osteoblasts. Compound-heterozygous mice lacking 1 copy of Pkd1 and Taz exhibited additive decrements in bone mass, impaired osteoblast-mediated bone formation, and enhanced bone marrow fat accumulation. Bone marrow stromal cells and osteoblasts derived from these mice showed impaired osteoblastogenesis and enhanced adipogenesis. Increased extracellular matrix stiffness and application of mechanical stretch to multipotent mesenchymal cells stimulated the nuclear translocation of the PC1 C-terminal tail/TAZ (PC1-CTT/TAZ) complex, leading to increased runt-related transcription factor 2-mediated (Runx2-mediated) osteogenic and decreased PPARγ-dependent adipogenic gene expression. Using structure-based virtual screening, we identified a compound predicted to bind to PC2 in the PC1:PC2 C-terminal tail region with helix:helix interaction. This molecule stimulated polycystin- and TAZ-dependent osteoblastogenesis and inhibited adipogenesis. Thus, we show that polycystins and TAZ integrate at the molecular level to reciprocally regulate osteoblast and adipocyte differentiation, indicating that the polycystins/TAZ complex may be a potential therapeutic target to increase bone mass.
Collapse
Affiliation(s)
- Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jerome Baudry
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA.,Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Li Cao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jinsong Huang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Hao Chen
- Department of Pharmaceutical Sciences and
| | | | - Wei Li
- Department of Pharmaceutical Sciences and
| | - Brittany Dong
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christopher M Waters
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jeremy C Smith
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA.,Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - L Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
94
|
Abstract
INTRODUCTION Polycystic kidney disease (PKD) is clinically and genetically heterogeneous and constitutes the most common heritable kidney disease. Most patients are affected by the autosomal dominant form (ADPKD) which generally is an adult-onset multisystem disorder. By contrast, the rarer recessive form ARPKD usually already manifests perinatally or in childhood. In some patients, however, ADPKD and ARPKD can phenotypically overlap with early manifestation in ADPKD and only late onset in ARPKD. Progressive fibrocystic renal changes are often accompanied by severe hepatobiliary changes or other extrarenal abnormalities. Areas covered: A reduced dosage of disease proteins disturbs cell homeostasis and explains a more severe clinical course in some PKD patients. Cystic kidney disease is also a common feature of other ciliopathies and genetic syndromes. Genetic diagnosis may guide clinical management and helps to avoid invasive measures and to detect renal and extrarenal comorbidities early in the clinical course. Expert Commentary: The broad phenotypic and genetic heterogeneity of cystic and polycystic kidney diseases make NGS a particularly powerful approach. Interpretation of data becomes the challenge and bench and bedside benefit from digitized multidisciplinary interrelationships.
Collapse
Affiliation(s)
- Carsten Bergmann
- a Center for Human Genetics , Bioscientia , Ingelheim , Germany.,b Department of Medicine , University Hospital Freiburg , Freiburg , Germany
| |
Collapse
|
95
|
Sha YK, Sha YW, Mei LB, Huang XJ, Wang X, Lin SB, Li L, Li P. Use of targeted sequence capture and high-throughput sequencing identifies a novel PKD1 mutation involved in adult polycystic kidney disease. Gene 2017; 634:1-4. [PMID: 28870863 DOI: 10.1016/j.gene.2017.08.040] [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: 06/25/2017] [Revised: 08/06/2017] [Accepted: 08/30/2017] [Indexed: 11/24/2022]
Abstract
Polycystic kidney disease (PKD) is a common inherited disease that is characterized by a progressive development of renal cysts. Approximately 85% of PKD cases are due to mutations in the polycystin 1 (PKD1) gene. Here, we report a pedigree containing nine patients with autosomal dominant PKD (ADPKD). Using targeted exome sequencing of PKD1 and PKD2 genes, we identified a novel heterozygous frameshift mutation c.3976_3977insCT (p.F1326Sfs*21) in the PKD1 gene that segregated between affected and unaffected family members. This mutation is currently not present in the 1000 Genomes Project nor ExAC databases and is therefore a novel PKD1 mutation involved in ADPKD. These results provide a novel sequence variant for the genetic analysis of this disease.
Collapse
Affiliation(s)
- Yan-Kun Sha
- Department of Nephrology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 361003, China
| | - Yan-Wei Sha
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Li-Bin Mei
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Xian-Jing Huang
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Xu Wang
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Shao-Bin Lin
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China.
| | - Ping Li
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China.
| |
Collapse
|
96
|
Ma M, Gallagher AR, Somlo S. Ciliary Mechanisms of Cyst Formation in Polycystic Kidney Disease. Cold Spring Harb Perspect Biol 2017; 9:a028209. [PMID: 28320755 PMCID: PMC5666631 DOI: 10.1101/cshperspect.a028209] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is a disease of defective tissue homeostasis resulting in active remodeling of nephrons and bile ducts to form fluid-filled sacs called cysts. The causal genes PKD1 and PKD2 encode transmembrane proteins polycystin 1 (PC1) and polycystin 2 (PC2), respectively. Together, the polycystins localize to the solitary primary cilium that protrudes from the apical surface of most kidney tubule cells and is thought to function as a privileged compartment that the cell uses for signal integration of sensory inputs. It has been proposed that PC1 and PC2 form a receptor-channel complex that detects external stimuli and transmit a local calcium-mediated signal, which may control a multitude of cellular processes by an as-yet unknown mechanism. Genetic studies using mouse models of cilia and polycystin dysfunction have shown that polycystins regulate an unknown cilia-dependent signal that is normally part of the homeostatic maintenance of nephron structure. ADPKD ensues when this pathway is dysregulated by absence of polycystins from intact cilia, but disruption of cilia also disrupts this signaling mechanism and ameliorates ADPKD even in the absence of polycystins. Understanding the role of cilia and ciliary signaling in ADPKD is challenging, but success will provide saltatory advances in our understanding of how tubule structure is maintained in healthy kidneys and how disruption of polycystin or cilia function leads to the pathological tissue remodeling process underlying ADPKD.
Collapse
Affiliation(s)
- Ming Ma
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8029
| | - Anna-Rachel Gallagher
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8029
| | - Stefan Somlo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8029
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06520-8029
| |
Collapse
|
97
|
Gao C, Zhang L, Zhang Y, Wallace DP, Lopez-Soler RI, Higgins PJ, Zhang W. Insights into cellular and molecular basis for urinary tract infection in autosomal-dominant polycystic kidney disease. Am J Physiol Renal Physiol 2017; 313:F1077-F1083. [PMID: 28794066 DOI: 10.1152/ajprenal.00279.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/24/2017] [Accepted: 08/07/2017] [Indexed: 12/27/2022] Open
Abstract
Urinary tract infection (UTI) is a broad term referring to an infection of the kidneys, ureters, bladder, and/or urethra. Because of its prevalence, frequent recurrence, and rising resistance to antibiotics, UTI has become a challenge in clinical practice. Autosomal-dominant polycystic kidney disease (ADPKD) is the most common monogenic disorder of the kidney and is characterized by the growth of fluid-filled cysts in both kidneys. Progressive cystic enlargement, inflammation, and interstitial fibrosis result in nephron loss with subsequent decline in kidney function. ADPKD patients frequently develop UTI; however, the cellular and molecular mechanisms responsible for the high UTI incidence in ADPKD patients remain virtually unaddressed. Emerging evidence suggests that α-intercalated cells (α-ICs) of the collecting ducts function in the innate immune defense against UTI. α-ICs inhibit bacterial growth by acidifying urine and secreting neutrophil gelatinase-associated lipocalin (NGAL) that chelates siderophore-containing iron. It is necessary to determine, therefore, if ADPKD patients with recurrent UTI have a reduced number and/or impaired function of α-ICs. Identification of the underlying cellular and molecular mechanisms may lead to the development of novel strategies to reduce UTI in ADPKD.
Collapse
Affiliation(s)
- Chao Gao
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Long Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Ye Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Darren P Wallace
- Departments of Internal Medicine and Physiology and The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Reynold I Lopez-Soler
- Department of Surgery, Division of Transplantation, Albany Medical College, Albany, New York
| | - Paul J Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York;
| |
Collapse
|
98
|
Fang Z, Xu S, Wang Y, Sun L, Feng Y, Guo Y, Li H, Jiang W. Pathogenicity analysis of novel variations in Chinese Han patients with polycystic kidney disease. Gene 2017; 626:433-441. [PMID: 28578020 DOI: 10.1016/j.gene.2017.05.046] [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: 02/22/2017] [Revised: 04/24/2017] [Accepted: 05/22/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Locus and allellic heterogeneity in polycystic kidney disease (PKD) is a great challenge in precision diagnosis. We aim to establish comprehensive methods to distinguish the pathogenic mutations from the variations in PKD1, PKD2 and PKHD1 genes in a limited time and lay the foundation for precisely prenatal diagnosis, preimplantation genetic diagnosis and presymptom diagnosis of PKD. METHODS Nested PCR combined with direct DNA sequencing were used to screen variations in PKD1, PKD2 and PKHD1 genes. The pathogenicity of de novel variations was assessed by the comprehensive methods including clinic data and literature review, databases query, analysis of co-segregation of the variants with the disease, variant frequency screening in the population, evolution conservation comparison, protein structure analysis and splice sites predictions. RESULTS 17 novel mutations from 15 Chinese Han families were clarified including 10 mutations in PKD1 gene and 7 mutations in PKHD1 gene. The novel mutations were classified as 4 definite pathogenic, 2 highly likely pathogenic, 4 likely pathogenic, 7 indeterminate by the comprehensive analysis. The results were verified the truth by the follow-up visits. CONCLUSIONS The comprehensive methods may be useful in distinguishing the pathogenic mutations from the variations in PKD1, PKD2 and PKHD1 genes for prenatal diagnosis and presymptom diagnosis of PKD. Our results also enriched PKD genes mutation spectrum and evolved possible genotype-phenotype correlations of Chinese Han population.
Collapse
Affiliation(s)
- Zishui Fang
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Shiyan Xu
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; ShenZhen People's Hospital, China
| | - Yonghua Wang
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Liwei Sun
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yi Feng
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yibin Guo
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hongyi Li
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Weiying Jiang
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
99
|
Giehl E, Lemos FO, Huang Y, Giordano FJ, Kuo IY, Ehrlich BE. Polycystin 2-dependent cardio-protective mechanisms revealed by cardiac stress. Pflugers Arch 2017; 469:1507-1517. [PMID: 28762163 DOI: 10.1007/s00424-017-2042-7] [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] [Received: 03/02/2017] [Revised: 06/08/2017] [Accepted: 07/20/2017] [Indexed: 01/19/2023]
Abstract
Although autosomal dominant polycystic kidney disease (ADPKD) is characterized by the development of multiple kidney cysts, the most frequent cause of death in ADPKD patients is cardiovascular disease. ADPKD is linked to mutations in PKD1 or pkd2, the genes that encode for the proteins polycystin 1 and polycystin 2 (PC1 and PC2, respectively). The cardiovascular complications have been assumed to be a consequence of renal hypertension and activation of renin/angiotensin/aldosterone (RAAS) pathway. However, the expression of PC1 and PC2 in cardiac tissue suggests additional direct effects of these proteins on cardiac function. We previously reported that zebrafish lacking PC2 develop heart failure, and that heterozygous Pkd2+/- mice are hypersensitive to acute β-adrenergic receptor (βAR) stimulation. Here, we investigate the effect of cardiac stress (prolonged continuous βAR stimulus) on Pkd2+/- mice. After βAR stimulation for 7 days, wild-type (WT) mice had increased left ventricular mass and natriuretic peptide (ANP and BNP) mRNA levels. The WT mice also had upregulated levels of PC2 and chromogranin B (CGB, an upstream regulator of BNP). Conversely, Pkd2+/- mice had increased left ventricular mass, but natriuretic peptide and CGB expression levels remained constant. Reversal of the increased cardiac mass was observed in WT mice 3 days after cessation of the βAR stimulation, but not in Pkd2+/- mice. We suggest that cardiac stress leads to upregulation of the PC2-CGB-BNP signaling axis, and this pathway regulates the production of cardio-protective natriuretic peptides. The lack of a PC2-dependent cardio-protective function may contribute to the severity of cardiac dysfunction in Pkd2+/- mice and in ADPKD patients.
Collapse
Affiliation(s)
- Esther Giehl
- Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Pharmacology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06520, USA
| | - Fernanda O Lemos
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06520, USA
| | - Yan Huang
- Department of Cardiology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06520, USA
| | - Frank J Giordano
- Department of Cardiology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06520, USA
| | - Ivana Y Kuo
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06520, USA
| | - Barbara E Ehrlich
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06520, USA. .,Department of Cellular and Molecular Physiology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06520, USA.
| |
Collapse
|
100
|
|