1
|
Bae Y, Lee S, Kim A, Lee S, Kim S, Park S, Noh J, Ryoo K, Yi G, Park J, Hwang EM. Astrocytic NHERF-1 Increases Seizure Susceptibility by Inhibiting Surface Expression of TREK-1. Glia 2025; 73:720-736. [PMID: 39543986 PMCID: PMC11845839 DOI: 10.1002/glia.24644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Mature hippocampal astrocytes exhibit a linear current-to-voltage (I-V) K+ membrane conductance called passive conductance. It is estimated to enable astrocytes to keep potassium homeostasis in the brain. We previously reported that the TWIK-1/TREK-1 heterodimeric channels are crucial for astrocytic passive conductance. However, the regulatory mechanism of these channels by other binding proteins remains elusive. Here, we identified Na+/H+ exchange regulator-1 (NHERF-1), a protein highly expressed in astrocytes, as a novel interaction partner for these channels. NHERF-1 endogenously bound to TWIK-1/TREK-1 in hippocampal cultured astrocytes. When NHERF-1 is overexpressed or silenced, surface expression and activity of TWIK-1/TREK-1 heterodimeric channels are inhibited or enhanced, respectively. Furthermore, we confirmed that reduced astrocytic passive conductance by NHERF-1 overexpressing in the hippocampus increases kainic acid (KA)-induced seizure sensitivity. Taken together, these results suggest that NHERF-1 is a key regulator of TWIK-1/TREK-1 heterodimeric channels in astrocytes and suppression of TREK-1 surface expression by NHERF-1 increases KA-induced seizure susceptibility via reduction of astrocytic passive conductance.
Collapse
Affiliation(s)
- Yeonju Bae
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Soomin Lee
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Ajung Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
| | - Shinae Lee
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- Department of bio and Brain Engineering, College of EngineeringKAISTDaejeonRepublic of Korea
| | - Seong‐Seop Kim
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Sunyoung Park
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Junyeol Noh
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Kanghyun Ryoo
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Gwan‐Su Yi
- Department of bio and Brain Engineering, College of EngineeringKAISTDaejeonRepublic of Korea
| | - Jae‐Yong Park
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
- Astrion, Inc.SeoulRepublic of Korea
| | - Eun Mi Hwang
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- Division of Bio‐Medical Science & Technology, KIST SchoolKorea University of Science and TechnologySeoulRepublic of Korea
| |
Collapse
|
2
|
Naciri Bennani H, Chtioui I, Allirot C, Somrani R, Jouve T, Rostaing L, Bourdat-Michel G. Effects of SLC34A3 or SLC34A1 variants on calcium and phosphorus homeostasis. Pediatr Nephrol 2025; 40:117-129. [PMID: 39256228 DOI: 10.1007/s00467-024-06505-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Variants in SLC34A1 and SLC34A2 genes, which encode co-transporters NaPi2a and NaPi2c, respectively, can lead to hypophosphatemia due to renal phosphate loss. This condition results in hypercalcitriolemia and hypercalciuria, leading to formation of kidney stones and nephrocalcinosis. Phenotype is highly variable. Management includes hyperhydration, dietary modifications, and/or phosphate supplementation. Thiazides and azoles may be used, but randomized studies are needed to confirm their clinical efficacy. METHODS We conducted a retrospective study in the pediatric nephrology unit at Grenoble University Hospital from January 2010 to December 2023. The study aimed to describe clinical and biological symptoms of patients with confirmed SLC34A1 and SLC34A3 gene variants and their outcomes. RESULTS A total of 11 patients (9 females) from 6 different families had variants in the SLC34A1 (5 patients) and SLC34A3 (6 patients) genes. Median age at diagnosis was 72 [1-108] months. Average follow-up duration was 8.1 ± 4.5 years. Presenting symptom was nephrocalcinosis (4 cases), followed by renal colic (3 cases). At diagnosis, 90% of patients had hypercalciuria and 45% had hypercalcitriolemia. Management included hyperhydration and dietary advice. All patients showed favorable outcomes with normal growth and school attendance. One patient with an SLC34A3 variant showed regression of nephrocalcinosis. Kidney function remained normal. CONCLUSION Clinical and biological manifestations of SLC34 gene variants are highly variable, even among siblings; therefore, management must be personalized. Hygienic and dietary measures (such as hyperhydration, a low sodium diet, and age-appropriate calcium intake) result in favorable outcomes in most cases. Use of azoles (e.g., fluconazole) appears to be a promising therapeutic option.
Collapse
Affiliation(s)
- Hamza Naciri Bennani
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
| | - Imane Chtioui
- Pediatric Department, Metropole Savoie Hospital Center, Chambéry, France
| | - Camille Allirot
- Pediatric Nephrology Department, Grenoble University Hospital, Grenoble, France
| | - Rim Somrani
- Pediatric Nephrology Department, Grenoble University Hospital, Grenoble, France
| | - Thomas Jouve
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
- Grenoble Alpes University, Grenoble, France
| | - Lionel Rostaing
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France.
- Grenoble Alpes University, Grenoble, France.
- Service de Néphrologie-Hémodialyse-Aphérèses Et Transplantation Rénale, CHU Grenoble-Alpes, Avenue Maquis du Grésivaudan, La Tronche, 38700, France.
| | | |
Collapse
|
3
|
Chen N, Zeng W, Luo Y, Dong M. Clinical and Molecular Cytogenetic Characterization of 2 Sibling Cases with 17q25 Duplication due to Unbalanced Translocation. Mol Syndromol 2024; 15:495-502. [PMID: 39634249 PMCID: PMC11614436 DOI: 10.1159/000538979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 04/17/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Partial 17q duplication is a rare chromosome abnormality. Features include severe psychomotor retardation, intellectual disability, facial dysmorphism, proximal limb shortness, and hyperlaxity of limb joints. Case Presentation The proband is a 7-year and 4-month-old boy with developmental delay, facial abnormality, joint laxity and scoliosis, ventriculomegaly, hydrocephalus, hypophosphatemia, and squint, while his older brother is a fetus who was aborted at 33rd week of gestation because of multiple malformations including ventriculomegaly and moderate hydrocephalus. Both siblings have features such as ventriculomegaly and hydrocephalus. Conclusion Here, we report 2 sibling cases with 17q25 duplication from a maternal translocation t(14;17). Our findings expanded the clinical spectra and described the fetal phenotype of 17q25 microduplication.
Collapse
Affiliation(s)
- Na Chen
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Wenshan Zeng
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Yuqin Luo
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Minyue Dong
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
4
|
Sneddon WB, Ramineni S, Van Doorn GE, Hepler JR, Friedman PA. Distinct and overlapping RGS14 and RGS12 actions regulate NPT2A-mediated phosphate transport. Biochem Biophys Res Commun 2024; 733:150700. [PMID: 39293332 DOI: 10.1016/j.bbrc.2024.150700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Parathyroid hormone (PTH) and fibroblast growth factor-23 (FGF23) control serum phosphate levels by downregulating the renal Na-phosphate transporter NPT2A, thereby decreasing phosphate absorption and augmenting urinary excretion. This mechanism requires NHERF1, a PDZ scaffold protein, and is governed by the regulator of G protein signaling-14 (RGS14), which harbors a carboxy-terminal PDZ ligand that binds NHERF1. RGS14 is part of a triad of structurally related RGS proteins that includes RGS12 and RGS10. Like RGS14, RGS12 contains a class 1 PDZ ligand. However, unlike RGS14, the larger RGS12 contains an upstream PDZ-binding domain. The studies outlined here examined and characterized the binding of RGS12 with NHERF1 and NPT2A and its function on hormone-regulated phosphate transport. Immunoblotting experiments revealed RGS12 C-terminal PDZ ligand binding to NHERF1. Further structural analysis disclosed that NPT2A engaged full-length RGS12 and the upstream fragment containing the PDZ domain. Neither the downstream RGS12 portion nor RGS14 interacted with NPT2A. PTH and FGF23 profoundly inhibited phosphate uptake in opossum kidney proximal tubule cells. Transfection with human RGS14, or human RGS12, abolished hormone-sensitive phosphate transport as reported for human proximal tubule cells. RGS12 inhibitory activity resides in the downstream region and is comparable to RGS14. The carboxy-terminal RGS12(667-1447) splice variant is prominently expressed in the kidney and may contribute to regulating hormone-sensitive phosphate transport.
Collapse
Affiliation(s)
- W Bruce Sneddon
- Laboratory for GPCR Biology, Departments of Pharmacology and Chemical Biology, USA
| | - Suneela Ramineni
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, USA
| | - G Emme Van Doorn
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, USA
| | - John R Hepler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, USA
| | - Peter A Friedman
- Laboratory for GPCR Biology, Departments of Pharmacology and Chemical Biology, USA; Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, USA.
| |
Collapse
|
5
|
Busselman BW, Ratnayake I, Terasaki MR, Thakkar VP, Ilyas A, Otterpohl KL, Zimmerman JL, Chandrasekar I. Actin cytoskeleton and associated myosin motors within the renal epithelium. Am J Physiol Renal Physiol 2024; 327:F553-F565. [PMID: 39052845 PMCID: PMC11483076 DOI: 10.1152/ajprenal.00078.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
This review highlights the complexity of renal epithelial cell membrane architectures and organelles through careful review of ultrastructural and physiological studies published over the past several decades. We also showcase the vital roles played by the actin cytoskeleton and actin-associated myosin motor proteins in regulating cell type-specific physiological functions within the cells of the renal epithelium. The purpose of this review is to provide a fresh conceptual framework to explain the structure-function relationships that exist between the actin cytoskeleton, organelle structure, and cargo transport within the mammalian kidney. With recent advances in technologies to visualize the actin cytoskeleton and associated proteins within intact kidneys, it has become increasingly imperative to reimagine the functional roles of these proteins in situ to provide a rationale for their unique, cell type-specific functions that are necessary to establish and maintain complex physiological processes.
Collapse
Affiliation(s)
- Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | | | - Mark R Terasaki
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| | - Vedant P Thakkar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Arooba Ilyas
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | - Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Jenna L Zimmerman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| |
Collapse
|
6
|
Liu XY, Song X, Czosnyka M, Robba C, Czosnyka Z, Summers JL, Yu HJ, Gao GY, Smielewski P, Guo F, Pang MJ, Ming D. Congenital hydrocephalus: a review of recent advances in genetic etiology and molecular mechanisms. Mil Med Res 2024; 11:54. [PMID: 39135208 PMCID: PMC11318184 DOI: 10.1186/s40779-024-00560-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/28/2024] [Indexed: 08/15/2024] Open
Abstract
The global prevalence rate for congenital hydrocephalus (CH) is approximately one out of every five hundred births with multifaceted predisposing factors at play. Genetic influences stand as a major contributor to CH pathogenesis, and epidemiological evidence suggests their involvement in up to 40% of all cases observed globally. Knowledge about an individual's genetic susceptibility can significantly improve prognostic precision while aiding clinical decision-making processes. However, the precise genetic etiology has only been pinpointed in fewer than 5% of human instances. More occurrences of CH cases are required for comprehensive gene sequencing aimed at uncovering additional potential genetic loci. A deeper comprehension of its underlying genetics may offer invaluable insights into the molecular and cellular basis of this brain disorder. This review provides a summary of pertinent genes identified through gene sequencing technologies in humans, in addition to the 4 genes currently associated with CH (two X-linked genes L1CAM and AP1S2, two autosomal recessive MPDZ and CCDC88C). Others predominantly participate in aqueduct abnormalities, ciliary movement, and nervous system development. The prospective CH-related genes revealed through animal model gene-editing techniques are further outlined, focusing mainly on 4 pathways, namely cilia synthesis and movement, ion channels and transportation, Reissner's fiber (RF) synthesis, cell apoptosis, and neurogenesis. Notably, the proper functioning of motile cilia provides significant impulsion for cerebrospinal fluid (CSF) circulation within the brain ventricles while mutations in cilia-related genes constitute a primary cause underlying this condition. So far, only a limited number of CH-associated genes have been identified in humans. The integration of genotype and phenotype for disease diagnosis represents a new trend in the medical field. Animal models provide insights into the pathogenesis of CH and contribute to our understanding of its association with related complications, such as renal cysts, scoliosis, and cardiomyopathy, as these genes may also play a role in the development of these diseases. Genes discovered in animals present potential targets for new treatments but require further validation through future human studies.
Collapse
Affiliation(s)
- Xiu-Yun Liu
- Medical School, Tianjin University, Tianjin, 300072, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, 300072, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300380, China
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Xin Song
- Medical School, Tianjin University, Tianjin, 300072, China
| | - Marek Czosnyka
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Chiara Robba
- San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, 16132, Genoa, Italy
| | - Zofia Czosnyka
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Jennifer Lee Summers
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Hui-Jie Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Guo-Yi Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Peter Smielewski
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Fang Guo
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Mei-Jun Pang
- Medical School, Tianjin University, Tianjin, 300072, China.
| | - Dong Ming
- Medical School, Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, 300072, China.
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300380, China.
| |
Collapse
|
7
|
Walker V. The Intricacies of Renal Phosphate Reabsorption-An Overview. Int J Mol Sci 2024; 25:4684. [PMID: 38731904 PMCID: PMC11083860 DOI: 10.3390/ijms25094684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
To maintain an optimal body content of phosphorus throughout postnatal life, variable phosphate absorption from food must be finely matched with urinary excretion. This amazing feat is accomplished through synchronised phosphate transport by myriads of ciliated cells lining the renal proximal tubules. These respond in real time to changes in phosphate and composition of the renal filtrate and to hormonal instructions. How they do this has stimulated decades of research. New analytical techniques, coupled with incredible advances in computer technology, have opened new avenues for investigation at a sub-cellular level. There has been a surge of research into different aspects of the process. These have verified long-held beliefs and are also dramatically extending our vision of the intense, integrated, intracellular activity which mediates phosphate absorption. Already, some have indicated new approaches for pharmacological intervention to regulate phosphate in common conditions, including chronic renal failure and osteoporosis, as well as rare inherited biochemical disorders. It is a rapidly evolving field. The aim here is to provide an overview of our current knowledge, to show where it is leading, and where there are uncertainties. Hopefully, this will raise questions and stimulate new ideas for further research.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton S016 6YD, UK
| |
Collapse
|
8
|
Friedman P, Mamonova T. The molecular sociology of NHERF1 PDZ proteins controlling renal hormone-regulated phosphate transport. Biosci Rep 2024; 44:BSR20231380. [PMID: 38465463 PMCID: PMC10987488 DOI: 10.1042/bsr20231380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/12/2024] Open
Abstract
Parathyroid hormone (PTH) and fibroblast growth factor-23 (FGF23) control extracellular phosphate levels by regulating renal NPT2A-mediated phosphate transport by a process requiring the PDZ scaffold protein NHERF1. NHERF1 possesses two PDZ domains, PDZ1 and PDZ2, with identical core-binding GYGF motifs explicitly recognizing distinct binding partners that play different and specific roles in hormone-regulated phosphate transport. The interaction of PDZ1 and the carboxy-terminal PDZ-binding motif of NPT2A (C-TRL) is required for basal phosphate transport. PDZ2 is a regulatory domain that scaffolds multiple biological targets, including kinases and phosphatases involved in FGF23 and PTH signaling. FGF23 and PTH trigger disassembly of the NHERF1-NPT2A complex through reversible hormone-stimulated phosphorylation with ensuing NPT2A sequestration, down-regulation, and cessation of phosphate absorption. In the absence of NHERF1-NPT2A interaction, inhibition of FGF23 or PTH signaling results in disordered phosphate homeostasis and phosphate wasting. Additional studies are crucial to elucidate how NHERF1 spatiotemporally coordinates cellular partners to regulate extracellular phosphate levels.
Collapse
Affiliation(s)
- Peter A. Friedman
- Laboratory for G Protein-Coupled Receptor Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, U.S.A
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, U.S.A
| | - Tatyana Mamonova
- Laboratory for G Protein-Coupled Receptor Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, U.S.A
| |
Collapse
|
9
|
Walker E, Hayes W, Bockenhauer D. Inherited non-FGF23-mediated phosphaturic disorders: A kidney-centric review. Best Pract Res Clin Endocrinol Metab 2024; 38:101843. [PMID: 38042745 DOI: 10.1016/j.beem.2023.101843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2023]
Abstract
Phosphate is freely filtered by the glomerulus and reabsorbed exclusively in the proximal tubule by two key transporters, NaPiIIA and NaPiIIC, encoded by SLC34A1 and SLC34A3, respectively. Regulation of these transporters occurs primarily through the hormone FGF23 and, to a lesser degree, PTH. Consequently, inherited non-FGF23 mediated phosphaturic disorders are due to generalised proximal tubular dysfunction, loss-of-function variants in SLC34A1 or SLC34A3 or excess PTH signalling. The corresponding disorders are Renal Fanconi Syndrome, Infantile Hypercalcaemia type 2, Hereditary Hypophosphataemic Rickets with Hypercalciuria and Familial Hyperparathyroidism. Several inherited forms of Fanconi renotubular syndrome (FRTS) have also been described with the underlying genes encoding for GATM, EHHADH, HNF4A and NDUFAF6. Here, we will review their pathophysiology, clinical manifestations and the implications for treatment from a kidney-centric perspective, focussing on those disorders caused by dysfunction of renal phosphate transporters. Moreover, we will highlight specific genetic aspects, as the availability of large population genetic databases has raised doubts about some of the originally proposed gene-disease associations concerning phosphate transporters or their associated proteins.
Collapse
Affiliation(s)
- Emma Walker
- Nephrology Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Wesley Hayes
- Nephrology Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Detlef Bockenhauer
- Nephrology Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; Department of Renal Medicine, University College London, London, UK.
| |
Collapse
|
10
|
Gupta DP, Bhusal A, Rahman MH, Kim JH, Choe Y, Jang J, Jung HJ, Kim UK, Park JS, Maeng LS, Suk K, Song GJ. EBP50 is a key molecule for the Schwann cell-axon interaction in peripheral nerves. Prog Neurobiol 2023; 231:102544. [PMID: 37940033 DOI: 10.1016/j.pneurobio.2023.102544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023]
Abstract
Peripheral nerve injury disrupts the Schwann cell-axon interaction and the cellular communication between them. The peripheral nervous system has immense potential for regeneration extensively due to the innate plastic potential of Schwann cells (SCs) that allows SCs to interact with the injured axons and exert specific repair functions essential for peripheral nerve regeneration. In this study, we show that EBP50 is essential for the repair function of SCs and regeneration following nerve injury. The increased expression of EBP50 in the injured sciatic nerve of control mice suggested a significant role in regeneration. The ablation of EBP50 in mice resulted in delayed nerve repair, recovery of behavioral function, and remyelination following nerve injury. EBP50 deficiency led to deficits in SC functions, including proliferation, migration, cytoskeleton dynamics, and axon interactions. The adeno-associated virus (AAV)-mediated local expression of EBP50 improved SCs migration, functional recovery, and remyelination. ErbB2-related proteins were not differentially expressed in EBP50-deficient sciatic nerves following injury. EBP50 binds and stabilizes ErbB2 and activates the repair functions to promote regeneration. Thus, we identified EBP50 as a potent SC protein that can enhance the regeneration and functional recovery driven by NRG1-ErbB2 signaling, as well as a novel regeneration modulator capable of potential therapeutic effects.
Collapse
Affiliation(s)
- Deepak Prasad Gupta
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic of Korea; Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Anup Bhusal
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jae-Hong Kim
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jaemyung Jang
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyun Jin Jung
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Un-Kyung Kim
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jin-Sung Park
- Department of Neurology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Lee-So Maeng
- Department of Hospital Pathology, Incheon St. Mary's Hospital College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Gyun Jee Song
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic of Korea; Department of Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea.
| |
Collapse
|
11
|
Sneddon WB, Friedman PA, Mamonova T. Mutations in an unrecognized internal NPT2A PDZ motif disrupt phosphate transport and cause congenital hypophosphatemia. Biochem J 2023; 480:685-699. [PMID: 37132631 PMCID: PMC10442799 DOI: 10.1042/bcj20230020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/04/2023]
Abstract
The Na+-dependent phosphate cotransporter-2A (NPT2A, SLC34A1) is a primary regulator of extracellular phosphate homeostasis. Its most prominent structural element is a carboxy-terminal PDZ ligand that binds Na+/H+ Exchanger Regulatory Factor-1 (NHERF1, SLC9A3R1). NHERF1, a multidomain PDZ protein, establishes NPT2A membrane localization and is required for hormone-inhibitable phosphate transport. NPT2A also possesses an uncharacterized internal PDZ ligand. Two recent clinical reports describe congenital hypophosphatemia in children harboring Arg495His or Arg495Cys variants within the internal PDZ motif. The wild-type internal 494TRL496 PDZ ligand binds NHERF1 PDZ2, which we consider a regulatory domain. Ablating the internal PDZ ligand with a 494AAA496 substitution blocked hormone-inhibitable phosphate transport. Complementary approaches, including CRISPR/Cas9 technology, site-directed mutagenesis, confocal microscopy, and modeling, showed that NPT2A Arg495His or Arg495Cys variants do not support PTH or FGF23 action on phosphate transport. Coimmunoprecipitation experiments indicate that both variants bind NHERF1 similarly to WT NPT2A. However, in contrast with WT NPT2A, NPT2A Arg495His, or Arg495Cys variants remain at the apical membrane and are not internalized in response to PTH. We predict that Cys or His substitution of the charged Arg495 changes the electrostatics, preventing phosphorylation of the upstream Thr494, interfering with phosphate uptake in response to hormone action, and inhibiting NPT2A trafficking. We advance a model wherein the carboxy-terminal PDZ ligand defines apical localization NPT2A, while the internal PDZ ligand is essential for hormone-triggered phosphate transport.
Collapse
Affiliation(s)
- W. Bruce Sneddon
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Peter A. Friedman
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Tatyana Mamonova
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| |
Collapse
|
12
|
Petzold F, Schönauer R, Werner A, Halbritter J. Clinical and Functional Assessment of Digenicity in Renal Phosphate Wasting. Nutrients 2023; 15:2081. [PMID: 37432176 DOI: 10.3390/nu15092081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 07/12/2023] Open
Abstract
Apart from increased fluid intake, patients with kidney stone disease (KSD) due to renal phosphate wasting require specific metaphylaxis. NaPi2a, NaPi2c, and NHERF1 regulate plasma phosphate concentration by reabsorbing phosphate in proximal kidney tubules and have been found altered in monogenic hypophosphatemia with a risk of KSD. In this study, we aimed at assessing the combined genetic alterations impacting NaPi2a, NaPi2c, and NHERF1. Therefore, we screened our hereditary KSD registry for cases of oligo- and digenicity, conducted reverse phenotyping, and undertook functional studies. As a result, we identified three patients from two families with digenic alterations in NaPi2a, NaPi2c, and NHERF1. In family 1, the index patient, who presented with severe renal calcifications and a bone mineralization disorder, carried digenic alterations affecting both NaPi transporter 2a and 2c. Functional analysis confirmed an additive genetic effect. In family 2, the index patient presented with kidney function decline, distinct musculature-related symptoms, and intracellular ATP depletion. Genetically, this individual was found to harbor variants in both NaPi2c and NHERF1 pointing towards genetic interaction. In summary, digenicity and gene dosage are likely to impact the severity of renal phosphate wasting and should be taken into account in terms of metaphylaxis through phosphate substitution.
Collapse
Affiliation(s)
- Friederike Petzold
- Division of Nephrology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Ria Schönauer
- Division of Nephrology, University of Leipzig Medical Center, 04103 Leipzig, Germany
- Department of Nephrology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Andreas Werner
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Jan Halbritter
- Division of Nephrology, University of Leipzig Medical Center, 04103 Leipzig, Germany
- Department of Nephrology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
13
|
Sneddon WB, Friedman PA, Mamonova T. Mutations in an unrecognized internal NPT2A PDZ motif disrupt phosphate transport causing congenital hypophosphatemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531332. [PMID: 36945373 PMCID: PMC10028803 DOI: 10.1101/2023.03.06.531332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The Na + -dependent phosphate cotransporter-2A (NPT2A, SLC34A1) is a primary regulator of extracellular phosphate homeostasis. Its most prominent structural element is a carboxy-terminal PDZ ligand that binds Na + /H + Exchanger Regulatory Factor-1 (NHERF1, SLC9A3R1). NHERF1, a multidomain PDZ protein,establishes NPT2A membrane localization and is required for hormone-sensitive phosphate transport. NPT2A also possesses an uncharacterized internal PDZ ligand. Two recent clinical reports describe congenital hypophosphatemia in children harboring Arg 495 His or Arg 495 Cys variants within the internal PDZ motif. The wild-type internal 494 TRL 496 PDZ ligand binds NHERF1 PDZ2, which we consider a regulatory domain. Ablating the internal PDZ ligand with a 494 AAA 496 substitution blocked hormone-sensitive phosphate transport. Complementary approaches, including CRISPR/Cas9 technology, site-directed mutagenesis, confocal microscopy, and modeling, showed that NPT2A Arg 495 His or Arg 495 Cys variants do not support PTH or FGF23 action on phosphate transport. Coimmunoprecipitation experiments indicate that both variants bind NHERF1 similarly to WT NPT2A. However, in contrast to WT NPT2A, NPT2A Arg 495 His or Arg 495 Cys variants remain at the apical membrane and are not internalized in response to PTH. We predict that Cys or His substitution of the charged Arg 495 changes the electrostatics, preventing phosphorylation of the upstream Thr 494 , interfering with phosphate uptake in response to hormone action, and inhibiting NPT2A trafficking. We advance a model wherein the carboxyterminal PDZ ligand defines apical localization NPT2A, while the internal PDZ ligand is essential for hormone-triggered phosphate transport.
Collapse
|
14
|
Van de Perre E, Bazin D, Estrade V, Bouderlique E, Wissing KM, Daudon M, Letavernier E. Randall’s plaque as the origin of idiopathic calcium oxalate stone formation: an update. CR CHIM 2022. [DOI: 10.5802/crchim.102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
15
|
Ford C, Burd CG. GOPC facilitates the sorting of syndecan-1 in polarized epithelial cells. Mol Biol Cell 2022; 33:ar86. [PMID: 35830596 PMCID: PMC9582621 DOI: 10.1091/mbc.e22-05-0165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 11/11/2022] Open
Abstract
The trans-Golgi network must coordinate sorting and secretion of proteins and lipids to intracellular organelles and the plasma membrane. During polarization of epithelial cells, changes in the lipidome and the expression and distribution of proteins contribute to the formation of apical and basolateral plasma membrane domains. Previous studies using HeLa cells show that the syndecan-1 transmembrane domain confers sorting within sphingomyelin-rich vesicles in a sphingomyelin secretion pathway. In polarized Madin-Darby canine kidney cells, we reveal differences in the sorting of syndecan-1, whereupon the correct trafficking of the protein is not dependent on its transmembrane domain and changes in sphingomyelin content of cells during polarization. Instead, we reveal that correct basolateral targeting of syndecan-1 requires a full-length PDZ motif in syndecan-1 and the PDZ domain golgin protein GOPC. Moreover, we reveal changes in Golgi morphology elicited by GOPC overexpression. These results suggest that the role of GOPC in sorting syndecan-1 is indirect and likely due to GOPC effects on Golgi organization.
Collapse
Affiliation(s)
- Charlotte Ford
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520
| | | |
Collapse
|
16
|
Friedman PA, Sneddon WB, Mamonova T, Montanez-Miranda C, Ramineni S, Harbin NH, Squires KE, Gefter JV, Magyar CE, Emlet DR, Hepler JR. RGS14 regulates PTH- and FGF23-sensitive NPT2A-mediated renal phosphate uptake via binding to the NHERF1 scaffolding protein. J Biol Chem 2022; 298:101836. [PMID: 35307350 PMCID: PMC9035407 DOI: 10.1016/j.jbc.2022.101836] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 02/06/2023] Open
Abstract
Phosphate homeostasis, mediated by dietary intake, renal absorption, and bone deposition, is incompletely understood because of the uncharacterized roles of numerous implicated protein factors. Here, we identified a novel role for one such element, regulator of G protein signaling 14 (RGS14), suggested by genome-wide association studies to associate with dysregulated Pi levels. We show that human RGS14 possesses a carboxy-terminal PDZ ligand required for sodium phosphate cotransporter 2a (NPT2A) and sodium hydrogen exchanger regulatory factor-1 (NHERF1)-mediated renal Pi transport. In addition, we found using isotope uptake measurements combined with bioluminescence resonance energy transfer assays, siRNA knockdown, pull-down and overlay assays, and molecular modeling that secreted proteins parathyroid hormone (PTH) and fibroblast growth factor 23 inhibited Pi uptake by inducing dissociation of the NPT2A-NHERF1 complex. PTH failed to affect Pi transport in cells expressing RGS14, suggesting that it suppresses hormone-sensitive but not basal Pi uptake. Interestingly, RGS14 did not affect PTH-directed G protein activation or cAMP formation, implying a postreceptor site of action. Further pull-down experiments and direct binding assays indicated that NPT2A and RGS14 bind distinct PDZ domains on NHERF1. We showed that RGS14 expression in human renal proximal tubule epithelial cells blocked the effects of PTH and fibroblast growth factor 23 and stabilized the NPT2A-NHERF1 complex. In contrast, RGS14 genetic variants bearing mutations in the PDZ ligand disrupted RGS14 binding to NHERF1 and subsequent PTH-sensitive Pi transport. In conclusion, these findings identify RGS14 as a novel regulator of hormone-sensitive Pi transport. The results suggest that changes in RGS14 function or abundance may contribute to the hormone resistance and hyperphosphatemia observed in kidney diseases.
Collapse
Affiliation(s)
- Peter A Friedman
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - W Bruce Sneddon
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Tatyana Mamonova
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Carolina Montanez-Miranda
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Suneela Ramineni
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nicholas H Harbin
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Katherine E Squires
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Julia V Gefter
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Clara E Magyar
- Department of Pathology and Laboratory Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - David R Emlet
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - John R Hepler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
17
|
Irsik DL, Bollag WB, Isales CM. Renal Contributions to Age-Related Changes in Mineral Metabolism. JBMR Plus 2021; 5:e10517. [PMID: 34693188 PMCID: PMC8520061 DOI: 10.1002/jbm4.10517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/28/2021] [Accepted: 05/09/2021] [Indexed: 11/10/2022] Open
Abstract
Aging results in a general decline in function in most systems. This is particularly true with respect to the skeleton and renal systems, impacting mineral homeostasis. Calcium and phosphate regulation requires tight coordination among the intestine, bone, parathyroid gland, and kidney. The role of the intestine is to absorb calcium and phosphate from the diet. The bone stores or releases calcium and phosphate depending on the body's needs. In response to low plasma ionized calcium concentration, the parathyroid gland produces parathyroid hormone, which modulates bone turnover. The kidney reabsorbs or excretes the minerals and serves as the final regulator of plasma concentration. Many hormones are involved in this process in addition to parathyroid hormone, including fibroblast growth factor 23 produced by the bone and calcitriol synthesized by the kidney. Sclerostin, calcitonin, osteoprotegerin, and receptor activator of nuclear factor‐κB ligand also contribute to tissue‐specific regulation. Changes in the function of organs due to aging or disease can perturb this balance. During aging, the intestine cannot absorb calcium efficiently due to decreased expression of key proteins. In the bone, the balance between bone formation and bone resorption tends toward the latter in older individuals. The kidney may not filter blood as efficiently in the later decades of life, and the expression of certain proteins necessary for mineral homeostasis declines with age. These changes often lead to dysregulation of organismal mineral homeostasis. This review will focus on how mineral homeostasis is impacted by aging with a particular emphasis on the kidney's role in this process. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Debra L Irsik
- Charlie Norwood VA Medical Center Augusta GA USA.,Department of Neuroscience and Regenerative Medicine Augusta University Augusta GA USA
| | - Wendy B Bollag
- Charlie Norwood VA Medical Center Augusta GA USA.,Department of Physiology Augusta University Augusta GA USA
| | - Carlos M Isales
- Department of Neuroscience and Regenerative Medicine Augusta University Augusta GA USA.,Division of Endocrinology, Department of Medicine Augusta University Augusta GA USA
| |
Collapse
|
18
|
Sun HL, Wu YW, Bian HG, Yang H, Wang H, Meng XM, Jin J. Function of Uric Acid Transporters and Their Inhibitors in Hyperuricaemia. Front Pharmacol 2021; 12:667753. [PMID: 34335246 PMCID: PMC8317579 DOI: 10.3389/fphar.2021.667753] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Disorders of uric acid metabolism may be associated with pathological processes in many diseases, including diabetes mellitus, cardiovascular disease, and kidney disease. These diseases can further promote uric acid accumulation in the body, leading to a vicious cycle. Preliminary studies have proven many mechanisms such as oxidative stress, lipid metabolism disorders, and rennin angiotensin axis involving in the progression of hyperuricaemia-related diseases. However, there is still lack of effective clinical treatment for hyperuricaemia. According to previous research results, NPT1, NPT4, OAT1, OAT2, OAT3, OAT4, URAT1, GLUT9, ABCG2, PDZK1, these urate transports are closely related to serum uric acid level. Targeting at urate transporters and urate-lowering drugs can enhance our understanding of hyperuricaemia and hyperuricaemia-related diseases. This review may put forward essential references or cross references to be contributed to further elucidate traditional and novel urate-lowering drugs benefits as well as provides theoretical support for the scientific research on hyperuricemia and related diseases.
Collapse
Affiliation(s)
- Hao-Lu Sun
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Yi-Wan Wu
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - He-Ge Bian
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Hui Yang
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Heng Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Juan Jin
- Department of Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
19
|
Bushau-Sprinkle AM, Barati MT, Zheng Y, Watson WH, Gagnon KB, Khundmiri SJ, Kitterman KT, Clark BJ, Siskind LJ, Doll MA, Brier ME, Coventry S, Lederer ED. Na/H Exchange Regulatory Factor 1 Deficient Mice Show Evidence of Oxidative Stress and Altered Cisplatin Pharmacokinetics. Antioxidants (Basel) 2021; 10:1036. [PMID: 34203453 PMCID: PMC8300832 DOI: 10.3390/antiox10071036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/22/2022] Open
Abstract
(1) Background: One third of patients who receive cisplatin develop an acute kidney injury. We previously demonstrated the Na/H Exchange Regulatory Factor 1 (NHERF1) loss resulted in increased kidney enzyme activity of the pentose phosphate pathway and was associated with more severe cisplatin nephrotoxicity. We hypothesized that changes in proximal tubule biochemical pathways associated with NHERF1 loss alters renal metabolism of cisplatin or response to cisplatin, resulting in exacerbated nephrotoxicity. (2) Methods: 2-4 month-old male wild-type and NHERF1 knock out littermate mice were treated with either vehicle or cisplatin (20 mg/kg dose IP), with samples taken at either 4, 24, or 72 h. Kidney injury was determined by urinary neutrophil gelatinase-associated lipocalin and histology. Glutathione metabolites were measured by HPLC and genes involved in glutathione synthesis were measured by qPCR. Kidney handling of cisplatin was assessed by a kidney cortex measurement of γ-glutamyl transferase activity, Western blot for γ-glutamyl transferase and cysteine S-conjugate beta lyase, and ICP-MS for platinum content. (3) Results: At 24 h knock out kidneys show evidence of greater tubular injury after cisplatin and exhibit a decreased reduced/oxidized glutathione ratio under baseline conditions in comparison to wild-type. KO kidneys fail to show an increase in γ-glutamyl transferase activity and experience a more rapid decline in tissue platinum when compared to wild-type. (4) Conclusions: Knock out kidneys show evidence of greater oxidative stress than wild-type accompanied by a greater degree of early injury in response to cisplatin. NHERF1 loss has no effect on the initial accumulation of cisplatin in the kidney cortex but is associated with an altered redox status which may alter the activity of enzymes involved in cisplatin metabolism.
Collapse
Affiliation(s)
- Adrienne M. Bushau-Sprinkle
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; (A.M.B.-S.); (Y.Z.); (W.H.W.); (L.J.S.); (M.A.D.); (M.E.B.)
| | - Michelle T. Barati
- Department of Medicine, Division of Nephrology, University of Louisville, Louisville, KY 40202, USA; (M.T.B.); (K.T.K.)
| | - Yuxuan Zheng
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; (A.M.B.-S.); (Y.Z.); (W.H.W.); (L.J.S.); (M.A.D.); (M.E.B.)
| | - Walter H. Watson
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; (A.M.B.-S.); (Y.Z.); (W.H.W.); (L.J.S.); (M.A.D.); (M.E.B.)
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40202, USA
| | - Kenneth B. Gagnon
- Division of Nephrology and Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Department of Medicine, Dallas, TX 75390, USA;
| | - Syed Jalal Khundmiri
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC 20059, USA;
| | - Kathleen T. Kitterman
- Department of Medicine, Division of Nephrology, University of Louisville, Louisville, KY 40202, USA; (M.T.B.); (K.T.K.)
| | - Barbara J. Clark
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA;
| | - Leah J. Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; (A.M.B.-S.); (Y.Z.); (W.H.W.); (L.J.S.); (M.A.D.); (M.E.B.)
| | - Mark A. Doll
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; (A.M.B.-S.); (Y.Z.); (W.H.W.); (L.J.S.); (M.A.D.); (M.E.B.)
| | - Michael E. Brier
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; (A.M.B.-S.); (Y.Z.); (W.H.W.); (L.J.S.); (M.A.D.); (M.E.B.)
- Department of Medicine, Division of Nephrology, University of Louisville, Louisville, KY 40202, USA; (M.T.B.); (K.T.K.)
| | - Susan Coventry
- Department of Pathology, University of Louisville, Louisville, KY 40202, USA;
- Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Eleanor D. Lederer
- Division of Nephrology and Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Department of Medicine, Dallas, TX 75390, USA;
- VA North Texas Health Sciences Center, Dallas, TX 75216, USA
| |
Collapse
|
20
|
Kammala AK, Bahal D, Yang C, Panettieri RA, Das R, Subramanian H. Na +/H + Exchanger Regulatory Factor 1 Mediates the Pathogenesis of Airway Inflammation in a Murine Model of House Dust Mite-Induced Asthma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2301-2311. [PMID: 33952618 PMCID: PMC8113128 DOI: 10.4049/jimmunol.2001199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/16/2021] [Indexed: 11/19/2022]
Abstract
Na+/H+ exchanger regulatory factor 1 (NHERF1), a class I PDZ-binding protein, regulates G protein-coupled receptor signaling in some cell types. NHERF1 also functions as a scaffolding protein and activates non-G protein-coupled receptor signaling pathways, thereby contributing to the pathogenesis of various diseases. Although we have previously shown that NHERF1 regulates mast cell functions, there is little information regarding the role of NHERF1 in other immune cells. How NHERF1 regulates the pathogenesis of allergic disease such as asthma also remains unknown. In the current study, we show that NHERF1 promotes allergic airway inflammation in a house dust mite extract (HDME)-induced mouse model of asthma. Specifically, HDME-specific serum IgE levels, airway leukocyte numbers, and goblet cell hyperplasia were reduced in NHERF1+/- mice as compared with NHERF1+/+ mice. Interestingly, the gene expression of inflammatory (IL-17a, IL-25, and IL-33) as well as T helper 2 (Th2) cytokines (IL-4, IL-5, and IL-13) and several chemokines that recruit eosinophils, neutrophils, and lymphocytes were also decreased in the lungs of NHERF1+/- mice exposed to HDME. Consistent with these observations, microRNAs regulating mucus production, inflammation, Th2 effector functions, and IL-13 expression were increased in the lungs of HDME-treated NHERF1+/- mice. Overall, our studies reveal a unique role for NHERF1 in regulating asthma pathogenesis, and further elucidation of the mechanisms through which NHERF1 modulates allergic inflammation will lead to the development of new therapeutic strategies for asthma.
Collapse
Affiliation(s)
- Ananth K Kammala
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Devika Bahal
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Canchai Yang
- Department of Physiology, Michigan State University, East Lansing, MI
| | | | - Rupali Das
- Department of Physiology, Michigan State University, East Lansing, MI
| | | |
Collapse
|
21
|
Vistrup-Parry M, Sneddon WB, Bach S, Strømgaard K, Friedman PA, Mamonova T. Multisite NHERF1 phosphorylation controls GRK6A regulation of hormone-sensitive phosphate transport. J Biol Chem 2021; 296:100473. [PMID: 33639163 PMCID: PMC8042174 DOI: 10.1016/j.jbc.2021.100473] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The type II sodium-dependent phosphate cotransporter (NPT2A) mediates renal phosphate uptake. The NPT2A is regulated by parathyroid hormone (PTH) and fibroblast growth factor 23, which requires Na+/H+ exchange regulatory factor-1 (NHERF1), a multidomain PDZ-containing phosphoprotein. Phosphocycling controls the association between NHERF1 and the NPT2A. Here, we characterize the critical involvement of G protein–coupled receptor kinase 6A (GRK6A) in mediating PTH-sensitive phosphate transport by targeted phosphorylation coupled with NHERF1 conformational rearrangement, which in turn allows phosphorylation at a secondary site. GRK6A, through its carboxy-terminal PDZ recognition motif, binds NHERF1 PDZ1 with greater affinity than PDZ2. However, the association between NHERF1 PDZ2 and GRK6A is necessary for PTH action. Ser162, a PKCα phosphorylation site in PDZ2, regulates the binding affinity between PDZ2 and GRK6A. Substitution of Ser162 with alanine (S162A) blocks the PTH action but does not disrupt the interaction between NHERF1 and the NPT2A. Replacement of Ser162 with aspartic acid (S162D) abrogates the interaction between NHERF1 and the NPT2A and concurrently PTH action. We used amber codon suppression to generate a phosphorylated Ser162(pSer162)-PDZ2 variant. KD values determined by fluorescence anisotropy indicate that incorporation of pSer162 increased the binding affinity to the carboxy terminus of GRK6A 2-fold compared with WT PDZ2. Molecular dynamics simulations predict formation of an electrostatic network between pSer162 and Asp183 of PDZ2 and Arg at position −1 of the GRK6A PDZ-binding motif. Our results suggest that PDZ2 plays a regulatory role in PTH-sensitive NPT2A-mediated phosphate transport and phosphorylation of Ser162 in PDZ2 modulates the interaction with GRK6A.
Collapse
Affiliation(s)
- Maria Vistrup-Parry
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - W Bruce Sneddon
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sofie Bach
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Peter A Friedman
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Tatyana Mamonova
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
22
|
Kowalczyk I, Lee C, Schuster E, Hoeren J, Trivigno V, Riedel L, Görne J, Wallingford JB, Hammes A, Feistel K. Neural tube closure requires the endocytic receptor Lrp2 and its functional interaction with intracellular scaffolds. Development 2021; 148:dev195008. [PMID: 33500317 PMCID: PMC7860117 DOI: 10.1242/dev.195008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022]
Abstract
Pathogenic mutations in the endocytic receptor LRP2 in humans are associated with severe neural tube closure defects (NTDs) such as anencephaly and spina bifida. Here, we have combined analysis of neural tube closure in mouse and in the African Clawed Frog Xenopus laevis to elucidate the etiology of Lrp2-related NTDs. Lrp2 loss of function impaired neuroepithelial morphogenesis, culminating in NTDs that impeded anterior neural plate folding and neural tube closure in both model organisms. Loss of Lrp2 severely affected apical constriction as well as proper localization of the core planar cell polarity (PCP) protein Vangl2, demonstrating a highly conserved role of the receptor in these processes, which are essential for neural tube formation. In addition, we identified a novel functional interaction of Lrp2 with the intracellular adaptor proteins Shroom3 and Gipc1 in the developing forebrain. Our data suggest that, during neurulation, motifs within the intracellular domain of Lrp2 function as a hub that orchestrates endocytic membrane removal for efficient apical constriction, as well as PCP component trafficking in a temporospatial manner.
Collapse
Affiliation(s)
- Izabela Kowalczyk
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Chanjae Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Elisabeth Schuster
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Josefine Hoeren
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Valentina Trivigno
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Levin Riedel
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Jessica Görne
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - John B Wallingford
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Annette Hammes
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Kerstin Feistel
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| |
Collapse
|
23
|
Mamonova T, Friedman PA. Noncanonical Sequences Involving NHERF1 Interaction with NPT2A Govern Hormone-Regulated Phosphate Transport: Binding Outside the Box. Int J Mol Sci 2021; 22:1087. [PMID: 33499384 PMCID: PMC7866199 DOI: 10.3390/ijms22031087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/27/2022] Open
Abstract
Na+/H+ exchange factor-1 (NHERF1), a multidomain PDZ scaffolding phosphoprotein, is required for the type II sodium-dependent phosphate cotransporter (NPT2A)-mediated renal phosphate absorption. Both PDZ1 and PDZ2 domains are involved in NPT2A-dependent phosphate uptake. Though harboring identical core-binding motifs, PDZ1 and PDZ2 play entirely different roles in hormone-regulated phosphate transport. PDZ1 is required for the interaction with the C-terminal PDZ-binding sequence of NPT2A (-TRL). Remarkably, phosphocycling at Ser290 distant from PDZ1, the penultimate step for both parathyroid hormone (PTH) and fibroblast growth factor-23 (FGF23) regulation, controls the association between NHERF1 and NPT2A. PDZ2 interacts with the C-terminal PDZ-recognition motif (-TRL) of G Protein-coupled Receptor Kinase 6A (GRK6A), and that promotes phosphorylation of Ser290. The compelling biological puzzle is how PDZ1 and PDZ2 with identical GYGF core-binding motifs specifically recognize distinct binding partners. Binding determinants distinct from the canonical PDZ-ligand interactions and located "outside the box" explain PDZ domain specificity. Phosphorylation of NHERF1 by diverse kinases and associated conformational changes in NHERF1 add more complexity to PDZ-binding diversity.
Collapse
Affiliation(s)
- Tatyana Mamonova
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA;
| | | |
Collapse
|
24
|
Koumakis E, Cormier C, Roux C, Briot K. The Causes of Hypo- and Hyperphosphatemia in Humans. Calcif Tissue Int 2021; 108:41-73. [PMID: 32285168 DOI: 10.1007/s00223-020-00664-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022]
Abstract
Phosphate homeostasis involves several major organs that are the skeleton, the intestine, the kidney, and parathyroid glands. Major regulators of phosphate homeostasis are parathormone, fibroblast growth factor 23, 1,25-dihydroxyvitamin D, which respond to variations of serum phosphate levels and act to increase or decrease intestinal absorption and renal tubular reabsorption, through the modulation of expression of transcellular transporters at the intestinal and/or renal tubular level. Any acquired or genetic dysfunction in these major organs or regulators may induce hypo- or hyperphosphatemia. The causes of hypo- and hyperphosphatemia are numerous. This review develops the main causes of acquired and genetic hypo- and hyperphosphatemia.
Collapse
Affiliation(s)
- Eugénie Koumakis
- Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Reference Center for Rare Genetic Bone Disorders, OSCAR Filière, Rheumatology Department, Cochin Hospital, AP-HP Centre-Paris University, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France.
| | - Catherine Cormier
- Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Reference Center for Rare Genetic Bone Disorders, OSCAR Filière, Rheumatology Department, Cochin Hospital, AP-HP Centre-Paris University, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Christian Roux
- Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Reference Center for Rare Genetic Bone Disorders, OSCAR Filière, Rheumatology Department, Cochin Hospital, AP-HP Centre-Paris University, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Karine Briot
- Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Reference Center for Rare Genetic Bone Disorders, OSCAR Filière, Rheumatology Department, Cochin Hospital, AP-HP Centre-Paris University, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France
| |
Collapse
|
25
|
Kammala AK, Syed M, Yang C, Occhiuto CJ, Subramanian H. A Critical Role for Na +/H + Exchanger Regulatory Factor 1 in Modulating FcεRI-Mediated Mast Cell Activation. THE JOURNAL OF IMMUNOLOGY 2020; 206:471-480. [PMID: 33361207 DOI: 10.4049/jimmunol.2000671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023]
Abstract
Mast cells are tissue-resident immune cells that play pivotal roles in initiating and amplifying allergic/anaphylactic reactions in humans. Their activation occurs via multiple mechanisms, which include cross-linking of the IgE-bound, high-affinity IgE receptors (FcεRI) by allergens or Ags and the binding of anaphylatoxins such as C3a to its receptor, C3aR. We have previously demonstrated that the Na+/H+ exchanger regulatory factor 1 (NHERF1) promotes C3aR functions in human mast cells. In the current study, we show that NHERF1 regulates mast cell response following FcεRI stimulation. Specifically, intracellular Ca2+ mobilization, activation of the MAPKs (ERK1/2 and P38), and production of cytokines (IL-13 and IL-6) following exposure to IgE/Ag were significantly reduced in mast cells from NHERF1+/‒ mice. In agreement with our in vitro data, mast cell-mediated passive cutaneous anaphylaxis and passive systemic anaphylaxis were reduced in NHERF1+/‒ mice and mast cell-deficient KitW-sh/W-sh mice engrafted with NHERF1+/‒ mast cells. Mechanistically, the levels of microRNAs (miRNAs) that regulate mast cell responses, miRNA 155-3p and miRNA 155-5p, were altered in mast cells from NHERF1+/‒ mice. Moreover, NHERF1 rapidly localized to the nucleus of mast cells following FcεRI stimulation. In summary, our results suggest that the NHERF1 acts as an adapter molecule and promotes IgE/Ag-induced mast cell activation. Further elucidating the mechanisms through which NHERF1 modulates mast cell responses will lend insights into the development of new therapeutic strategies to target mast cells during anaphylaxis or other allergic diseases.
Collapse
Affiliation(s)
- Ananth K Kammala
- Department of Physiology, Michigan State University, East Lansing, MI 48824
| | - Meesum Syed
- Department of Physiology, Michigan State University, East Lansing, MI 48824
| | - Canchai Yang
- Department of Physiology, Michigan State University, East Lansing, MI 48824
| | | | | |
Collapse
|
26
|
NHERF1 Loss Upregulates Enzymes of the Pentose Phosphate Pathway in Kidney Cortex. Antioxidants (Basel) 2020; 9:antiox9090862. [PMID: 32937931 PMCID: PMC7554817 DOI: 10.3390/antiox9090862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
(1) Background: We previously showed Na/H exchange regulatory factor 1 (NHERF1) loss resulted in increased susceptibility to cisplatin nephrotoxicity. NHERF1-deficient cultured proximal tubule cells and proximal tubules from NHERF1 knockout (KO) mice exhibit altered mitochondrial protein expression and poor survival. We hypothesized that NHERF1 loss results in changes in metabolic pathways and/or mitochondrial dysfunction, leading to increased sensitivity to cisplatin nephrotoxicity. (2) Methods: Two to 4-month-old male wildtype (WT) and KO mice were treated with vehicle or cisplatin (20 mg/kg dose IP). After 72 h, kidney cortex homogenates were utilized for metabolic enzyme activities. Non-treated kidneys were used to isolate mitochondria for mitochondrial respiration via the Seahorse XF24 analyzer. Non-treated kidneys were also used for LC-MS analysis to evaluate kidney ATP abundance, and electron microscopy (EM) was utilized to evaluate mitochondrial morphology and number. (3) Results: KO mouse kidneys exhibit significant increases in malic enzyme and glucose-6 phosphate dehydrogenase activity under baseline conditions but in no other gluconeogenic or glycolytic enzymes. NHERF1 loss does not decrease kidney ATP content. Mitochondrial morphology, number, and area appeared normal. Isolated mitochondria function was similar between WT and KO. Conclusions: KO kidneys experience a shift in metabolism to the pentose phosphate pathway, which may sensitize them to the oxidative stress imposed by cisplatin.
Collapse
|
27
|
Deciphering ion transporters, kinases and PDZ-adaptor molecules that mediate guanylate cyclase C agonist-dependent intestinal fluid loss in vivo. Biochem Pharmacol 2020; 178:114040. [DOI: 10.1016/j.bcp.2020.114040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/13/2020] [Indexed: 01/12/2023]
|
28
|
Gordon RJ, Li D, Doyle D, Zaritsky J, Levine MA. Digenic Heterozygous Mutations in SLC34A3 and SLC34A1 Cause Dominant Hypophosphatemic Rickets with Hypercalciuria. J Clin Endocrinol Metab 2020; 105:dgaa217. [PMID: 32311027 PMCID: PMC7448300 DOI: 10.1210/clinem/dgaa217] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Hypophosphatemia and metabolic bone disease are associated with hereditary hypophosphatemic rickets with hypercalciuria (HHRH) due to biallelic mutations of SLC34A3 encoding the NPT2C sodium-phosphate cotransporter and nephrolithiasis/osteoporosis, hypophosphatemic 1 (NPHLOP1) due to monoallelic mutations in SLC34A1 encoding the NPT2A sodium-phosphate cotransporter. OBJECTIVE To identify a genetic cause of apparent dominant transmission of HHRH. DESIGN AND SETTING Retrospective and prospective analysis of clinical and molecular characteristics of patients studied in 2 academic medical centers. METHODS We recruited 4 affected and 3 unaffected members of a 4-generation family in which the proband presented with apparent HHRH. We performed clinical examinations, biochemical and radiological analyses, and molecular studies of genomic DNA. RESULTS The proband and her affected sister and mother carried pathogenic heterozygous mutations in 2 related genes, SLC34A1 (exon 13, c.1535G>A; p.R512H) and SLC34A3 (exon 13, c.1561dupC; L521Pfs*72). The proband and her affected sister inherited both gene mutations from their mother, while their clinically less affected brother, father, and paternal grandmother carried only the SLC34A3 mutation. Renal phosphate-wasting exhibited both a gene dosage-effect and an age-dependent attenuation of severity. CONCLUSIONS We describe a kindred with autosomal dominant hypophosphatemic rickets in which whole exome analysis identified digenic heterozygous mutations in SLC34A1 and SLC34A3. Subjects with both mutations were more severely affected than subjects carrying only one mutation. These findings highlight the challenges of assigning causality to plausible genetic variants in the next generation sequencing era.
Collapse
Affiliation(s)
- Rebecca J Gordon
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Division of Endocrinology and Diabetes and the Center for Bone Health, The Children’s Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Dong Li
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Daniel Doyle
- Division of Pediatric Endocrinology, Sidney Kimmel Medical College of Thomas Jefferson University and Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Joshua Zaritsky
- Division of Pediatric Nephrology, Sidney Kimmel Medical College of Thomas Jefferson University and Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Michael A Levine
- Division of Endocrinology and Diabetes and the Center for Bone Health, The Children’s Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Abstract
Kidney stone disease (nephrolithiasis) is a common problem that can be associated with alterations in urinary solute composition including hypercalciuria. Studies suggest that the prevalence of monogenic kidney stone disorders, including renal tubular acidosis with deafness, Bartter syndrome, primary hyperoxaluria and cystinuria, in patients attending kidney stone clinics is ∼15%. However, for the majority of individuals, nephrolithiasis has a multifactorial aetiology involving genetic and environmental factors. Nonetheless, the genetic influence on stone formation in these idiopathic stone formers remains considerable and twin studies estimate a heritability of >45% for nephrolithiasis and >50% for hypercalciuria. The contribution of polygenic influences from multiple loci have been investigated by genome-wide association and candidate gene studies, which indicate that a number of genes and molecular pathways contribute to the risk of stone formation. Genetic approaches, studying both monogenic and polygenic factors in nephrolithiasis, have revealed that the following have important roles in the aetiology of kidney stones: transporters and channels; ions, protons and amino acids; the calcium-sensing receptor (a G protein-coupled receptor) signalling pathway; and the metabolic pathways for vitamin D, oxalate, cysteine, purines and uric acid. These advances, which have increased our understanding of the pathogenesis of nephrolithiasis, will hopefully facilitate the future development of targeted therapies for precision medicine approaches in patients with nephrolithiasis.
Collapse
Affiliation(s)
- Sarah A Howles
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
30
|
Abstract
Phosphate is an essential nutrient for life and is a critical component of bone formation, a major signaling molecule, and structural component of cell walls. Phosphate is also a component of high-energy compounds (i.e., AMP, ADP, and ATP) and essential for nucleic acid helical structure (i.e., RNA and DNA). Phosphate plays a central role in the process of mineralization, normal serum levels being associated with appropriate bone mineralization, while high and low serum levels are associated with soft tissue calcification. The serum concentration of phosphate and the total body content of phosphate are highly regulated, a process that is accomplished by the coordinated effort of two families of sodium-dependent transporter proteins. The three isoforms of the SLC34 family (SLC34A1-A3) show very restricted tissue expression and regulate intestinal absorption and renal excretion of phosphate. SLC34A2 also regulates the phosphate concentration in multiple lumen fluids including milk, saliva, pancreatic fluid, and surfactant. Both isoforms of the SLC20 family exhibit ubiquitous expression (with some variation as to which one or both are expressed), are regulated by ambient phosphate, and likely serve the phosphate needs of the individual cell. These proteins exhibit similarities to phosphate transporters in nonmammalian organisms. The proteins are nonredundant as mutations in each yield unique clinical presentations. Further research is essential to understand the function, regulation, and coordination of the various phosphate transporters, both the ones described in this review and the phosphate transporters involved in intracellular transport.
Collapse
Affiliation(s)
- Nati Hernando
- University of Zurich-Irchel, Institute of Physiology, Zurich, Switzerland; Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky; and Robley Rex VA Medical Center, Louisville, Kentucky
| | - Kenneth Gagnon
- University of Zurich-Irchel, Institute of Physiology, Zurich, Switzerland; Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky; and Robley Rex VA Medical Center, Louisville, Kentucky
| | - Eleanor Lederer
- University of Zurich-Irchel, Institute of Physiology, Zurich, Switzerland; Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky; and Robley Rex VA Medical Center, Louisville, Kentucky
| |
Collapse
|
31
|
Song GJ, Gupta DP, Rahman MH, Park HT, Al Ghouleh I, Bisello A, Lee MG, Park JY, Park HH, Jun JH, Chung KW, Choi BO, Suk K. Loss-of-function of EBP50 is a new cause of hereditary peripheral neuropathy: EBP50 functions in peripheral nerve system. Glia 2020; 68:1794-1809. [PMID: 32077526 DOI: 10.1002/glia.23805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022]
Abstract
Finding causative genetic mutations is important in the diagnosis and treatment of hereditary peripheral neuropathies. This study was conducted to find new genes involved in the pathophysiology of hereditary peripheral neuropathy. We identified a new mutation in the EBP50 gene, which is co-segregated with neuropathic phenotypes, including motor and sensory deficit in a family with Charcot-Marie-Tooth disease. EBP50 is known to be important for the formation of microvilli in epithelial cells, and the discovery of this gene mutation allowed us to study the function of EBP50 in the nervous system. EBP50 was strongly expressed in the nodal and paranodal regions of sciatic nerve fibers, where Schwann cell microvilli contact the axolemma, and at the growth tips of primary Schwann cells. In addition, EBP50 expression was decreased in mouse models of peripheral neuropathy. Knockout mice were used to study EBP50 function in the peripheral nervous system. Interestingly motor function deficit and abnormal histology of nerve fibers were observed in EBP50+/- heterozygous mice at 12 months of age, but not 3 months. in vitro studies using Schwann cells showed that NRG1-induced AKT activation and migration were significantly reduced in cells overexpressing the I325V mutant of EBP50 or cells with knocked-down EBP50 expression. In conclusion, we show for the first time that loss of function due to EBP50 gene deficiency or mutation can cause peripheral neuropathy.
Collapse
Affiliation(s)
- Gyun Jee Song
- Department of Medical Science, Institute for Bio-Medical Convergence, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Republic of Korea
| | - Deepak Prasad Gupta
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hwan Tae Park
- Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Imad Al Ghouleh
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Alessandro Bisello
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Maan-Gee Lee
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, Republic of Korea
| | - Hyun Ho Park
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Jin Hyun Jun
- Department of Senior Healthcare, BK21 Plus Program, Graduate School of Eulji University, Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Republic of Korea
| | - Ki Wha Chung
- Department of Biological Sciences, Kongju National University, Gongju, Republic of Korea
| | - Byung-Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
32
|
Abstract
Over the past 25 years, successive cloning of SLC34A1, SLC34A2 and SLC34A3, which encode the sodium-dependent inorganic phosphate (Pi) cotransport proteins 2a-2c, has facilitated the identification of molecular mechanisms that underlie the regulation of renal and intestinal Pi transport. Pi and various hormones, including parathyroid hormone and phosphatonins, such as fibroblast growth factor 23, regulate the activity of these Pi transporters through transcriptional, translational and post-translational mechanisms involving interactions with PDZ domain-containing proteins, lipid microdomains and acute trafficking of the transporters via endocytosis and exocytosis. In humans and rodents, mutations in any of the three transporters lead to dysregulation of epithelial Pi transport with effects on serum Pi levels and can cause cardiovascular and musculoskeletal damage, illustrating the importance of these transporters in the maintenance of local and systemic Pi homeostasis. Functional and structural studies have provided insights into the mechanism by which these proteins transport Pi, whereas in vivo and ex vivo cell culture studies have identified several small molecules that can modify their transport function. These small molecules represent potential new drugs to help maintain Pi homeostasis in patients with chronic kidney disease - a condition that is associated with hyperphosphataemia and severe cardiovascular and skeletal consequences.
Collapse
|
33
|
Bushau-Sprinkle AM, Lederer ED. New roles of the Na +/H + exchange regulatory factor 1 scaffolding protein: a review. Am J Physiol Renal Physiol 2020; 318:F804-F808. [PMID: 31984791 DOI: 10.1152/ajprenal.00467.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Na+/H+ exchange regulatory factor 1 (NHERF1), a member of a PDZ scaffolding protein family, was first identified as an organizer of membrane-bound protein complexes composed of hormone receptors, signal transduction pathways, and electrolyte and mineral transporters and channels. NHERF1 is involved in the regulation of Na+/H+ exchanger 3, Na+-dependent phosphate transporter 2a, and Na+-K+-ATPase through its ability to scaffold these transporters to the plasma membrane, allowing regulation of these protein complexes with their associated hormone receptors. Recently, NHERF1 has received increased interest in its involvement in a variety of functions, including cell structure and trafficking, tumorigenesis and tumor behavior, inflammatory responses, and tissue injury. In this review, we highlight the evidence for the expansive role of NHERF1 in cell biology and speculate on the implications for renal physiology and pathophysiology.
Collapse
Affiliation(s)
- Adrienne M Bushau-Sprinkle
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky.,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| | - Eleanor D Lederer
- Division of Nephrology, Department of Medicine, University of Louisville, Louisville, Kentucky.,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| |
Collapse
|
34
|
Abstract
Parathyroid hormone (PTH), PTH-related peptide (PTHrP), PTHR, and their cognate G protein-coupled receptor play defining roles in the regulation of extracellular calcium and phosphate metabolism and in controlling skeletal growth and repair. Acting through complex signaling mechanisms that in many instances proceed in a tissue-specific manner, precise control of these processes is achieved. A variety of direct and indirect disease processes, along with genetic anomalies, can cause these schemes to become dysfunctional. Here, we review the basic components of this regulatory network and present both the well-established elements and emerging findings and concepts with the overall objective to provide a framework for understanding the elementary aspects of how PTH and PTHrP behave and as a call to encourage further investigation that will yield more comprehensive understanding of the physiological and pathological steps at play, with a goal toward novel therapeutic interventions.
Collapse
|
35
|
Abstract
Ruminants (Ruminantia) are among the most successful herbivorous mammals, exhibiting wide-ranging morphological and ecological characteristics (such as headgear and multichambered stomach) and including various key livestock species (e.g., cattle, buffalo, yak, sheep, and goat). Understanding their evolution is of great significance not only in scientific research but also in applications potential for human society. The rapid growth of genomic resources provides unprecedented opportunities to dissect the evolutionary histories and molecular mechanisms underlying the distinct characteristics of ruminants. Here we summarize our current understanding of the genetic, morphological, and ecological diversity of ruminants and provide prospects for future studies.
Collapse
Affiliation(s)
- Bao Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming Yunnan 650204, China
| | - Le Chen
- Center for Ecological and Environmental Sciences, Northwestern Polytechnical University, Xi'an Shaanxi 710072, China, E-mail:chen_
| | - Wen Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China, E-mail:.,Center for Ecological and Environmental Sciences, Northwestern Polytechnical University, Xi'an Shaanxi 710072, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming Yunnan 650223, China
| |
Collapse
|
36
|
Martins JS, Liu ES, Sneddon WB, Friedman PA, Demay MB. 1,25-Dihydroxyvitamin D Maintains Brush Border Membrane NaPi2a and Attenuates Phosphaturia in Hyp Mice. Endocrinology 2019; 160:2204-2214. [PMID: 31237611 PMCID: PMC6735734 DOI: 10.1210/en.2019-00186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022]
Abstract
Phosphate homeostasis is critical for many cellular processes and is tightly regulated. The sodium-dependent phosphate cotransporter, NaPi2a, is the major regulator of urinary phosphate reabsorption in the renal proximal tubule. Its activity is dependent upon its brush border localization that is regulated by fibroblast growth factor 23 (FGF23) and PTH. High levels of FGF23, as are seen in the Hyp mouse model of human X-linked hypophosphatemia, lead to renal phosphate wasting. Long-term treatment of Hyp mice with 1,25-dihydroxyvitamin D (1,25D) or 1,25D analogues has been shown to improve renal phosphate wasting in the setting of increased FGF23 mRNA expression. Studies were undertaken to define the cellular and molecular basis for this apparent FGF23 resistance. 1,25D increased FGF23 protein levels in the cortical bone and circulation of Hyp mice but did not impair FGF23 cleavage. 1,25D attenuated urinary phosphate wasting as early as one hour postadministration, without suppressing FGF23 receptor/coreceptor expression. Although 1,25D treatment induced expression of early growth response 1, an early FGF23 responsive gene required for its phosphaturic effects, it paradoxically enhanced renal phosphate reabsorption and NaPi2a protein expression in renal brush border membranes (BBMs) within one hour. The Na-H+ exchange regulatory factor 1 (NHERF1) is a scaffolding protein thought to anchor NaPi2a to the BBM. Although 1,25D did not alter NHERF1 protein levels acutely, it enhanced NHERF1-NaPi2a interactions in Hyp mice. 1,25D also prevented the decrease in NHERF1/NaPi2a interactions in PTH-treated wild-type mice. Thus, these investigations identify a novel role for 1,25D in the hormonal regulation of renal phosphate handling.
Collapse
Affiliation(s)
- Janaina S Martins
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Eva S Liu
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Division of Endocrinology Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
| | - W Bruce Sneddon
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Peter A Friedman
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
37
|
|
38
|
Jeong J, Kim W, Hens J, Dann P, Schedin P, Friedman PA, Wysolmerski JJ. NHERF1 Is Required for Localization of PMCA2 and Suppression of Early Involution in the Female Lactating Mammary Gland. Endocrinology 2019; 160:1797-1810. [PMID: 31087002 PMCID: PMC6619491 DOI: 10.1210/en.2019-00230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
Abstract
Prior studies have demonstrated that the calcium pump, plasma membrane calcium ATPase 2 (PMCA2), mediates calcium transport into milk and prevents mammary epithelial cell death during lactation. PMCA2 also regulates cell proliferation and cell death in breast cancer cells, in part by maintaining the receptor tyrosine kinase ErbB2/HER2 within specialized plasma membrane domains. Furthermore, the regulation of PMCA2 membrane localization and activity in breast cancer cells requires its interaction with the PDZ domain-containing scaffolding molecule sodium-hydrogen exchanger regulatory factor (NHERF) 1. In this study, we asked whether NHERF1 also interacts with PMCA2 in normal mammary epithelial cells during lactation. Our results demonstrate that NHERF1 expression is upregulated during lactation and that it interacts with PMCA2 at the apical membrane of secretory luminal epithelial cells. Similar to PMCA2, NHERF1 expression is rapidly reduced by milk stasis after weaning. Examining lactating NHERF1 knockout (KO) mice showed that NHERF1 contributes to the proper apical location of PMCA2, for proper apical-basal polarity in luminal epithelial cells, and that it participates in the suppression of Stat3 activation and the prevention of premature mammary gland involution. Additionally, we found that PMCA2 also interacts with the closely related scaffolding molecule, NHERF2, at the apical membrane, which likely maintains PMCA2 at the plasma membrane of mammary epithelial cells in lactating NHERF1KO mice. Based on these data, we conclude that, during lactation, NHERF1 is required for the proper expression and apical localization of PMCA2, which, in turn, contributes to preventing the premature activation of Stat3 and the lysosome-mediated cell death pathway that usually occur only early in mammary involution.
Collapse
Affiliation(s)
- Jaekwang Jeong
- Section of Endocrinology and Metabolism, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Wonnam Kim
- Division of Pharmacology, College of Korean Medicine, Semyung University, Jecheon, Republic of Korea
| | - Julie Hens
- Section of Endocrinology and Metabolism, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Pamela Dann
- Section of Endocrinology and Metabolism, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Pepper Schedin
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Peter A Friedman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John J Wysolmerski
- Section of Endocrinology and Metabolism, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Correspondence: John J. Wysolmerski, MD, Section of Endocrinology and Metabolism, Department of Internal Medicine, TAC S123a, Yale University School of Medicine, 333 Cedar Street, FMT 102, Box 208020, New Haven, Connecticut 06520. E-mail:
| |
Collapse
|
39
|
Ghimire S, Terhzaz S, Cabrero P, Romero MF, Davies SA, Dow JAT. Targeted renal knockdown of Na +/H + exchanger regulatory factor Sip1 produces uric acid nephrolithiasis in Drosophila. Am J Physiol Renal Physiol 2019; 317:F930-F940. [PMID: 31364377 DOI: 10.1152/ajprenal.00551.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nephrolithiasis is one of the most common kidney diseases, with poorly understood pathophysiology, but experimental study has been hindered by lack of experimentally tractable models. Drosophila melanogaster is a useful model organism for renal diseases because of genetic and functional similarities of Malpighian (renal) tubules with the human kidney. Here, we demonstrated function of the sex-determining region Y protein-interacting protein-1 (Sip1) gene, an ortholog of human Na+/H+ exchanger regulatory factor (NHERF1), in Drosophila Malpighian tubules and its impact on nephrolithiasis. Abundant birefringent calculi were observed in Sip1 mutant flies, and the phenotype was also observed in renal stellate cell-specific RNA interference Sip1 knockdown in otherwise normal flies, confirming a renal etiology. This phenotype was abolished in rosy mutant flies (which model human xanthinuria) and by the xanthine oxidase inhibitor allopurinol, suggesting that the calculi were of uric acid. This was confirmed by direct biochemical assay for urate. Stones rapidly dissolved when the tubule was bathed in alkaline media, suggesting that Sip1 knockdown was acidifying the tubule. SIP1 was shown to collocate with Na+/H+ exchanger isoform 2 (NHE2) and with moesin in stellate cells. Knockdown of NHE2 specifically to the stellate cells also increased renal uric acid stone formation, and so a model was developed in which SIP1 normally regulates NHE2 activity and luminal pH, ultimately leading to uric acid stone formation. Drosophila renal tubules may thus offer a useful model for urate nephrolithiasis.
Collapse
Affiliation(s)
- Saurav Ghimire
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Selim Terhzaz
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Pablo Cabrero
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Michael F Romero
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Shireen A Davies
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Julian A T Dow
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
40
|
Bhattacharya S, Stanley CB, Heller WT, Friedman PA, Bu Z. Dynamic structure of the full-length scaffolding protein NHERF1 influences signaling complex assembly. J Biol Chem 2019; 294:11297-11310. [PMID: 31171716 PMCID: PMC6643037 DOI: 10.1074/jbc.ra119.008218] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/05/2019] [Indexed: 01/14/2023] Open
Abstract
The Na+/H+ exchange regulatory cofactor 1 (NHERF1) protein modulates the assembly and intracellular trafficking of several transmembrane G protein-coupled receptors (GPCRs) and ion transport proteins with the membrane-cytoskeleton adapter protein ezrin. Here, we applied solution NMR and small-angle neutron scattering (SANS) to structurally characterize full-length NHERF1 and disease-associated variants that are implicated in impaired phosphate homeostasis. Using NMR, we mapped the modular architecture of NHERF1, which is composed of two structurally-independent PDZ domains that are connected by a flexible, disordered linker. We observed that the ultra-long and disordered C-terminal tail of NHERF1 has a type 1 PDZ-binding motif that interacts weakly with the proximal, second PDZ domain to form a dynamically autoinhibited structure. Using ensemble-optimized analysis of SANS data, we extracted the molecular size distribution of structures from the extensive conformational space sampled by the flexible chain. Our results revealed that NHERF1 is a diffuse ensemble of variable PDZ domain configurations and a disordered C-terminal tail. The joint NMR/SANS data analyses of three disease variants (L110V, R153Q, and E225K) revealed significant differences in the local PDZ domain structures and in the global conformations compared with the WT protein. Furthermore, we show that the substitutions affect the affinity and kinetics of NHERF1 binding to ezrin and to a C-terminal peptide from G protein-coupled receptor kinase 6A (GRK6A). These findings provide important insight into the modulation of the intrinsic flexibility of NHERF1 by disease-associated point mutations that alter the dynamic assembly of signaling complexes.
Collapse
Affiliation(s)
| | - Christopher B Stanley
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37830
| | - William T Heller
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37830
| | - Peter A Friedman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Zimei Bu
- Department of Chemistry and Biochemistry, City College of New York, New York, New York 10031
| |
Collapse
|
41
|
Gorvin CM, Loh NY, Stechman MJ, Falcone S, Hannan FM, Ahmad BN, Piret SE, Reed AA, Jeyabalan J, Leo P, Marshall M, Sethi S, Bass P, Roberts I, Sanderson J, Wells S, Hough TA, Bentley L, Christie PT, Simon MM, Mallon AM, Schulz H, Cox RD, Brown MA, Huebner N, Brown SD, Thakker RV. Mice with a Brd4 Mutation Represent a New Model of Nephrocalcinosis. J Bone Miner Res 2019; 34:1324-1335. [PMID: 30830987 PMCID: PMC6658219 DOI: 10.1002/jbmr.3695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 01/26/2019] [Accepted: 02/05/2019] [Indexed: 12/30/2022]
Abstract
Nephrolithiasis (NL) and nephrocalcinosis (NC), which comprise renal calcification of the collecting system and parenchyma, respectively, have a multifactorial etiology with environmental and genetic determinants and affect ∼10% of adults by age 70 years. Studies of families with hereditary NL and NC have identified >30 causative genes that have increased our understanding of extracellular calcium homeostasis and renal tubular transport of calcium. However, these account for <20% of the likely genes that are involved, and to identify novel genes for renal calcification disorders, we investigated 1745 12-month-old progeny from a male mouse that had been treated with the chemical mutagen N-ethyl-N-nitrosourea (ENU) for radiological renal opacities. This identified a male mouse with renal calcification that was inherited as an autosomal dominant trait with >80% penetrance in 152 progeny. The calcification consisted of calcium phosphate deposits in the renal papillae and was associated with the presence of the urinary macromolecules osteopontin and Tamm-Horsfall protein, which are features found in Randall's plaques of patients with NC. Genome-wide mapping located the disease locus to a ∼30 Mbp region on chromosome 17A3.3-B3 and whole-exome sequence analysis identified a heterozygous mutation, resulting in a missense substitution (Met149Thr, M149T), in the bromodomain-containing protein 4 (BRD4). The mutant heterozygous (Brd4+/M149T ) mice, when compared with wild-type (Brd4+/+ ) mice, were normocalcemic and normophosphatemic, with normal urinary excretions of calcium and phosphate, and had normal bone turnover markers. BRD4 plays a critical role in histone modification and gene transcription, and cDNA expression profiling, using kidneys from Brd4+/M149T and Brd4+/+ mice, revealed differential expression of genes involved in vitamin D metabolism, cell differentiation, and apoptosis. Kidneys from Brd4+/M149T mice also had increased apoptosis at sites of calcification within the renal papillae. Thus, our studies have established a mouse model, due to a Brd4 Met149Thr mutation, for inherited NC. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Caroline M Gorvin
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Nellie Y Loh
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Michael J Stechman
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Sara Falcone
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Fadil M Hannan
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Bushra N Ahmad
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Sian E Piret
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Anita Ac Reed
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jeshmi Jeyabalan
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul Leo
- Translational Genomics Group, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT) at Translational Research Institute, Brisbane, Australia
| | - Mhairi Marshall
- Translational Genomics Group, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT) at Translational Research Institute, Brisbane, Australia
| | - Siddharth Sethi
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Paul Bass
- Department of Cellular Pathology, Royal Free Hospital, London, UK
| | - Ian Roberts
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford, UK
| | - Jeremy Sanderson
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Sara Wells
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Tertius A Hough
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Liz Bentley
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Paul T Christie
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Michelle M Simon
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Ann-Marie Mallon
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Herbert Schulz
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Roger D Cox
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Matthew A Brown
- Translational Genomics Group, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT) at Translational Research Institute, Brisbane, Australia
| | | | - Steve D Brown
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
42
|
Chen L, Qiu Q, Jiang Y, Wang K, Lin Z, Li Z, Bibi F, Yang Y, Wang J, Nie W, Su W, Liu G, Li Q, Fu W, Pan X, Liu C, Yang J, Zhang C, Yin Y, Wang Y, Zhao Y, Zhang C, Wang Z, Qin Y, Liu W, Wang B, Ren Y, Zhang R, Zeng Y, da Fonseca RR, Wei B, Li R, Wan W, Zhao R, Zhu W, Wang Y, Duan S, Gao Y, Zhang YE, Chen C, Hvilsom C, Epps CW, Chemnick LG, Dong Y, Mirarab S, Siegismund HR, Ryder OA, Gilbert MTP, Lewin HA, Zhang G, Heller R, Wang W. Large-scale ruminant genome sequencing provides insights into their evolution and distinct traits. Science 2019; 364:364/6446/eaav6202. [DOI: 10.1126/science.aav6202] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/16/2019] [Indexed: 12/17/2022]
Abstract
The ruminants are one of the most successful mammalian lineages, exhibiting morphological and habitat diversity and containing several key livestock species. To better understand their evolution, we generated and analyzed de novo assembled genomes of 44 ruminant species, representing all six Ruminantia families. We used these genomes to create a time-calibrated phylogeny to resolve topological controversies, overcoming the challenges of incomplete lineage sorting. Population dynamic analyses show that population declines commenced between 100,000 and 50,000 years ago, which is concomitant with expansion in human populations. We also reveal genes and regulatory elements that possibly contribute to the evolution of the digestive system, cranial appendages, immune system, metabolism, body size, cursorial locomotion, and dentition of the ruminants.
Collapse
|
43
|
E6 proteins from high-risk HPV, low-risk HPV, and animal papillomaviruses activate the Wnt/β-catenin pathway through E6AP-dependent degradation of NHERF1. PLoS Pathog 2019; 15:e1007575. [PMID: 31002735 PMCID: PMC6493770 DOI: 10.1371/journal.ppat.1007575] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/01/2019] [Accepted: 03/25/2019] [Indexed: 11/19/2022] Open
Abstract
High-risk human papillomavirus (HPV) E6 proteins associate with the cellular ubiquitin ligase E6-Associated Protein (E6AP), and then recruit both p53 and certain cellular PDZ proteins for ubiquitination and degradation by the proteasome. Low-risk HPV E6 proteins also associate with E6AP, yet fail to recruit p53 or PDZ proteins; their E6AP-dependent targets have so far been uncharacterized. We found a cellular PDZ protein called Na+/H+ Exchanger Regulatory Factor 1 (NHERF1) is targeted for degradation by both high and low-risk HPV E6 proteins as well as E6 proteins from diverse non-primate mammalian species. NHERF1 was degraded by E6 in a manner dependent upon E6AP ubiquitin ligase activity but independent of PDZ interactions. A novel structural domain of E6, independent of the p53 recognition domain, was necessary to associate with and degrade NHERF1, and the NHERF1 EB domain was required for E6-mediated degradation. Degradation of NHERF1 by E6 activated canonical Wnt/β-catenin signaling, a key pathway that regulates cell growth and proliferation. Expression levels of NHERF1 increased with increasing cell confluency. This is the first study in which a cellular protein has been identified that is targeted for degradation by both high and low-risk HPV E6 as well as E6 proteins from diverse animal papillomaviruses. This suggests that NHERF1 plays a role in regulating squamous epithelial growth and further suggests that the interaction of E6 proteins with NHERF1 could be a common therapeutic target for multiple papillomavirus types. Papillomaviruses cause benign squamous epithelial tumors through the action of virally encoded oncoproteins termed E6 and E7, which are classified as either high or low-risk based upon the propensity of the tumor to evolve into cancer. E6 proteins from both high and low-risk HPVs interact with a cellular ubiquitin ligase called E6AP. High-risk E6 proteins hijack E6AP ubiquitin ligase activity to target p53 for degradation. Degradation targets of the low-risk E6 proteins in complex with E6AP have not been described. Here, we describe a protein called NHERF1 that is targeted for degradation by both high and low-risk E6 proteins, as well as E6 proteins from diverse animal species. Degradation of NHERF1 resulted in activation of an oncogenic cellular signaling pathway called Wnt. Identification of NHERF1 as a highly conserved E6 degradation target could inform therapies directed against both low-risk HPVs and cancer-inducing high-risk HPVs.
Collapse
|
44
|
Wagner CA, Rubio-Aliaga I, Hernando N. Renal phosphate handling and inherited disorders of phosphate reabsorption: an update. Pediatr Nephrol 2019; 34:549-559. [PMID: 29275531 DOI: 10.1007/s00467-017-3873-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 01/12/2023]
Abstract
Renal phosphate handling critically determines plasma phosphate and whole body phosphate levels. Filtered phosphate is mostly reabsorbed by Na+-dependent phosphate transporters located in the brush border membrane of the proximal tubule: NaPi-IIa (SLC34A1), NaPi-IIc (SLC34A3), and Pit-2 (SLC20A2). Here we review new evidence for the role and relevance of these transporters in inherited disorders of renal phosphate handling. The importance of NaPi-IIa and NaPi-IIc for renal phosphate reabsorption and mineral homeostasis has been highlighted by the identification of mutations in these transporters in a subset of patients with infantile idiopathic hypercalcemia and patients with hereditary hypophosphatemic rickets with hypercalciuria. Both diseases are characterized by disturbed calcium homeostasis secondary to elevated 1,25-(OH)2 vitamin D3 as a consequence of hypophosphatemia. In vitro analysis of mutated NaPi-IIa or NaPi-IIc transporters suggests defective trafficking underlying disease in most cases. Monoallelic pathogenic mutations in both SLC34A1 and SLC34A3 appear to be very frequent in the general population and have been associated with kidney stones. Consistent with these findings, results from genome-wide association studies indicate that variants in SLC34A1 are associated with a higher risk to develop kidney stones and chronic kidney disease, but underlying mechanisms have not been addressed to date.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland. .,National Center for Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland.
| | - Isabel Rubio-Aliaga
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.,National Center for Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Nati Hernando
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.,National Center for Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| |
Collapse
|
45
|
Levi M, Gratton E. Visualizing the regulation of SLC34 proteins at the apical membrane. Pflugers Arch 2019; 471:533-542. [PMID: 30613865 PMCID: PMC6436987 DOI: 10.1007/s00424-018-02249-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 10/27/2022]
Abstract
The cloning of the renal NaPi-2a (SLC34A1) and NaPi-2c (SLC34A3) phosphate transporters has made it possible to characterize the molecular and biophysical regulation of renal proximal tubular reabsorption of inorganic phosphate (Pi). Dietary factors, such as Pi and K, and several hormones and phosphatonins, including parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), and glucocorticoids, regulate the transporters through various transcriptional, translational, and post-translational mechanisms that involve acute trafficking via endocytosis or exocytosis, interactions with PDZ domain proteins, lipid microdomains, and diffusion and clustering in the apical brush border membrane. The visualization of these trafficking events by means of novel microscopy techniques that includes fluorescence lifetime imaging microscopy (FLIM), Förster resonance energy transfer (FRET), fluctuation correlation spectroscopy (FCS), and modulation tracking (MT), is the primary focus of this review.
Collapse
Affiliation(s)
- Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, USA.
| | - Enrico Gratton
- Department of Biomedical Engineering, Laboratory for Fluorescence Dynamics, University of California at Irvine, Irvine, CA, USA
| |
Collapse
|
46
|
Bushau-Sprinkle A, Barati M, Conklin C, Dupre T, Gagnon KB, Khundmiri SJ, Clark B, Siskind L, Doll MA, Rane M, Brier M, Coventry S, Lederer ED. Loss of the Na +/H + Exchange Regulatory Factor 1 Increases Susceptibility to Cisplatin-Induced Acute Kidney Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1190-1200. [PMID: 30926337 DOI: 10.1016/j.ajpath.2019.02.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/12/2019] [Accepted: 02/25/2019] [Indexed: 12/19/2022]
Abstract
Na+/H+ exchange regulatory cofactor (NHERF)-1, a scaffolding protein, anchors multiple membrane proteins in renal proximal tubules. Cultured proximal tubule cells deficient in Nherf1 and proximal tubules from Nherf1-deficient mice exhibit aberrant trafficking. Nherf1-deficient cells also exhibit an altered transcription pattern and worse survival. These observations suggest that NHERF1 loss increases susceptibility to acute kidney injury (AKI). Male and female wild-type C57BL/6J and Nherf1 knockout mice were treated with saline or cisplatin (20 mg/kg dose i.p.) to induce AKI and were euthanized after 72 hours. Blood and urine were collected for assessments of blood urea nitrogen and neutrophil gelatinase-associated lipocalin, respectively. Kidneys were harvested for histology (hematoxylin and eosin, periodic acid-Schiff) and terminal deoxynucleotidyl transferase dUTP nick end labeling assay, Kim1 mRNA assessment, and Western blot analysis for cleaved caspase 3. Cisplatin treatment was associated with significantly greater severity of AKI in knockout compared with wild-type mice, as demonstrated by semiquantitative injury score (2.8 versus 1.89, P < 0.001), blood urea nitrogen (151.8 ± 17.2 mg/dL versus 97.8 ± 10.1 mg/dL, P < 0.05), and neutrophil gelatinase-associated lipocalin urine protein (55.6 ± 21.3 μg/mL versus 2.7 ± 0.53 μg/mL, P < 0.05). Apoptosis markers were significantly increased in cisplatin-treated Nherf1 knockout and wild-type mice compared to respective controls. These data suggest that NHERF1 loss increases susceptibility to AKI.
Collapse
Affiliation(s)
| | - Michelle Barati
- Department of Medicine, Division of Nephrology, University of Louisville, Louisville, Kentucky
| | - Caryl Conklin
- Department of Medicine, Division of Nephrology, University of Louisville, Louisville, Kentucky
| | - Tess Dupre
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Kenneth B Gagnon
- Department of Medicine, Division of Nephrology, University of Louisville, Louisville, Kentucky
| | - Syed J Khundmiri
- Department of Physiology and Biophysics, Howard University, Washington, DC
| | - Barbara Clark
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky
| | - Leah Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Mark A Doll
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Madhavi Rane
- Department of Medicine, Division of Nephrology, University of Louisville, Louisville, Kentucky
| | - Michael Brier
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky; Department of Medicine, Division of Nephrology, University of Louisville, Louisville, Kentucky
| | - Susan Coventry
- Department of Pathology, University of Louisville, Louisville, Kentucky; Department of Pediatrics, University of Louisville, Louisville, Kentucky
| | - Eleanor D Lederer
- Department of Medicine, Division of Nephrology, University of Louisville, Louisville, Kentucky; Robley Rex VA Medical Center, Louisville, Kentucky.
| |
Collapse
|
47
|
Kim KU, Pang WK, Kang S, Ryu DY, Song WH, Rahman MS, Kwon WS, Pang MG. Sperm solute carrier family 9 regulator 1 is correlated with boar fertility. Theriogenology 2019; 126:254-260. [DOI: 10.1016/j.theriogenology.2018.12.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/06/2018] [Accepted: 12/13/2018] [Indexed: 11/15/2022]
|
48
|
Zhang Q, Xiao K, Paredes JM, Mamonova T, Sneddon WB, Liu H, Wang D, Li S, McGarvey JC, Uehling D, Al-Awar R, Joseph B, Jean-Alphonse F, Orte A, Friedman PA. Parathyroid hormone initiates dynamic NHERF1 phosphorylation cycling and conformational changes that regulate NPT2A-dependent phosphate transport. J Biol Chem 2019; 294:4546-4571. [PMID: 30696771 PMCID: PMC6433080 DOI: 10.1074/jbc.ra119.007421] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/25/2019] [Indexed: 12/30/2022] Open
Abstract
Na+-H+ exchanger regulatory factor-1 (NHERF1) is a PDZ protein that scaffolds membrane proteins, including sodium-phosphate co-transport protein 2A (NPT2A) at the plasma membrane. NHERF1 is a phosphoprotein with 40 Ser and Thr residues. Here, using tandem MS analysis, we characterized the sites of parathyroid hormone (PTH)-induced NHERF1 phosphorylation and identified 10 high-confidence phosphorylation sites. Ala replacement at Ser46, Ser162, Ser181, Ser269, Ser280, Ser291, Thr293, Ser299, and Ser302 did not affect phosphate uptake, but S290A substitution abolished PTH-dependent phosphate transport. Unexpectedly, Ser290 was rapidly dephosphorylated and rephosphorylated after PTH stimulation, and we found that protein phosphatase 1α (PP1α), which binds NHERF1 through a conserved VxF/W PP1 motif, dephosphorylates Ser290 Mutating 257VPF259 eliminated PP1 binding and blunted dephosphorylation. Tautomycetin blocked PP1 activity and abrogated PTH-sensitive phosphate transport. Using fluorescence lifetime imaging (FLIM), we observed that PTH paradoxically and transiently elevates intracellular phosphate. Added phosphate blocked PP1α-mediated Ser290 dephosphorylation of recombinant NHERF1. Hydrogen-deuterium exchange MS revealed that β-sheets in NHERF1's PDZ2 domain display lower deuterium uptake than those in the structurally similar PDZ1, implying that PDZ1 is more cloistered. Dephosphorylated NHERF1 exhibited faster exchange at C-terminal residues suggesting that NHERF1 dephosphorylation precedes Ser290 rephosphorylation. Our results show that PP1α and NHERF1 form a holoenzyme and that a multiprotein kinase cascade involving G protein-coupled receptor kinase 6A controls the Ser290 phosphorylation status of NHERF1 and regulates PTH-sensitive, NPT2A-mediated phosphate uptake. These findings reveal how reversible phosphorylation modifies protein conformation and function and the biochemical mechanisms underlying PTH control of phosphate transport.
Collapse
Affiliation(s)
- Qiangmin Zhang
- From the Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology
| | - Kunhong Xiao
- From the Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology.,Vascular Medicine Institute, and.,Biomedical Mass Spectrometry Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - José M Paredes
- the Department of Physical Chemistry, Faculty of Pharmacy, University of Granada, 18071-Granada, Spain
| | - Tatyana Mamonova
- From the Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology
| | - W Bruce Sneddon
- From the Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology
| | - Hongda Liu
- From the Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology
| | - Dawei Wang
- From the Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology
| | - Sheng Li
- the Department of Medicine, University of California San Diego, La Jolla, California 92093, and
| | - Jennifer C McGarvey
- From the Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology
| | - David Uehling
- the Department of Drug Discovery, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Rima Al-Awar
- the Department of Drug Discovery, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Babu Joseph
- the Department of Drug Discovery, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | | | - Angel Orte
- the Department of Physical Chemistry, Faculty of Pharmacy, University of Granada, 18071-Granada, Spain
| | - Peter A Friedman
- From the Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, .,Department of Structural Biology
| |
Collapse
|
49
|
Abstract
Calcium kidney stones are common worldwide. Most are idiopathic and composed of calcium oxalate. Calcium phosphate is present in around 80% and may initiate stone formation. Stone production is multifactorial with a polygenic genetic contribution. Phosphaturia is found frequently among stone formers but until recently received scant attention. This review examines possible mechanisms for the phosphaturia and its relevance to stone formation from a wide angle. There is a striking lack of clinical data. Phosphaturia is associated, but not correlated, with hypercalciuria, increased 1,25 dihydroxy-vitamin D [1,25 (OH)2D], and sometimes evidence of disturbances in proximal renal tubular function. Phosphate reabsorption in the proximal renal tubules requires tightly regulated interaction of many proteins. Paracellular flow through intercellular tight junctions is the major route of phosphate absorption from the intestine and can be reduced therapeutically in hyperphosphatemic patients. In monogenic defects stones develop when phosphaturia is associated with hypercalciuria, generally explained by increased 1,25 (OH)2D production in response to hypophosphatemia. Calcification does not occur in disorders with increased FGF23 when phosphaturia occurs in isolation and 1,25 (OH)2D is suppressed. Candidate gene studies have identified mutations in the phosphate transporters, but in few individuals. One genome-wide study identified a polymorphism of the phosphate transporter gene SLC34A4 associated with stones. Others did not find mutations obviously linked to phosphate reabsorption. Future genetic studies should have a wide trawl and should focus initially on groups of patients with clearly defined phenotypes. The global data should be pooled.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.
| |
Collapse
|
50
|
Jacquillet G, Unwin RJ. Physiological regulation of phosphate by vitamin D, parathyroid hormone (PTH) and phosphate (Pi). Pflugers Arch 2019; 471:83-98. [PMID: 30393837 PMCID: PMC6326012 DOI: 10.1007/s00424-018-2231-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/20/2018] [Accepted: 10/25/2018] [Indexed: 01/05/2023]
Abstract
Inorganic phosphate (Pi) is an abundant element in the body and is essential for a wide variety of key biological processes. It plays an essential role in cellular energy metabolism and cell signalling, e.g. adenosine and guanosine triphosphates (ATP, GTP), and in the composition of phospholipid membranes and bone, and is an integral part of DNA and RNA. It is an important buffer in blood and urine and contributes to normal acid-base balance. Given its widespread role in almost every molecular and cellular function, changes in serum Pi levels and balance can have important and untoward effects. Pi homoeostasis is maintained by a counterbalance between dietary Pi absorption by the gut, mobilisation from bone and renal excretion. Approximately 85% of total body Pi is present in bone and only 1% is present as free Pi in extracellular fluids. In humans, extracellular concentrations of inorganic Pi vary between 0.8 and 1.2 mM, and in plasma or serum Pi exists in both its monovalent and divalent forms (H2PO4- and HPO42-). In the intestine, approximately 30% of Pi absorption is vitamin D regulated and dependent. To help maintain Pi balance, reabsorption of filtered Pi along the renal proximal tubule (PT) is via the NaPi-IIa and NaPi-IIc Na+-coupled Pi cotransporters, with a smaller contribution from the PiT-2 transporters. Endocrine factors, including, vitamin D and parathyroid hormone (PTH), as well as newer factors such as fibroblast growth factor (FGF)-23 and its coreceptor α-klotho, are intimately involved in the control of Pi homeostasis. A tight regulation of Pi is critical, since hyperphosphataemia is associated with increased cardiovascular morbidity in chronic kidney disease (CKD) and hypophosphataemia with rickets and growth retardation. This short review considers the control of Pi balance by vitamin D, PTH and Pi itself, with an emphasis on the insights gained from human genetic disorders and genetically modified mouse models.
Collapse
Affiliation(s)
- Grégory Jacquillet
- Centre for Nephrology, University College London (UCL), Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Robert J Unwin
- Centre for Nephrology, University College London (UCL), Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK.
- AstraZeneca IMED ECD CVRM R&D, Gothenburg, Sweden.
| |
Collapse
|