1
|
Chambers BE, Weaver NE, Lara CM, Nguyen TK, Wingert RA. (Zebra)fishing for nephrogenesis genes. Tissue Barriers 2024; 12:2219605. [PMID: 37254823 PMCID: PMC11042071 DOI: 10.1080/21688370.2023.2219605] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/14/2023] [Indexed: 06/01/2023] Open
Abstract
Kidney disease is a devastating condition affecting millions of people worldwide, where over 100,000 patients in the United States alone remain waiting for a lifesaving organ transplant. Concomitant with a surge in personalized medicine, single-gene mutations, and polygenic risk alleles have been brought to the forefront as core causes of a spectrum of renal disorders. With the increasing prevalence of kidney disease, it is imperative to make substantial strides in the field of kidney genetics. Nephrons, the core functional units of the kidney, are epithelial tubules that act as gatekeepers of body homeostasis by absorbing and secreting ions, water, and small molecules to filter the blood. Each nephron contains a series of proximal and distal segments with explicit metabolic functions. The embryonic zebrafish provides an ideal platform to systematically dissect the genetic cues governing kidney development. Here, we review the use of zebrafish to discover nephrogenesis genes.
Collapse
Affiliation(s)
- Brooke E. Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Nicole E. Weaver
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Caroline M. Lara
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| |
Collapse
|
2
|
Nurcombe ZW, Hehr CL, McFarlane S. Plexina4 and cell survival in the developing zebrafish hindbrain. Dev Dyn 2023; 252:1323-1337. [PMID: 37283310 DOI: 10.1002/dvdy.633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Growth factors are important in the developing and mature nervous system to support the survival of neurons. Developmental signaling molecules are known for their roles in controlling neurogenesis and neural circuit formation. Whether or not these molecules also have roles in cell survival in the developing nervous system is poorly understood. Plexins are a family of transmembrane receptors that bind Semaphorin ligands and are known to function in the guidance of developing axons and blood vessels. RESULTS In embryonic zebrafish, plexina4 is expressed widely in the brain, becoming largely restricted to the hindbrain as neurogenesis and differentiation proceed. Apoptosis is increased in the embryonic hindbrain of a plexina4ca307/ca307 CRISPR mutant. Based on the literature, we tested the secreted heat shock protein, Clusterin, as a candidate ligand to mediate cell survival through Plexina4. clusterin is expressed by the floor plate of the embryonic zebrafish hindbrain, in proximity to plexina4-expressing hindbrain cells. Morpholino-mediated knockdown of Clusterin increases cell apoptosis in the hindbrain, with additional cell death observed in epistasis experiments where Clusterin is knocked down in a plexina4 mutant background. CONCLUSIONS Our data suggest that Plexina4 promotes cell survival in the developing zebrafish hindbrain, likely through a pathway independent of Clusterin.
Collapse
Affiliation(s)
- Zachary W Nurcombe
- Department Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Graduate Program in Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Carrie Lynn Hehr
- Department Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sarah McFarlane
- Department Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
3
|
Rbaibi Y, Long KR, Shipman KE, Ren Q, Baty CJ, Kashlan OB, Weisz OA. Megalin, cubilin, and Dab2 drive endocytic flux in kidney proximal tubule cells. Mol Biol Cell 2023; 34:ar74. [PMID: 37126375 PMCID: PMC10295476 DOI: 10.1091/mbc.e22-11-0510] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/27/2023] [Accepted: 04/17/2023] [Indexed: 05/02/2023] Open
Abstract
The kidney proximal tubule (PT) elaborates a uniquely high-capacity apical endocytic pathway to retrieve albumin and other proteins that escape the glomerular filtration barrier. Megalin and cubilin/amnionless (CUBAM) receptors engage Dab2 in these cells to mediate clathrin-dependent uptake of filtered ligands. Knockout of megalin or Dab2 profoundly inhibits apical endocytosis and is believed to atrophy the endocytic pathway. We generated CRISPR/Cas9 knockout (KO) clones lacking cubilin, megalin, or Dab2 expression in highly differentiated PT cells and determined the impact on albumin internalization and endocytic pathway function. KO of each component had different effects on the concentration dependence of albumin uptake as well its distribution within PT cells. Reduced uptake of a fluid phase marker was also observed, with megalin KO cells having the most dramatic decline. Surprisingly, protein levels and distribution of key endocytic proteins were preserved in KO PT cell lines and in megalin KO mice, despite the reduced endocytic activity. Our data highlight specific functions of megalin, cubilin, and Dab2 in apical endocytosis and demonstrate that these proteins drive endocytic flux without compromising the physical integrity of the apical endocytic pathway. Our studies suggest a novel model to explain how these components coordinate endocytic uptake in PT cells.
Collapse
Affiliation(s)
- Youssef Rbaibi
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Kimberly R. Long
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Katherine E. Shipman
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Qidong Ren
- School of Medicine, Tsinghua University, Beijing, China, 100084
| | - Catherine J. Baty
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Ossama B. Kashlan
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Ora A. Weisz
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| |
Collapse
|
4
|
Williams DM, Gungordu L, Jackson-Crawford A, Lowe M. Assessment of endocytic traffic and Ocrl function in the developing zebrafish neuroepithelium. J Cell Sci 2022; 135:276669. [PMID: 35979861 PMCID: PMC9592051 DOI: 10.1242/jcs.260339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/11/2022] [Indexed: 12/05/2022] Open
Abstract
Endocytosis allows cells to internalise a wide range of molecules from their environment and to maintain their plasma membrane composition. It is vital during development and for maintenance of tissue homeostasis. The ability to visualise endocytosis in vivo requires suitable assays to monitor the process. Here, we describe imaging-based assays to visualise endocytosis in the neuroepithelium of living zebrafish embryos. Injection of fluorescent tracers into the brain ventricles followed by live imaging was used to study fluid-phase or receptor-mediated endocytosis, for which we used receptor-associated protein (RAP, encoded by Lrpap1) as a ligand for low-density lipoprotein receptor-related protein (LRP) receptors. Using dual-colour imaging combined with expression of endocytic markers, it is possible to track the progression of endocytosed tracers and to monitor trafficking dynamics. Using these assays, we reveal a role for the Lowe syndrome protein Ocrl in endocytic trafficking within the neuroepithelium. We also found that the RAP-binding receptor Lrp2 (encoded by lrp2a) appears to contribute only partially to neuroepithelial RAP endocytosis. Altogether, our results provide a basis to track endocytosis within the neuroepithelium in vivo and support a role for Ocrl in this process. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Daniel M Williams
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Lale Gungordu
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Anthony Jackson-Crawford
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
5
|
Retrospective Multicenter Study on Outcome Measurement for Dyskinesia Improvement in Parkinson's Disease Patients with Pallidal and Subthalamic Nucleus Deep Brain Stimulation. Brain Sci 2022; 12:brainsci12081054. [PMID: 36009117 PMCID: PMC9405623 DOI: 10.3390/brainsci12081054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 11/17/2022] Open
Abstract
Deep brain stimulation (DBS) is an effective treatment for dyskinesia in patients with Parkinson’s disease (PD), among which the therapeutic targets commonly used include the subthalamic nucleus (STN) and the globus pallidus internus (GPi). Levodopa-induced dyskinesia (LID) is one of the common motor complications arising in PD patients on chronic treatment with levodopa. In this article, we retrospectively evaluated the outcomes of LID with the Unified Dyskinesia Rating Scale (UDysRS) in patients who underwent DBS in multiple centers with a GPi or an STN target. Meanwhile, the Med off MDS-Unified Parkinson’s Disease Rating Scale (MDS-UPDRS-Ⅲ) and the levodopa equivalent daily dose (LEDD) were also observed as secondary indicators. PD patients with a GPi target showed a more significant improvement in the UDysRS compared with an STN target (92.9 ± 16.7% vs. 66.0 ± 33.6%, p < 0.0001). Both the GPi and the STN showed similar improvement in Med off UPDRS-III scores (49.8 ± 22.6% vs. 52.3 ± 29.5%, p = 0.5458). However, the LEDD was obviously reduced with the STN target compared with the GPi target (44.6 ± 28.1% vs. 12.2 ± 45.8%, p = 0.006).
Collapse
|
6
|
Morgan J, Yarwood R, Starborg T, Yan G, Lowe M. Pacsin2 is required for endocytosis in the zebrafish pronephric tubule. Biol Open 2022; 11:275521. [PMID: 35616009 PMCID: PMC9235069 DOI: 10.1242/bio.059150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/19/2022] [Indexed: 11/25/2022] Open
Abstract
Endocytosis mediates the cellular uptake of numerous molecules from the extracellular space and is a fundamentally important process. In the renal proximal tubule, the scavenger receptor megalin and its co-receptor cubilin mediate endocytosis of low molecular weight proteins from the renal filtrate. However, the extent to which megalin endocytosis relies on different components of the trafficking machinery remains relatively poorly defined in vivo. In this study, we identify a functional requirement for the F-BAR protein pacsin2 in endocytosis in the renal proximal tubule of zebrafish larvae. Pacsin2 is expressed throughout development and in all zebrafish tissues, similar to the mammalian orthologue. Within renal tubular epithelial cells, pacsin2 is enriched at the apical pole where it is localised to endocytic structures. Loss of pacsin2 results in reduced endocytosis within the proximal tubule, which is accompanied by a reduction in the abundance of megalin and endocytic organelles. Our results indicate that pacsin2 is required for efficient endocytosis in the proximal tubule, where it likely cooperates with other trafficking machinery to maintain endocytic uptake and recycling of megalin. Summary: We identify a role for the F-BAR protein pacsin2 in endocytosis in the renal tubule of zebrafish larvae.
Collapse
Affiliation(s)
- Joseph Morgan
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Rebecca Yarwood
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Tobias Starborg
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Guanhua Yan
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
7
|
Wu QX, Liu HQ, Wang YJ, Chen TC, Wei ZY, Chang JH, Chen TH, Seema J, Lin EC. Chemical Exchange Saturation Transfer (CEST) Signal at −1.6 ppm and Its Application for Imaging a C6 Glioma Model. Biomedicines 2022; 10:biomedicines10061220. [PMID: 35740241 PMCID: PMC9219881 DOI: 10.3390/biomedicines10061220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 02/01/2023] Open
Abstract
The chemical exchange saturation transfer (CEST) signal at −1.6 ppm is attributed to the choline methyl on phosphatidylcholines and results from the relayed nuclear Overhauser effect (rNOE), that is, rNOE(−1.6). The formation of rNOE(−1.6) involving the cholesterol hydroxyl is shown in liposome models. We aimed to confirm the correlation between cholesterol content and rNOE(−1.6) in cell cultures, tissues, and animals. C57BL/6 mice (N = 9) bearing the C6 glioma tumor were imaged in a 7 T MRI scanner, and their rNOE(−1.6) images were cross-validated through cholesterol staining with filipin. Cholesterol quantification was obtained using an 18.8-T NMR spectrometer from the lipid extracts of the brain tissues from another group of mice (N = 3). The cholesterol content in the cultured cells was manipulated using methyl-β-cyclodextrin and a complex of cholesterol and methyl-β-cyclodextrin. The rNOE(−1.6) of the cell homogenates and their cholesterol levels were measured using a 9.4-T NMR spectrometer. The rNOE(−1.6) signal is hypointense in the C6 tumors of mice, which matches the filipin staining results, suggesting that their tumor region is cholesterol deficient. The tissue extracts also indicate less cholesterol and phosphatidylcholine contents in tumors than in normal brain tissues. The amplitude of rNOE(−1.6) is positively correlated with the cholesterol concentration in the cholesterol-manipulated cell cultures. Our results indicate that the cholesterol dependence of rNOE(−1.6) occurs in cell cultures and solid tumors of C6 glioma. Furthermore, when the concentration of phosphatidylcholine is carefully considered, rNOE(−1.6) can be developed as a cholesterol-weighted imaging technique.
Collapse
Affiliation(s)
- Qi-Xuan Wu
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62102, Taiwan; (Q.-X.W.); (H.-Q.L.); (Y.-J.W.); (Z.-Y.W.); (J.-H.C.); (T.-H.C.)
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (T.-C.C.); (J.S.)
| | - Hong-Qing Liu
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62102, Taiwan; (Q.-X.W.); (H.-Q.L.); (Y.-J.W.); (Z.-Y.W.); (J.-H.C.); (T.-H.C.)
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (T.-C.C.); (J.S.)
| | - Yi-Jiun Wang
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62102, Taiwan; (Q.-X.W.); (H.-Q.L.); (Y.-J.W.); (Z.-Y.W.); (J.-H.C.); (T.-H.C.)
| | - Tsai-Chen Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (T.-C.C.); (J.S.)
| | - Zi-Ying Wei
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62102, Taiwan; (Q.-X.W.); (H.-Q.L.); (Y.-J.W.); (Z.-Y.W.); (J.-H.C.); (T.-H.C.)
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (T.-C.C.); (J.S.)
| | - Jung-Hsuan Chang
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62102, Taiwan; (Q.-X.W.); (H.-Q.L.); (Y.-J.W.); (Z.-Y.W.); (J.-H.C.); (T.-H.C.)
| | - Ting-Hao Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62102, Taiwan; (Q.-X.W.); (H.-Q.L.); (Y.-J.W.); (Z.-Y.W.); (J.-H.C.); (T.-H.C.)
| | - Jaya Seema
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (T.-C.C.); (J.S.)
| | - Eugene C. Lin
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62102, Taiwan; (Q.-X.W.); (H.-Q.L.); (Y.-J.W.); (Z.-Y.W.); (J.-H.C.); (T.-H.C.)
- Center for Nano Bio-Detection, National Chung Cheng University, Chiayi 62102, Taiwan
- Correspondence: ; Tel.: +886-5-272-0411 (ext. 66418); Fax: +886-5-272-1040
| |
Collapse
|
8
|
Edwards A, Long KR, Baty CJ, Shipman KE, Weisz OA. Modeling normal and nephrotic axial uptake of albumin and other filtered proteins along the proximal tubule. J Physiol 2022; 600:1933-1952. [PMID: 35178707 PMCID: PMC9012691 DOI: 10.1113/jp282885] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/10/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS We used new and published data to develop a mathematical model that predicts the profile of albumin uptake in the mouse proximal tubule (PT) in normal and nephrotic states, and partially accounts for competitive inhibition of uptake by normally filtered and pathologic ligands. Three pathways, consisting of high-affinity uptake by cubilin receptors, low-affinity uptake by megalin receptors, and fluid phase uptake, contribute to the overall retrieval of filtered proteins. The axial profile and efficiency of protein uptake depend on the initial filtrate composition and the individual protein affinities for megalin and cubilin. Under normal conditions, the majority of albumin is retrieved in S1 but shifts to S2 under nephrotic conditions. Other proteins exhibit different uptake profiles. Our model explains how tubular proteinuria can occur despite a large excess in potential PT uptake capacity. ABSTRACT Recent studies indicate that filtered albumin is retrieved in the proximal tubule (PT) via three pathways: receptor-mediated endocytosis via cubilin (high affinity) and megalin (low affinity), and fluid-phase uptake. Expression of megalin is required to maintain all three pathways, making it challenging to determine their respective contributions. Moreover, uptake of filtered molecules varies between the sub-segments (S1, S2, and S3) that make up the PT. Here we used new and published data to develop a mathematical model that predicts the rates of albumin uptake in mouse PT sub-segments in normal and nephrotic states, and partially accounts for competition by β2-microglobulin (β2m) and Immunoglobulin G (IgG). Our simulations indicate that receptor-mediated, rather than fluid-phase uptake, accounts for the vast majority of ligand recovery. Our model predicts that ∼75% of normally filtered albumin is reabsorbed via cubilin; however, megalin-mediated uptake predominates under nephrotic conditions. Our results also suggest that ∼80% of albumin is normally recovered in S1, whereas nephrotic conditions or knockout of cubilin shifts the bulk of albumin uptake to S2. The model predicts β2m and IgG axial recovery profiles qualitatively similar to those of albumin under normal conditions. In contrast with albumin however, the bulk of IgG and β2m uptake still occurs in S1 under nephrotic conditions. Overall, our model provides a kinetic rationale for why tubular proteinuria can occur even though a large excess in potential PT uptake capacity exists, and suggests testable predictions to expand our understanding of the recovery profile of filtered proteins along the PT. Abstract figure legend. Data from mouse models and from cultured proximal tubule (PT) cells were used to create a mathematical model that predicts the uptake profile of albumin and other filtered ligands along the mouse PT in normal and nephrotic states. The distinct contributions of cubilin receptors (magenta), megalin receptors (green), and fluid phase uptake (blue) to total albumin retrieval (black) in S1, S2, and S3 subsegments of the PT are delineated. Under normal conditions, albumin is primarily recovered in the S1 segment by cubilin, whereas the majority is retrieved in S2 under nephrotic conditions. Other proteins exhibit strikingly different uptake profiles. Our model explains how the distribution and capacity of high-affinity and low-affinity uptake pathways enable uptake of albumin over a broad range of filtered concentrations, and how tubular proteinuria can occur despite a large excess in potential PT uptake capacity. Created with BioRender.com. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Aurélie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Kimberly R Long
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Catherine J Baty
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Katherine E Shipman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Ora A Weisz
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| |
Collapse
|
9
|
Long KR, Rbaibi Y, Bondi CD, Ford BR, Poholek AC, Boyd-Shiwarski CR, Tan RJ, Locker JD, Weisz OA. Cubilin-, megalin-, and Dab2-dependent transcription revealed by CRISPR/Cas9 knockout in kidney proximal tubule cells. Am J Physiol Renal Physiol 2022; 322:F14-F26. [PMID: 34747197 PMCID: PMC8698540 DOI: 10.1152/ajprenal.00259.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023] Open
Abstract
The multiligand receptors megalin (Lrp2) and cubilin (Cubn) and their endocytic adaptor protein Dab2 (Dab2) play essential roles in maintaining the integrity of the apical endocytic pathway of proximal tubule (PT) cells and have complex and poorly understood roles in the development of chronic kidney disease. Here, we used RNA-sequencing and CRISPR/Cas9 knockout (KO) technology in a well-differentiated cell culture model to identify PT-specific transcriptional changes that are directly consequent to the loss of megalin, cubilin, or Dab2 expression. KO of Lrp2 had the greatest transcriptional effect, and nearly all genes whose expression was affected in Cubn KO and Dab2 KO cells were also changed in Lrp2 KO cells. Pathway analysis and more granular inspection of the altered gene profiles suggested changes in pathways with immunomodulatory functions that might trigger the pathological changes observed in KO mice and patients with Donnai-Barrow syndrome. In addition, differences in transcription patterns between Lrp2 and Dab2 KO cells suggested the possibility that altered spatial signaling by aberrantly localized receptors contributes to transcriptional changes upon the disruption of PT endocytic function. A reduction in transcripts encoding sodium-glucose cotransporter isoform 2 was confirmed in Lrp2 KO mouse kidney lysates by quantitative PCR analysis. Our results highlight the role of megalin as a master regulator and coordinator of ion transport, metabolism, and endocytosis in the PT. Compared with the studies in animal models, this approach provides a means to identify PT-specific transcriptional changes that are directly consequent to the loss of these target genes.NEW & NOTEWORTHY Megalin and cubilin receptors together with their adaptor protein Dab2 represent major components of the endocytic machinery responsible for efficient uptake of filtered proteins by the proximal tubule (PT). Dab2 and megalin expression have been implicated as both positive and negative modulators of kidney disease. We used RNA sequencing to knock out CRISPR/Cas9 cubilin, megalin, and Dab2 in highly differentiated PT cells to identify PT-specific changes that are directly consequent to knockout of each component.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Agenesis of Corpus Callosum/genetics
- Agenesis of Corpus Callosum/metabolism
- Agenesis of Corpus Callosum/pathology
- Animals
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- CRISPR-Associated Protein 9/genetics
- CRISPR-Cas Systems
- Cells, Cultured
- Databases, Genetic
- Gene Knockout Techniques
- Gene Regulatory Networks
- Hearing Loss, Sensorineural/genetics
- Hearing Loss, Sensorineural/metabolism
- Hearing Loss, Sensorineural/pathology
- Hernias, Diaphragmatic, Congenital/genetics
- Hernias, Diaphragmatic, Congenital/metabolism
- Hernias, Diaphragmatic, Congenital/pathology
- Humans
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Low Density Lipoprotein Receptor-Related Protein-2/genetics
- Low Density Lipoprotein Receptor-Related Protein-2/metabolism
- Male
- Mice, Knockout
- Monodelphis
- Myopia/genetics
- Myopia/metabolism
- Myopia/pathology
- Proteinuria/genetics
- Proteinuria/metabolism
- Proteinuria/pathology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Renal Tubular Transport, Inborn Errors/genetics
- Renal Tubular Transport, Inborn Errors/metabolism
- Renal Tubular Transport, Inborn Errors/pathology
- Transcription, Genetic
- Mice
Collapse
Affiliation(s)
- Kimberly R Long
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Youssef Rbaibi
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Corry D Bondi
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - B Rhodes Ford
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Amanda C Poholek
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Cary R Boyd-Shiwarski
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Roderick J Tan
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joseph D Locker
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ora A Weisz
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Krohn P, Rega LR, Harvent M, Festa BP, Taranta A, Luciani A, Dewulf J, Cremonesi A, Camassei FD, Hanson JVM, Gerth-Kahlert C, Emma F, Berquez M, Devuyst O. OUP accepted manuscript. Hum Mol Genet 2022; 31:2262-2278. [PMID: 35137071 PMCID: PMC9262394 DOI: 10.1093/hmg/ddac033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/21/2022] [Accepted: 02/02/2022] [Indexed: 11/14/2022] Open
Abstract
Recessive mutations in the CTNS gene encoding the lysosomal transporter cystinosin cause cystinosis, a lysosomal storage disease leading to kidney failure and multisystem manifestations. A Ctns knockout mouse model recapitulates features of cystinosis, but the delayed onset of kidney manifestations, phenotype variability and strain effects limit its use for mechanistic and drug development studies. To provide a better model for cystinosis, we generated a Ctns knockout rat model using CRISPR/Cas9 technology. The Ctns−/− rats display progressive cystine accumulation and crystal formation in multiple tissues including kidney, liver and thyroid. They show an early onset and progressive loss of urinary solutes, indicating generalized proximal tubule dysfunction, with development of typical swan-neck lesions, tubulointerstitial fibrosis and kidney failure, and decreased survival. The Ctns−/− rats also present crystals in the cornea, and bone and liver defects, as observed in patients. Mechanistically, the loss of cystinosin induces a phenotype switch associating abnormal proliferation and dedifferentiation, loss of apical receptors and transporters, and defective lysosomal activity and autophagy in the cells. Primary cultures of proximal tubule cells derived from the Ctns−/− rat kidneys confirmed the key changes caused by cystine overload, including reduced endocytic uptake, increased proliferation and defective lysosomal dynamics and autophagy. The novel Ctns−/− rat model and derived proximal tubule cell system provide invaluable tools to investigate the pathogenesis of cystinosis and to accelerate drug discovery.
Collapse
Affiliation(s)
- Patrick Krohn
- Institute of Physiology, University of Zurich, Zurich 8057, Switzerland
| | - Laura Rita Rega
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
| | - Marianne Harvent
- Institute of Physiology, University of Zurich, Zurich 8057, Switzerland
| | | | - Anna Taranta
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
| | | | - Joseph Dewulf
- Department of Laboratory Medicine, Cliniques universitaires Saint Luc, UCLouvain, Brussels 1200, Belgium
- Department of Biochemistry, de Duve Institute, UCLouvain, Brussels 1200, Belgium
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich 8032, Switzerland
| | | | - James V M Hanson
- Department of Ophthalmology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Christina Gerth-Kahlert
- Department of Ophthalmology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Francesco Emma
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
- Department of Pediatric Subspecialties, Division of Nephrology, Children’s Hospital Bambino Gesù, IRCCS, Rome 00165, Italy
| | - Marine Berquez
- To whom correspondence should be addressed at: University of Zurich, Mechanisms of Inherited Kidney Disorders Group, Winterthurerstrasse 190, Zurich 8057, Switzerland. Tel: +41 (0)44 635 51 07; (Marine Berquez); Tel: +41 (0)44 635 50 82; Fax: +41 (0)44 635 68 14; (Olivier Devuyst)
| | - Olivier Devuyst
- To whom correspondence should be addressed at: University of Zurich, Mechanisms of Inherited Kidney Disorders Group, Winterthurerstrasse 190, Zurich 8057, Switzerland. Tel: +41 (0)44 635 51 07; (Marine Berquez); Tel: +41 (0)44 635 50 82; Fax: +41 (0)44 635 68 14; (Olivier Devuyst)
| |
Collapse
|
11
|
Oltrabella F, Jackson-Crawford A, Yan G, Rixham S, Starborg T, Lowe M. IPIP27A cooperates with OCRL to support endocytic traffic in the zebrafish pronephric tubule. Hum Mol Genet 2021; 31:1183-1196. [PMID: 34673953 DOI: 10.1093/hmg/ddab307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 01/08/2023] Open
Abstract
Endocytosis is a fundamentally important process through which material is internalized into cells from the extracellular environment. In the renal proximal tubule, endocytosis of the abundant scavenger receptor megalin and its co-receptor cubilin play a vital role in retrieving low molecular weight proteins from the renal filtrate. Although we know much about megalin and its ligands, the machinery and mechanisms by which the receptor is trafficked through the endosomal system remain poorly defined. In this study, we show that Ipip27A, an interacting partner of the Lowe syndrome protein OCRL, is required for endocytic traffic of megalin within the proximal renal tubule of zebrafish larvae. Knockout of Ipip27A phenocopies the endocytic phenotype seen upon loss of OCRL, with a deficit in uptake of both fluid-phase and protein cargo, which is accompanied by a reduction in megalin abundance and altered endosome morphology. Rescue and co-depletion experiments indicate that Ipip27A functions together with OCRL to support proximal tubule endocytosis. The results therefore identify Ipip27A as a new player in endocytic traffic in the proximal tubule in vivo and support the view that defective endocytosis underlies the renal tubulopathy in Lowe syndrome and Dent-2 disease.
Collapse
Affiliation(s)
- Francesca Oltrabella
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.,Medical Scientific Liaison - Nephrology, Astellas Pharma, Via Dante, 20123 Milano, Italy
| | - Anthony Jackson-Crawford
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.,Department of Blood Sciences, Grange University Hospital, Llanyravon, Gwent, NP44 8YN
| | - Guanhua Yan
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Sarah Rixham
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Tobias Starborg
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.,Rosalind Franklin Institute, Harwell Campus, Didcot, OX11 0FA, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
12
|
Luciani A, Schumann A, Berquez M, Chen Z, Nieri D, Failli M, Debaix H, Festa BP, Tokonami N, Raimondi A, Cremonesi A, Carrella D, Forny P, Kölker S, Diomedi Camassei F, Diaz F, Moraes CT, Di Bernardo D, Baumgartner MR, Devuyst O. Impaired mitophagy links mitochondrial disease to epithelial stress in methylmalonyl-CoA mutase deficiency. Nat Commun 2020; 11:970. [PMID: 32080200 PMCID: PMC7033137 DOI: 10.1038/s41467-020-14729-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/28/2020] [Indexed: 01/09/2023] Open
Abstract
Deregulation of mitochondrial network in terminally differentiated cells contributes to a broad spectrum of disorders. Methylmalonic acidemia (MMA) is one of the most common inherited metabolic disorders, due to deficiency of the mitochondrial methylmalonyl-coenzyme A mutase (MMUT). How MMUT deficiency triggers cell damage remains unknown, preventing the development of disease-modifying therapies. Here we combine genetic and pharmacological approaches to demonstrate that MMUT deficiency induces metabolic and mitochondrial alterations that are exacerbated by anomalies in PINK1/Parkin-mediated mitophagy, causing the accumulation of dysfunctional mitochondria that trigger epithelial stress and ultimately cell damage. Using drug-disease network perturbation modelling, we predict targetable pathways, whose modulation repairs mitochondrial dysfunctions in patient-derived cells and alleviate phenotype changes in mmut-deficient zebrafish. These results suggest a link between primary MMUT deficiency, diseased mitochondria, mitophagy dysfunction and epithelial stress, and provide potential therapeutic perspectives for MMA.
Collapse
Affiliation(s)
- Alessandro Luciani
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland.
| | - Anke Schumann
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland
- Division of Metabolism and Children's Research Center, University Children's Hospital, 8032, Zurich, Switzerland
| | - Marine Berquez
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland
| | - Zhiyong Chen
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland
| | - Daniela Nieri
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland
| | - Mario Failli
- Department of Biomedicine, University of Eastern Finland, 70211, Kuopio, Finland
| | - Huguette Debaix
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland
| | - Beatrice Paola Festa
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland
| | - Natsuko Tokonami
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland
| | - Andrea Raimondi
- San Raffaele Scientific Institute, Experimental Imaging Center, 20132, Milan, Italy
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, 8032, Zurich, Switzerland
| | - Diego Carrella
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078, Naples, Italy
| | - Patrick Forny
- Division of Metabolism and Children's Research Center, University Children's Hospital, 8032, Zurich, Switzerland
| | - Stefan Kölker
- Division of Inherited Metabolic Diseases, University Children's Hospital Heidelberg, 69120, Heidelberg, Germany
| | | | - Francisca Diaz
- Department of Neurology, University of Miami Miller School of Medicine, 33136, Miami, FL, USA
| | - Carlos T Moraes
- Department of Neurology, University of Miami Miller School of Medicine, 33136, Miami, FL, USA
| | - Diego Di Bernardo
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078, Naples, Italy
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital, 8032, Zurich, Switzerland
| | - Olivier Devuyst
- Institute of Physiology and NCCR Kidney.CH, University of Zurich, 8057, Zurich, Switzerland.
- Division of Nephrology, Cliniques Universitaires Saint-Luc, 1040, Brussels, Belgium.
| |
Collapse
|
13
|
Chen Z, Luciani A, Mateos JM, Barmettler G, Giles RH, Neuhauss SCF, Devuyst O. Transgenic zebrafish modeling low-molecular-weight proteinuria and lysosomal storage diseases. Kidney Int 2019; 97:1150-1163. [PMID: 32061435 DOI: 10.1016/j.kint.2019.11.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 10/16/2019] [Accepted: 11/01/2019] [Indexed: 12/12/2022]
Abstract
Epithelial cells lining the proximal tubule of the kidney reabsorb and metabolize most of the filtered low-molecular-weight proteins through receptor-mediated endocytosis and lysosomal processing. Congenital and acquired dysfunctions of the proximal tubule are consistently reflected by the inappropriate loss of solutes including low-molecular-weight proteins in the urine. The zebrafish pronephros shares individual functional segments with the human nephron, including lrp2a/megalin-dependent endocytic transport processes of the proximal tubule. Although the zebrafish has been used as a model organism for toxicological studies and drug discovery, there is no available assay that allows large-scale assessment of proximal tubule function in larval or adult stages. Here we establish a transgenic Tg(lfabp::½vdbp-mCherry) zebrafish line expressing in the liver the N-terminal region of vitamin D-binding protein coupled to the acid-insensitive, red monomeric fluorescent protein mCherry (½vdbp-mCherry). This low-molecular-weight protein construct is secreted into the bloodstream, filtered through the glomerulus, reabsorbed by receptor-mediated endocytosis and processed in the lysosomes of proximal tubule cells of the fish. Thus, our proof-of-concept studies using zebrafish larvae knockout for lrp2a and clcn7 or exposed to known nephrotoxins (gentamicin and cisplatin) demonstrate that this transgenic line is useful to monitor low-molecular-weight proteinuria and lysosomal processing. This represents a powerful new model organism for drug screening and studies of nephrotoxicity.
Collapse
Affiliation(s)
- Zhiyong Chen
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - José María Mateos
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Gery Barmettler
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Rachel H Giles
- Department of Nephrology and Hypertension, Hubrecht Institute, Utrecht, The Netherlands; University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
14
|
Gliozzi ML, Espiritu EB, Shipman KE, Rbaibi Y, Long KR, Roy N, Duncan AW, Lazzara MJ, Hukriede NA, Baty CJ, Weisz OA. Effects of Proximal Tubule Shortening on Protein Excretion in a Lowe Syndrome Model. J Am Soc Nephrol 2019; 31:67-83. [PMID: 31676724 DOI: 10.1681/asn.2019020125] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 09/24/2019] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Lowe syndrome (LS) is an X-linked recessive disorder caused by mutations in OCRL, which encodes the enzyme OCRL. Symptoms of LS include proximal tubule (PT) dysfunction typically characterized by low molecular weight proteinuria, renal tubular acidosis (RTA), aminoaciduria, and hypercalciuria. How mutant OCRL causes these symptoms isn't clear. METHODS We examined the effect of deleting OCRL on endocytic traffic and cell division in newly created human PT CRISPR/Cas9 OCRL knockout cells, multiple PT cell lines treated with OCRL-targeting siRNA, and in orcl-mutant zebrafish. RESULTS OCRL-depleted human cells proliferated more slowly and about 10% of them were multinucleated compared with fewer than 2% of matched control cells. Heterologous expression of wild-type, but not phosphatase-deficient, OCRL prevented the accumulation of multinucleated cells after acute knockdown of OCRL but could not rescue the phenotype in stably edited knockout cell lines. Mathematic modeling confirmed that reduced PT length can account for the urinary excretion profile in LS. Both ocrl mutant zebrafish and zebrafish injected with ocrl morpholino showed truncated expression of megalin along the pronephric kidney, consistent with a shortened S1 segment. CONCLUSIONS Our data suggest a unifying model to explain how loss of OCRL results in tubular proteinuria as well as the other commonly observed renal manifestations of LS. We hypothesize that defective cell division during kidney development and/or repair compromises PT length and impairs kidney function in LS patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Nairita Roy
- Department of Pathology, McGowan Institute for Regenerative Medicine, and Pittsburgh Liver Research Center, Pittsburgh, Pennsylvania
| | - Andrew W Duncan
- Department of Pathology, McGowan Institute for Regenerative Medicine, and Pittsburgh Liver Research Center, Pittsburgh, Pennsylvania
| | - Matthew J Lazzara
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia; and
| | - Neil A Hukriede
- Department of Developmental Biology, and.,Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Ora A Weisz
- Renal-Electrolyte Division, Department of Medicine,
| |
Collapse
|
15
|
Zhu T, Zhang F, Li H, He Y, Zhang G, Huang N, Guo M, Li X. Long-term icariin treatment ameliorates cognitive deficits via CD4 + T cell-mediated immuno-inflammatory responses in APP/PS1 mice. Clin Interv Aging 2019; 14:817-826. [PMID: 31190768 PMCID: PMC6511656 DOI: 10.2147/cia.s208068] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Alzheimer’s disease (AD) is the most common neurodegenerative disorder that also involves neuroinflammation in addition to many other features. Icariin (ICA) as one of the active ingredients of Chinese herbal medicine has the immunomodulating function. This study aimed to investigate the immunotherapeutic potential of ICA on AD. Methods: APP/PS1 mice and wild type C57BL/6 mice were subjected to orally ICA administration (60 mg/kg/d) for 8 months. Then, the ethological and biochemical experiments, such as Morris water maze assay, Aβ ELISA, blood T cell flow cytometry, and plasma and brain cytokines array, were conducted to evaluate the effects of ICA administration. Results: ICA significantly improved spatial learning and memory retention in APP/PS1 mice. Long-term application of ICA could also reduce hippocampus Aβ deposition, modulate the differentiation of CD4+ T cells, and modulate the release of inflammatory cytokines in plasma and brain tissue. Conclusion: ICA shows the neuroprotective effects via modulating the CD4+ T lymphocyte-related immuno-inflammatory responses in APP/PS1 mice and may be a promising drug against AD progression.
Collapse
Affiliation(s)
- Tianrui Zhu
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Feng Zhang
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Heng Li
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Yi He
- Department of Neurology, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710021, People's Republic of China
| | - Guitao Zhang
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Nana Huang
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Mingming Guo
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, People's Republic of China
| | - Xiaohong Li
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| |
Collapse
|
16
|
Tian D, Shi X, Zhao Y, Peng X, Zou L, Xu L, Ma Y, Wen Y, Faulhaber-Walter R, Chen L. The effect of A1 adenosine receptor in diabetic megalin loss with caspase-1/IL18 signaling. Diabetes Metab Syndr Obes 2019; 12:1583-1596. [PMID: 31695457 PMCID: PMC6717852 DOI: 10.2147/dmso.s215531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/16/2019] [Indexed: 12/18/2022] Open
Abstract
PURPOSE In our previous study, exacerbation of albuminuria was observed in A1 adenosine receptor knockout (A1AR-/-) mice with diabetic nephropathy (DN), but the mechanism was unclear. Here, we investigated the relationship of megalin loss and albuminuria, to identify the protective effect of A1AR in megalin loss associated albuminuria by inhibiting pyroptosis-related caspase-1/IL-18 signaling of DN. METHODS We successfully collected DN patients' samples and built diabetes mice models induced by streptozotocin. Megalin, cubilin, and A1AR expression were detected in kidney tissue samples from DN patients and mice through immunohistochemical and immunofluorescent staining. A1AR, caspase-1, interleukin-18 (IL-18) expression were analyzed using Western blotting in wild-type and A1AR -/- mice. Human renal proximal tubular epithelial cells (PTC) were cultured with high glucose to observe the effect of A1AR agonist and antagonist on caspase-1/IL-18 and megalin injury. RESULTS The loss of megalin, co-localized with A1AR at PTC, was associated with the level of albuminuria in diabetic patients and mice. The injury of megalin-cubilin was accompanied with the A1AR upregulation (1.30±0.1 vs 0.98±0.2, P=0.042), the caspase-1 (1.33±0.1 vs 1.0±0.2, P=0.036), and IL-18 (1.26±0.2 vs 0.96±0.2, P=0.026) signaling activation in mice with DN. More severe pathological injury, 24 hrs urine albumin excretion (170.8±4.1 μg/d vs 132.0±2.9 μg/d vs 17.9±2.8 μg/d, P<0.001) and megalin-cubilin loss were observed in A1AR -/- DN mice with more pronounced caspase-1 (1.52±0.03 vs 1.20±0.01, P=0.017) and IL-18 (1.42±0.02 vs 1.21±0.02, P=0.018) secretion. High glucose could stimulate the secretion of caspase-1 (1.72 times, P≤0.01) and IL-18 (1.64 times, P≤0.01), which was abolished by A1AR agonist and aggravated by A1AR antagonist. CONCLUSION A1AR played a protective role in proximal tubular megalin loss associated albuminuria by inhibiting the pyroptosis-related caspase-1/IL-18 signaling in DN.
Collapse
Affiliation(s)
- Dongli Tian
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| | - Xiaoxiao Shi
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| | - Yumo Zhao
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| | - Xiaoyan Peng
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| | - Linfeng Zou
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| | - Lubin Xu
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| | - Ying Ma
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| | - Yubin Wen
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| | | | - Limeng Chen
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
- Correspondence: Limeng ChenDepartment of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Road 1 Shuaifuyuan, Wangfujing Street, Beijing100730, People’s Republic of ChinaTel +86 106 915 5351Fax +86 106 915 5058Email
| |
Collapse
|
17
|
Elmonem MA, Berlingerio SP, van den Heuvel LP, de Witte PA, Lowe M, Levtchenko EN. Genetic Renal Diseases: The Emerging Role of Zebrafish Models. Cells 2018; 7:cells7090130. [PMID: 30200518 PMCID: PMC6162634 DOI: 10.3390/cells7090130] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 12/14/2022] Open
Abstract
The structural and functional similarity of the larval zebrafish pronephros to the human nephron, together with the recent development of easier and more precise techniques to manipulate the zebrafish genome have motivated many researchers to model human renal diseases in the zebrafish. Over the last few years, great advances have been made, not only in the modeling techniques of genetic diseases in the zebrafish, but also in how to validate and exploit these models, crossing the bridge towards more informative explanations of disease pathophysiology and better designed therapeutic interventions in a cost-effective in vivo system. Here, we review the significant progress in these areas giving special attention to the renal phenotype evaluation techniques. We further discuss the future applications of such models, particularly their role in revealing new genetic diseases of the kidney and their potential use in personalized medicine.
Collapse
Affiliation(s)
- Mohamed A Elmonem
- Department of Pediatric Nephrology & Development and Regeneration, University Hospitals Leuven, KU Leuven-University of Leuven, Herestraat 49, Box 817, 3000 Leuven, Belgium.
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, 11628 Cairo, Egypt.
| | - Sante Princiero Berlingerio
- Department of Pediatric Nephrology & Development and Regeneration, University Hospitals Leuven, KU Leuven-University of Leuven, Herestraat 49, Box 817, 3000 Leuven, Belgium.
| | - Lambertus P van den Heuvel
- Department of Pediatric Nephrology & Development and Regeneration, University Hospitals Leuven, KU Leuven-University of Leuven, Herestraat 49, Box 817, 3000 Leuven, Belgium.
- Department of Pediatric Nephrology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Peter A de Witte
- Laboratory for Molecular Bio-Discovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven-University of Leuven, 3000 Leuven, Belgium.
| | - Martin Lowe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
| | - Elena N Levtchenko
- Department of Pediatric Nephrology & Development and Regeneration, University Hospitals Leuven, KU Leuven-University of Leuven, Herestraat 49, Box 817, 3000 Leuven, Belgium.
| |
Collapse
|
18
|
Schütte-Nütgen K, Edeling M, Mendl G, Krahn MP, Edemir B, Weide T, Kremerskothen J, Michgehl U, Pavenstädt H. Getting a Notch closer to renal dysfunction: activated Notch suppresses expression of the adaptor protein Disabled-2 in tubular epithelial cells. FASEB J 2018; 33:821-832. [PMID: 30052485 DOI: 10.1096/fj.201800392rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Reactivation of Notch signaling in kidneys of animal models and patients with chronic kidney disease (CKD) has been shown to contribute to epithelial injury and fibrosis development. Here, we investigated the mechanisms of Notch-induced injury in renal epithelial cells. We performed genome-wide transcriptome analysis to identify Notch target genes using an in vitro system of cultured tubular epithelial cells expressing the intracellular domain of Notch1. One of the top downregulated genes was Disabled-2 ( Dab2). With the use of Drosophila nephrocytes as a model system, we found that Dab (the Drosophila homolog of Dab2) knockdown resulted in a significant filtration defect, indicating that loss of Dab2 plays a functional role in kidney disease development. We showed that Dab2 expression in cultured tubular epithelial cells is involved in endocytic regulation and that it also protects cells from TGF-β-induced epithelial-to-mesenchymal transition. In vivo correlation studies indicated its additional role in renal ischemia-induced injury. Together, these data suggest that Dab2 plays a versatile role in the kidney and may impact on acute and CKDs.-Schütte-Nütgen, K., Edeling, M., Mendl, G., Krahn, M. P., Edemir, B., Weide, T., Kremerskothen, J., Michgehl, U., Pavenstädt, H. Getting a Notch closer to renal dysfunction: activated Notch suppresses expression of the adaptor protein Disabled-2 in tubular epithelial cells.
Collapse
Affiliation(s)
| | - Maria Edeling
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | - Gudrun Mendl
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | - Michael P Krahn
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | - Bayram Edemir
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and.,Department of Hematology and Oncology, Internal Medicine IV, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Thomas Weide
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | | | - Ulf Michgehl
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | - Hermann Pavenstädt
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| |
Collapse
|
19
|
Endocytic receptor LRP2/megalin—of holoprosencephaly and renal Fanconi syndrome. Pflugers Arch 2017; 469:907-916. [DOI: 10.1007/s00424-017-1992-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 12/31/2022]
|
20
|
Nielsen R, Christensen EI, Birn H. Megalin and cubilin in proximal tubule protein reabsorption: from experimental models to human disease. Kidney Int 2017; 89:58-67. [PMID: 26759048 DOI: 10.1016/j.kint.2015.11.007] [Citation(s) in RCA: 341] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/17/2015] [Accepted: 08/19/2015] [Indexed: 01/19/2023]
Abstract
Proximal tubule protein uptake is mediated by 2 receptors, megalin and cubilin. These receptors rescue a variety of filtered ligands, including biomarkers, essential vitamins, and hormones. Receptor gene knockout animal models have identified important functions of the receptors and have established their essential role in modulating urinary protein excretion. Rare genetic syndromes associated with dysfunction of these receptors have been identified and characterized, providing additional information on the importance of these receptors in humans. Using various disease models in combination with receptor gene knockout, the implications of receptor dysfunction in acute and chronic kidney injury have been explored and have pointed to potential new roles of these receptors. Based on data from animal models, this paper will review current knowledge on proximal tubule endocytic receptor function and regulation, and their role in renal development, protein reabsorption, albumin uptake, and normal renal physiology. These findings have implications for the pathophysiology and diagnosis of proteinuric renal diseases. We will examine the limitations of the different models and compare the findings to phenotypic observations in inherited human disorders associated with receptor dysfunction. Furthermore, evidence from receptor knockout mouse models as well as human observations suggesting a role of protein receptors for renal disease will be discussed in light of conditions such as chronic kidney disease, diabetes, and hypertension.
Collapse
Affiliation(s)
- Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Henrik Birn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
21
|
Abstract
Cells lining the proximal tubule (PT) of the kidney are highly specialized for apical endocytosis of filtered proteins and small bioactive molecules from the glomerular ultrafiltrate to maintain essentially protein-free urine. Compromise of this pathway results in low molecular weight (LMW) proteinuria that can progress to end-stage kidney disease. This review describes our current understanding of the endocytic pathway and the multiligand receptors that mediate LMW protein uptake in PT cells, how these are regulated in response to physiologic cues, and the molecular basis of inherited diseases characterized by LMW proteinuria.
Collapse
Affiliation(s)
- Megan L Eshbach
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; ,
| | - Ora A Weisz
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; ,
| |
Collapse
|
22
|
Kusakabe M, Ishikawa A, Ravinet M, Yoshida K, Makino T, Toyoda A, Fujiyama A, Kitano J. Genetic basis for variation in salinity tolerance between stickleback ecotypes. Mol Ecol 2016; 26:304-319. [DOI: 10.1111/mec.13875] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/30/2016] [Accepted: 09/07/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Makoto Kusakabe
- Atmosphere and Ocean Research Institute; The University of Tokyo; Kashiwanoha 5-1-5 Kashiwa Chiba 277-8564 Japan
- Department of Biological Science; Faculty of Science; Shizuoka University; 836 Ohya, Suruga-ku Shizuoka 422-8529 Japan
| | - Asano Ishikawa
- Division of Ecological Genetics; National Institute of Genetics; Yata 1111 Mishima Shizuoka 411-8540 Japan
| | - Mark Ravinet
- Division of Ecological Genetics; National Institute of Genetics; Yata 1111 Mishima Shizuoka 411-8540 Japan
- Centre for Ecological and Evolutionary Synthesis; University of Oslo; P.O. Box 1066 Blindern Oslo NO-0316 Oslo Norway
| | - Kohta Yoshida
- Division of Ecological Genetics; National Institute of Genetics; Yata 1111 Mishima Shizuoka 411-8540 Japan
| | - Takashi Makino
- Department of Ecology and Evolutionary Biology; Graduate School of Life Sciences; Tohoku University; Sendai Miyagi 980-8578 Japan
| | - Atsushi Toyoda
- Comparative Genomics Laboratory; National Institute of Genetics; Yata 1111 Mishima Shizuoka 411-8540 Japan
| | - Asao Fujiyama
- Comparative Genomics Laboratory; National Institute of Genetics; Yata 1111 Mishima Shizuoka 411-8540 Japan
| | - Jun Kitano
- Division of Ecological Genetics; National Institute of Genetics; Yata 1111 Mishima Shizuoka 411-8540 Japan
| |
Collapse
|
23
|
Nasrallah R, Fast EM, Solaimani P, Knezevic K, Eliades A, Patel R, Thambyrajah R, Unnikrishnan A, Thoms J, Beck D, Vink CS, Smith A, Wong J, Shepherd M, Kent D, Roychoudhuri R, Paul F, Klippert J, Hammes A, Willnow T, Göttgens B, Dzierzak E, Zon LI, Lacaud G, Kouskoff V, Pimanda JE. Identification of novel regulators of developmental hematopoiesis using Endoglin regulatory elements as molecular probes. Blood 2016; 128:1928-1939. [PMID: 27554085 PMCID: PMC5064716 DOI: 10.1182/blood-2016-02-697870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 08/15/2016] [Indexed: 12/13/2022] Open
Abstract
Enhancers are the primary determinants of cell identity, and specific promoter/enhancer combinations of Endoglin (ENG) have been shown to target blood and endothelium in the embryo. Here, we generated a series of embryonic stem cell lines, each targeted with reporter constructs driven by specific promoter/enhancer combinations of ENG, to evaluate their discriminative potential and value as molecular probes of the corresponding transcriptome. The Eng promoter (P) in combination with the -8/+7/+9-kb enhancers, targeted cells in FLK1 mesoderm that were enriched for blast colony forming potential, whereas the P/-8-kb enhancer targeted TIE2+/c-KIT+/CD41- endothelial cells that were enriched for hematopoietic potential. These fractions were isolated using reporter expression and their transcriptomes profiled by RNA-seq. There was high concordance between our signatures and those from embryos with defects at corresponding stages of hematopoiesis. Of the 6 genes that were upregulated in both hemogenic mesoderm and hemogenic endothelial fractions targeted by the reporters, LRP2, a multiligand receptor, was the only gene that had not previously been associated with hematopoiesis. We show that LRP2 is indeed involved in definitive hematopoiesis and by doing so validate the use of reporter gene-coupled enhancers as probes to gain insights into transcriptional changes that facilitate cell fate transitions.
Collapse
Affiliation(s)
- Rabab Nasrallah
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Eva M Fast
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA; Howard Hughes Medical Institute, Stem Cell Program and Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Parham Solaimani
- Erasmus Medical Center Stem Cell Institute, Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kathy Knezevic
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Alexia Eliades
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Rahima Patel
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Roshana Thambyrajah
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Ashwin Unnikrishnan
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Julie Thoms
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Dominik Beck
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Chris S Vink
- Erasmus Medical Center Stem Cell Institute, Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands; Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Aileen Smith
- Department of Haematology, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Jason Wong
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Mairi Shepherd
- Department of Haematology, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - David Kent
- Department of Haematology, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Rahul Roychoudhuri
- The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Fabian Paul
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; and
| | - Julia Klippert
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; and
| | - Annette Hammes
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; and
| | - Thomas Willnow
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; and
| | - Bertie Göttgens
- Department of Haematology, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Elaine Dzierzak
- Erasmus Medical Center Stem Cell Institute, Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands; Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Leonard I Zon
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA; Howard Hughes Medical Institute, Stem Cell Program and Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - George Lacaud
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Valerie Kouskoff
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - John E Pimanda
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia; Department of Haematology, The Prince of Wales Hospital, Sydney, Australia
| |
Collapse
|
24
|
Christ A, Herzog K, Willnow TE. LRP2, an auxiliary receptor that controls sonic hedgehog signaling in development and disease. Dev Dyn 2016; 245:569-79. [PMID: 26872844 DOI: 10.1002/dvdy.24394] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 12/31/2022] Open
Abstract
To fulfill their multiple roles in organ development and adult tissue homeostasis, hedgehog (HH) morphogens act through their receptor Patched (PTCH) on target cells. However, HH actions also require HH binding proteins, auxiliary cell surface receptors that agonize or antagonize morphogen signaling in a context-dependent manner. Here, we discuss recent findings on the LDL receptor-related protein 2 (LRP2), an exemplary HH binding protein that modulates sonic hedgehog activities in stem and progenitor cell niches in embryonic and adult tissues. LRP2 functions are crucial for developmental processes in a number of tissues, including the brain, the eye, and the heart, and defects in this receptor pathway are the cause of devastating congenital diseases in humans. Developmental Dynamics 245:569-579, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Katja Herzog
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| |
Collapse
|
25
|
Wan X, Lee MS, Zhou W. Dosage-dependent role of Rac1 in podocyte injury. Am J Physiol Renal Physiol 2016; 310:F777-F784. [PMID: 26792065 DOI: 10.1152/ajprenal.00381.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/19/2016] [Indexed: 01/03/2023] Open
Abstract
Activation of small GTPase Rac1 in podocytes is associated with rodent models of kidney injury and familial nephrotic syndrome. Induced Rac1 activation in podocytes in transgenic mice results in rapid transient proteinuria and foot process effacement, but not glomerular sclerosis. Thus it remains an open question whether abnormal activation of Rac1 in podocytes is sufficient to cause permanent podocyte damage. Using a number of transgenic zebrafish models, we showed that moderate elevation of Rac1 activity in podocytes did not impair the glomerular filtration barrier but aggravated metronidazole-induced podocyte injury, while inhibition of Rac1 activity ameliorated metronidazole-induced podocyte injury. Furthermore, a further increase in Rac1 activity in podocytes was sufficient to cause proteinuria and foot process effacement, which resulted in edema and lethality in juvenile zebrafish. We also found that activation of Rac1 in podocytes significantly downregulated the expression of nephrin and podocin, suggesting an adverse effect of Rac1 on slit diaphragm protein expression. Taken together, our data have demonstrated a causal link between excessive Rac1 activity and podocyte injury in a dosage-dependent manner, and transgenic zebrafish of variable Rac1 activities in podocytes may serve as useful animal models for the study of Rac1-related podocytopathy.
Collapse
Affiliation(s)
- Xiaoyang Wan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Mi-Sun Lee
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Weibin Zhou
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
26
|
Mushirobira Y, Mizuta H, Luo W, Todo T, Hara A, Reading BJ, Sullivan CV, Hiramatsu N. Molecular cloning and partial characterization of a low‐density lipoprotein receptor‐related protein 13 (Lrp13) involved in vitellogenin uptake in the cutthroat trout (
Oncorhynchus clarki
). Mol Reprod Dev 2015; 82:986-1000. [DOI: 10.1002/mrd.22579] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/30/2015] [Indexed: 01/01/2023]
Affiliation(s)
- Yuji Mushirobira
- Graduate School of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Hiroko Mizuta
- Graduate School of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Wenshu Luo
- Graduate School of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Takashi Todo
- Faculty of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Akihiko Hara
- Faculty of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Benjamin J. Reading
- Department of Applied EcologyNorth Carolina State UniversityRaleighNorth Carolina
| | | | | |
Collapse
|
27
|
Cases O, Joseph A, Obry A, Santin MD, Ben-Yacoub S, Pâques M, Amsellem-Levera S, Bribian A, Simonutti M, Augustin S, Debeir T, Sahel JA, Christ A, de Castro F, Lehéricy S, Cosette P, Kozyraki R. Foxg1-Cre Mediated Lrp2 Inactivation in the Developing Mouse Neural Retina, Ciliary and Retinal Pigment Epithelia Models Congenital High Myopia. PLoS One 2015; 10:e0129518. [PMID: 26107939 PMCID: PMC4480972 DOI: 10.1371/journal.pone.0129518] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/08/2015] [Indexed: 12/11/2022] Open
Abstract
Myopia is a common ocular disorder generally due to increased axial length of the eye-globe. Its extreme form high myopia (HM) is a multifactorial disease leading to retinal and scleral damage, visual impairment or loss and is an important health issue. Mutations in the endocytic receptor LRP2 gene result in Donnai-Barrow (DBS) and Stickler syndromes, both characterized by HM. To clearly establish the link between Lrp2 and congenital HM we inactivated Lrp2 in the mouse forebrain including the neural retina and the retinal and ciliary pigment epithelia. High resolution in vivo MRI imaging and ophthalmological analyses showed that the adult Lrp2-deficient eyes were 40% longer than the control ones mainly due to an excessive elongation of the vitreal chamber. They had an apparently normal intraocular pressure and developed chorioretinal atrophy and posterior scleral staphyloma features reminiscent of human myopic retinopathy. Immunomorphological and ultrastructural analyses showed that increased eye lengthening was first observed by post-natal day 5 (P5) and that it was accompanied by a rapid decrease of the bipolar, photoreceptor and retinal ganglion cells, and eventually the optic nerve axons. It was followed by scleral thinning and collagen fiber disorganization, essentially in the posterior pole. We conclude that the function of LRP2 in the ocular tissues is necessary for normal eye growth and that the Lrp2-deficient eyes provide a unique tool to further study human HM.
Collapse
Affiliation(s)
- Olivier Cases
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
| | - Antoine Joseph
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
| | - Antoine Obry
- CNRS, UMR_6270, PISSARO Proteomics Platform, Institute for Research and Innovation in Biomedicine, Rouen University Hospital, Rouen, F-76821, France
- INSERM, U905, PISSARO Proteomics Platform, Institute for Research and Innovation in Biomedicine, Rouen University Hospital, Rouen, F-76821, France
| | | | - Sirine Ben-Yacoub
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
| | - Michel Pâques
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
- Centre Hospitalier National d’Ophthalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France
| | - Sabine Amsellem-Levera
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
| | - Ana Bribian
- Grupo de Neurobiologia del Desarollo-GNDe, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Manuel Simonutti
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
| | - Sébastien Augustin
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
| | | | - José Alain Sahel
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
- Centre Hospitalier National d’Ophthalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France
| | - Annabel Christ
- Max-Delbrück-Center for Molecular Medicine, Berlin, D-13125, Germany
| | - Fernando de Castro
- Grupo de Neurobiologia del Desarollo-GNDe, Hospital Nacional de Parapléjicos, Toledo, Spain
| | | | - Pascal Cosette
- CNRS, UMR_6270, PISSARO Proteomics Platform, Institute for Research and Innovation in Biomedicine, Rouen University Hospital, Rouen, F-76821, France
| | - Renata Kozyraki
- INSERM, U968, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, Paris, F-75012, France
- CNRS, UMR_7210, Paris, F-75012, France
- * E-mail:
| |
Collapse
|
28
|
Oltrabella F, Pietka G, Ramirez IBR, Mironov A, Starborg T, Drummond IA, Hinchliffe KA, Lowe M. The Lowe syndrome protein OCRL1 is required for endocytosis in the zebrafish pronephric tubule. PLoS Genet 2015; 11:e1005058. [PMID: 25838181 PMCID: PMC4383555 DOI: 10.1371/journal.pgen.1005058] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 02/07/2015] [Indexed: 02/03/2023] Open
Abstract
Lowe syndrome and Dent-2 disease are caused by mutation of the inositol 5-phosphatase OCRL1. Despite our increased understanding of the cellular functions of OCRL1, the underlying basis for the renal tubulopathy seen in both human disorders, of which a hallmark is low molecular weight proteinuria, is currently unknown. Here, we show that deficiency in OCRL1 causes a defect in endocytosis in the zebrafish pronephric tubule, a model for the mammalian renal tubule. This coincides with a reduction in levels of the scavenger receptor megalin and its accumulation in endocytic compartments, consistent with reduced recycling within the endocytic pathway. We also observe reduced numbers of early endocytic compartments and enlarged vacuolar endosomes in the sub-apical region of pronephric cells. Cell polarity within the pronephric tubule is unaffected in mutant embryos. The OCRL1-deficient embryos exhibit a mild ciliogenesis defect, but this cannot account for the observed impairment of endocytosis. Catalytic activity of OCRL1 is required for renal tubular endocytosis and the endocytic defect can be rescued by suppression of PIP5K. These results indicate for the first time that OCRL1 is required for endocytic trafficking in vivo, and strongly support the hypothesis that endocytic defects are responsible for the renal tubulopathy in Lowe syndrome and Dent-2 disease. Moreover, our results reveal PIP5K as a potential therapeutic target for Lowe syndrome and Dent-2 disease. Phosphoinositide lipids are key regulators of cellular physiology and consequently enzymes that generate or remove these lipids are of fundamental importance. Mutation of one such enzyme, called OCRL1, causes two disorders in humans, Lowe syndrome and Dent-2 disease. However, the underlying mechanisms remain poorly defined. Here, we demonstrate that OCRL1 regulates endocytosis, the process by which cells internalize material from their extracellular environment. Importantly, this is demonstrated in a physiologically relevant tissue in vivo, namely the zebrafish renal tubule. Defective endocytosis can explain the renal symptoms seen in Lowe syndrome and Dent-2 patients. We also report that defects in cell polarity or cilia formation cannot explain the renal symptoms. This study not only increases our understanding of the endocytic pathway, it also provides a mechanistic explanation for the renal defects observed in Lowe syndrome and Dent-2 patients.
Collapse
Affiliation(s)
| | - Grzegorz Pietka
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Aleksandr Mironov
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Toby Starborg
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Iain A Drummond
- Nephrology Division, Massachusetts General Hospital and Department of Genetics, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | | | - Martin Lowe
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
McMahon GM, Olden M, Garnaas M, Yang Q, Liu X, Hwang SJ, Larson MG, Goessling W, Fox CS. Sequencing of LRP2 reveals multiple rare variants associated with urinary trefoil factor-3. J Am Soc Nephrol 2014; 25:2896-905. [PMID: 24876117 DOI: 10.1681/asn.2013111240] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Novel biomarkers are being investigated to identify patients with kidney disease. We measured a panel of 13 urinary biomarkers in participants from the Offspring Cohort of the Framingham Heart Study. Using an Affymetrix chip with imputation to 2.5 M single-nucleotide polymorphisms (SNPs), we conducted a GWAS of these biomarkers (n=2640) followed by exonic sequencing and genotyping. Functional studies in zebrafish were used to investigate histologic correlation with renal function. Across all 13 biomarkers, there were 97 significant SNPs at three loci. Lead SNPs at each locus were rs6555820 (P=6.7×10(-49); minor allele frequency [MAF]=0.49) in HAVCR1 (associated with kidney injury molecule-1), rs7565788 (P=2.15×10(-16); MAF=0.22) in LRP2 (associated with trefoil factor 3 [TFF3]), and rs11048230 (P=4.77×10(-8); MAF=0.10) in an intergenic region near RASSF8 (associated with vascular endothelial growth factor). Validation in the CKDGen Consortium (n=67,093) showed that only rs7565788 at LRP2, which encodes megalin, was associated with eGFR (P=0.003). Sequencing of exons 16-72 of LRP2 in 200 unrelated individuals at extremes of urinary TFF3 levels identified 197 variants (152 rare; MAF<0.05), 31 of which (27 rare) were nonsynonymous. In aggregate testing, rare variants were associated with urinary TFF3 levels (P=0.003), and the lead GWAS signal was not explained by these variants. Knockdown of LRP2 in zebrafish did not alter the renal phenotype in static or kidney injury models. In conclusion, this study revealed common variants associated with urinary levels of TFF3, kidney injury molecule-1, and vascular endothelial growth factor and identified a cluster of rare variants independently associated with TFF3.
Collapse
Affiliation(s)
- Gearoid M McMahon
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Divisions of Nephrology
| | - Matthias Olden
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | | | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Xuan Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Martin G Larson
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | | | | | - Caroline S Fox
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Endocrinology and Metabolism, Brigham and Women's Hospital, Boston, Massachusetts; and
| |
Collapse
|
30
|
Mehta ZB, Pietka G, Lowe M. The cellular and physiological functions of the Lowe syndrome protein OCRL1. Traffic 2014; 15:471-87. [PMID: 24499450 PMCID: PMC4278560 DOI: 10.1111/tra.12160] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/03/2014] [Accepted: 02/05/2014] [Indexed: 12/17/2022]
Abstract
Phosphoinositide lipids play a key role in cellular physiology, participating in a wide array of cellular processes. Consequently, mutation of phosphoinositide-metabolizing enzymes is responsible for a growing number of diseases in humans. Two related disorders, oculocerebrorenal syndrome of Lowe (OCRL) and Dent-2 disease, are caused by mutation of the inositol 5-phosphatase OCRL1. Here, we review recent advances in our understanding of OCRL1 function. OCRL1 appears to regulate many processes within the cell, most of which depend upon coordination of membrane dynamics with remodeling of the actin cytoskeleton. Recently developed animal models have managed to recapitulate features of Lowe syndrome and Dent-2 disease, and revealed new insights into the underlying mechanisms of these disorders. The continued use of both cell-based approaches and animal models will be key to fully unraveling OCRL1 function, how its loss leads to disease and, importantly, the development of therapeutics to treat patients.
Collapse
Affiliation(s)
- Zenobia B Mehta
- Faculty of Life Sciences, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK; Current address: Faculty of Medicine, Imperial College, London, UK
| | | | | |
Collapse
|
31
|
Willnow TE, Christ A, Hammes A. Endocytic receptor-mediated control of morphogen signaling. Development 2013; 139:4311-9. [PMID: 23132241 DOI: 10.1242/dev.084467] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Receptor-mediated endocytosis provides a mechanism by which cells take up signaling molecules from the extracellular space. Recent studies have shown that one class of endocytic receptors, the low-density lipoprotein receptor-related proteins (LRPs), is of particular relevance for embryonic development. In this Primer, we describe how LRPs constitute central pathways that modulate morphogen presentation to target tissues and cellular signal reception, and how LRP dysfunction leads to developmental disturbances in many species.
Collapse
Affiliation(s)
- Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, D-13125 Berlin, Germany.
| | | | | |
Collapse
|
32
|
Gerlach GF, Wingert RA. Kidney organogenesis in the zebrafish: insights into vertebrate nephrogenesis and regeneration. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:559-85. [PMID: 24014448 DOI: 10.1002/wdev.92] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vertebrates form a progressive series of up to three kidney organs during development-the pronephros, mesonephros, and metanephros. Each kidney derives from the intermediate mesoderm and is comprised of conserved excretory units called nephrons. The zebrafish is a powerful model for vertebrate developmental genetics, and recent studies have illustrated that zebrafish and mammals share numerous similarities in nephron composition and physiology. The zebrafish embryo forms an architecturally simple pronephros that has two nephrons, and these eventually become a scaffold onto which a mesonephros of several hundred nephrons is constructed during larval stages. In adult zebrafish, the mesonephros exhibits ongoing nephrogenesis, generating new nephrons from a local pool of renal progenitors during periods of growth or following kidney injury. The characteristics of the zebrafish pronephros and mesonephros make them genetically tractable kidney systems in which to study the functions of renal genes and address outstanding questions about the mechanisms of nephrogenesis. Here, we provide an overview of the formation and composition of these zebrafish kidney organs, and discuss how various zebrafish mutants, gene knockdowns, and transgenic models have created frameworks in which to further delineate nephrogenesis pathways.
Collapse
Affiliation(s)
- Gary F Gerlach
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | | |
Collapse
|
33
|
Current world literature. Curr Opin Pediatr 2012; 24:277-84. [PMID: 22414891 DOI: 10.1097/mop.0b013e328351e459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
34
|
Sherpa T, Hunter SS, Frey RA, Robison BD, Stenkamp DL. Retinal proliferation response in the buphthalmic zebrafish, bugeye. Exp Eye Res 2011; 93:424-436. [PMID: 21723280 DOI: 10.1016/j.exer.2011.06.001.retinal] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 05/23/2011] [Accepted: 06/05/2011] [Indexed: 05/23/2023]
Abstract
The zebrafish retina regenerates in response to acute retinal lesions, replacing damaged neurons with new neurons. In this study we test the hypothesis that chronic stress to inner retinal neurons also triggers a retinal regeneration response in the bugeye zebrafish. Mutations in the lrp2 gene in zebrafish are associated with a progressive eye phenotype (bugeye) that models several risk factors for human glaucoma including buphthalmos (enlarged eyes), elevated intraocular pressure (IOP), and upregulation of genes related to retinal ganglion cell pathology. The retinas of adult bugeye zebrafish showed high rates of ongoing proliferation which resulted in the production of a small number of new retinal neurons, particularly photoreceptors. A marker of mechanical cell stress, Hsp27, was strongly expressed in inner retinal neurons and glia of bugeye retinas. The more enlarged eyes of individual bugeye zebrafish showed disrupted retinal lamination, and a persistent reduced density of neurons in the ganglion cell layer (GCL), although total numbers of GCL neurons were higher than in control eyes. Despite the presence of a proliferative response to damage, the adult bugeye zebrafish remained behaviorally blind. These findings suggest the existence of an unsuccessful regenerative response to a persistent pathological condition in the bugeye zebrafish.
Collapse
Affiliation(s)
- Tshering Sherpa
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844-3051, USA
| | | | | | | | | |
Collapse
|
35
|
Sherpa T, Hunter SS, Frey RA, Robison BD, Stenkamp DL. Retinal proliferation response in the buphthalmic zebrafish, bugeye. Exp Eye Res 2011; 93:424-36. [PMID: 21723280 DOI: 10.1016/j.exer.2011.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 05/23/2011] [Accepted: 06/05/2011] [Indexed: 10/18/2022]
Abstract
The zebrafish retina regenerates in response to acute retinal lesions, replacing damaged neurons with new neurons. In this study we test the hypothesis that chronic stress to inner retinal neurons also triggers a retinal regeneration response in the bugeye zebrafish. Mutations in the lrp2 gene in zebrafish are associated with a progressive eye phenotype (bugeye) that models several risk factors for human glaucoma including buphthalmos (enlarged eyes), elevated intraocular pressure (IOP), and upregulation of genes related to retinal ganglion cell pathology. The retinas of adult bugeye zebrafish showed high rates of ongoing proliferation which resulted in the production of a small number of new retinal neurons, particularly photoreceptors. A marker of mechanical cell stress, Hsp27, was strongly expressed in inner retinal neurons and glia of bugeye retinas. The more enlarged eyes of individual bugeye zebrafish showed disrupted retinal lamination, and a persistent reduced density of neurons in the ganglion cell layer (GCL), although total numbers of GCL neurons were higher than in control eyes. Despite the presence of a proliferative response to damage, the adult bugeye zebrafish remained behaviorally blind. These findings suggest the existence of an unsuccessful regenerative response to a persistent pathological condition in the bugeye zebrafish.
Collapse
Affiliation(s)
- Tshering Sherpa
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844-3051, USA
| | | | | | | | | |
Collapse
|