1
|
Gorman BR, Voloudakis G, Igo RP, Kinzy T, Halladay CW, Bigdeli TB, Zeng B, Venkatesh S, Cooke Bailey JN, Crawford DC, Markianos K, Dong F, Schreiner PA, Zhang W, Hadi T, Anger MD, Stockwell A, Melles RB, Yin J, Choquet H, Kaye R, Patasova K, Patel PJ, Yaspan BL, Jorgenson E, Hysi PG, Lotery AJ, Gaziano JM, Tsao PS, Fliesler SJ, Sullivan JM, Greenberg PB, Wu WC, Assimes TL, Pyarajan S, Roussos P, Peachey NS, Iyengar SK. Genome-wide association analyses identify distinct genetic architectures for age-related macular degeneration across ancestries. Nat Genet 2024; 56:2659-2671. [PMID: 39623103 DOI: 10.1038/s41588-024-01764-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 04/22/2024] [Indexed: 12/12/2024]
Abstract
To effectively reduce vision loss due to age-related macular generation (AMD) on a global scale, knowledge of its genetic architecture in diverse populations is necessary. A critical element, AMD risk profiles in African and Hispanic/Latino ancestries, remains largely unknown. We combined data in the Million Veteran Program with five other cohorts to conduct the first multi-ancestry genome-wide association study of AMD and discovered 63 loci (30 novel). We observe marked cross-ancestry heterogeneity at major risk loci, especially in African-ancestry populations which demonstrate a primary signal in a major histocompatibility complex class II haplotype and reduced risk at the established CFH and ARMS2/HTRA1 loci. Dissecting local ancestry in admixed individuals, we find significantly smaller marginal effect sizes for CFH risk alleles in African ancestry haplotypes. Broadening efforts to include ancestrally distinct populations helped uncover genes and pathways that boost risk in an ancestry-dependent manner and are potential targets for corrective therapies.
Collapse
Affiliation(s)
- Bryan R Gorman
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters Veterans Affairs Medical Center, New York/New Jersey VA Health Care Network, Bronx, NY, USA
| | - Robert P Igo
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Tyler Kinzy
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher W Halladay
- Center of Innovation in Long Term Services and Supports, VA Providence Healthcare System, Providence, RI, USA
| | - Tim B Bigdeli
- Research Service, VA New York Harbor Healthcare System, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Biao Zeng
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanan Venkatesh
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters Veterans Affairs Medical Center, New York/New Jersey VA Health Care Network, Bronx, NY, USA
| | - Jessica N Cooke Bailey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics & Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Dana C Crawford
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics & Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Kyriacos Markianos
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
| | - Frederick Dong
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Patrick A Schreiner
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Wen Zhang
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tamer Hadi
- Eye Clinic, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Ophthalmology and Visual Sciences, University Hospitals Eye Institute, Cleveland, OH, USA
| | - Matthew D Anger
- Eye Clinic, VA Western NY Healthcare System, Buffalo, NY, USA
- Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
| | - Amy Stockwell
- Department of Human Genetics, Genentech, South San Francisco, CA, USA
| | - Ronald B Melles
- Department of Ophthalmology, Kaiser Permanente Northern California, Redwood City, CA, USA
| | - Jie Yin
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Rebecca Kaye
- Southampton Eye Unit, University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Karina Patasova
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Praveen J Patel
- National Institute for Health and Care Research Biomedical Research Centre, Moorfields Eye Hospital National Health Service Foundation Trust, London, UK
- Institute of Ophthalmology, University College London, London, UK
| | - Brian L Yaspan
- Department of Human Genetics, Genentech, South San Francisco, CA, USA
| | | | - Pirro G Hysi
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- UCL Great Ormond Street Institute of Child Health, King's College London, London, UK
- Sørlandet Sykehus Arendal, Arendal Hospital, Arendal, Norway
| | - Andrew J Lotery
- Southampton Eye Unit, University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - J Michael Gaziano
- Million Veteran Program Coordinating Center, VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip S Tsao
- VA Palo Alto Epidemiology Research and Information Center for Genomics, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven J Fliesler
- Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
- Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
- Biochemistry, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
- Graduate Program in Neurosciences, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
| | - Jack M Sullivan
- Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
- Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
- Graduate Program in Neurosciences, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
| | - Paul B Greenberg
- Section of Ophthalmology, VA Providence Healthcare System, Providence, RI, USA
- Division of Ophthalmology, Alpert Medical School, Brown University, Providence, RI, USA
| | - Wen-Chih Wu
- Section of Cardiology, Medical Service, VA Providence Healthcare System, Providence, RI, USA
- Division of Cardiology, Department of Medicine, Alpert Medical School, Brown University, Providence, RI, USA
| | - Themistocles L Assimes
- VA Palo Alto Epidemiology Research and Information Center for Genomics, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Precision Medicine and Translational Therapeutics, VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters Veterans Affairs Medical Center, New York/New Jersey VA Health Care Network, Bronx, NY, USA.
| | - Neal S Peachey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA.
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| | - Sudha K Iyengar
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA.
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA.
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA.
- Department of Genetics & Genome Sciences, Case Western Reserve University, Cleveland, OH, USA.
- Department of Ophthalmology and Visual Sciences, University Hospitals Eye Institute, Cleveland, OH, USA.
| |
Collapse
|
2
|
Mazade R, Palumaa T, Pardue MT. Insights Into Myopia from Mouse Models. Annu Rev Vis Sci 2024; 10:10.1146/annurev-vision-102122-102059. [PMID: 38635876 PMCID: PMC11615738 DOI: 10.1146/annurev-vision-102122-102059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Animal models are critical for understanding the initiation and progression of myopia, a refractive condition that causes blurred distance vision. The prevalence of myopia is rapidly increasing worldwide, and myopia increases the risk of developing potentially blinding diseases. Current pharmacological, optical, and environmental interventions attenuate myopia progression in children, but it is still unclear how this occurs or how these interventions can be improved to increase their protective effects. To optimize myopia interventions, directed mechanistic studies are needed. The mouse model is well-suited to these studies because of its well-characterized visual system and the genetic experimental tools available, which can be combined with pharmacological and environmental manipulations for powerful investigations of causation. This review describes aspects of the mouse visual system that support its use as a myopia model and presents genetic, pharmacological, and environmental studies that significantly contribute to our understanding of the mechanisms that underlie myopigenesis.
Collapse
Affiliation(s)
- Reece Mazade
- 1Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, USA; , ,
| | - Teele Palumaa
- 1Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, USA; , ,
- 2Institute of Genomics, University of Tartu, Tartu, Estonia
- 3Eye Clinic, East Tallinn Central Hospital, Tallinn, Estonia
| | - Machelle T Pardue
- 1Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, USA; , ,
- 4Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Ahmad S, Wright KN, VonCannon JL, Ferrario CM, Ola MS, Choudhary M, Malek G, Gustafson JR, Sappington RM. Internalization of Angiotensin-(1-12) in Adult Retinal Pigment Epithelial-19 Cells. J Ocul Pharmacol Ther 2023; 39:290-299. [PMID: 36944130 PMCID: PMC10178934 DOI: 10.1089/jop.2022.0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/12/2023] [Indexed: 03/23/2023] Open
Abstract
Purpose: Angiotensin-(1-12) [Ang-(1-12)] serves as a primary substrate to generate angiotensin II (Ang II) by angiotensin-converting enzyme and/or chymase suggests it may be an unrecognized source of Ang II-mediated microvascular complication in hypertension-mediated retinopathy. We investigated Ang-(1-12) expression and internalization in adult retinal pigment epithelial-19 (ARPE-19) cultured cells. We performed the internalization of Ang-(1-12) in ARPE-19 cells in the presence of a highly specific monoclonal antibody (mAb) developed against the C-terminal end of the Ang-(1-12) sequence. Methods: All experiments were performed in confluent ARPE-19 cells (passage 28-35). We employed high-performance liquid chromatography to purify radiolabeled, 125I-Ang-(1-12) and immuno-neutralization with Ang-(1-12) mAb to demonstrate Ang-(1-12)'s internalization in ARPE-19 cells. Internalization was also demonstrated by immunofluorescence (IF) method. Results: These procedures revealed internalization of an intact 125I-Ang-(1-12) in ARPE-19 cells. A significant reduction (∼53%, P < 0.0001) in 125I-Ang-(1-12) internalization was detected in APRE-19 cells in the presence of the mAb. IF staining experiments further confirms internalization of Ang-(1-12) into the cells from the extracellular culture medium. No endogenous expression was detected in the ARPE-19 cells. An increased intensity of IF staining was detected in cells exposed to 1.0 μM Ang-(1-12) compared with 0.1 μM. Furthermore, we found hydrolysis of Ang-(1-12) into Ang II by ARPE-19 cells' plasma membranes. Conclusions: Intact Ang-(1-12) peptide is internalized from the extracellular spaces in ARPE-19 cells and metabolized into Ang II. The finding that a selective mAb blocks cellular internalization of Ang-(1-12) suggests alternate therapeutic approaches to prevent/reduce the RPE cells Ang II burden.
Collapse
Affiliation(s)
- Sarfaraz Ahmad
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Kendra N. Wright
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jessica L. VonCannon
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Carlos M. Ferrario
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Mohammad S. Ola
- Department of Biochemistry, King Saud University, Riyadh, Saudi Arabia
| | - Mayur Choudhary
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Goldis Malek
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jenna R. Gustafson
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Rebecca M. Sappington
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
4
|
Zeitz C, Roger JE, Audo I, Michiels C, Sánchez-Farías N, Varin J, Frederiksen H, Wilmet B, Callebert J, Gimenez ML, Bouzidi N, Blond F, Guilllonneau X, Fouquet S, Léveillard T, Smirnov V, Vincent A, Héon E, Sahel JA, Kloeckener-Gruissem B, Sennlaub F, Morgans CW, Duvoisin RM, Tkatchenko AV, Picaud S. Shedding light on myopia by studying complete congenital stationary night blindness. Prog Retin Eye Res 2023; 93:101155. [PMID: 36669906 DOI: 10.1016/j.preteyeres.2022.101155] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/20/2023]
Abstract
Myopia is the most common eye disorder, caused by heterogeneous genetic and environmental factors. Rare progressive and stationary inherited retinal disorders are often associated with high myopia. Genes implicated in myopia encode proteins involved in a variety of biological processes including eye morphogenesis, extracellular matrix organization, visual perception, circadian rhythms, and retinal signaling. Differentially expressed genes (DEGs) identified in animal models mimicking myopia are helpful in suggesting candidate genes implicated in human myopia. Complete congenital stationary night blindness (cCSNB) in humans and animal models represents an ON-bipolar cell signal transmission defect and is also associated with high myopia. Thus, it represents also an interesting model to identify myopia-related genes, as well as disease mechanisms. While the origin of night blindness is molecularly well established, further research is needed to elucidate the mechanisms of myopia development in subjects with cCSNB. Using whole transcriptome analysis on three different mouse models of cCSNB (in Gpr179-/-, Lrit3-/- and Grm6-/-), we identified novel actors of the retinal signaling cascade, which are also novel candidate genes for myopia. Meta-analysis of our transcriptomic data with published transcriptomic databases and genome-wide association studies from myopia cases led us to propose new biological/cellular processes/mechanisms potentially at the origin of myopia in cCSNB subjects. The results provide a foundation to guide the development of pharmacological myopia therapies.
Collapse
Affiliation(s)
- Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.
| | - Jérome E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, Saclay, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France
| | | | | | - Juliette Varin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, APHP, Paris, France
| | | | - Nassima Bouzidi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Frederic Blond
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Stéphane Fouquet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Vasily Smirnov
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Ajoy Vincent
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Elise Héon
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France; Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Robert M Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Andrei V Tkatchenko
- Oujiang Laboratory, Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health, Wenzhou, China; Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, New York, NY, USA
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
5
|
Yu P, Wang Y, Li Z, Jin H, Li LL, Han X, Wang ZW, Yang XL, Li XY, Zhang XJ, Zhou L, Gui JF. Causal gene identification and desirable trait recreation in goldfish. SCIENCE CHINA LIFE SCIENCES 2022; 65:2341-2353. [DOI: 10.1007/s11427-022-2194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022]
|
6
|
Megalin and Vitamin D Metabolism—Implications in Non-Renal Tissues and Kidney Disease. Nutrients 2022; 14:nu14183690. [PMID: 36145066 PMCID: PMC9506339 DOI: 10.3390/nu14183690] [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: 08/10/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Megalin is an endocytic receptor abundantly expressed in proximal tubular epithelial cells and other calciotropic extrarenal cells expressing vitamin D metabolizing enzymes, such as bone and parathyroid cells. The receptor functions in the uptake of the vitamin D-binding protein (DBP) complexed to 25 hydroxyvitamin D3 (25(OH)D3), facilitating the intracellular conversion of precursor 25(OH)D3 to the active 1,25 dihydroxyvitamin D3 (1,25(OH)2D3). The significance of renal megalin-mediated reabsorption of 25(OH)D3 and 1,25(OH)2D3 has been well established experimentally, and other studies have demonstrated relevant roles of extrarenal megalin in regulating vitamin D homeostasis in mammary cells, fat, muscle, bone, and mesenchymal stem cells. Parathyroid gland megalin may regulate calcium signaling, suggesting intriguing possibilities for megalin-mediated cross-talk between calcium and vitamin D regulation in the parathyroid; however, parathyroid megalin functionality has not been assessed in the context of vitamin D. Within various models of chronic kidney disease (CKD), megalin expression appears to be downregulated; however, contradictory results have been observed between human and rodent models. This review aims to provide an overview of the current knowledge of megalin function in the context of vitamin D metabolism, with an emphasis on extrarenal megalin, an area that clearly requires further investigation.
Collapse
|
7
|
Wang Y, Chen X, Jiang T, Gu Y, Zhang X, Yuan W, Zhao A, Li R, Wang Z, Hu Z, Liu H. Expanding the phenotypic spectrum of mutations in LRP2: a novel candidate gene of non-syndromic familial comitant strabismus. J Transl Med 2021; 19:495. [PMID: 34872573 PMCID: PMC8647414 DOI: 10.1186/s12967-021-03155-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022] Open
Abstract
Background Comitant strabismus (CS) is a heterogeneous disorder that is a major contributing factor to unilateral childhood-onset visual impairment. Studies have confirmed that genetic factors play an important role in the development of CS. The aim of this study was to identify the genetic cause of non-syndromic familial CS. Methods Fourteen unrelated CS families were recruited for the study. Twelve affected and 2 unaffected individuals from a large four-generation family (CS08) were selected to perform whole genome-wide linkage analysis. Parallel whole-exome sequencing (WES) was conducted in the same family (9 patients and 1 unaffected member) and 31 additional CS cases from 13 other unrelated families. Sanger sequencing was used to determine whether any of the remaining variants co-segregated with the disease phenotype in the corresponding family. Results Based on linkage analysis, CS in family CS08 mapped to a novel region of 34.17 centimorgan (cM) on chromosome 2q22.3-2q32.1 between markers D2S151 and D2S364, with a maximum log odds (LOD) score of 3.54 (theta = 0) at D2S142. Parallel WES identified a heterozygous variant, LRP2 c.335 A > G (p.Q112R), located in such a linkage interval that completely co-segregated with the disease in the family. Furthermore, another novel heterozygous variant (c.7274A > G, p.D2425G) in LRP2 that co-segregated was detected in 2 additional affected individuals from another unrelated family by WES. Both variants are predicted to be damaging by PolyPhen-2, SIFT and MutationTaster, and were absent in 100 ethnically matched normal controls. Conclusion LRP2 is a novel candidate genetic cause of non-syndromic familial CS. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03155-z.
Collapse
Affiliation(s)
- Yue Wang
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China.,Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China
| | - Xuejuan Chen
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China.,Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China
| | - Tao Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China
| | - Yayun Gu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China
| | - Xiaohan Zhang
- Department of Ophthalmology, Wuxi Children's Hospital, Wuxi, China
| | - Wenwen Yuan
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Andi Zhao
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Rui Li
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Zijin Wang
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China. .,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China.
| | - Hu Liu
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China.
| |
Collapse
|
8
|
Higham A, Hildebrand GD, Graham-Evans KAJ, Gilbert RD, Horton R, Hunt D, Shears D, Patel CK. Ectopic vortex veins and varices in Donnai Barrow syndrome. Ophthalmic Genet 2021; 43:248-252. [PMID: 34704885 DOI: 10.1080/13816810.2021.1992787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Donnai Barrow Syndrome (DBS) is a rare, multi-system autosomal recessively inherited disorder of relevance to ophthalmologists. To aim to describe the ocular phenotype using multimodal imaging for two cases of genetically confirmed DBS and compare against the published phenotype. MATERIALS AND METHODS Retrospective case series of two unrelated patients with DBS and review of the literature. Both cases were referred to our tertiary unit for laser prophylaxis against retinal detachment. RESULTS There was extreme high myopia greater than 20 dioptres without rhegmatogenous retinal detachment (RRD). Anterior segment features included iris transillumination and ciliary body hypoplasia. Posterior segment changes included previously described changes of optic nerve hypoplasia and a strikingly abnormal appearance of the fundus consisting of multiple bilateral giant posterior vortex veins (PVV). The mouse model shows a similar phenotype. CONCLUSIONS Ectopic vortex veins of the choroid expand the phenotype of DBS and can be helpful in distinguishing the differential diagnosis of high myopia in children. Posterior vortex veins have been described in adult high myopia as acquired but our cases suggest that they could be congenital. Orbital manipulation and hypotony during surgery should be avoided to minimise complications. The evidence to recommend prophylactic laser retinopexy in these cases is inconclusive, and overall we recommend that conservative management should be considered using wide-angle retinal imaging in the clinic.
Collapse
Affiliation(s)
- Aisling Higham
- Ophthalmology Department, John Radcliffe Hospital, Oxford University Hospitals, Oxford, UK
| | | | | | - Rodney D Gilbert
- Nephrology Department, Southampton Children's Hospital, Southampton, England.,Faculty of Medicine, University of Southampton, Southampton, UK
| | - Rachel Horton
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - David Hunt
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Deborah Shears
- Oxford Centre for Genomic Medicine, Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Chetan Kantibhai Patel
- Ophthalmology Department, John Radcliffe Hospital, Oxford University Hospitals, Oxford, UK.,Ophthalmology Department, Great Ormond Street Hospital for Children, London, England
| |
Collapse
|
9
|
Bryniarski MA, Zhao B, Chaves LD, Mikkelsen JH, Yee BM, Yacoub R, Shen S, Madsen M, Morris ME. Immunoglobulin G Is a Novel Substrate for the Endocytic Protein Megalin. AAPS JOURNAL 2021; 23:40. [PMID: 33677748 DOI: 10.1208/s12248-021-00557-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/07/2021] [Indexed: 11/30/2022]
Abstract
Therapeutic immunoglobulin G (IgG) antibodies comprise the largest class of protein therapeutics. Several factors that influence their overall disposition have been well-characterized, including target-mediated mechanics and convective flow. What remains poorly defined is the potential for non-targeted entry into various tissues or cell types by means of uptake via cell surface receptors at those sites. Megalin and cubilin are large endocytic receptors whose cooperative function plays important physiological roles at the tissues in which they are expressed. One such example is the kidney, where loss of either results in significant declines in proximal tubule protein reabsorption. Due to their diverse ligand profile and broad tissue expression, megalin and cubilin represent potential candidates for receptor-mediated uptake of IgG into various epithelia. Therefore, the objective of the current work was to determine if IgG was a novel ligand of megalin and/or cubilin. Direct binding was measured for human IgG with both megalin and the cubilin/amnionless complex. Additional work focusing on the megalin-IgG interaction was then conducted to build upon these findings. Cell uptake studies using megalin ligands for competitive inhibition or proximal tubule cells stably transduced with megalin-targeted shRNA constructs supported a role for megalin in the endocytosis of human IgG. Furthermore, a pharmacokinetic study using transgenic mice with a kidney-specific mosaic knockout of megalin demonstrated increased urinary excretion of human IgG in megalin knockout mice when compared to wild-type controls. These findings indicate that megalin is capable of binding and internalizing IgG via a high affinity interaction.
Collapse
Affiliation(s)
- Mark A Bryniarski
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 445 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Bei Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 445 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Lee D Chaves
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 445 Pharmacy Building, Buffalo, New York, 14214-8033, USA.,Department of Internal Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | | | - Benjamin M Yee
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 445 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Rabi Yacoub
- Department of Internal Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Shichen Shen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 445 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Mette Madsen
- Department of Biomedicine, Aarhus University, 8000, Aarhus C., Denmark
| | - Marilyn E Morris
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 445 Pharmacy Building, Buffalo, New York, 14214-8033, USA.
| |
Collapse
|
10
|
Christ A, Marczenke M, Willnow TE. LRP2 controls sonic hedgehog-dependent differentiation of cardiac progenitor cells during outflow tract formation. Hum Mol Genet 2020; 29:3183-3196. [PMID: 32901292 PMCID: PMC7689296 DOI: 10.1093/hmg/ddaa200] [Citation(s) in RCA: 13] [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: 06/30/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Conotruncal malformations are a major cause of congenital heart defects in newborn infants. Recently, genetic screens in humans and in mouse models have identified mutations in LRP2, a multi-ligand receptor, as a novel cause of a common arterial trunk, a severe form of outflow tract (OFT) defect. Yet, the underlying mechanism why the morphogen receptor LRP2 is essential for OFT development remained unexplained. Studying LRP2-deficient mouse models, we now show that LRP2 is expressed in the cardiac progenitor niche of the anterior second heart field (SHF) that contributes to the elongation of the OFT during separation into aorta and pulmonary trunk. Loss of LRP2 in mutant mice results in the depletion of a pool of sonic hedgehog-dependent progenitor cells in the anterior SHF due to premature differentiation into cardiomyocytes as they migrate into the OFT myocardium. Depletion of this cardiac progenitor cell pool results in aberrant shortening of the OFT, the likely cause of CAT formation in affected mice. Our findings identified the molecular mechanism whereby LRP2 controls the maintenance of progenitor cell fate in the anterior SHF essential for OFT separation, and why receptor dysfunction is a novel cause of conotruncal malformation.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Maike Marczenke
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| |
Collapse
|
11
|
Charlton JR, Tan W, Daouk G, Teot L, Rosen S, Bennett KM, Cwiek A, Nam S, Emma F, Jouret F, Oliveira JP, Tranebjærg L, Frykholm C, Mane S, Hildebrandt F, Srivastava T, Storm T, Christensen EI, Nielsen R. Beyond the tubule: pathological variants of LRP2, encoding the megalin receptor, result in glomerular loss and early progressive chronic kidney disease. Am J Physiol Renal Physiol 2020; 319:F988-F999. [PMID: 33103447 DOI: 10.1152/ajprenal.00295.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pathogenic variants in the LRP2 gene, encoding the multiligand receptor megalin, cause a rare autosomal recessive syndrome: Donnai-Barrow/Facio-Oculo-Acoustico-Renal (DB/FOAR) syndrome. Because of the rarity of the syndrome, the long-term consequences of the tubulopathy on human renal health have been difficult to ascertain, and the human clinical condition has hitherto been characterized as a benign tubular condition with asymptomatic low-molecular-weight proteinuria. We investigated renal function and morphology in a murine model of DB/FOAR syndrome and in patients with DB/FOAR. We analyzed glomerular filtration rate in mice by FITC-inulin clearance and clinically characterized six families, including nine patients with DB/FOAR and nine family members. Urine samples from patients were analyzed by Western blot analysis and biopsy materials were analyzed by histology. In the mouse model, we used histological methods to assess nephrogenesis and postnatal renal structure and contrast-enhanced magnetic resonance imaging to assess glomerular number. In megalin-deficient mice, we found a lower glomerular filtration rate and an increase in the abundance of injury markers, such as kidney injury molecule-1 and N-acetyl-β-d-glucosaminidase. Renal injury was validated in patients, who presented with increased urinary kidney injury molecule-1, classical markers of chronic kidney disease, and glomerular proteinuria early in life. Megalin-deficient mice had normal nephrogenesis, but they had 19% fewer nephrons in early adulthood and an increased fraction of nephrons with disconnected glomerulotubular junction. In conclusion, megalin dysfunction, as present in DB/FOAR syndrome, confers an increased risk of progression into chronic kidney disease.
Collapse
Affiliation(s)
- Jennifer R Charlton
- Division of Nephrology, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Weizhen Tan
- Division of Nephrology, Massachusetts General Hospital for Children, Boston, Massachusetts
| | - Ghaleb Daouk
- Division of Nephrology, Massachusetts General Hospital for Children, Boston, Massachusetts
| | - Lisa Teot
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Seymour Rosen
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts.,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Kevin M Bennett
- Department of Radiology, Washington University in Saint Louis, St. Louis, Missouri
| | - Aleksandra Cwiek
- Division of Nephrology, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Sejin Nam
- Department of Physics, University of Hawai'i at Manoa, Manoa, Hawai'i
| | - Francesco Emma
- Division of Nephrology, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital- Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - François Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Unit of Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - João Paulo Oliveira
- Service of Medical Genetics, São João University Hospital Centre and Faculty of Medicine, University of Porto and i3S-Institute for Health Research and Innovation, Porto, Portugal
| | - Lisbeth Tranebjærg
- Department of Clinical Genetics, Rigshospitalet/The Kennedy Centre, Copenhagen, Denmark.,Institute of Clinical Medicine, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| | - Carina Frykholm
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Shrikant Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Tina Storm
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Storm T, Burgoyne T, Futter CE. Membrane trafficking in the retinal pigment epithelium at a glance. J Cell Sci 2020; 133:133/16/jcs238279. [PMID: 32855284 DOI: 10.1242/jcs.238279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The retinal pigment epithelium (RPE) is a highly specialised pigmented monolayer sandwiched between the choroid and the photoreceptors in the retina. Key functions of the RPE include transport of nutrients to the neural retina, removal of waste products and water from the retina to the blood, recycling of retinal chromophores, absorption of scattered light and phagocytosis of the tips of the photoreceptor outer segments. These functions place a considerable membrane trafficking burden on the RPE. In this Cell Science at a Glance article and the accompanying poster, we focus on RPE-specific adaptations of trafficking pathways. We outline mechanisms underlying the polarised expression of membrane proteins, melanosome biogenesis and movement, and endocytic trafficking, as well as photoreceptor outer segment phagocytosis and degradation. We also briefly discuss theories of how dysfunction in trafficking pathways contributes to retinal disease.
Collapse
Affiliation(s)
- Tina Storm
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Thomas Burgoyne
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Clare E Futter
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| |
Collapse
|
13
|
Storm T, Burgoyne T, Dunaief JL, Christensen EI, Futter C, Nielsen R. Selective Ablation of Megalin in the Retinal Pigment Epithelium Results in Megaophthalmos, Macromelanosome Formation and Severe Retina Degeneration. Invest Ophthalmol Vis Sci 2019; 60:322-330. [PMID: 30665232 PMCID: PMC6343679 DOI: 10.1167/iovs.18-25667] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Mutations in the megalin-encoding gene, LRP2, cause high myopia as seen in patients suffering from Donnai-Barrow/facio-oculo-acoustico-renal syndrome. Megalin is present in both the nonpigmented epithelium of the ciliary body and in the RPE. In this study, we set out to establish an animal model to study the mechanisms underlying the ocular phenotype and to establish if high myopia/megaophthalmos is induced by postnatal megalin-deficiency in the RPE. Methods Postnatal RPE-specific deletion of megalin was generated by crossing mice bearing a homozygous loxP-flanked Lrp2 allele with transgenic mice expressing the Cre recombinase driven by the BEST1 promotor. The model was investigated by immunohistologic techniques, and transmission electron microscopy. Results Mice with postnatal RPE-specific loss of megalin developed a megaophthalmos phenotype with dramatic increase in ocular size and severe retinal thinning associated with compromised vision. This phenotype was present at postnatal day 14, indicating rapid development in the period from onset of BEST1 promotor activity at postnatal day 10. Additionally, RPE melanosomes exhibited abnormal size and morphology, suggested by electron tomography to be caused by fusion events between multiple melanosomes. Conclusions Postnatal loss of megalin in the RPE induces dramatic and rapid ocular growth and retinal degeneration compatible with the high myopia observed in Donnai-Barrow patients. The morphologic changes of RPE melanosomes, believed to be largely inert and fully differentiated at birth, suggested a continued plasticity of mature melanosomes and a requirement for megalin to maintain their number and morphology.
Collapse
Affiliation(s)
- Tina Storm
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | | | - Joshua L Dunaief
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Erik I Christensen
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - Clare Futter
- UCL Institute of Ophthalmology, London, United Kingdom
| | - Rikke Nielsen
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Abstract
Cell-to-cell communication is fundamental for embryo development and subsequent tissue homeostasis. This communication is often mediated by a small number of signaling pathways in which a secreted ligand binds to the surface of a target cell, thereby activating signal transduction. In vertebrate neural development, these signaling mechanisms are repeatedly used to obtain different and context-dependent outcomes. Part of the versatility of these communication mechanisms depends on their finely tuned regulation that controls timing, spatial localization, and duration of the signaling. The existence of secreted antagonists, which prevent ligand–receptor interaction, is an efficient mechanism to regulate some of these pathways. The Hedgehog family of signaling proteins, however, activates a pathway that is controlled largely by the positive or negative activity of membrane-bound proteins such as Cdon, Boc, Gas1, or Megalin/LRP2. In this review, we will use the development of the vertebrate retina, from its early specification to neurogenesis, to discuss whether there is an advantage to the use of such regulators, pointing to unresolved or controversial issues.
Collapse
Affiliation(s)
- Viviana Gallardo
- Centro de Biología Molecular , CSIC-UAM, Madrid, 28049, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular , CSIC-UAM, Madrid, 28049, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
| |
Collapse
|
15
|
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: 3.0] [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]
|
16
|
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: 313] [Impact Index Per Article: 44.7] [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
|
17
|
Storm T, Christensen EI, Christensen JN, Kjaergaard T, Uldbjerg N, Larsen A, Honoré B, Madsen M. Megalin Is Predominantly Observed in Vesicular Structures in First and Third Trimester Cytotrophoblasts of the Human Placenta. J Histochem Cytochem 2016; 64:769-784. [PMID: 27798286 DOI: 10.1369/0022155416672210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/30/2016] [Indexed: 12/31/2022] Open
Abstract
The membrane receptor megalin is crucial for normal fetal development. Besides its expression in the developing fetus, megalin is also expressed in the human placenta. Similar to its established function in the kidney proximal tubules, placental megalin has been proposed to mediate uptake of vital nutrients. However, details of megalin expression, subcellular localization, and function in the human placenta remain to be established. By immunohistochemical analyses of first trimester and term human placenta, we showed that megalin is predominantly expressed in cytotrophoblasts, the highly proliferative cells in placenta. Only limited amounts of megalin could be detected in syncytiotrophoblasts and least in term placenta syncytiotrophoblasts. Immunocytochemical analyses furthermore showed that placental megalin associates with structures of the endolysosomal apparatus. Combined, our results clearly place placental megalin in the context of endocytosis and trafficking of ligands. However, due to the limited expression of megalin in syncytiotrophoblasts, especially in term placenta, it appears that the main role for placental megalin is not to mediate uptake of nutrients from the maternal bloodstream, as previously proposed. In contrast, our results point toward novel and complex functions for megalin in the cytotrophoblasts. Thus, we propose that the perception of placental megalin localization and function should be revised.
Collapse
Affiliation(s)
- Tina Storm
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark (TS, EIC, JNC, TK, AL, BH, MM)
| | - Erik I Christensen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark (TS, EIC, JNC, TK, AL, BH, MM)
| | - Julie Nelly Christensen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark (TS, EIC, JNC, TK, AL, BH, MM)
| | - Tine Kjaergaard
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark (TS, EIC, JNC, TK, AL, BH, MM)
| | - Niels Uldbjerg
- Department of Clinical Medicine-Obstetrics and Gynaecology, Aarhus University Hospital, Skejby, Denmark (NU)
| | - Agnete Larsen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark (TS, EIC, JNC, TK, AL, BH, MM)
| | - Bent Honoré
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark (TS, EIC, JNC, TK, AL, BH, MM)
| | | |
Collapse
|
18
|
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: 3.4] [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
|
19
|
Lu X, Elizondo RA, Nielsen R, Christensen EI, Yang J, Hammock BD, Watsky MA. Vitamin D in Tear Fluid. Invest Ophthalmol Vis Sci 2015; 56:5880-7. [PMID: 26348637 DOI: 10.1167/iovs.15-17177] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine the source(s) of vitamin D in tear fluid and examine the expression of the endocytic proteins and putative vitamin D transporters megalin and cubilin in lacrimal and Harderian glands. METHODS Wild-type, heterozygous, and vitamin D receptor (VDR) knockout C57BL/6 mice were used, with a subset of knockout mice fed a replenishment diet for some studies. Mouse lacrimal and Harderian glands from each group were used to measure megalin and cubilin by RT-PCR, Western blot, and immunohistochemistry. New Zealand white rabbits were used to collect lacrimal and accessory gland fluid for vitamin D mass spectroscopy measurements. RESULTS Ten-week-old knockout mice were significantly (P < 0.05) smaller than wild-type mice. Real-time PCR and Western blot showed decreased expression of megalin and cubilin in select VDR knockout mouse groups. Immunohistochemistry showed apical duct cell megalin staining and weaker megalin staining in VDR knockout mice compared with controls. Vitamin D2 was more prevalent in rabbit lacrimal and accessory gland fluid than vitamin D3, and greater amounts of Vitamin D2 were found in in tear fluid obtained directly from lacrimal and accessory glands as compared with plasma concentrations. CONCLUSIONS This is the first study to demonstrate the presence of megalin and cubilin in lacrimal and accessory glands responsible for producing tear fluid. The results strengthen the hypothesis that megalin and cubilin are likely involved in the secretory pathway of vitamin D into tear fluid by the duct cells.
Collapse
Affiliation(s)
- Xiaowen Lu
- Department of Cell Biology and Anatomy Georgia Regents University, Augusta, Georgia, United States
| | - Rodolfo A Elizondo
- Department of Cell Biology and Anatomy Georgia Regents University, Augusta, Georgia, United States
| | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Jun Yang
- Department of Entomology and Nematology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, California, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, California, United States
| | - Mitchell A Watsky
- Department of Cell Biology and Anatomy Georgia Regents University, Augusta, Georgia, United States
| |
Collapse
|
20
|
Christ A, Christa A, Klippert J, Eule JC, Bachmann S, Wallace VA, Hammes A, Willnow TE. LRP2 Acts as SHH Clearance Receptor to Protect the Retinal Margin from Mitogenic Stimuli. Dev Cell 2015; 35:36-48. [PMID: 26439398 DOI: 10.1016/j.devcel.2015.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 08/21/2015] [Accepted: 09/07/2015] [Indexed: 01/17/2023]
Abstract
During forebrain development, LRP2 promotes morphogen signaling as an auxiliary SHH receptor. However, in the developing retina, LRP2 assumes the opposing function, mediating endocytic clearance of SHH and antagonizing morphogen action. LRP2-mediated clearance prevents spread of SHH activity from the central retina into the retinal margin to protect quiescent progenitor cells in this niche from mitogenic stimuli. Loss of LRP2 in mice increases the sensitivity of the retinal margin for SHH, causing expansion of the retinal progenitor cell pool and hyperproliferation of this tissue. Our findings document the ability of LRP2 to act, in a context-dependent manner, as activator or inhibitor of the SHH pathway. Our current findings uncovered LRP2 activity as the molecular mechanism imposing quiescence of the retinal margin in the mammalian eye and suggest SHH-induced proliferation of the retinal margin as cause of the large eye phenotype observed in mouse models and patients with LRP2 defects.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.
| | - Anna Christa
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Julia Klippert
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - J Corinna Eule
- Small Animal Clinic, Free University Berlin, 14163 Berlin, Germany
| | - Sebastian Bachmann
- Institute for Vegetative Anatomy, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Valerie A Wallace
- Toronto Western Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Annette Hammes
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.
| |
Collapse
|
21
|
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: 3.1] [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
|
22
|
In-depth phenotyping of a Donnai-Barrow patient helps clarify proximal tubule dysfunction. Pediatr Nephrol 2015; 30:1027-31. [PMID: 25822460 DOI: 10.1007/s00467-014-3037-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/11/2014] [Accepted: 12/16/2014] [Indexed: 10/23/2022]
Abstract
BACKGROUND The megalin/cubilin/amnionless complex is essential for albumin and low molecular weight (LMW) protein reabsorption by renal proximal tubules (PT). Mutations of the LRP2 gene encoding megalin cause autosomal recessive Donnai-Barrow/facio-oculo-acoustico-renal syndrome (DB/FOAR), which is characterized by LMW proteinuria. The pathophysiology of DB/FOAR-associated PT dysfunction remains unclear. CLINICAL CASE A 3-year-old girl presented with growth retardation and proteinuria. Clinical examination was unremarkable, except for a still-opened anterior fontanel and myopia. Psychomotor development was delayed. At 6, she developed sensorineural hearing loss. Hypertelorism was noted when she turned 12. Blood analyses, including renal function parameters, were normal. Urine sediment was bland. Proteinuria was significant and included albumin and LMW proteins. Immunoblotting analyses detected cubilin and type 3 carbonic anhydrase (CA3) in the urine. Renal ultrasound was unremarkable. Optical examination of a renal biopsy did not disclose any tubular or glomerular abnormality. Electron microscopy revealed that PT apical endocytic apparatus was significantly less developed. Immunostaining for megalin showed a faint signal in PT cytosol contrasting with the distribution of cubilin at the apical membrane. The diagnostic procedure led to identifying two mutations of the LRP2 gene. CONCLUSIONS The functional loss of megalin in DB/FOAR causes PT dysfunction characterized by increased urinary shedding of CA3 and cubilin.
Collapse
|
23
|
Andersen RK, Hammer K, Hager H, Christensen JN, Ludvigsen M, Honoré B, Thomsen MBH, Madsen M. Melanoma tumors frequently acquire LRP2/megalin expression, which modulates melanoma cell proliferation and survival rates. Pigment Cell Melanoma Res 2015; 28:267-80. [PMID: 25585665 DOI: 10.1111/pcmr.12352] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/12/2015] [Indexed: 12/13/2022]
Abstract
We show that the multiligand receptor megalin, known to mediate uptake and trafficking of nutrients and signaling molecules, is frequently expressed in malignant melanoma samples. Expression of megalin-encoding mRNA was investigated in 65 samples of nevi, melanomas, and melanoma metastases and was observed in more than 60% of the malignant samples, while only in 20% of the benign counterparts. Megalin expression in nevus and melanoma samples was additionally investigated by immunohistochemistry, which confirmed our mRNA-based observations. We furthermore show that a panel of tumor-derived melanoma cell lines express LRP2/megalin endogenously. In these cells, megalin is internalized from the cell surface and localizes extensively to intracellular vesicles, confirming receptor activity and pointing toward association with the endocytic apparatus. Groundbreaking, our results indicate that sustained megalin expression in melanoma cells is crucial for cell maintenance, as siRNA-mediated reduction in melanoma cell expression of LRP2/megalin significantly decreases melanoma cell proliferation and survival rates.
Collapse
Affiliation(s)
- Rikke K Andersen
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | | | | | | | | | | | | | | |
Collapse
|