1
|
Finch RH, Vitry G, Siew K, Walsh SB, Beheshti A, Hardiman G, da Silveira WA. Spaceflight causes strain-dependent gene expression changes in the kidneys of mice. NPJ Microgravity 2025; 11:11. [PMID: 40133368 PMCID: PMC11937539 DOI: 10.1038/s41526-025-00465-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Incidence of kidney stones in astronauts is a major risk factor associated with long-term missions, caused by increased blood calcium levels due to bone demineralisation triggered by microgravity and space radiation. Transcriptomic changes have been observed in tissues during spaceflight, including the kidney. We analysed kidney transcriptome patterns in two different strains of mice flown on the International Space Station, C57BL/6J and BALB/c. Here we show a link between spaceflight and transcriptome patterns associated with dysregulation of lipid and extracellular matrix metabolism and altered transforming growth factor-beta signalling. A stronger response was seen in C57BL/6J mice than BALB/c. Genetic differences in hyaluronan metabolism between strains may confer protection against extracellular matrix remodelling through the downregulation of epithelial-mesenchymal transition. We intend for our findings to contribute to the development of new countermeasures against kidney disease in astronauts and people here on Earth.
Collapse
Affiliation(s)
- Rebecca H Finch
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK
| | - Geraldine Vitry
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Department of Oncology, 3970 Reservoir Rd, NW, New Research Building EP11, Washington, DC, 20057, USA
| | - Keith Siew
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Stephen B Walsh
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Afshin Beheshti
- Center for Space Biomedicine, McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Gary Hardiman
- Faculty of Medicine, Health and Life Sciences, Institute for Global Food Security (IGFS), School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
- Department of Medicine, Medical University of South Carolina, MSC 403, 171 Ashley Ave Suite 419, Charleston, SC, 29425, USA
| | - Willian A da Silveira
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK.
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France.
- School of Science, Engineering and Environment. University of Salford, Manchester, M5 4WT, UK.
| |
Collapse
|
2
|
Song C, Wang K, Li L, Hu L, Bai J, Zhao L, Liu C, Li S. Analysis of candidate variants in a Chinese family with monozygotic twins with keratoconus: a case report. Ophthalmic Genet 2025; 46:31-39. [PMID: 39544142 DOI: 10.1080/13816810.2024.2427295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 04/30/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Keratoconus (KC) is an asymmetrical bilateral corneal ectasia, of which the pathogenesis is unknown. Moreover, genetic factors play an important role. We reported ophthalmic findings in a Chinese family with monozygotic twins with KC to describe the clinical features and identify genetic variants. METHODS Comprehensive ophthalmic clinical assessments and examinations, including history, slit-lamp biomicroscopy, best-corrected visual acuity, corneal topography, anterior segment optical coherence tomography, and corneal biomechanics, were carried out on the twins and their parents. Whole-genome sequencing (WGS) was performed to identify variants in this family. SIFT, PolyPhen2, MutationTaster, and CADD were used to predict the effect of amino acid substitutions on the affected protein. RESULTS The twins presented typical KC features. However, their mother did not meet the criteria for a KC diagnosis but exhibited KC subclinical manifestations. After screening, 12 potentially pathogenic variants in 10 genes were identified in both twins and emerged as candidate variants for this family. These genes included 1 previously reported KC-associated variant (ZNF469, c.4384 G>A); 8 variants in 6 KC-associated genes (GRHPR, c.337 G>A, c.862_863del; COL6A1, c.920 G>A; FLG, c.8753C>G; HSPG2, c.9503C>T; KRT82, c.1306 G>A; SCN9A, c.5702_5706del, c.641 G>A); and 3 variants in 3 non-KC-associated genes (PDE6G, c.6C>A; HAL, c.1724C>T; AGBL1, c.2381 G>A). CONCLUSIONS The accumulation of these potentially pathogenic variants in twins may have caused KC in these twins. These results expand the spectrum of KC candidate variants and provide a basis for further studies on KC.
Collapse
Affiliation(s)
- Chunyuan Song
- Aier Eye Hospital, Jinan University, Guangzhou, China
| | - Kehua Wang
- Department of Ophthalmology, Changsha Aier Eye Hospital, Changsha, China
| | - Ling Li
- Department of Ophthalmology, Beijing Aier-Intech Eye Hospital, Beijing, China
- Department of Ophthalmology, Aier Corneal Institute, Beijing, China
| | - Luping Hu
- Department of Ophthalmology, Beijing Aier-Intech Eye Hospital, Beijing, China
| | - Jie Bai
- Department of Ophthalmology, Beijing Aier-Intech Eye Hospital, Beijing, China
- Department of Ophthalmology, Aier Corneal Institute, Beijing, China
| | - Lin Zhao
- Department of Ophthalmology, Beijing Aier-Intech Eye Hospital, Beijing, China
- Department of Ophthalmology, Aier Corneal Institute, Beijing, China
| | - Chang Liu
- Department of Ophthalmology, Beijing Aier-Intech Eye Hospital, Beijing, China
- Department of Ophthalmology, Aier Corneal Institute, Beijing, China
| | - Shaowei Li
- Aier Eye Hospital, Jinan University, Guangzhou, China
- Department of Ophthalmology, Beijing Aier-Intech Eye Hospital, Beijing, China
- Department of Ophthalmology, Aier Corneal Institute, Beijing, China
- Aier Eye Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
3
|
Finch RH, Vitry G, Siew K, Walsh SB, Behesti A, Hardiman G, da Silveira WA. Spaceflight causes strain dependent gene expression changes associated with lipid and extracellular matrix dysregulation in the mouse kidney in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.584781. [PMID: 38559158 PMCID: PMC10979940 DOI: 10.1101/2024.03.13.584781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
To explore new worlds we must ensure humans can survive and thrive in the space environment. Incidence of kidney stones in astronauts is a major risk factor associated with long term missions, caused by increased blood calcium levels due to bone demineralisation triggered by microgravity and space radiation. Transcriptomic changes have been observed in other tissues during spaceflight, including the kidney. We analysed kidney transcriptome patterns in two different strains of mice flown on the International Space Station, C57BL/6J and BALB/c. Here we show a link between spaceflight and transcriptome patterns associated with dysregulation of lipid and extracellular matrix metabolism and altered transforming growth factor-beta signalling. A stronger response was seen in C57BL/6J mice than BALB/c. Genetic differences in hyaluronan metabolism between strains may confer protection against extracellular matrix remodelling through downregulation of epithelial-mesenchymal transition. We intend for our findings to contribute to development of new countermeasures against kidney disease in astronauts and people here on Earth.
Collapse
|
4
|
Huang L, Zou J, Zhang Y, Gu J, Wu J, Zhang C. Human umbilical cord mesenchymal stem cell therapy for renal dysfunction in Alport syndrome: protocol for an open-label, single-arm trial in China. BMJ Open 2024; 14:e075138. [PMID: 38490657 PMCID: PMC10946359 DOI: 10.1136/bmjopen-2023-075138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 02/01/2024] [Indexed: 03/17/2024] Open
Abstract
INTRODUCTION Alport syndrome (AS) is one of the most common fatal hereditary renal diseases in human, with a high risk of progressing to end-stage renal disease without effective treatments. Mesenchymal stem cells (MSCs) have recently emerged as a promising therapeutic strategy for chronic kidney disease. However, the safety and therapeutic potential of MSC transfusion for patients with AS are still need to be confirmed. Therefore, we have designed a clinical trial to evaluate the hypothesis that intravenous infusion of human umbilical cord-derived MSC (hUC-MSC) is safe, feasible, and well-tolerated in children with AS. METHODS AND ANALYSIS We report the protocol of the first prospective, open-label, single-arm clinical trial to evaluate the safety and preliminary efficacy of hUC-MSC transfusion in children with early-stage AS. Paediatric patients diagnosed with AS who have persistent albuminuria will be candidates for screening. Twelve eligible patients are planned to recruit and will receive hUC-MSC infusions under close safety monitoring, and complete the efficacy assessments at scheduled follow-up visits. The primary endpoints include the occurrence of adverse events to assess safety and the albuminuria level for efficacy evaluation. Secondary endpoint assessments are based on haematuria and glomerular filtration measurements. Each patient's efficacy endpoints will be evaluated against their baseline levels. Additionally, the underlying mechanism of hUC-MSC therapy will be explored through transcriptomic and proteomic analysis of blood and urine samples. ETHICS AND DISSEMINATION The protocol (V.1.0, date 17 January 2015) was approved by the institutional review board of the Affiliated Taihe Hospital of Hubei University of Medicine (ethical approval 03 March 2015). Written informed consent will be obtained from the patient and/or guardians before study specific process. In addition to publication in a peer-reviewed scientific journal, a lay summary of study will be available for participants and the public on the Chinese Organization for Rare Disorders website (http://www.cord.org.cn/). TRIAL REGISTRATION NUMBER ISRCTN62094626.
Collapse
Affiliation(s)
- Li Huang
- Department of Pharmacy, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Jun Zou
- Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | | | | | - Jianlong Wu
- Department of Pharmacy, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Che Zhang
- Department of Pharmacy, South China Hospital, Medical School, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
5
|
Zhu V, Huang T, Wang D, Colville D, Mack H, Savige J. Ocular manifestations of the genetic causes of focal and segmental glomerulosclerosis. Pediatr Nephrol 2024; 39:655-679. [PMID: 37578539 PMCID: PMC10817844 DOI: 10.1007/s00467-023-06073-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 08/15/2023]
Abstract
Genetic forms of focal and segmental glomerulosclerosis (FSGS) often have extra-renal manifestations. This study examined FSGS-associated genes from the Genomics England Renal proteinuria panel for reported and likely ocular features. Thirty-two of the 55 genes (58%) were associated with ocular abnormalities in human disease, and a further 12 (22%) were expressed in the retina or had an eye phenotype in mouse models. The commonest genes affected in congenital nephrotic syndrome (NPHS1, NPHS2, WT1, LAMB2, PAX2 but not PLCE1) may have ocular manifestations . Many genes affected in childhood-adolescent onset FSGS (NPHS1, NPHS2, WT1, LAMB2, SMARCAL1, NUP107 but not TRPC6 or PLCE1) have ocular features. The commonest genes affected in adult-onset FSGS (COL4A3-COL4A5, GLA ) have ocular abnormalities but not the other frequently affected genes (ACTN4, CD2AP, INF2, TRPC6). Common ocular associations of genetic FSGS include cataract, myopia, strabismus, ptosis and retinal atrophy. Mitochondrial forms of FSGS (MELAS, MIDD, Kearn's Sayre disease) are associated with retinal atrophy and inherited retinal degeneration. Some genetic kidney diseases (CAKUT, ciliopathies, tubulopathies) that result in secondary forms of FSGS also have ocular features. Ocular manifestations suggest a genetic basis for FSGS, often help identify the affected gene, and prompt genetic testing. In general, ocular abnormalities require early evaluation by an ophthalmologist, and sometimes, monitoring or treatment to improve vision or prevent visual loss from complications. In addition, the patient should be examined for other syndromic features and first degree family members assessed.
Collapse
Affiliation(s)
- Victor Zhu
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Tess Huang
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - David Wang
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Deb Colville
- Department of Surgery, Royal Victorian Eye and Ear Hospital, The University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Heather Mack
- Department of Surgery, Royal Victorian Eye and Ear Hospital, The University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Judy Savige
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia.
| |
Collapse
|
6
|
Majithia S, Chong CCY, Chee ML, Yu M, Soh ZD, Thakur S, Lavanya R, Rim TH, Nusinovici S, Koh V, Sabanayagam C, Cheng CY, Tham YC. Associations between Chronic Kidney Disease and Thinning of Neuroretinal Layers in Multiethnic Asian and White Populations. OPHTHALMOLOGY SCIENCE 2024; 4:100353. [PMID: 37869020 PMCID: PMC10587624 DOI: 10.1016/j.xops.2023.100353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/22/2023] [Accepted: 06/14/2023] [Indexed: 10/24/2023]
Abstract
Purpose To evaluate the relationships between chronic kidney disease (CKD) with retinal nerve fiber layer (RNFL) and ganglion cell-inner plexiform layer (GCIPL) thickness profiles of eyes in Asian and White populations. Design Cross-sectional analysis. Participants A total of 5066 Asian participants (1367 Malays, 1772 Indians, and 1927 Chinese) from the Singapore Epidemiology of Eye Diseases Study (SEED) were included, consisting of 9594 eyes for peripapillary RNFL analysis and 8661 eyes for GCIPL analysis. Additionally, 45 064 White participants (87 649 eyes) from the United Kingdom Biobank (UKBB) were included for both macular RNFL analysis and GCIPL analysis. Methods Nonglaucoma participants aged ≥ 40 years with complete data for estimated glomerular filtration rate (eGFR) were included from both SEED and UKBB. In SEED, peripapillary RNFL and GCIPL thickness were measured by Cirrus HD-OCT 4000. In UKBB, macular RNFL and GCIPL were measured by Topcon 3D-OCT 1000 Mark II. Chronic kidney disease was defined as eGFR < 60 ml/min/1.73 m2 in both data sets. To evaluate the associations between kidney function status with RNFL and GCIPL thickness profiles, multivariable linear regression with generalized estimating equation models were performed in SEED and UKBB data sets separately. Main Outcome Measures Average peripapillary and macular RNFL thickness and macular GCIPL thickness. Results In SEED, after adjusting for age, gender, ethnicity, systolic blood pressure, antihypertensive medication, diabetes, hyperlipidemia, body mass index, smoking status, and intraocular pressure, presence of CKD (β = -1.31; 95% confidence interval [CI], -2.37 to -0.26; P = 0.015) and reduced eGFR (per 10 ml/min/1.73 m2; β = -0.32; 95% CI, -0.50 to -0.13; P = 0.001) were associated with thinner average peripapillary RNFL. Presence of CKD (β = -1.63; 95% CI, -2.42 to -0.84) and reduced eGFR (per 10 ml/min/1.73 m2; β = -0.30; 95% CI, -0.44 to -0.16) were consistently associated with thinner GCIPL in SEED (all P < 0.001). In UKBB, after adjusting for the above-mentioned covariates (except ethnicity), reduced eGFR (per 10 ml/min/1.73 m2; β = -0.06; 95% CI, -0.10 to -0.01; P = 0.008) was associated with thinner macular RNFL and CKD (β = -0.62; 95% CI, -1.16 to -0.08; P = 0.024) was associated with thinner average GCIPL. Conclusion We consistently observed associations between CKD and thinning of RNFL and GCIPL across Asian and White populations' eyes. These findings further suggest that compromised kidney function is associated with RNFL and GCIPL thinning. Financial Disclosures The author(s) have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Shivani Majithia
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | | | - Miao Li Chee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Marco Yu
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
| | - Zhi Da Soh
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Sahil Thakur
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Raghavan Lavanya
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Tyler Hyungtaek Rim
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
| | - Simon Nusinovici
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Victor Koh
- Centre for Innovation & Precision Eye Health, Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Ophthalmology, National University Hospital, Singapore
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
- Centre for Innovation & Precision Eye Health, Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yih-Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
- Centre for Innovation & Precision Eye Health, Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
7
|
Evans W, Richardson-May J, Arora R. Multimodal Imaging in Unusual Alport Retinopathy. Cureus 2024; 16:e52768. [PMID: 38389645 PMCID: PMC10882256 DOI: 10.7759/cureus.52768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Alport syndrome, a rare genetic condition, can manifest various ocular abnormalities. This case report presents a unique instance of Alport syndrome where bilateral reduced visual acuity led to cataract surgery and subsequent central serous chorioretinopathy due to steroid treatment. By utilizing multiple imaging modalities, we aim to illustrate classical and atypical findings, addressing a literature gap and sharing our experience for educational purposes.
Collapse
Affiliation(s)
- William Evans
- Ophthalmology, Salisbury District Hospital, Salisbury, GBR
| | | | - Rashi Arora
- Ophthalmology, Salisbury District Hospital, Salisbury, GBR
| |
Collapse
|
8
|
Greferath U, Fletcher E, Savige J, Mack HG. Drusen and Other Retinal Findings in People With IgA Glomerulonephritis. Am J Ophthalmol 2024; 257:247-253. [PMID: 37757996 DOI: 10.1016/j.ajo.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE Retinal drusen have been described in people with IgA nephropathy. We examined the frequency of drusen in IgA nephropathy and compared their location and composition with those for drusen in age-related macular degeneration. DESIGN Immunohistological case series of eyes of patients with IgA nephropathy, and a comparison eye with age-related macular degeneration. METHODS Donor eyes from 4 individuals (3 male, 1 female, aged 40-80 years) with biopsy-proven IgA nephropathy and kidney failure were examined for the presence of drusen, and location and composition using antibodies for vitronectin, IgA, IgM, IgG, C3, and C1q. Results were compared with those for drusen in macular degeneration without IgA nephropathy. RESULTS All 4 donors had sparse, subretinal pigment epithelium drusen of 55-65 mm diameter that stained for vitronectin but not for IgA or complement. All donors had retinal capillaries and choriocapillaris staining for IgA. The youngest donor (female, 40) had rare deposits in the outer nuclear layer that stained for IgA, but not for vitronectin. The oldest donor (male, 82) had large cystlike spaces in the inner nuclear and plexiform layers, and smaller cysts in the outer nuclear layer, with no staining for IgA or complement. CONCLUSIONS Retinal drusen are uncommon in IgA nephropathy, even with kidney failure. Drusen in IgA nephropathy resemble drusen found in age-related macular degeneration. IgA-staining deposits in the outer nuclear layer were likely due to systemic deposition of IgA and complement activation. The nature of cystic spaces is unknown. Further analysis of the retinas of people with glomerulonephritis is recommended.
Collapse
Affiliation(s)
- Ursula Greferath
- University of Melbourne, Parkville, Victoria, Australia (U.G., E.F., J.S., H.G.M.)
| | - Erica Fletcher
- University of Melbourne, Parkville, Victoria, Australia (U.G., E.F., J.S., H.G.M.)
| | - Judy Savige
- University of Melbourne, Parkville, Victoria, Australia (U.G., E.F., J.S., H.G.M.)
| | - Heather G Mack
- University of Melbourne, Parkville, Victoria, Australia (U.G., E.F., J.S., H.G.M.).
| |
Collapse
|
9
|
Yang G, Mack H, Harraka P, Colville D, Savige J. Ocular manifestations of the genetic renal tubulopathies. Ophthalmic Genet 2023; 44:515-529. [PMID: 37702059 DOI: 10.1080/13816810.2023.2253901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/26/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND The genetic tubulopathies are rare and heterogenous disorders that are often difficult to identify. This study examined the tubulopathy-causing genes for ocular associations that suggested their genetic basis and, in some cases, the affected gene. METHODS Sixty-seven genes from the Genomics England renal tubulopathy panel were reviewed for ocular features, and for retinal expression in the Human Protein Atlas and an ocular phenotype in mouse models in the Mouse Genome Informatics database. The genes resulted in disease affecting the proximal tubules (n = 24); the thick ascending limb of the loop of Henle (n = 10); the distal convoluted tubule (n = 15); or the collecting duct (n = 18). RESULTS Twenty-five of the tubulopathy-associated genes (37%) had ocular features reported in human disease, 49 (73%) were expressed in the retina, although often at low levels, and 16 (24%) of the corresponding mouse models had an ocular phenotype. Ocular abnormalities were more common in genes affected in the proximal tubulopathies (17/24, 71%) than elsewhere (7/43, 16%). They included structural features (coloboma, microphthalmia); refractive errors (myopia, astigmatism); crystal deposition (in oxalosis, cystinosis) and sclerochoroidal calcification (in Bartter, Gitelman syndromes). Retinal atrophy was common in the mitochondrial-associated tubulopathies. Structural abnormalities and crystal deposition were present from childhood, but sclerochoroidal calcification typically occurred after middle age. CONCLUSIONS Ocular abnormalities are uncommon in the genetic tubulopathies but may be helpful in recognizing the underlying genetic disease. The retinal expression and mouse phenotype data suggest that further ocular associations may become apparent with additional reports. Early identification may be necessary to monitor and treat visual complications.
Collapse
Affiliation(s)
- GeFei Yang
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Royal Melbourne Hospital, Parkville, Australia
| | - Heather Mack
- Department of Surgery (Ophthalmology), The University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Philip Harraka
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Royal Melbourne Hospital, Parkville, Australia
| | - Deb Colville
- Department of Surgery (Ophthalmology), The University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Judy Savige
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
10
|
Jadon T, Sadda S, Singh G, Narayan P, Chhablani J, Venkatesh P. NOVEL RETINAL IMAGING ABNORMALITIES IN ALPORT SYNDROME. Retin Cases Brief Rep 2023; 17:499-503. [PMID: 37643031 DOI: 10.1097/icb.0000000000001265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
PURPOSE The purpose of this study was to report a novel observation during retinal screening of a child with Alport syndrome. METHODS This was a review of case record and imaging files. RESULTS Clinical examination of the retina and standard color fundus photography revealed no abnormality. However, distinct and identical wrinkling of the temporal macula (fingerprint sign) in both eyes was noted on Optos pseudocolor images of the retina. On optical coherence tomography, there were corresponding "saw-tooth" corrugations in the middle layers of the retina. En face images further highlighted the characteristic nature of this unusual observation. CONCLUSION Fingerprint sign in the retina, a heretofore undescribed feature, is reported in a child with biopsy confirmed Alport syndrome.
Collapse
Affiliation(s)
| | - Srinivas Sadda
- Doheny Eye Institute, Los Angeles, California
- Department of Ophthalmology, University of California-Los Angeles, Los Angeles, California
| | | | | | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
11
|
Tauqeer Z, O'Neil EC, Brucker AJ, Aleman TS. NPHP1 FULL DELETION CAUSES NEPHRONOPHTHISIS AND A CONE-ROD DYSTROPHY. Retin Cases Brief Rep 2023; 17:352-358. [PMID: 36913617 DOI: 10.1097/icb.0000000000001208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To describe in detail the structural and functional phenotypes of a patient with cone-rod dystrophy associated with a full deletion of the NPHP1 gene. METHODS A 30-year-old man with a history of end-stage renal disease presented with progressive vision loss in early adulthood prompting evaluation for retinal disease. Ophthalmic evaluation was performed including visual fields, electroretinography, spectral domain optical coherence tomography and short-wavelength and near-infrared fundus autofluorescence imaging. RESULTS The visual acuity was 20/60 in each eye. Fundus examination revealed a subtle bull's-eye maculopathy confirmed with fundus autofluorescence. Spectral domain optical coherence tomography demonstrated perifoveal loss of the outer retinal layers with structural preservation further peripherally. Static perimetry confirmed the loss of cone greater than rod sensitivities in a manner that colocalized to structural findings. Electroretinography revealed decreased cone- and rod-mediated responses. Genetic testing confirmed a homozygous whole-gene deletion of the NPHP1 gene. CONCLUSION NPHP1 -associated retinal degeneration may present as a cone-rod dystrophy in addition to the previously reported rod-predominant phenotypes and can notably be associated with systemic abnormalities, including renal disease. Our work further expands on the growing literature describing the retinal disease associated with systemic ciliopathies.
Collapse
Affiliation(s)
| | - Erin C O'Neil
- Scheie Eye Institute; and
- The Center for Advanced Retinal and Ocular Therapeutics, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Tomas S Aleman
- Scheie Eye Institute; and
- The Center for Advanced Retinal and Ocular Therapeutics, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Wang D, Trevillian P, May S, Diakumis P, Wang Y, Colville D, Bahlo M, Greferath U, Fletcher E, Young B, Mack HG, Savige J. KCTD1 and Scalp-Ear-Nipple ('Finlay-Marks') syndrome may be associated with myopia and Thin basement membrane nephropathy through an effect on the collagen IV α3 and α4 chains. Ophthalmic Genet 2023; 44:19-27. [PMID: 36579937 DOI: 10.1080/13816810.2022.2144900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Scalp-Ear-Nipple syndrome is caused by pathogenic KCTD1 variants and characterised by a scalp defect, prominent ears, and rudimentary breasts. We describe here further clinical associations in the eye and kidney. METHODS Fifteen affected members from two unrelated families with p.(Ala30Glu) or p.(Pro31Leu) in KCTD1 were examined for ocular and renal abnormalities. The relevant proteins were studied in the eye and kidney, and the mutation consequences determined from mouse knockout models. RESULTS Five males and 10 females with a median age of 40 years (range 1-70) with pathogenic variants p.(Ala30Glu) (n = 12) or p.(Pro31Leu) (n = 3) in KCTD1 were studied. Of the 6 who underwent detailed ophthalmic examination, 5 (83%) had low myopic astigmatism, the mean spherical equivalent of 10 eyes was 2.38D, and one (17%) had hypermetropic astigmatism. One female had a divergent strabismus.Five individuals had renal cysts (5/15, 33%), with renal biopsy in one demonstrating a thinned glomerular basement membrane identical to that seen in Thin basement membrane nephropathy (AD Alport syndrome).In the eye, KCTD1 and its downstream targets, TFAP2, and the collagen IV α3 and α4 chains localised to the cornea and near the retinal amacrine cells. In the kidney, all these proteins except TFAP2 were expressed in the podocytes and distal tubules. TFAP2B and COL4A4 knockout mice also had kidney cysts, and COL4A3 and COL4A4 knockout mice had myopia. CONCLUSION Individuals with a pathogenic KCTD1 variant may have low myopic astigmatism and represent a further rare genetic cause for a thinned glomerular basement membrane.
Collapse
Affiliation(s)
- Dongmao Wang
- Department of Medicine (Northern Health and Melbourne Health), University of Melbourne, Melbourne, Australia
| | - Paul Trevillian
- Department of Nephrology, John Hunter Hospital, Newcastle, Australia
| | - Stephen May
- Renal Unit, Tamworth Hospital, Tamworth, Australia
| | - Peter Diakumis
- Department of Bioinformatics, Walter and Eliza Hall Institute, Parkville, Australia
| | - Yanyan Wang
- Department of Medicine (Northern Health and Melbourne Health), University of Melbourne, Melbourne, Australia
| | - Deb Colville
- Department of Medicine (Northern Health and Melbourne Health), University of Melbourne, Melbourne, Australia
| | - Melanie Bahlo
- Department of Bioinformatics, Walter and Eliza Hall Institute, Parkville, Australia
| | - Una Greferath
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia
| | - Erica Fletcher
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia
| | - Barbara Young
- Department of Pathology John Hunter Hospital, Newcastle, Australia
| | - Heather G Mack
- Department of Ophthalmology, Royal Victorian Eye and Ear Hospital, University of Melbourne, East Melbourne, Australia
| | - Judy Savige
- Department of Medicine (Northern Health and Melbourne Health), University of Melbourne, Melbourne, Australia
| |
Collapse
|
13
|
Increased end-stage renal disease risk in age-related macular degeneration: a nationwide cohort study with 10-year follow-up. Sci Rep 2023; 13:183. [PMID: 36604459 PMCID: PMC9814881 DOI: 10.1038/s41598-022-26964-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
Common etiologies between age-related macular degeneration (AMD) and kidney disease advocate a close link between AMD and end-stage renal disease (ESRD). However, the risk of ESRD in people with AMD was not reported. Here, we investigated the association between AMD and the risk of ESRD by using a nationwide, population-based cohort data in Korea. 4,206,862 participants aged 50 years or older were categorized by presence of AMD and visual disability. Risk of ESRD was the primary outcome. Cox regression hazard model was used to examine the hazard ratios (HRs) with adjustment for potential confounders. Stratified analyses by age, sex, baseline kidney function, and cardiometabolic comorbidities were performed. During the mean 9.95 years of follow-up, there were 21,759 incident ESRD events (0.52%). AMD was associated with 33% increased risk of ESRD (adjusted HR [aHR] 1.33, 95% confidence interval [CI] 1.24-1.44), and the risk was even higher when accompanied by visual disability (aHR 2.05, 95% CI 1.68-2.50) than when not (aHR 1.26, 95% CI 1.17-1.37). Age, baseline kidney function, and cardiometabolic comorbidities significantly interact between AMD and the risk of ESRD. Our findings have clinical implications on disease prevention and risk factor management of ESRD in patients with AMD.
Collapse
|
14
|
GUNN's DOTS AS INDICATORS OF RENAL FUNCTION, FINDINGS FROM THE TONGREN HEALTH CARE STUDY. Retina 2022; 42:789-796. [PMID: 35350051 DOI: 10.1097/iae.0000000000003354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE To investigate the prevalence of Gunn's dots (GDs) and associated systemic factors in adult Chinese. METHODS A cross-sectional study enrolling participants older than 45 years from a community-based study. Gunn's dots were evaluated using fundus photography, and associated systemic factors were analyzed. Patients with any retinal or optic neuropathy were excluded. RESULTS The study included 4,118 participants (mean age: 58.3 ± 9.9 years; male: 1,699/41.3%). Gunn's dots were found in 931 participants, with a prevalence of 22.6 ± 0.8% (95% confidence interval [CI]: 21.3-23.9). Systemic factors associated with a higher GD prevalence were younger age (odds ratio [OR]: 0.92; 95% CI: 0.91-0.93; P < 0.001), higher estimated glomerular filtration rate (eGFR) (OR: 1.01; 95% CI: 1.001-1.02; P = 0.022), and higher serum concentration of triglycerides (OR: 1.08; 95% CI: 1.004-1.16; P = 0.040). The GD prevalence was 3.5 (OR = 3.46; 95% CI: 1.06-11.35) and 4.4 (OR = 4.37; 95% CI: 1.27-15.09) times greater for participants with an eGFR of ≥90 mL/minute/1.73 m2 and an eGFR of ≥100 mL/minute/1.73 m2, respectively, as compared with participants with an eGFR of <60 mL/minute/1.73 m2. CONCLUSION The GD prevalence (mean: 22.6%) was associated with younger age, higher eGFR, and higher serum triglyceride concentrations. The presence of GDs may serve as indicators of healthy renal function.
Collapse
|
15
|
Savige J, Lipska-Zietkiewicz BS, Watson E, Hertz JM, Deltas C, Mari F, Hilbert P, Plevova P, Byers P, Cerkauskaite A, Gregory M, Cerkauskiene R, Ljubanovic DG, Becherucci F, Errichiello C, Massella L, Aiello V, Lennon R, Hopkinson L, Koziell A, Lungu A, Rothe HM, Hoefele J, Zacchia M, Martic TN, Gupta A, van Eerde A, Gear S, Landini S, Palazzo V, al-Rabadi L, Claes K, Corveleyn A, Van Hoof E, van Geel M, Williams M, Ashton E, Belge H, Ars E, Bierzynska A, Gangemi C, Renieri A, Storey H, Flinter F. Guidelines for Genetic Testing and Management of Alport Syndrome. Clin J Am Soc Nephrol 2022; 17:143-154. [PMID: 34930753 PMCID: PMC8763160 DOI: 10.2215/cjn.04230321] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Genetic testing for pathogenic COL4A3-5 variants is usually undertaken to investigate the cause of persistent hematuria, especially with a family history of hematuria or kidney function impairment. Alport syndrome experts now advocate genetic testing for persistent hematuria, even when a heterozygous pathogenic COL4A3 or COL4A4 is suspected, and cascade testing of their first-degree family members because of their risk of impaired kidney function. The experts recommend too that COL4A3 or COL4A4 heterozygotes do not act as kidney donors. Testing for variants in the COL4A3-COL4A5 genes should also be performed for persistent proteinuria and steroid-resistant nephrotic syndrome due to suspected inherited FSGS and for familial IgA glomerulonephritis and kidney failure of unknown cause.
Collapse
Affiliation(s)
- Judy Savige
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Parkville, Victoria, Australia
| | | | - Elizabeth Watson
- South West Genetic Laboratory Hub, North Bristol Trust, Bristol, United Kingdom
| | - Jens Michael Hertz
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Constantinos Deltas
- Center of Excellence in Biobanking and Biomedical Research, University of Cyprus Medical School, Nicosia, Cyprus
| | - Francesca Mari
- Department of Medical Biotechnology, Medical Genetics, University of Siena, Siena, Italy
| | - Pascale Hilbert
- Departement de Biologie Moleculaire, Institute de Pathologie et de Genetique, Gosselies, Belgium
| | - Pavlina Plevova
- Department of Medical Genetics, University Hospital of Ostrava, Ostrava, Czech Republic
- Department of Biomedical Sciences, University Hospital of Ostrava, Ostrava, Czech Republic
| | - Peter Byers
- Department of Pathology, University of Washington, Seattle, Washington
- Department of Medicine (Medical Genetics), University of Washington, Seattle, Washington
| | - Agne Cerkauskaite
- Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Martin Gregory
- Division of Nephrology, Department of Medicine, University of Utah Health, Salt Lake City, Utah
| | - Rimante Cerkauskiene
- Clinic of Pediatrics, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Danica Galesic Ljubanovic
- Department of Pathology, University of Zagreb, School of Medicine, Dubrava University Hospital, Zagreb, Croatia
| | | | | | - Laura Massella
- Division of Nephrology and Dialysis, Bambino Gesù Children's Hospital, Rome, Italy
| | - Valeria Aiello
- Department of Experimental Diagnostic and Specialty Medicine, Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Louise Hopkinson
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Ania Koziell
- School of Immunology and Microbial Sciences, Faculty of Life Sciences, King's College London, London, United Kingdom
| | - Adrian Lungu
- Pediatric Nephrology Department, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Julia Hoefele
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
| | | | | | - Asheeta Gupta
- Birmingham Children’s Hospital, Birmingham, United Kingdom
| | | | | | - Samuela Landini
- Medical Genetics Unit, Department of Clinical and Experimental Biomedical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Viviana Palazzo
- Medical Genetics Unit, Meyer Children's University Hospital, Florence, Italy
| | - Laith al-Rabadi
- Health Sciences Centre, University of Utah, Salt Lake City, Utah
| | - Kathleen Claes
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Anniek Corveleyn
- Center for Human Genetics, University Hospitals and Katholieke Universiteit Leuven, Leuven, Belgium
| | - Evelien Van Hoof
- Center for Human Genetics, University Hospitals and Katholieke Universiteit Leuven, Leuven, Belgium
| | - Micheel van Geel
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Maggie Williams
- Bristol Genetics Laboratory Pathology Sciences, Southmead Hospital, Southmead, United Kingdom
| | - Emma Ashton
- North East Thames Regional Genetics Laboratory, Great Ormond Street Hospital, London, United Kingdom
| | - Hendica Belge
- Institut de Pathologie et de Génétique, Center for Human Genetics, Gosselies, Belgium
| | - Elisabet Ars
- Molecular Biology Laboratory, Fundacio Puigvert, Instituto de Investigaciones Biomédicas Sant Pau, Universitat Autonoma de Barcelona, Instituto de Investigación Carlos III, Barcelona, Spain
| | - Agnieszka Bierzynska
- Bristol Renal Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Concetta Gangemi
- Division of Nephrology and Dialysis, University Hospital of Verona, Verona, Italy
| | - Alessandra Renieri
- Department of Medical Biotechnology, Medical Genetics, University of Siena, Siena, Italy
| | - Helen Storey
- Molecular Genetics, Viapath Laboratories, Guy’s Hospital, London, United Kingdom
| | - Frances Flinter
- Department of Clinical Genetics, Guy’s and St. Thomas’ National Health Service Foundation Trust, London, United Kingdom
| |
Collapse
|
16
|
Mack HG, Colville DJ, Harraka P, Savige JA, Invernizzi A, Fraser-Bell S. Retinal findings in glomerulonephritis. Clin Exp Optom 2021; 105:474-486. [PMID: 34877922 DOI: 10.1080/08164622.2021.2003691] [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: 10/19/2022] Open
Abstract
The complement system is part of the innate immune system activated by three distinct pathways: classical, lectin and alternative. It is also involved in retinal development and homoeostasis. Dense deposit disease is a rare renal disease associated with mutations in Complement factor H and overactivity of the alternative complement pathway. As well as glomerulonephritis, many affected individuals have retinal drusen and may be at risk of vision loss due to macular atrophy or choroidal neovascularisation. We discuss the reclassification of dense deposit disease as a type of C3 glomerulonephropathy, and hypothesise on the mechanisms of retinal abnormalities. Drusen have also been described in individuals with other types of glomerulonephritis involving abnormalities of the classical (membranoproliferative glomerulonephritis type 1) or lectin (IgA nephropathy, lupus nephritis) complement pathways. Although drusen are found in abnormalities of all three complement pathways, the age at onset, aetiology, and the threat to vision differs. This review describes drusen and other retinal abnormalities associated with the glomerulonephritides due to abnormal activation in each of the three complement activation pathways, and provides the first report of drusen occurring in a patient with the recently reclassified C3 glomerulonephritis with homozygous variant V62I in complement factor H. Optometric management of young patients presenting with retinal drusen is discussed, and complement-based therapies for visual loss are reviewed.
Collapse
Affiliation(s)
- Heather G Mack
- Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, Australia.,Department of Ophthalmology, Melbourne Health, Melbourne, Australia.,Centre for Eye Research, University of Melbourne, Melbourne, Australia
| | - Deborah J Colville
- Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, Australia.,Department of Ophthalmology, Melbourne Health, Melbourne, Australia
| | - Phillip Harraka
- Department of Medicine (Northern), University of Melbourne, Melbourne, Australia
| | - Judith Anne Savige
- Department of Medicine (Northern), University of Melbourne, Melbourne, Australia
| | - Alessandro Invernizzi
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy
| | | |
Collapse
|
17
|
Yahalom C, Volovelsky O, Macarov M, Altalbishi A, Alsweiti Y, Schneider N, Hanany M, Khan MI, Cremers FPM, Anteby I, Banin E, Sharon D, Khateb S. SENIOR-LØKEN SYNDROME: A Case Series and Review of the Renoretinal Phenotype and Advances of Molecular Diagnosis. Retina 2021; 41:2179-2187. [PMID: 33512896 DOI: 10.1097/iae.0000000000003138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE To report genetic and clinical findings in a case series of 10 patients from eight unrelated families diagnosed with Senior-Løken syndrome. METHODS A retrospective study of patients with Senior-Løken syndrome. Data collected included clinical findings electroretinography and ocular imaging. Genetic analysis was based on molecular inversion probes, whole-exome sequencing (WES), and Sanger sequencing. RESULTS All patients who underwent electrophysiology (8/10) had widespread photoreceptor degeneration. Genetic analysis revealed two mutations in NPHP1, two mutations in NPHP4, and two mutations in IQCB1 (NPHP5). Five of the six mutations identified in the current study were found in a single family each in our cohort. The IQCB1-p.R461* mutation has been identified in 3 families. Patients harboring mutations in IQCB1 were diagnosed with Leber congenital amaurosis, while patients with NPHP4 and NPHP1 mutations showed early and sector retinitis pigmentosa, respectively. Full-field electroretinography was extinct for 6 of 10 patients, moderately decreased for two, and unavailable for another 2 subjects. Renal involvement was evident in 7/10 patients at the time of diagnosis. Kidney function was normal (based on serum creatinine) in patients younger than 10 years. Mutations in IQCB1 were associated with high hypermetropia, whereas mutations in NPHP4 were associated with high myopia. CONCLUSION Patients presenting with infantile inherited retinal degeneration are not universally screened for renal dysfunction. Modern genetic tests can provide molecular diagnosis at an early age and therefore facilitate early diagnosis of renal disease with recommended periodic screening beyond childhood and family planning.
Collapse
Affiliation(s)
- Claudia Yahalom
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oded Volovelsky
- Pediatric Nephrology Unit, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Macarov
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alaa Altalbishi
- St John of Jerusalem Eye Hospital Group, East Jerusalem, Israel
| | - Yahya Alsweiti
- St John of Jerusalem Eye Hospital Group, East Jerusalem, Israel
| | - Nina Schneider
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mor Hanany
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Muhammad Imran Khan
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands ; and
| | - Frans P M Cremers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands ; and
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Irene Anteby
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Banin
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dror Sharon
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samer Khateb
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
18
|
Currant H, Hysi P, Fitzgerald TW, Gharahkhani P, Bonnemaijer PWM, Senabouth A, Hewitt AW, Atan D, Aung T, Charng J, Choquet H, Craig J, Khaw PT, Klaver CCW, Kubo M, Ong JS, Pasquale LR, Reisman CA, Daniszewski M, Powell JE, Pébay A, Simcoe MJ, Thiadens AAHJ, van Duijn CM, Yazar S, Jorgenson E, MacGregor S, Hammond CJ, Mackey DA, Wiggs JL, Foster PJ, Patel PJ, Birney E, Khawaja AP. Genetic variation affects morphological retinal phenotypes extracted from UK Biobank optical coherence tomography images. PLoS Genet 2021; 17:e1009497. [PMID: 33979322 PMCID: PMC8143408 DOI: 10.1371/journal.pgen.1009497] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 05/24/2021] [Accepted: 03/18/2021] [Indexed: 12/15/2022] Open
Abstract
Optical Coherence Tomography (OCT) enables non-invasive imaging of the retina and is used to diagnose and manage ophthalmic diseases including glaucoma. We present the first large-scale genome-wide association study of inner retinal morphology using phenotypes derived from OCT images of 31,434 UK Biobank participants. We identify 46 loci associated with thickness of the retinal nerve fibre layer or ganglion cell inner plexiform layer. Only one of these loci has been associated with glaucoma, and despite its clear role as a biomarker for the disease, Mendelian randomisation does not support inner retinal thickness being on the same genetic causal pathway as glaucoma. We extracted overall retinal thickness at the fovea, representative of foveal hypoplasia, with which three of the 46 SNPs were associated. We additionally associate these three loci with visual acuity. In contrast to the Mendelian causes of severe foveal hypoplasia, our results suggest a spectrum of foveal hypoplasia, in part genetically determined, with consequences on visual function.
Collapse
Affiliation(s)
- Hannah Currant
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Pirro Hysi
- School of Life Course Sciences, Section of Ophthalmology, King’s College London, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Tomas W. Fitzgerald
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Puya Gharahkhani
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Pieter W. M. Bonnemaijer
- Department of Ophthalmology, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- The Rotterdam Eye Hospital, Rotterdam, The Netherlands
| | - Anne Senabouth
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, Australia
| | - Alex W. Hewitt
- Menzies Institute for Medical Research, School of Medicine, University of Tasmania, Tasmania, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | | | | | - Denize Atan
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Bristol Eye Hospital, University Hospitals Bristol & Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jason Charng
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, The University of Western Australia, Perth, Australia
| | - Hélène Choquet
- Kaiser Permanente Northern California Division of Research, Oakland, California, United States of America
| | - Jamie Craig
- Department of Ophthalmology, Flinders University, Flinders Medical Centre, Bedford Park, Australia
| | - Peng T. Khaw
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Ophthalmology Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Jue-Sheng Ong
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Louis R. Pasquale
- Eye and Vision Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Charles A. Reisman
- Topcon Healthcare Solutions R&D, Oakland, New Jersey, United States of America
| | - Maciej Daniszewski
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Australia
| | - Joseph E. Powell
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, Australia
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, Australia
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Mark J. Simcoe
- Department of Ophthalmology, Kings College London, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
| | | | - Cornelia M. van Duijn
- Nuffield Department Of Population Health, University of Oxford, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Oxford, United Kingdom
| | - Seyhan Yazar
- Garvan-Weizmann Centre for Single Cell Genomics, Garvan Institute of Medical Research, Sydney, Australia
| | - Eric Jorgenson
- Kaiser Permanente Northern California Division of Research, Oakland, California, United States of America
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Chris J. Hammond
- School of Life Course Sciences, Section of Ophthalmology, King’s College London, London, United Kingdom
| | - David A. Mackey
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, The University of Western Australia, Perth, Australia
| | - Janey L. Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Paul J. Foster
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Praveen J. Patel
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Ewan Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Anthony P. Khawaja
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| |
Collapse
|
19
|
Lin J, Hu J, Schlotterer A, Wang J, Kolibabka M, Awwad K, Dietrich N, Breitschopf K, Wohlfart P, Kannt A, Lorenz K, Feng Y, Popp R, Hoffmann S, Fleming I, Hammes HP. Protective effect of Soluble Epoxide Hydrolase Inhibition in Retinal Vasculopathy associated with Polycystic Kidney Disease. Am J Cancer Res 2020; 10:7857-7871. [PMID: 32685025 PMCID: PMC7359083 DOI: 10.7150/thno.43154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: Vasoregression secondary to glial activation develops in various retinal diseases, including retinal degeneration and diabetic retinopathy. Photoreceptor degeneration and subsequent retinal vasoregression, characterized by pericyte loss and acellular capillary formation in the absence diabetes, are also seen in transgenic rats expressing the polycystic kidney disease (PKD) gene. Activated Müller glia contributes to retinal vasodegeneration, at least in part via the expression of the soluble epoxide hydrolase (sEH). Given that an increase in sEH expression triggered vascular destabilization in diabetes, and that vasoregression is similar in diabetic mice and PKD rats, the aim of the present study was to determine whether sEH inhibition could prevent retinal vasoregression in the PKD rat. Methods: One-month old male homozygous transgenic PKD rats were randomly allocated to receive vehicle or a sEH inhibitor (sEH-I; Sar5399, 30 mg/kg) for four weeks. Wild-type Sprague-Dawley (SD) littermates received vehicle as controls. Retinal sEH expression and activity were measured by Western blotting and LC-MS, and vasoregression was quantified in retinal digestion preparations. Microglial activation and immune response cytokines were assessed by immunofluorescence and quantitative PCR, respectively. 19,20-dihydroxydocosapentaenoic acid (19,20-DHDP) mediated Notch signaling, microglial activation and migration were assessed in vivo and in vitro. Results: This study demonstrates that sEH expression and activity were increased in PKD retinae, which led to elevated production of 19,20-DHDP and the depression of Notch signaling. The latter changes elicited pericyte loss and the recruitment of CD11b+/CD74+ microglia to the perivascular region. Microglial activation increased the expression of immune-response cytokines, and reduced levels of Notch3 and delta-like ligand 4 (Dll4). Treatment with Sar5399 decreased 19,20-DHDP generation and increased Notch3 expression. Sar5399 also prevented vasoregression by reducing pericyte loss and suppressed microglial activation as well as the expression of immune-response cytokines. Mechanistically, the activation of Notch signaling by Dll4 maintained a quiescent microglial cell phenotype, i.e. reduced both the surface presentation of CD74 and microglial migration. In contrast, in retinal explants, 19,20-DHDP and Notch inhibition both promoted CD74 expression and reversed the Dll4-induced decrease in migration. Conclusions: Our data indicate that 19,20-DHDP-induced alterations in Notch-signaling result in microglia activation and pericyte loss and contribute to retinal vasoregression in polycystic kidney disease. Moreover, sEH inhibition can ameliorate vasoregression through reduced activity of inflammatory microglia. sEH inhibition is thus an attractive new therapeutic approach to prevent retinal vasoregression.
Collapse
|
20
|
Ayine P, Selvaraju V, Venkatapoorna CMK, Geetha T. Parental Feeding Practices in Relation to Maternal Education and Childhood Obesity. Nutrients 2020; 12:nu12041033. [PMID: 32283764 PMCID: PMC7231200 DOI: 10.3390/nu12041033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 11/24/2022] Open
Abstract
Parental beliefs, attitudes, and feeding practices play a vital role in childhood obesity. This study aimed to assess parental perceptions, concerns about weight, feeding practices using the Child Feeding Questionnaire (CFQ), and its association with body mass index (BMI) and maternal education in elementary school children. Participants aged 6–10 years (n = 169) were recruited and anthropometric measurements were obtained. Pearson’s correlation and hierarchical linear regression analysis were used to examine the association between BMI z-score and the seven factors of the CFQ. The BMI z-score was significantly associated with parental perceived child weight and concern about child weight. The BMI z-score had a significant negative association with parents pressuring children to eat. Parents of obese children reported significantly higher (p < 0.001) levels of perceived child weight (β = 0.312) and concern (β = 0.320) about their child’s weight compared to the normal weight and overweight groups. Parents of overweight children showed considerably less (β = −0.224; p < 0.005) pressuring towards their children to eat as compared to parents of normal weight children. Additionally, we found that the parental feeding practice (pressure to eat) was only dependent upon maternal education. The path analysis indicates that maternal education has a mediating effect on BMI z-score and pressure to eat is related to BMI z-score through maternal education. The findings demonstrate the association between the parents’ perceptions, concerns, and pressure to eat with BMI z-score of elementary school-aged children. Only the parental feeding practice pressure to eat was dependent upon the maternal education.
Collapse
Affiliation(s)
- Priscilla Ayine
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (P.A.); (V.S.); (C.M.K.V.)
| | - Vaithinathan Selvaraju
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (P.A.); (V.S.); (C.M.K.V.)
| | - Chandra M. K. Venkatapoorna
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (P.A.); (V.S.); (C.M.K.V.)
| | - Thangiah Geetha
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (P.A.); (V.S.); (C.M.K.V.)
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
- Correspondence: ; Tel.: +1-334-844-7418
| |
Collapse
|
21
|
Tham YC, Chee ML, Dai W, Lim ZW, Majithia S, Siantar R, Thakur S, Rim T, Cheung CY, Sabanayagam C, Aung T, Wong TY, Cheng CY. Profiles of Ganglion Cell-Inner Plexiform Layer Thickness in a Multi-Ethnic Asian Population: The Singapore Epidemiology of Eye Diseases Study. Ophthalmology 2020; 127:1064-1076. [PMID: 32197910 DOI: 10.1016/j.ophtha.2020.01.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/31/2019] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
PURPOSE To examine the normative profile and determinants of macular ganglion cell-inner plexiform layer (GCIPL) thickness based on spectral-domain OCT (SD-OCT) in a nonglaucoma, multi-ethnic Asian population. DESIGN Population-based, cross-sectional study. PARTICIPANTS Ethnic Chinese, Malay, and Indian adults aged ≥40 years recruited from the Singapore Epidemiology of Eye Diseases Study. METHODS All participants underwent standardized examinations. The GCIPL thickness was measured using Cirrus HD-OCT (Carl Zeiss Meditec, Dublin, CA). Participants with glaucoma or poor-quality scans were excluded. Eye-specific data were used. Associations of ocular and systemic factors with GCIPL thickness parameters were investigated using multivariable linear regression with generalized estimating equation models to account for correlation between both eyes. MAIN OUTCOME MEASURES GCIPL thickness. RESULTS A total of 4464 participants (7520 eyes) consisting of 1625 Chinese, 1212 Malay, and 1627 Indian adults contributed to this analysis. Average GCIPL thickness was 82.6±6.1 μm in Chinese, 81.5±6.8 μm in Malays, and 78.0±6.9 μm in Indians (P < 0.001 by analysis of variance). The 5th percentile limit of average GCIPL thickness was 72 μm in Chinese, 70 μm in Malays, and 67 μm in Indians. In multivariable analysis adjusting for age, gender, axial length, presence of cataract, OCT signal strength, disc area, hypertension, diabetes, and hyperlipidemia, eyes of Indians were observed to have 3.43 μm thinner GCIPL on average compared with Chinese (P < 0.001) and 3.36 μm thinner GCIPL compared with Malays (P < 0.001). In addition, older age (per decade; β = -2.51), female (β = -1.57), longer axial length (per mm; β = -1.54), and presence of chronic kidney disease (β = -1.49) were significantly associated with thinner average GCIPL (all P ≤ 0.008). Larger optic disc area (per mm2; β = 0.78; P < 0.001) was associated with thicker GCIPL. These factors were consistently observed to be significant for superior and inferior hemisphere GCIPL thickness. CONCLUSIONS GCIPL thickness profiles were significantly thinner in Indians compared with Chinese and Malays. Our findings further highlight the need of a more refined, ethnic-specific normative database for GCIPL thickness, which in turn may improve the detection and diagnosis of glaucoma in Asians.
Collapse
Affiliation(s)
- Yih-Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
| | - Miao Li Chee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Wei Dai
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Zhi Wei Lim
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Shivani Majithia
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | | | - Sahil Thakur
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Tyler Rim
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
| | - Carol Y Cheung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
22
|
Gupta V, Jamil M, Luthra S, Puthalath AS. Alport syndrome with bilateral simultaneous anterior and posterior lenticonus with severe temporal macular thinning. BMJ Case Rep 2019; 12:e229554. [PMID: 31420426 PMCID: PMC6700549 DOI: 10.1136/bcr-2019-229554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2019] [Indexed: 11/04/2022] Open
Abstract
Alport syndrome (AS) is a hereditary disease with various modes of inheritance, X-linked being the the most common. Anterior lenticonus is the characteristic abnormality along with perimacular and peripheral fleck retinopathy. Our two cases of AS had simultaneous anterior and posterior lenticonus with severe temporal macular thinning on optical coherence tomography with no specific renal symptomatology and were diagnosed as AS without any invasive renal biopsy. First patient was a 19-year-old man who presented with compound myopia due to bilateral anterior and posterior lenticonus with perimacular fleck retinopathy and lozenge sign and bilateral moderate sensorineural hearing loss (SNHL). Second patient was a 24-year-old man who presented with difficulty in vision due to bilateral anterior and posterior lenticonus with bilateral severe SNHL. Our cases emphasise the crucial role of an ophthalmologist in diagnosing AS before the onset of renal symptoms and prompting further nephrological work-up in the patient or the carrier.
Collapse
Affiliation(s)
- Vinita Gupta
- Ophthalmology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Mahsa Jamil
- Ophthalmology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Saurabh Luthra
- Ophthalmology, Drishti Eye Institute, Dehradun, Uttarakhand, India
| | - Athul S Puthalath
- Ophthalmology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| |
Collapse
|
23
|
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are worldwide public health problems affecting millions of people and have rapidly increased in prevalence in recent years. Due to the multiple causes of renal failure, many animal models have been developed to advance our understanding of human nephropathy. Among these experimental models, rodents have been extensively used to enable mechanistic understanding of kidney disease induction and progression, as well as to identify potential targets for therapy. In this review, we discuss AKI models induced by surgical operation and drugs or toxins, as well as a variety of CKD models (mainly genetically modified mouse models). Results from recent and ongoing clinical trials and conceptual advances derived from animal models are also explored.
Collapse
Affiliation(s)
- Yin-Wu Bao
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China. .,Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Yuan Yuan
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China. .,Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Jiang-Hua Chen
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China.
| | - Wei-Qiang Lin
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China. .,Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China
| |
Collapse
|
24
|
Midterm Outcome of Kidney Transplantation From Donors With Thin Basement Membrane Nephropathy. Transplantation 2018; 102:e180-e184. [DOI: 10.1097/tp.0000000000002089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
25
|
Chen Y, Colville D, Ierino F, Symons A, Savige J. Temporal retinal thinning and the diagnosis of Alport syndrome and Thin basement membrane nephropathy. Ophthalmic Genet 2017; 39:208-214. [PMID: 29172845 DOI: 10.1080/13816810.2017.1401088] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Alport syndrome is an inherited disease characterized by renal failure, hearing loss, and ocular abnormalities, including temporal retinal thinning. This study compared retinal thinning in Alport syndrome and other renal diseases. METHODS Alport syndrome was diagnosed on renal biopsy and genetic testing. Subjects underwent optical coherence tomography (OCT) (Spectralis OCT, Heidelberg Instruments). Retinal thinning was determined from horizontal macular OCT scans through the foveal center using the formula: Temporal thickness index (TTI) = (nasal - temporal thickness) ÷ nasal thickness × 100%, and compared with the normal range for each age group. Statistical analysis was performed using Student's t test, Mann-Whitney U test, and ROC analysis (SPPS, IBM). RESULTS The mean temporal retinal thickness index was 12.4 ± 5.2% in men (n = 19) and 7.4 ± 1.4% in women (n = 28) with X-linked Alport syndrome; 13.1 ± 4.5% (n = 4) in recessive disease; 6.4 ± 2.2% (n = 5) in Thin basement membrane nephropathy; and 6.3 ± 3.3% (n = 14) in other renal diseases. Thinning was worse in men than women with X-linked disease (p < 0.01), and worse in men who developed early onset renal failure (R2 = 0.75). Temporal retinal thinning was 84% sensitive for men with X-linked Alport syndrome and 67% specific (AUC = 0.83) compared with other renal diseases. CONCLUSIONS Retinal temporal thinning is diagnostic for X-linked Alport syndrome in men and distinguishes them this condition from Thin basement membrane nephropathy, but only in men (p = 0.002). Temporal retinal thinning may also identify men and women with the rarer autosomal recessive disease.
Collapse
Affiliation(s)
- Yan Chen
- a Department of Medicine, Melbourne Health and Northern Health , The University of Melbourne , Melbourne , Australia
| | - Deb Colville
- a Department of Medicine, Melbourne Health and Northern Health , The University of Melbourne , Melbourne , Australia
| | - Francesco Ierino
- b Department of Nephrology , Austin Health, Heidelberg , Heidelberg , Australia
| | - Andrew Symons
- c Department of Ophthalmology and Surgery , Melbourne Health , Melbourne , Australia
| | - Judy Savige
- a Department of Medicine, Melbourne Health and Northern Health , The University of Melbourne , Melbourne , Australia
| |
Collapse
|
26
|
Affiliation(s)
- Heather G Mack
- Department of Ophthalmology (Royal Victorian Eye and Ear Hospital and Melbourne Health) and Department of Surgery (Melbourne Health), The University of Melbourne, Melbourne, VIC, Australia
| | | |
Collapse
|
27
|
Kelly YP, Patil A, Wallis L, Murray S, Kant S, Kaballo MA, Casserly L, Doyle B, Dorman A, O'Kelly P, Conlon PJ. Outcomes of kidney transplantation in Alport syndrome compared with other forms of renal disease. Ren Fail 2016; 39:290-293. [PMID: 27917694 PMCID: PMC6014522 DOI: 10.1080/0886022x.2016.1262266] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Introduction: Alport syndrome is an inherited renal disease characterized by hematuria, renal failure, hearing loss and a lamellated glomerular basement membrane. Patients with Alport syndrome who undergo renal transplantation have been shown to have patient and graft survival rates similar to or better than those of patients with other renal diseases. Methods: In this national case series, based in Beaumont Hospital Dublin, we studied the cohort of patients who underwent renal transplantation over the past 33 years, recorded prospectively in the Irish Renal Transplant Registry, and categorized them according to the presence or absence of Alport syndrome. The main outcomes assessed were patient and renal allograft survival. Results: Fifty-one patients diagnosed with Alport syndrome in Beaumont Hospital received 62 transplants between 1982 and 2014. The comparison group of non-Alport patients comprised 3430 patients for 3865 transplants. Twenty-year Alport patient survival rate was 70.2%, compared to 44.8% for patients with other renal diseases (p = .01). Factors associated with patient survival included younger age at transplantation as well as differences in recipient sex, donor age, cold ischemia time, and episodes of acute rejection. Twenty-year graft survival was 46.8% for patients with Alport syndrome compared to 30.2% for those with non-Alport disease (p = .11). Conclusions: Adjusting for baseline differences between the groups, patients with end-stage kidney disease (ESKD) due to Alport syndrome have similar patient and graft survival to those with other causes of ESKD. This indicates that early diagnosis and management can lead to favorable outcomes for this patient cohort.
Collapse
Affiliation(s)
- Yvelynne P Kelly
- a Department of Nephrology , Beaumont Hospital , Dublin , Ireland
| | - Anish Patil
- b Royal College of Surgeons in Ireland , Dublin , Ireland
| | - Luke Wallis
- b Royal College of Surgeons in Ireland , Dublin , Ireland
| | - Susan Murray
- c Department of Nephrology , University Hospital Galway , Galway , Ireland
| | - Saumitra Kant
- d Department of Nephrology , Cork University Hospital , Cork , Ireland
| | - Mohammed A Kaballo
- e Department of Nephrology , University Hospital Limerick , Dooradoyle , Limerick , Ireland
| | - Liam Casserly
- e Department of Nephrology , University Hospital Limerick , Dooradoyle , Limerick , Ireland
| | - Brendan Doyle
- f Department of Pathology , Beaumont Hospital , Dublin , Ireland
| | - Anthony Dorman
- f Department of Pathology , Beaumont Hospital , Dublin , Ireland
| | - Patrick O'Kelly
- a Department of Nephrology , Beaumont Hospital , Dublin , Ireland
| | - Peter J Conlon
- a Department of Nephrology , Beaumont Hospital , Dublin , Ireland.,b Royal College of Surgeons in Ireland , Dublin , Ireland
| |
Collapse
|
28
|
Song D, Kanu LN, Li Y, Kelly KL, Bhuyan RK, Aleman T, Morgan JIW, Dunaief JL. AMD-like retinopathy associated with intravenous iron. Exp Eye Res 2016; 151:122-33. [PMID: 27565570 DOI: 10.1016/j.exer.2016.08.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 07/28/2016] [Accepted: 08/17/2016] [Indexed: 12/19/2022]
Abstract
Iron accumulation in the retina is associated with the development of age-related macular degeneration (AMD). IV iron is a common method to treat iron deficiency anemia in adults, and its retinal manifestations have not hitherto been identified. To assess whether IV iron formulations can be retina-toxic, we generated a mouse model for iron-induced retinal damage. Male C57BL/6J mice were randomized into groups receiving IV iron-sucrose (+Fe) or 30% sucrose (-Fe). Iron levels in neurosensory retina (NSR), retinal pigment epithelium (RPE), and choroid were assessed using immunofluorescence, quantitative PCR, and the Perls' iron stain. Iron levels were most increased in the RPE and choroid while levels in the NSR did not differ significantly in +Fe mice compared to controls. Eyes from +Fe mice shared histological features with AMD, including Bruch's membrane (BrM) thickening with complement C3 deposition, as well as RPE hypertrophy and vacuolization. This focal degeneration correlated with areas of high choroidal iron levels. Ultrastructural analysis provided further detail of the RPE/photoreceptor outer segment vacuolization and Bruch's membrane thickening. Findings were correlated with a clinical case of a 43-year-old patient who developed numerous retinal drusen, the hallmark of AMD, within 11 months of IV iron therapy. Our results suggest that IV iron therapy may have the potential to induce or exacerbate a form of retinal degeneration. This retinal degeneration shares features with AMD, indicating the need for further study of AMD risk in patients receiving IV iron treatment.
Collapse
Affiliation(s)
- Delu Song
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Levi N Kanu
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Yafeng Li
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristen L Kelly
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Rupak K Bhuyan
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Tomas Aleman
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica I W Morgan
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua L Dunaief
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Khan KN, Mahroo OA, Khan RS, Mohamed MD, McKibbin M, Bird A, Michaelides M, Tufail A, Moore AT. Differentiating drusen: Drusen and drusen-like appearances associated with ageing, age-related macular degeneration, inherited eye disease and other pathological processes. Prog Retin Eye Res 2016; 53:70-106. [DOI: 10.1016/j.preteyeres.2016.04.008] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 04/24/2016] [Accepted: 04/27/2016] [Indexed: 12/11/2022]
|
30
|
Joly D, Béroud C, Grünfeld JP. Rare inherited disorders with renal involvement-approach to the patient. Kidney Int 2015; 87:901-8. [PMID: 25651365 DOI: 10.1038/ki.2014.432] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/14/2014] [Accepted: 08/21/2014] [Indexed: 12/18/2022]
Abstract
The list of rare inherited disorders with renal involvement is rapidly growing. Many are single gene diseases affecting children, but cases are not restricted to pediatrics and diagnosis is often difficult and delayed. The expanding use of next-generation sequencing techniques is expected to discover new diseases, to challenge the definition of rarity, to accelerate and shake up our diagnostic paradigms, to promote 'deep phenotyping', and ultimately improve disease ontology. Rare renal diseases are exemplary of a transition from pediatric to adult-type care and pluridisciplinary approach, necessitating cooperation between geneticists, nephropediatricians, adult nephrologists, other physicians, nurses, social workers, and dietitians. They have raised new ethical issues, not only in genetic counseling, but also in public health, regarding equity, and distribution of care. Patient's organizations have grown and have been very active to promote information and research. Efforts are underway to create interoperable patient's registries and ultimately worldwide networks gathering patients, researchers, clinicians, pharmaceutical industry, and health authorities. Progress in genetics and pathophysiological mechanisms will hopefully increase the number of efficient orphan medicinal products. Finally, new frontiers set by rare nephropathies may improve the understanding, treatments, and outcomes of more frequent renal diseases.
Collapse
Affiliation(s)
- Dominique Joly
- 1] Service de Néphrologie, Université Paris-Descartes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France [2] INSERM U845, Hôpital Necker-Enfants Malades, Paris, France
| | - Christophe Béroud
- 1] Aix-Marseille Université, Marseille, France [2] Département de Génétique Médicale, Hôpital d'enfants Timone, AP-HM, Marseille, France
| | - Jean-Pierre Grünfeld
- Service de Néphrologie, Université Paris-Descartes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| |
Collapse
|
31
|
Mallett A, Tang W, Clayton PA, Stevenson S, McDonald SP, Hawley CM, Badve SV, Boudville N, Brown FG, Campbell SB, Johnson DW. End-stage kidney disease due to Alport syndrome: outcomes in 296 consecutive Australia and New Zealand Dialysis and Transplant Registry cases. Nephrol Dial Transplant 2014; 29:2277-86. [DOI: 10.1093/ndt/gfu254] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
32
|
Johnson AC, Lee JW, Harmon AC, Morris Z, Wang X, Fratkin J, Rapp JP, Gomez-Sanchez E, Garrett MR. A mutation in the start codon of γ-crystallin D leads to nuclear cataracts in the Dahl SS/Jr-Ctr strain. Mamm Genome 2013; 24:95-104. [PMID: 23404175 PMCID: PMC3628938 DOI: 10.1007/s00335-013-9447-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
Abstract
Cataracts are a major cause of blindness. The most common forms of cataracts are age- and UV-related and develop mostly in the elderly, while congenital cataracts appear at birth or in early childhood. The Dahl salt-sensitive (SS/Jr) rat is an extensively used model of salt-sensitive hypertension that exhibits concomitant renal disease. In the mid-1980s, cataracts appeared in a few animals in the Dahl S colony, presumably the result of a spontaneous mutation. The mutation was fixed and bred to establish the SS/Jr-Ctr substrain. The SS/Jr-Ctr substrain has been used exclusively by a single investigator to study the role of steroids and hypertension. Using a classical positional cloning approach, we localized the cataract gene with high resolution to a less than 1-Mbp region on chromosome 9 using an F1(SS/Jr-Ctr × SHR) × SHR backcross population. The 1-Mbp region contained only 13 genes, including 4 genes from the γ-crystallins (Cryg) gene family, which are known to play a role in cataract formation. All of the γ-crystallins were sequenced and a novel point mutation in the start codon (ATG → GTG) of the Crygd gene was identified. This led to the complete absence of the CRYGD protein in the eyes of the SS/Jr-Ctr strain. In summary, the identification of the genetic cause in this novel cataract model may provide an opportunity to better understand the development of cataracts, particularly in the context of hypertension.
Collapse
Affiliation(s)
- Ashley C. Johnson
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Jonathan W. Lee
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Ashlyn C. Harmon
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Zaliya Morris
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Xuexiang Wang
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Jonathan Fratkin
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216
| | | | - Elise Gomez-Sanchez
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
- GV(Sonny) Montgomery VAMC
| | - Michael R. Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|
33
|
Savige J, Gregory M, Gross O, Kashtan C, Ding J, Flinter F. Expert guidelines for the management of Alport syndrome and thin basement membrane nephropathy. J Am Soc Nephrol 2013; 24:364-75. [PMID: 23349312 DOI: 10.1681/asn.2012020148] [Citation(s) in RCA: 226] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Few prospective, randomized controlled clinical trials address the diagnosis and management of patients with Alport syndrome or thin basement membrane nephropathy. Adult and pediatric nephrologists and geneticists from four continents whose clinical practice focuses on these conditions have developed the following guidelines. The 18 recommendations are based on Level D (Expert opinion without explicit critical appraisal, or based on physiology, bench research, or first principles-National Health Service category) or Level III (Opinions of respected authorities, based on clinical experience, descriptive studies, or reports of expert committees-U.S. Preventive Services Task Force) evidence. The recommendations include the use of genetic testing as the gold standard for the diagnosis of Alport syndrome and the demonstration of its mode of inheritance; the need to identify and follow all affected members of a family with X-linked Alport syndrome, including most mothers of affected males; the treatment of males with X-linked Alport syndrome and individuals with autosomal recessive disease with renin-angiotensin system blockade, possibly even before the onset of proteinuria; discouraging the affected mothers of males with X-linked Alport syndrome from renal donation because of their own risk of kidney failure; and consideration of genetic testing to exclude X-linked Alport syndrome in some individuals with thin basement membrane nephropathy. The authors recognize that as evidence emerges, including data from patient registries, these guidelines will evolve further.
Collapse
Affiliation(s)
- Judy Savige
- Department of Medicine (Northern Health), University of Melbourne, Melbourne, Australia.
| | | | | | | | | | | |
Collapse
|
34
|
Retinal Manifestations of Renal and Connective Tissue Diseases. Clin Ophthalmol 2012; 52:93-105. [DOI: 10.1097/iio.0b013e31823bbb32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
35
|
Deva R, Alias MA, Colville D, Tow FKNFH, Ooi QL, Chew S, Mohamad N, Hutchinson A, Koukouras I, Power DA, Savige J. Vision-threatening retinal abnormalities in chronic kidney disease stages 3 to 5. Clin J Am Soc Nephrol 2011; 6:1866-71. [PMID: 21784818 PMCID: PMC3359545 DOI: 10.2215/cjn.10321110] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 04/21/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES Retinal abnormalities are common in inherited and acquired renal disease. This study determined the prevalence of retinal abnormalities in chronic kidney disease (CKD) stages 3 to 5. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS One hundred fifty patients with CKD stages 3 to 5 and 150 age- and gender-matched hospital patients with CKD stages 1 to 2 underwent bilateral retinal photography. These images were reviewed for incidental abnormalities, microvascular (Wong and Mitchell classification) and diabetic retinopathy (Airlie House criteria), and macular degeneration (Seddon classification). RESULTS Three (2%) patients with CKD stages 3 to 5 had retinal features characteristic of inherited renal disease (atrophy in Myopathy, Encephalopathy, Lactic Acidosis, Stroke-like episodes [MELAS] syndrome; and 2 with drusen in dense deposit disease). Fifty-nine (39%) patients had moderate-severe microvascular retinopathy (hemorrhages, exudates, etc.) compared with 19 (13%) with CKD stages 1 to 2. Forty-one (28%) had moderate-severe diabetic retinopathy (microaneurysms, exudates, etc.) compared with 16 (11%) with CKD stages 1 to 2. Ten (7%) had severe macular degeneration (geographic atrophy, hemorrhage, exudates, membranes) compared with one (1%) with CKD stages 1 to 2. Renal failure was an independent risk factor for microvascular retinopathy, diabetic retinopathy, and macular degeneration. Eleven (7.3%) patients with renal failure and one (0.7%) with CKD stages 1 to 2 had previously unrecognized vision-threatening retinal abnormalities that required immediate ophthalmologic attention. CONCLUSIONS Retinal abnormalities are common in CKD stages 3 to 5, and are more severe and more likely to threaten vision than in hospital patients with CKD stages 1 to 2.
Collapse
Affiliation(s)
- Rajeev Deva
- The University of Melbourne, Department of Medicine, Northern Health, Cooper Street, Melbourne, Victoria 3076 Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|